Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
STABLE MODULATORS OF GAMMA-C-CYTOKINE ACTIVITY
Document Type and Number:
WIPO Patent Application WO/2018/187499
Kind Code:
A1
Abstract:
Disclosed herein are stable peptide antagonists based on the consensus yc-subunit binding site to inhibit the activity of yc-cytokines. Such peptide antagonists are capable of inhibiting the activity of multiple yc-cytokine family members. The yc-family cytokines are associated with important human diseases, such as leukemia, autoimmune diseases, collagen diseases, diabetes mellitus, skin diseases, degenerative neuronal diseases and graft-versus-host disease (GvHD). Thus, inhibitors of yc-cytokine activity are valuable therapeutic and cosmetic agents as well as research tools.

Inventors:
DOERR NICHOLAS (US)
AL-MAWSAWI LAITH Q (US)
AZIMI NAZLI (US)
Application Number:
PCT/US2018/026125
Publication Date:
October 11, 2018
Filing Date:
April 04, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BIONIZ LLC (US)
International Classes:
C07K7/50; A61K8/64; A61K38/00; C07K1/107; C07K1/113; C07K7/06; C07K7/08; C07K14/54; C07K14/55
Foreign References:
US20130217858A12013-08-22
US7786072B22010-08-31
Other References:
See also references of EP 3606940A4
Attorney, Agent or Firm:
JUANG, Agnes (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A composite peptide comprising amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises an amino acid sequence D/E-F-L-E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2), wherein X denotes any amino acid, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element.

2. The composite peptide of claim 1, wherein the at least two alpha-alkenyl substituted amino acids are linked to form the at least one intra-peptide hydrocarbon linker element by ring closing metathesis, wherein the ring closing metathesis is catalyzed by Grubb's catalyst.

3. The composite peptide of claim 1, wherein an amino acid in the composite peptide is selected from the group consisting of natural amino acids, non-natural amino acids, (D) stereochemical configuration amino acids, (L) stereochemical configuration amino acids, (R) stereochemical configuration amino acids and (S) stereochemical configuration amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are selected from the group consisting of R-propenylalanine (CAS: 288617-76-5; R3Ala), S-propenylalanine (CAS: 288617-71-0; S3Ala), D-allylglycine (CAS: 170642-28-1; D3Gly), L-allylglycine (CAS: 146549-21-5; L3Gly), R-pentenylalanine (CAS: 288617-77-6; R5Ala), S- pentenylalanine (CAS: 288617-73-2; S5Ala), R-pentenylglycine (CAS: 1093645-21-6; R5Gly), S-pentenylglycine (CAS: 856412-22-1; S5Gly), R-butenylalanine (CAS: 1311933-

82- 0; R4Ala), S-butenylalanine (CAS: 288617-72-1; S4Ala), R-butenylglycine (CAS: 865352-21-2; R4Gly), S-butenylglycine (CAS: 851909-08-5; S4Gly), R-hexenylalanine (CAS: 288617-78-7; R6Ala), S-hexenylalanine (CAS: 288617-74-3; S6Ala), R- hexenylglycine (CAS: 1208226-88-3; R6Gly), S-hexenylglycine (CAS: 1251904-51-4; S6Gly), R-heptenylalanine (CAS: 1311933-84-2; R7Ala), S-heptenylalanine (CAS: 1311933-

83- 1; S7Ala), R-heptenylglycine (CAS: 1262886-63-4; R7Gly), S-heptenylglycine (CAS: 1058705-57-9; S7Gly), di- substituted bis-propenylglycine (CAS: 1311992-97-8; bis3Gly), di- substituted bis-pentenylglycine (CAS: 1068435-19-7; bis5Gly), di-substituted bis- butenylglycine (bis4Gly), di-substituted bis-hexenylglycine (bis6Gly), di-substituted bis- heptenylglycine (bis7Gly), R-octenylalanine (CAS: 945212-26-0; R8Ala), S-octenylalanine (CAS: 288617-75-4; S8Ala), R-octenylglycine (CAS: 1191429-20-5; R8Gly), and S- octenylglycine (CAS: 1262886-64-5; S8Gly).

4. The composite peptide of claim 1, wherein the at least two alpha-alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by n-2 amino acids, wherein n represents the number of amino acids encompassed by the intra- peptide linkage.

5. The composite peptide of claim 1, wherein when the at least two alpha-alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by three amino acids, the at least one intra-peptide hydrocarbon linker element spans a single a- helical turn of the composite peptide.

6. The composite peptide of claim 5, wherein when the composite peptide comprises one or more non-contiguous single a-helical turns, the amino acid positions that correlate with a single α-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the single α-helical turn and i+4 is the last amino acid position of the single α-helical turn, and wherein amino acid positions i and i+4 comprise alpha-alkenyl substituted amino acids.

7. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R-propenylalanine (CAS: 288617-76-5; R3Ala), S-propenylalanine (CAS: 288617-71-0; S3Ala), D-allylglycine (CAS: 170642-28-1; D3Gly), and L-allylglycine (CAS: 146549-21-5; L3Gly), and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R- pentenylalanine (CAS: 288617-77-6; R5Ala), S-pentenylalanine (CAS: 288617-73-2; S5Ala), R-pentenylglycine (CAS: 1093645-21-6; R5Gly), and S-pentenylglycine (CAS: 856412-22-1; S5Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 1.

8. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 2.

9. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R-butenylalanine (CAS: 1311933-82-0; R4Ala), S-butenylalanine (CAS: 288617-72-1; S4Ala), R-butenylglycine (CAS: 865352-21-2; R4Gly), and S-butenylglycine (CAS: 851909-08-5; S4Gly), and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R4Ala, S4Ala, R4Gly, andS4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 3.

10. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 4.

11. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R-hexenylalanine (CAS: 288617-78-7; R6Ala), S-hexenylalanine (CAS: 288617-74-3; S6Ala), R-hexenylglycine (CAS: 1208226-88-3; R6Gly), and S-hexenylglycine (CAS: 1251904-51-4; S6Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 5.

12. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 6.

13. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R-heptenylalanine (CAS: 1311933-84-2; R7Ala), S-heptenylalanine (CAS: 1311933-83-1; S7Ala), R-heptenylglycine (CAS: 1262886-63-4; R7Gly), and S- heptenylglycine (CAS: 1058705-57-9; S7Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 7.

14. The composite peptide of claim 6, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 8.

15. The composite peptide of claim 5, wherein when the composite peptide comprises two or more contiguous single a-helical turns, the amino acid positions that correlate with the first single α-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the first single α-helical turn and i+4 is the last amino acid position of the first single a- helical turn, and the amino acid positions that correlate with the second single α-helical turn of the composite peptide correspond to amino acid positions i+4 and i+8 of the composite peptide, where i+4 is the first amino acid position of the second single α-helical turn and i+8 is the last amino acid position of the second single α-helical turn, and wherein amino acid positions i, i+4 and i+8 comprise alpha-alkenyl substituted amino acids.

16. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-propenylglycine (CAS: 1311992-97-8; bis3Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 9.

17. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-pentenylglycine (CAS: 1068435-19-7; bis5Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 10.

18. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-butenylglycine (bis4Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 11.

19. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis5Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 12.

20. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis4Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R6Ala, S6Ala, R6Gly, or S6Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 13.

21. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-hexenylglycine (bis6Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 14.

22. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis3Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 15.

23. The composite peptide of claim 15, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-heptenylglycine (bis7Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 16.

24. The composite peptide of claim 1, wherein when the at least two alpha-alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by six residues, the at least one intra-peptide hydrocarbon linker element spans a double oc- helical turn of the composite peptide.

25. The composite peptide of claim 24, wherein when the composite peptide comprises one or more non-contiguous double oc-helical turns, the amino acid positions that correlate with a double oc-helical turn of the composite peptide correspond to amino acid positions i and i+7 of the composite peptide, where i is the first amino acid position of the double a-helical turn and i+7 is the last amino acid position of the double a-helical turn, and wherein amino acid positions i and i+7 comprise alpha-alkenyl substituted amino acids.

26. The composite peptide of claim 25, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R-octenylalanine (CAS: 945212-26-0; R8Ala), S-octenylalanine (CAS: 288617-75-4; S8Ala), R-octenylglycine (CAS: 1191429-20-5; R8Gly), and S- octenylglycine (CAS: 1262886-64-5; S8Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 17.

27. The composite peptide of claim 25, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 18.

28. The composite peptide of claim 25, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 19.

29. The composite peptide of claim 25, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 20.

30. The composite peptide of claim 24, wherein when the composite peptide comprises two or more contiguous double α-helical turns, the amino acid positions that correlate with the first double α-helical turn of the composite peptide correspond to amino acid positions i and i+7 of the composite peptide, where i is the first amino acid position of the first double α-helical turn and i+7 is the last amino acid position of the first double a- helical turn, and the amino acid positions that correlate with the second double α-helical turn of the composite peptide correspond to amino acid positions i+7 and i+14 of the composite peptide, where i+7 is the first amino acid position of the second double a-helical turn and i+14 is the last amino acid position of the second double α-helical turn, and wherein amino acid positions i, i+7 and i+14 comprise alpha-alkenyl substituted amino acids.

31. The composite peptide of claim 30, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis5Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 21.

32. The composite peptide of claim 30, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis6Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 22.

33. The composite peptide of claim 30, wherein when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis7Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 23.

34. The composite peptide of any one of claims 1-33, wherein a derivative of the composite peptide comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide.

35. The composite peptide of any one of claims 1-34, wherein the composite peptide inhibits the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21.

36. The composite peptide of any one of claims 1-35, further comprising a signal peptide.

37. The composite peptide of any one of claims 1-36, further comprising one or more additional moieties conjugated at the N-terminus, C-terminus or a side residue of the composite peptide.

38. The composite peptide of claim 37, wherein the one or more additional moieties are selected from the group consisting of bovine serum albumin (BSA), albumin, Keyhole Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion, and Poly Ethylene Glycol (PEG).

39. The composite peptide of any one of claims 1-38, wherein the composite peptide comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises the amino acid sequence I-K-E-F- L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), and wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element.

40. The composite peptide of any one of claims 1-38, wherein the composite peptide comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises the amino acid sequence P-K-E-F- L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), and wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element.

41. The composite peptide of any one of claims 1-38, wherein the composite peptide inhibits a cell growth promoting activity of IL-15, IL-21, or a combination thereof.

42. The composite peptide of any one of claims 1-38, wherein the composite peptide inhibits a STAT5 signaling activity of IL-15.

43. The composite peptide of any one of claims 1-38, wherein the composite peptide inhibits a STAT3 signaling activity of IL-21.

44. The composite peptide of any one of claims 1-38, wherein the gastric stability of the composite peptide having at least one intra-peptide hydrocarbon linker element is enhanced compared to an unmodified composite peptide without an intra-peptide hydrocarbon linker element.

45. A pharmaceutical composition comprising:

a therapeutically effective amount of a composite peptide, or a derivative thereof; and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3.

46. The pharmaceutical composition of claim 45, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21.

47. A method of preventing or treating a yc-cytokine-mediated disease, the method comprising

administering to a subject in need thereof, a pharmaceutical composition comprising:

a therapeutically effective amount of a composite peptide, or a derivative thereof; and

a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element,

wherein the composite peptide or derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21,

wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide, thereby preventing or treating the yc-cytokine-mediated disease.

48. The method of claim 47, wherein the yc-cytokine-mediated disease is selected from the group consisting of CD4-leukemia, CD8-leukemia, LGL-leukemia, systemic lupus erythematosis, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, graft-versus-host disease (GvHD), inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease), Systemic Lupus Erythematosus, and alopecia areata.

49. A method of preventing or treating an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease, the method comprising:

administering to a subject in need thereof, a pharmaceutical composition comprising:

a therapeutically effective amount of a composite peptide, or a derivative thereof; and

a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element,

wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21,

wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide,

thereby preventing or treating an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease.

50. The method of claim 49, wherein the HAM/TSP associated disease is selected from the group consisting of Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neeoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, and myositis.

51. A method of preventing or treating an inflammatory respiratory disease, the method comprising:

administering to a subject in need thereof, a pharmaceutical composition comprising:

a therapeutically effective amount of a composite peptide, or a derivative thereof; and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element,

wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21,

wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide,

thereby preventing or treating an inflammatory respiratory disease.

52. The method of claim 51, wherein the inflammatory respiratory disease is selected from the group consisting of asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis.

53. A method of preventing or treating a cosmetic condition, the method comprising:

administering to a subject in need thereof, a pharmaceutical composition comprising:

a therapeutically effective amount of a composite peptide, or a derivative thereof; and

a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21,

wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide,

thereby preventing or treating a cosmetic condition.

54. The method of claim 53, wherein the wherein the cosmetic condition is selected from the group consisting of acne, hair loss, sunburn, nail maintenance, and appearance of aging.

55. A kit for preventing or treating a condition in a patient, wherein the condition is a jc cytokine-mediated disease, an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease, an inflammatory respiratory disease, a cosmetic condition, or a combination thereof, the kit comprising a pharmaceutical composition, wherein the pharmaceutical composition comprises:

a therapeutically effective amount of a composite peptide or a derivative thereof; and

a pharmaceutically acceptable carrier, diluent, excipient or combination thereof;

wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element,

wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, and

wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide.

56. The kit of claim 55, wherein the condition is one or more of CD4 leukemia, CD8 leukemia, LGL leukemia, systemic lupus erythematosus, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uveitis, inflammation of the eye, graft-versus- host disease (GvHD), inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease), Systemic Lupus Erythematosus, alopecia areata, Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neeoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, myositis, asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis, acne, hair loss, sunburn, nail maintenance, or appearance of aging.

57. The composite peptide of any one of claims 1-44, wherein the one or more carbon-carbon double bonds present in the intra-peptide hydrocarbon linker are utilized for one or more organic chemical reactions to add one or more additional chemical functionalities.

58. The composite peptide of claim 57, wherein the one or more organic chemical reactions comprises an alkene reaction.

59. The composite peptide of claim 58, wherein the alkene reaction is selected from the group consisting of hydroboration, oxymercuration, hydration, chlorination, bromination, addition of HF, HBr, HC1 or HI, dihydroxylation, epoxidation, hydrogenation, and cyclopropanation.

60. The composite peptide of claim 59, wherein one or more additional chemical functionalities can be added subsequent to the alkene reaction wherein the one or more additional chemical functionalities comprise a covalent addition of one or more chemical group substituents, wherein the covalent addition of one or more chemical group substituents comprises nucleophilic reactions with epoxide and hydroxyl groups.

61. The composite peptide of claim 60, wherein the one or more additional chemical functionalities are selected from the group consisting of biotin, radioisotopes, therapeutic agents, rapamycin, vinblastine, taxol, non-protein fluorescent chemical groups, FrrC, hydrazide, rhodamine, maleimide, protein fluorescent groups, GFP, YFP, and mCherry.

Description:
STABLE MODULATORS OF GAMMA-C-CYTOKINE ACTIVITY

PRIORITY AND CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application No. 62/483,210, filed on April 7, 2017, which is hereby incorporated by reference in its entirety.

SEQUENCE LISTING IN ELECTRONIC FORMAT

[0002] 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 BION010WOSEQLIST.txt, created and last saved on March 30, 2018, which is 60,939 bytes 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

Field

[0003] Some embodiments relate to peptide antagonists of yc-family cytokines, a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. Some embodiments also relate to the therapeutic uses of such peptides for the treatment of certain human diseases. The present embodiments also relate to the cosmeceutical applications of such peptides. Description of target diseases, cosmeceutical applications, as well as methods of administration, production, and commercialization of the peptides are disclosed.

Description of the Related Art

[0004] Cytokines are a diverse group of soluble factors that mediate various cell functions, such as, growth, functional differentiation, and promotion or prevention of programmed cell death (apoptotic cell death). Cytokines, unlike hormones, are not produced by specialized glandular tissues, but can be produced by a wide variety of cell types, such as epithelial, stromal or immune cells. [0005] The yc-family cytokines are a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. These cytokines are critically required for the early development of T cells in the thymus as well as their homeostasis in the periphery.

SUMMARY

[0006] In some embodiments, a composite peptide comprising amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises an amino acid sequence D/E-F-L-E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2), wherein X denotes any amino acid, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element is provided.

[0007] In some embodiments of the composite peptide, the at least two alpha- alkenyl substituted amino acids are linked to form the at least one intra-peptide hydrocarbon linker element by ring closing metathesis, wherein the ring closing metathesis is catalyzed by Grubb's catalyst.

[0008] In some embodiments, an amino acid in the composite peptide is selected from the group consisting of natural amino acids, non-natural amino acids, (D) stereochemical configuration amino acids, (L) stereochemical configuration amino acids, (R) stereochemical configuration amino acids and (S) stereochemical configuration amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are selected from the group consisting of R-propenylalanine (CAS: 288617-76-5; R3Ala), S-propenylalanine (CAS: 288617-71-0; S3Ala), D-allylglycine (CAS: 170642-28-1; D3Gly), L-allylglycine (CAS: 146549-21-5; L3Gly), R-pentenylalanine (CAS: 288617-77-6; R5Ala), S-pentenylalanine (CAS: 288617-73-2; S5Ala), R-pentenylglycine (CAS: 1093645-21-6; R5Gly), S- pentenylglycine (CAS: 856412-22-1; S5Gly), R-butenylalanine (CAS: 1311933-82-0; R4Ala), S-butenylalanine (CAS: 288617-72-1; S4Ala), R-butenylglycine (CAS: 865352-21- 2; R4Gly), S-butenylglycine (CAS: 851909-08-5; S4Gly), R-hexenylalanine (CAS: 288617- 78-7; R6Ala), S-hexenylalanine (CAS: 288617-74-3; S6Ala), R-hexenylglycine (CAS: 1208226-88-3; R6Gly), S-hexenylglycine (CAS: 1251904-51-4; S6Gly), R-heptenylalanine (CAS: 1311933-84-2; R7Ala), S-heptenylalanine (CAS: 1311933-83-1; S7Ala), R- heptenylglycine (CAS: 1262886-63-4; R7Gly), S-heptenylglycine (CAS: 1058705-57-9; S7Gly), di-substituted bis-propenylglycine (CAS: 1311992-97-8; bis3Gly), di- substituted bis- pentenylglycine (CAS: 1068435-19-7; bis5Gly), di-substituted bis-butenylglycine (bis4Gly), di-substituted bis-hexenylglycine (bis6Gly), di-substituted bis-heptenylglycine (bis7Gly), R- octenylalanine (CAS: 945212-26-0; R8Ala), S-octenylalanine (CAS: 288617-75-4; S8Ala), R-octenylglycine (CAS: 1191429-20-5; R8Gly), and S-octenylglycine (CAS: 1262886-64-5; S8Gly).

[0009] In some embodiments of the composite peptide, the at least two alpha- alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by n-2 amino acids, wherein n represents the number of amino acids encompassed by the intra-peptide linkage.

[0010] In some embodiments of the composite peptide, when the at least two alpha-alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by three amino acids, the at least one intra-peptide hydrocarbon linker element spans a single a-helical turn of the composite peptide.

[0011] In some embodiments of the composite peptide, when the composite peptide comprises one or more non-contiguous single α-helical turns, the amino acid positions that correlate with a single α-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the single α-helical turn and i+4 is the last amino acid position of the single a- helical turn, and wherein amino acid positions i and i+4 comprise alpha-alkenyl substituted amino acids.

[0012] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R- propenylalanine (CAS: 288617-76-5; R3Ala), S-propenylalanine (CAS: 288617-71-0; S3Ala), D-allylglycine (CAS: 170642-28-1; D3Gly), and L-allylglycine (CAS: 146549-21-5; L3Gly), and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R-pentenylalanine (CAS: 288617-77-6; R5Ala), S-pentenylalanine (CAS: 288617-73-2; S5Ala), R-pentenylglycine (CAS: 1093645-21-6; R5Gly), and S- pentenylglycine (CAS: 856412-22-1; S5Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 1. [0013] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 2.

[0014] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R-butenylalanine (CAS: 1311933-82-0; R4Ala), S-butenylalanine (CAS: 288617-72-1 ; S4Ala), R- butenylglycine (CAS: 865352-21-2; R4Gly), and S-butenylglycine (CAS: 851909-08-5; S4Gly), and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R4Ala, S4Ala, R4Gly, andS4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 3.

[0015] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 4.

[0016] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R-hexenylalanine (CAS: 288617-78-7; R6Ala), S- hexenylalanine (CAS: 288617-74-3; S6Ala), R-hexenylglycine (CAS: 1208226-88-3; R6Gly), and S-hexenylglycine (CAS: 1251904-51-4; S6Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 5.

[0017] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 6.

[0018] In some embodiments of the composite peptide, the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R-heptenylalanine (CAS: 1311933-84-2; R7Ala), S- heptenylalanine (CAS: 1311933-83-1; S7Ala), R-heptenylglycine (CAS: 1262886-63-4; R7Gly), and S-heptenylglycine (CAS: 1058705-57-9; S7Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 7.

[0019] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, and the alpha-alkenyl substituted amino acid at position i+4 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 8.

[0020] In some embodiments of the composite peptide, when the composite peptide comprises two or more contiguous single a-helical turns, the amino acid positions that correlate with the first single α-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the first single α-helical turn and i+4 is the last amino acid position of the first single a- helical turn, and the amino acid positions that correlate with the second single α-helical turn of the composite peptide correspond to amino acid positions i+4 and i+8 of the composite peptide, where i+4 is the first amino acid position of the second single α-helical turn and i+8 is the last amino acid position of the second single α-helical turn, and wherein amino acid positions i, i+4 and i+8 comprise alpha-alkenyl substituted amino acids.

[0021] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-propenylglycine (CAS: 1311992-97-8; bis3Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 9.

[0022] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-pentenylglycine (CAS: 1068435-19-7; bis5Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 10. [0023] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-butenylglycine (bis4Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 11.

[0024] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis5Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 12.

[0025] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis4Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R6Ala, S6Ala, R6Gly, or S6Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 13.

[0026] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-hexenylglycine (bis6Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R4Ala, S4Ala, R4Gly, and S4Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 14.

[0027] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the alpha-alkenyl substituted amino acid at position i+4 is bis3Gly, and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring- closing metathesis is represented by Formula 15.

[0028] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the alpha-alkenyl substituted amino acid at position i+4 is di-substituted bis-he tenylglycine (bis7Gly), and the alpha-alkenyl substituted amino acid at position i+8 is selected from the group consisting of R3Ala, S3Ala, D3Gly, and L3Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 16.

[0029] In some embodiments of the composite peptide, when the at least two alpha-alkenyl substituted amino acids linked by the at least one intra-peptide hydrocarbon are separated by six residues, the at least one intra-peptide hydrocarbon linker element spans a double a-helical turn of the composite peptide.

[0030] In some embodiments of the composite peptide, when the composite peptide comprises one or more non-contiguous double α-helical turns, the amino acid positions that correlate with a double α-helical turn of the composite peptide correspond to amino acid positions i and i+7 of the composite peptide, where i is the first amino acid position of the double α-helical turn and i+7 is the last amino acid position of the double a- helical turn, and wherein amino acid positions i and i+7 comprise alpha-alkenyl substituted amino acids.

[0031] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R-octenylalanine (CAS: 945212-26-0; R8Ala), S-octenylalanine (CAS: 288617-75-4; S8Ala), R-octenylglycine (CAS: 1191429-20-5; R8Gly), and S- octenylglycine (CAS: 1262886-64-5; S8Gly), the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 17.

[0032] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R5Ala, S5Ala, R5Gly, and S5Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 18.

[0033] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position i is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 19. [0034] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position z is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, and the alpha-alkenyl substituted amino acid at position i+7 is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 20.

[0035] In some embodiments of the composite peptide, when the composite peptide comprises two or more contiguous double a-helical turns, the amino acid positions that correlate with the first double α-helical turn of the composite peptide correspond to amino acid positions z and z ' +7 of the composite peptide, where z is the first amino acid position of the first double α-helical turn and z+7 is the last amino acid position of the first double α-helical turn, and the amino acid positions that correlate with the second double a- helical turn of the composite peptide correspond to amino acid positions z+7 and i+14 of the composite peptide, where z+7 is the first amino acid position of the second double a-helical turn and i+14 is the last amino acid position of the second double α-helical turn, and wherein amino acid positions z, i+7 and i+14 comprise alpha-alkenyl substituted amino acids.

[0036] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position z is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis5Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R8Ala, S8Ala, R8Gly, and S8Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 21.

[0037] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position z is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis6Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R7Ala, S7Ala, R7Gly, and S7Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 22.

[0038] In some embodiments of the composite peptide, when the alpha-alkenyl substituted amino acid at position z is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the alpha-alkenyl substituted amino acid at position i+7 is bis7Gly, and the alpha-alkenyl substituted amino acid at position i+14 is selected from the group consisting of R6Ala, S6Ala, R6Gly, and S6Gly, the hydrocarbon linker element formed by the ring-closing metathesis is represented by Formula 23.

[0039] In some embodiments, a derivative of the composite peptide comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide.

[0040] In some embodiments, the composite peptide inhibits the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21.

[0041] In some embodiments, the composite peptide further comprises a signal peptide. In some embodiments, the composite peptide further comprises one or more additional moieties conjugated at the N-terminus, C-terminus or a side residue of the composite peptide. In some embodiments, the composite peptide further comprises the one or more additional moieties are selected from the group consisting of bovine serum albumin (BSA), albumin, Keyhole Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion, and Poly Ethylene Glycol (PEG).

[0042] In some embodiments, the composite peptide comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises the amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N- T-S (SEQ ID NO: 1), and wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element.

[0043] In some embodiments, the composite peptide comprises amino acid sequences of at least two interleukin (IL) protein gamma-c-box D-helix regions, wherein the composite peptide comprises the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I- H-Q-S-L-S (SEQ ID NO: 3), and wherein the composite peptide comprises at least two alpha- alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element.

[0044] In some embodiments, the composite peptide the composite peptide inhibits a cell growth promoting activity of IL-15, IL-21, or a combination thereof. In some embodiments, the composite peptide inhibits a STAT5 signaling activity of IL-15. In some embodiments, the composite peptide inhibits a STAT3 signaling activity of IL-21.

[0045] In some embodiments, a pharmaceutical composition comprising a therapeutically effective amount of a composite peptide, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof is provided. In some embodiments of the pharmaceutical composition, the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3. In some embodiments of the pharmaceutical composition, the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21.

[0046] In some embodiments, a method of preventing or treating a yc-cytokine- mediated disease is provided. In some embodiments, the method comprises administering to a subject in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of a composite peptide, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide, thereby preventing or treating the yc-cytokine-mediated disease.

[0047] In some embodiments, the yc-cytokine-mediated disease is selected from the group consisting of CD4-leukemia, CD8-leukemia, LGL-leukemia, systemic lupus erythematosis, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, graft-versus-host disease (GvHD), inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease), Systemic Lupus Erythematosus, and alopecia areata.

[0048] In some embodiments, a method of preventing or treating an HTLV-1- associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease is provided. In some embodiments, the method comprises administering to a subject in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of a composite peptide, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide, thereby preventing or treating an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease.

[0049] In some embodiments, the HAM/TSP associated disease is selected from the group consisting of Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neeoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, and myositis.

[0050] In some embodiments, a method of preventing or treating an inflammatory respiratory disease is provided. In some embodiments, the method comprises administering to a subject in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of a composite peptide, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide, thereby preventing or treating an inflammatory respiratory disease.

[0051] In some embodiments, the inflammatory respiratory disease is selected from the group consisting of asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis.

[0052] In some embodiments, a method of preventing or treating a cosmetic condition is provided. In some embodiments, the method comprises administering to a subject in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of a composite peptide, or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide, thereby preventing or treating a cosmetic condition.

[0053] In some embodiments, the cosmetic condition is selected from the group consisting of acne, hair loss, sunburn, nail maintenance, and appearance of aging.

[0054] In some embodiments, a kit for preventing or treating a condition in a patient is provided. In some embodiments of the kit, the condition is a yc cytokine-mediated disease, an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease, an inflammatory respiratory disease, a cosmetic condition, or a combination thereof. In some embodiments, the kit comprises a pharmaceutical composition, wherein the pharmaceutical composition comprising a therapeutically effective amount of a composite peptide or a derivative thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof, wherein the composite peptide is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, wherein the composite peptide comprises at least two alpha-alkenyl substituted amino acids, and wherein the at least two alpha-alkenyl substituted amino acids are linked via at least one intra-peptide hydrocarbon linker element, wherein the composite peptide or the derivative thereof modulates the activity of one or more yc-cytokines selected from the group consisting of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, and wherein the derivative thereof comprises an amino acid sequence that shares about 50% to about 99% identity with the composite peptide.

[0055] In some embodiments of the kit, the condition is one or more of CD4 leukemia, CD8 leukemia, LGL leukemia, systemic lupus erythematosus, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uveitis, inflammation of the eye, graft-versus- host disease (GvHD), inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease), Systemic Lupus Erythematosus, alopecia areata, Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neeoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, myositis, asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis, acne, hair loss, sunburn, nail maintenance, or appearance of aging.

[0056] In some embodiments of the composite peptide, the one or more carbon- carbon double bonds present in the intra-peptide hydrocarbon linker are utilized for one or more organic chemical reactions to add one or more additional chemical functionalities. In some embodiments of the composite peptide, the one or more organic chemical reactions comprises an alkene reaction. In some embodiments of the composite peptide, the alkene reaction is selected from the group consisting of hydroboration, oxymercuration, hydration, chlorination, bromination, addition of HF, HBr, HC1 or HI, dihydroxylation, epoxidation, hydrogenation, and cyclopropanation. In some embodiments of the composite peptide, one or more additional chemical functionalities can be added subsequent to the alkene reaction wherein the one or more additional chemical functionalities comprise a covalent addition of one or more chemical group substituents, wherein the covalent addition of one or more chemical group substituents comprises nucleophilic reactions with epoxide and hydroxyl groups. In some embodiments of the composite peptide, the one or more additional chemical functionalities are selected from the group consisting of biotin, radioisotopes, therapeutic agents, rapamycin, vinblastine, taxol, non-protein fluorescent chemical groups, FITC, hydrazide, rhodamine, maleimide, protein fluorescent groups, GFP, YFP, and mCherry.

BRIEF DESCRIPTION OF THE DRAWINGS

[0057] FIG. 1A shows an alignment of the D-helix region of human yc-cytokine family members.

[0058] FIG. IB depicts the yc-box (SEQ ID NO: 10) and IL-2/IL-15 box (SEQ ID NO: 11) motifs which give rise to the consensus sequence around the D-helix region of the yc-cytokines.

[0059] FIG. 2 depicts a diagramed representation of the biochemical properties of amino acids.

[0060] FIG. 3A shows inhibition of IL-15 and IL-9 activity by ΒΝΖ-γ in a PT-18 proliferation assay. [0061] FIG. 3B shows a proliferation assay of CTLL-2 cells grown in the presence of IL-2 or IL-15 and 0, 0.1, 1 or 10 μΜ ΒΝΖ-γ.

[0062] FIG. 3C shows inhibition of IL-15 and IL-21 activity by SEQ ID NO: 3 in a NK92 proliferation assay.

[0063] FIG. 3D shows a proliferation assay of NK92 cells grown in the presence of IL-2, IL-15, or IL-21 and a μΜ dose response of SEQ ID NO: 3.

[0064] FIG. 4A shows inhibition of IL-15 -mediated tyrosine-phosphorylation of STAT5 by ΒΝΖ-γ.

[0065] FIG. 4B shows inhibition of IL-15-mediated phosphorylation of STAT5, but not IL-2-mediated phosphorylation of STAT5 in CTLL-2 cells by SEQ ID NO: 3, and inhibition of IL-21 -mediated phosphorylation of STAT3 in NK92 cells by SEQ ID NO: 3.

[0066] FIG. 5A shows an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood. T-cell proliferation is inhibited by addition of ΒΝΖ-γ.

[0067] FIG. 5B shows the population of CD4+CD25+ cells in an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood is diminished after adding BNZ-yto the culture.

[0068] FIG. 5C shows the population of CD4+Ki67 cells in an ex vivo T-cell proliferation assay using HAM/TSP peripheral blood is reduced after adding ΒΝΖ-γ to the culture.

[0069] FIG. 5D shows the percent of live cells by Guava staining in an ex vivo T- cell proliferation assay using HAM/TSP peripheral blood is not impacted after adding ΒΝΖ-γ to the culture.

[0070] FIG. 6 shows the alignment of the sequence of SEQ ID NO: 3 to the D-helix regions of different human yc-cytokine family members. The shaded areas represent amino acid sequences of the human yc-cytokine family members that are identical to their corresponding amino acids in the sequence of SEQ ID NO: 3.

[0071] FIG. 7 shows the relative inhibition of IL-15 activity by a series of custom peptide derivatives of SEQ ID NO: 3 containing an intra-peptide hydrocarbon linker element spanning a single helical turn (amino acid positions i and i+4) or two helical turns (amino acid positions i and i+7) in the alpha-helical secondary structure of each custom peptide derivative as compared to the unmodified SEQ ID NO: 3 in a NK92 proliferation assay. [0072] FIG. 8A shows a time-course protease stability measurement of unmodified SEQ ID NO: 3 in comparison with a representative custom peptide derivative of SEQ ID NO: 3 containing one hydrocarbon linker element (SEQ ID NO: 39), and another representative custom peptide derivative of SEQ ID NO: 3 containing two hydrocarbon linker elements and certain amino acid positions in the (D) stereochemical configuration (SEQ ID NO: 57) in simulated intestinal fluid over 60 minutes.

[0073] FIG. 8B shows a time-course protease stability measurement of a representative custom peptide derivative of SEQ ID NO: 3 containing one hydrocarbon linker element (SEQ ID NO: 39) in comparison with a representative custom peptide derivative of SEQ ID NO: 3 containing two hydrocarbon linker elements (SEQ ID NO: 83) in simulated intestinal fluid over 120 minutes.

[0074] FIG. 9 shows inhibition of IL-15 and IL-21 induced gene transcription of interferon gamma (IFNg) by SEQ ID NO: 83 (10 μΜ), but not that of the non-yc cytokine IL- 12 induction of IFNg gene transcription in the human NK92 cell line.

DETAILED DESCRIPTION

Overview

[0075] More than 100 cytokines have been identified so far and are considered to have developed by means of gene duplications from a pool of primordial genes (See Bazan, J.F. 1990, Immunol. Today 11:350-354). In support of this view, it is common for a group of cytokines to share a component in their multi-subunit receptor system. The most well- documented shared cytokine subunit in T cells is the common γ subunit (yc- subunit).

[0076] The yc-subunit is shared by 6 known cytokines (Interleukin-2 (IL-2), Interleukin-4 (IL-4), Interleukin-7 (IL-7), Interleukin-9 (IL-9), Interleukin-15 (IL-15), and Interleukin-21 (IL-21), collectively called the "YC-cytokines" or "yc-family cytokines") and plays an indispensable role in transducing cell activation signals for all these cytokines. Additionally, for each of the yc-cytokines, there are one or two private cytokine- specific receptor subunits that when complexed with the yc-subunit, give rise to a fully functional receptor. (See Rochman et al., 2009, Nat Rev Immunol. 9: 480-90.)

[0077] The yc-family cytokines are a group of mammalian cytokines that are mainly produced by epithelial, stromal and immune cells and control the normal and pathological activation of a diverse array of lymphocytes. These cytokines are critically required for the early development of T cells in the thymus as well as their homeostasis in the periphery. For example, in the absence of the yc-subunit, T, B and NK cells do not develop in mice. (See Sugamura et al., 1996, Annu. Rev. Immunol.14: 179-205).

[0078] The yc-cytokines are important players in the development of the lymphoid cells that constitute the immune system, particularly T, B, and NK cells. Further, yc-cytokines have been implicated in various human diseases. Thus, factors that inhibit yc-cytokine activity would provide useful tools to elucidate the developmental mechanism of subsets of lymphocytes and to treat immune disorders and yc-cytokine-mediated diseases.

[0079] Germ line depletion of the genes encoding the yc-subunit in mice or mutations of yc-subunit in humans are known to cause severe combined immunodeficiency (SCID) by disrupting the normal appearance or function of NK, T, and B cells. The importance of the yc-subunit in the signal transduction of the yc-cytokines, IL-2, -4, -7, -9, 15, -21, is indicated in studies demonstrating the lack of response of lymphocytes from these mice and human patients to the yc-cytokines (reviewed in Sugamura et al., 1995 Adv. Immunol. 59:225-277). This indicates that disruption of the interaction between the yc-subunit and a yc-cytokine would efficiently block the intracellular signaling events by the yc-cytokine family members. Therefore antagonist peptides according to the present embodiments are expected to effectively block the pathogenic changes in humans suffering from the diseases mediated by misregulation of the yc-cytokine family members.

[0080] As an alternative to antibody-mediated approaches for modulating the activity of individual yc-cytokines, Applicants have devised novel, low molecular weight compounds herein referred to as "Simul-Block", which suppress the activity of multiple yc-cytokines. These low molecular weight compounds, which include both chemicals and peptides, are less immunogenic than antibodies. These properties distinguish Simul-Block as a more efficient strategy for mediating yc-cytokine activity in clinical interventions.

Pathologies Associated with the yc-Cytokines

[0081] Recent studies have indicated that dysregulation of expression and dysfunction of the yc-cytokines could lead to a wide variety of human immunologic and hematopoietic diseases. IL-2

[0082] While IL-2 was historically considered a prototype T cell growth factor, the generation of a knockout mouse lacking IL-2 expression revealed that IL-2 is not critical for the growth or development of conventional T cells in vivo. Over-expression of IL-2, however, leads to a preferential expansion of a subset of T-cells; the regulatory T cells (T-regs). (See Antony et al., 2006, J. Immunol. 176:5255-66.) T-regs suppress the immune responses of other cells and thus act to maintain peripheral tolerance (reviewed in Sakaguchi et al., 2008, Cell 133:775-87). Breakdown of peripheral tolerance is thought to cause autoimmune diseases in humans.

[0083] Thus, the immunosuppressive function of T-regs is thought to prevent the development of autoimmune diseases (See Sakaguchi et al., 2008, Cell 133:775-87). T-regs have also been implicated in cancer, where solid tumors and hematologic malignancies have been associated with elevated numbers of T-regs (See De Rezende et al., 2010, Arch. Immunol. Ther. Exp. 58: 179-190).

IL-4

[0084] IL-4 is a non-redundant cytokine involved in the differentiation of T helper cells into the Th2 (T-helper type 2) subset, which promotes the differentiation of premature B cells into IgE-producing plasma cells. IgE levels are elevated in allergic asthma. Thus, IL-4 is implicated in the development of allergic Asthma. Antibodies targeting IL-4 can be used to treat or even prevent the onset of allergic asthma. (See Le Buanec et al., 2007, Vaccine 25:7206-16.)

IL-7

[0085] IL-7 is essential for B cell development and the early development of T cells in the thymus. In mice, the abnormal expression of IL-7 causes T-cell-associated leukemia. (See Fisher et al., 1993, Leukemia 2:S66-68.) However, in humans, misregulation of IL-7 does not appear to cause T-cell-associated leukemia. In humans, up-regulation of IL-7 either alone or in combination with another yc-cytokine family member, IL-15, has been implicated in Large Granular Lymphocyte (LGL) leukemia. IL-9

[0086] The role of IL-9 is still rather uncharacterized compared to other yc-cytokine family members. Mice depleted of the IL-9 gene appear normal and do not lack any subsets of cells in the lymphoid and hematopoietic compartments. Recent studies, however, reveal an in vivo role for IL-9 in the generation of Thl7 (T-helper induced by interleukin-17) cells (See Littman et al., 2010, Cell 140(6):845-58; and Nowak et al., 2009, J. Exp. Med. 206: 1653-60).

IL-15

[0087] IL-15 is critically involved in the development of NK cells, NK-T cells, some subsets of intraepithelial lymphocytes (IELs), γδ-Τ cells, and memory-phenotype CD8 T-cells (See Waldmann, 2007, J. Clin. Immunol. 27: 1-18; and Tagaya et al., 1996, EMBO J. 15:4928-39.) Over-expression of IL-15 in mice leads to the development of NK-T cell and CD 8 cell type T cell leukemia (See Fehniger et al., 2001, J. Exp. Med. 193:219-31 ; Sato et al. 2011 Blood 117:4032-40). These experimentally induced leukemias appear similar to LGL (large-granular lymphocyte) leukemia in humans, since in both instances the leukemic cells express CD8 antigen.

[0088] It is also suspected that IL-15 -mediated autocrine mechanisms may be involved in the leukemic transformation of CD4 T lymphocytes. (See Azimi et al., 1998, Proc. Natl. Acad. Sci. 95:2452-7; Azimi et al., 1999, J. Immunol. 163:4064-72; Azimi et al., 2000, AIDS Res. Hum. Retroviruses 16: 1717-22; and Azimi et al., 2001, Proc. Natl. Acad. Sci. 98: 14559-64). For example, CD4-tropic HTLV-I, which causes Adult T cell leukemia in humans, induces autocrine growth of virus-transformed T cells through the production of IL-15 and IL-15RCC (Azimi et al., 1998, Proc. Natl. Acad. Sci. 95:2452-7).

[0089] In addition to leukemic transformation, recent studies implicate IL-15 in the pathological development of Celiac disease (CD), an autoimmune disease. IL-15 is known to stimulate the differentiation of NK, CD8 and intestinal intraepithelial lymphocyte (IEL) cells into lymphokine-activated killer (LAK) cells by inducing the expression of cytolytic enzymes (i.e., Granzyme and Perforin) as well as interferon-γ. Celiac Disease (denoted CD from herein) is an immune-mediated enteropathy that is triggered by the consumption of gluten-containing food in individuals that express specific HLA-DQ alleles. [0090] The prevalence of this disease is 1% in the western population. The only current treatment for CD is the complete elimination of gluten from the patient's diet. The pathology of CD is mainly caused by extensive damage to the intestinal mucosa, which is caused by activated CD8 T cells that have infiltrated to the intestinal lamina propria. These CD8 T cells appear to be activated through mechanisms involving IL-15. One recent publication demonstrated in mice that ectopic over-expression of IL-15 by enterocytes leads to the development of enteropathy, which closely resembles the lesions in CD patients. Neutralization of IL-15 activity dramatically diminished the pathological changes. Thus, an intervention blocking the activation of CD 8 T cells by IL-15 appears to provide an alternative strategy in managing CD to the conventional gluten-free diet.

IL-21

[0091] IL-21 is the most recently discovered member of the yc-family. Unlike other family members, IL-21 does not appear to have potent growth-promoting effects. Instead, IL-21 is thought to function more as a differentiation factor than a factor controlling cellular proliferation (See Tagaya, 2010, J. Leuk. Biol. 87:13-15).

Current Strategies for Treating yc-Cytokine-Mediated Disorders

[0092] As yc-cytokines are thought to be involved in numerous human diseases, several methods of treating yc-cytokine-implicated diseases by inhibiting yc-cytokine family activities have been proposed. These methods include the use of cytokine- specific monoclonal antibodies to neutralize the targeted cytokine's activity in vivo; use of monoclonal antibodies targeting the private cytokine-specific receptor subunits (subunits other than the shared yc-subunit) to selectively inhibit cytokine activity; and use of chemical inhibitors that block the downstream intracellular cytokine signal transduction pathway.

[0093] While cytokine-specific antibodies are often the first choice in designing therapeutics, cytokines that share receptor components display overlapping functions (See Paul, W.E., 1989, Cell 57:521-24) and more than one cytokine can co-operate to cause a disease (See Examples described herein). Thus, approaches involving neutralization of a single cytokine may not be effective in the treatment of cytokine-implicated human diseases.

[0094] Strategies for designing therapeutics that inhibit the function of multiple cytokines via antibodies which recognize a shared receptor component have also been proposed. However, the multi-subunit nature of cytokine receptor systems and the fact that functional receptors for a single cytokine can assume different configurations makes this approach difficult.

[0095] For example, a functional IL-15 receptor can be either IL-15R /yc or E_--15Ra/ /yc. (See Dubois et al., 2002, Immunity 17:537-47.) An antibody against the IL-15R receptor (ΤΜβΙ), is an efficient inhibitor of the IL-15 function, but only when the IL-15Ra molecule is absent from the receptor complex. (See Tanaka et al., 1991, J. Immunol. 147:2222-28.) Thus, the effectiveness of a monoclonal anti-receptor antibody, whether raised against a shared or a private subunit, can be context-dependent and is unpredictable in vivo.

[0096] Although clinical use of monoclonal antibodies against biologically active factors or receptors associated with the pathogenesis of diseases is an established practice, there are few demonstrations of successful outcomes. Moreover, establishment of a clinically-suited monoclonal antibody treatment is a long and difficult process, with the successful generation of a neutralizing antibody largely a matter of luck. For example, due to the critical importance of the yc-subunit in mediating signaling by yc-family cytokines, many attempts to generate polyclonal and monoclonal antibodies against the yc-subunit have been made and there exist many commercial antibodies recognizing the yc-subunit in mice and in humans. Curiously, however, none of these anti-yc- subunit antibodies block the function of the yc-cytokines.

[0097] Another problem with the therapeutic use of monoclonal antibodies is that monoclonal antibodies are usually generated by immunizing rodents with human proteins, so the generated antibody is a foreign protein and thus highly immunogenic. To circumvent this problem, the amino acid sequence of the monoclonal antibody is molecularly modified so that the antibody molecule is recognized as a human immunoglobulin (a process called humanization), but this process requires time and expense.

Targeting JAK3, as an Existing Alternative Example for the Inhibition of Multiple yc-cytokines

[0098] The interaction between the yc-subunit and a yc-cytokine leads to the activation of an intracellular protein tyrosine kinase called Janus kinase 3 (Jak3). Jak3, in turn, phosphorylates multiple signaling molecules including STAT5, and PI3 kinase. The interaction of the yc-subunit and Jak3 is very specific. In fact, there is no other receptor molecule that recruits Jak3 for signal transduction. (See O'Shea, 2004, Ann. Rheum. Dis. 63:(suppl. II):ii67-7.) Thus, the inhibition of cytokine signaling through the yc-subunit can be accomplished by blocking the activity of Jak3 kinase. Accordingly, multiple chemical inhibitors that target the kinase activity of Jak3 have been introduced to the market. (See Pesu et al., 2008, Immunol. Rev. 223:132-142.) One such example is CP690,550.

[0099] The major shortcoming of these protein kinase inhibitors is the lack of specificity to Jak3 kinase. These drugs intercept the binding of ATP (adenosine-triphosphate) molecules to Jak3 kinase, a common biochemical reaction for many protein kinases, and thus tend to block the action of multiple intracellular protein kinases that are unrelated to Jak3 kinase whose actions are critically needed for the well-being of normal cells in various tissues. Thus, more specific inhibitors of signaling through the yc-subunit are needed.

[0100] There is therefore a great need for an alternative strategy for treating yc-cytokine-implicated diseases.

Discovery of the yc-box

[0101] The C-terminus (the D-helix) of the yc-cytokines contains the proposed site for interacting with the common yc-subunit of the multi-unit cytokine receptors. (Bernard et al., 2004 J. Biol. Chem. 279:24313-21.) Comparison of the biochemical properties of the amino acids of all yc-cytokines identified in mice and humans revealed that the chemical nature of the amino acids, for example, hydrophobicity, hydrophilicity, base/acidic nature, are conserved, if not identical, at many positions in the D-helix across the members of the yc-cytokine family.

[0102] In contrast, the sequence of IL-13, which is related to the yc-cytokine IL-4, but does not bind to the yc-subunit, does not exhibit significant homology in the D-helix region to the yc-cytokines, suggesting that the sequence homology in the D-helix region is correlated with binding to the yc-subunit. As shown in FIG. 1A, alignment of the amino acid sequences of the D-helix region of yc-cytokine family members in humans reveals a motif of moderate sequence homology in these cytokines referred to herein as "the yc-box".

[0103] The yc-box (SEQ ID NO: 10) comprises 19 amino acids where out of the 19 positions, positions 4, 5, and 13 are fully conserved as Phenylalanine, Leucine, and Glutamine, respectively. Less conservation is observed at positions 6, 7 and 11 of the yc-box where the amino acid is one of two or three related amino acids that share physico-chemical properties: position 6 may be occupied by the polar amino acids Glutamate, Asparagine or Glutamine; non-polar amino acids Serine or Arginine can occupy position 7; and position 11 is occupied by either of the non-polar aliphatic amino acids Leucine or Isoleucine. Positions 9 and 16 may be occupied by the either the non-polar amino acid Isoleucine or the polar amino acid Lysine. See FIG. IB. Some differences in the amino acid composition of the yc-box are observed at positions 9 and 16 amongst subfamilies of the yc-cytokines. Comparison of the yc-cytokines across species indicates that Isoleucine is often present at the 9 and 16 positions in the IL-2/15 subfamily, whereas the other yc-family members often possess Lysine in these positions. Not wishing to be bound by a particular theory, Isoleucine and Lysine are biochemically different and thus may impart specific conformational differences between the IL-2/15 subfamily and other yc-cytokines.

[0104] Conservation of the yc-box motif between yc-cytokines is supported by findings that a Glutamine (Gin, Q) residue located in the D-helix region is critical for the binding of the yc-cytokines to the yc-subunit. (Bernard et al., 2004 J. Biol. Chem. 279: 24313-21.)

Peptide Inhibitors of yc-Cytokine Activity

[0105] The activity of yc-family cytokines may be blocked by disrupting the interaction between the yc-cytokine and the yc-subunit, for example by introducing a competitive inhibitor which can interact with the yc-subunit without stimulating signaling through the multi-subunit cytokine receptors. Not to be bound by a particular theory, the conserved yc-box motif, which participates in binding of the yc-family cytokines to the yc-subunit, presents a core base amino acid sequence which can be utilized to design peptide inhibitors of yc-cytokine signaling.

[0106] Provided herein are stable composite peptides, and compositions, kits and/or systems thereof to modulate yc-cytokine signaling. The terms "composite peptide," "oligopeptide," "polypeptide," "peptide," and "protein" can be used interchangeably when referring to the "custom peptide derivatives" provided in accordance with the present embodiments and can be used to designate a series of amino acid residues of any length. The peptides of the present embodiments may be linear or cyclic. The peptides of the present embodiments may include natural amino acids, non-natural amino acids amino acids in (D) stereochemical configuration, amino acids in (L) stereochemical configuration, amino acids in (R) stereochemical configuration, amino acids in (S) stereochemical configuration, or a combination thereof.

[0107] The core yc-box amino acid sequence comprises: D/E-F-L-E/Q/N-S/R-X- I/K-X-L/I-X-Q (SEQ ID NO: 2) (where X denotes any amino acid). At least some embodiments described herein relate to custom peptide derivatives of the core yc-box amino acid sequence which can inhibit the activity of one or more yc-cytokines. Custom peptide derivatives include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to the core yc-box amino acid sequence. Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of the core yc-box. For example, amino acids with similar physico-chemical properties would include Phenylalanine, Tyrosine, Tryptophan, and Histidine, which are aromatic amino acids. FIG. 2 shows a diagrammed representation of amino acids with similar physico-chemical properties which may be may be substituted for the amino acids comprising the core yc-box. Peptide derivatives of the core yc-box may be 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length. In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/pep tides.

[0108] Some embodiments described herein relate to enhancing the formation of and/or stabilizing one or more helical secondary structures of custom peptide derivatives of the core yc-box. In some embodiments, a helical secondary structure of a custom peptide derivative of the core yc-box is enhanced and/or stabilized by incorporating one or more intra- peptide hydrocarbon linker elements (often referred to as "staples" in the literature).

[0109] As used herein, the term "staples," "hydrocarbon," "hydrocarbon linker- element," "hydrocarbon linker chain," "linker element," "linker chain" or "linker" refers to any carbon-containing chemical chain (straight or branched) comprising at least 1 to about 20 carbon atoms. The carbon-containing chemical chain may have one or more chemical group substituents covalently attached within the chain. The carbon-containing chemical chain may have one or more carbon-carbon double bonds in either Z or E geometric configurations, or a combination of Z and E geometric configurations. The carbon-containing chemical chain may also have one or more carbon-carbon triple bonds. In some embodiments, the carbon- containing chemical chain may have one or more carbon-carbon double bonds in either Z or E geometric configurations, or a combination of Z and E geometric configurations and may additionally have one or more carbon-carbon triple bonds. The carbon-containing chemical chain may also covalently incorporate halogen atom substitutions (one or more of fluorine, chlorine, bromine, and/or iodine) according to the chemical valences allowed by the halogen atom. The carbon containing chemical chain may covalently incorporate heteroatom substitutions (one or more of oxygen, nitrogen, and/or sulfur) according to the chemical valences allowed by the heteroatom. The carbon-containing chemical chain may covalently incorporate, without limitations, one or more carbon aromatic ring systems (non-limiting examples include phenyl, naphthyl, anthracyl, etc.) with or without substituents, one or more heteroaryl ring systems (non-limiting examples include furan, thiophene, pyrrole, pyridine, pyran, oxazine, thiazine, pyrimidine, pyridazine, pyrazine, thiine, pyrazole, imidazole, triazole, indole, quinolone, isoxazole, oxazole, isothiazole, thiazole, etc.) with or without substituents, one or more non-aromatic carbon ring systems (non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.) with or without substituents, and one or more heterocyclyl ring systems (non-limiting examples include ethylene oxide, ethylene imine, ethylene sulfide, tetrahydrofuran, tetrahydrothiophene, tetrahydropyran, tetrahydrothiopyran, dioxane, pyrrolidine, piperidine, piperazine, morpholine, etc.) with our without substituents. Suitable substituents, without limitation, include one or more of alkyl, alkaryl, aryl, aralkyl, alkoxy, thioalkoxy, aryloxy, haloalkyl, halo, oxo, nitro, hydroxy, mercapto, carboxy, alkylcarbonyl, alkoxycarbonyl, alkanesulfonyl, amino, amido, azido, cyano, PEG, affinity labels, targeting moiety, fatty-acid derived acyl group, biotin, radioisotopes, therapeutic agents (non-limiting examples include rapamycin, vinblastine, taxol, etc.), non-protein fluorescent chemical groups (non-limiting examples include FITC, hydrazide, rhodamine, maleimide, etc.), and protein fluorescent groups (non-limiting examples include GFP, YFP, mCherry, etc.).

[0110] In some embodiments, one or more linker elements can join any two or more separate custom peptide derivatives of the present disclosure. In some embodiments, when one or more linker elements join any two or more separate custom peptide derivatives, the linker elements are referred to as inter-peptide linker elements.

[0111] In some embodiments, core yc-box custom peptide derivatives comprise two or more oc-alkenyl substituted amino acids. In some embodiments, the two or more oc- alkenyl substituted amino acids are linked via one or more intra-peptide hydrocarbon linker elements incorporated at the a-alkenyl substituted amino acids. In some embodiments, the a- alkenyl substituted amino acids are utilized to catalyze the formation of an intra-peptide hydrocarbon linker element by ring-closing metathesis during peptide synthesis. Intra-peptide linker elements join separate amino acids on the same sequence of a custom peptide derivative of the present disclosure. In some embodiments, the peptides of the present disclosure are linear or cyclic. In some embodiments, one or more peptides have a combination of inter-peptide and intra-peptide hydrocarbon linkers. It will be appreciated by one of ordinary skill in the art that any combination of linear and cyclic peptides and any combination and number of inter-peptide and intra-peptide hydrocarbon linkers are possible. For example, a circular peptide may have one or more intra-peptide linkers linking two substituted amino acids (e.g., α-alkenyl substituted amino acids) within the circular peptide. The circular peptide may additional be linked via one or more inter-peptide linkers to another peptide comprising one or more substituted amino acids (e.g., α-alkenyl substituted amino acids).

[0112] Non-limiting examples of α-alkenyl substituted amino acids include R- propenylalanine (CAS: 288617-76-5; R3Ala), S-propenylalanine (CAS: 288617-71-0; S3Ala), D-allylglycine (CAS: 170642-28-1 ; D3Gly), L-allylglycine (CAS: 146549-21-5; L3Gly), R-pentenylalanine (CAS: 288617-77-6; R5Ala), S-pentenylalanine (CAS: 288617- 73-2; S5Ala), R-pentenylglycine (CAS: 1093645-21-6; R5Gly), S-pentenylglycine (CAS: 856412-22-1; S5Gly), R-butenylalanine (CAS: 1311933-82-0; R4Ala), S-butenylalanine (CAS: 288617-72-1; S4Ala), R-butenylglycine (CAS: 865352-21-2; R4Gly), S- butenylglycine (CAS: 851909-08-5; S4Gly), R-hexenylalanine (CAS: 288617-78-7; R6Ala), S-hexenylalanine (CAS: 288617-74-3; S6Ala), R-hexenylglycine (CAS: 1208226-88-3; R6Gly), S-hexenylglycine (CAS: 1251904-51-4; S6Gly), R-heptenylalanine (CAS: 1311933- 84-2; R7Ala), S-heptenylalanine (CAS: 1311933-83-1; S7Ala), R-heptenylglycine (CAS: 1262886-63-4; R7Gly), S-heptenylglycine (CAS: 1058705-57-9; S7Gly), di-substituted bis- propenylglycine (CAS: 1311992-97-8; bis3Gly), di-substituted bis-pentenylglycine (CAS: 1068435-19-7; bis5Gly), di-substituted bis-butenylglycine (bis4Gly), di-substituted bis- hexenylglycine (bis6Gly), di-substituted bis-heptenylglycine (bis7Gly), R-octenylalanine (CAS: 945212-26-0; R8Ala), S-octenylalanine (CAS: 288617-75-4; S8Ala), R-octenylglycine (CAS: 1191429-20-5; R8Gly), and S-octenylglycine (CAS: 1262886-64-5; S8Gly) (See TABLE 1). Other amino acid substitutions are also contemplated and within the scope of this disclosure. [0113] It will be understood by one of ordinary skill in the art that any amino acid at any of the positions in the embodiments of the peptides disclosed herein can be a substituted amino acid (e.g., an oc-alkenyl substituted amino acid). In some embodiments, other modifications of amino acids that allow for the formation of one or more inter-peptide and/or intra-peptide linkages (e.g., via hydrocarbon linker elements) are also contemplated.

[0114] Intra-peptide hydrocarbon linker elements have been shown to increase stability of peptide(s) by decreasing the susceptibility of the peptide(s) to proteolytic digestion (reviewed in Walensky and Bird, 2014, J. Med. Chem. 57:6275-88, which is hereby incorporated by reference in its entirety).Thus, in some embodiments, intra-peptide hydrocarbon linker elements decrease the susceptibility of peptides to degradation by serine proteases, cysteine proteases, threonine proteases, aspartic proteases, glutamic proteases, metalloproteases, asparagine peptide lyases or a combination thereof. In some embodiments, intra-peptide hydrocarbon linker elements improve the biological activity of certain peptides derived from alpha-helical regions by stabilizing the bioactive secondary structure of peptide. In some embodiments, a biological activity corresponds to inhibition of the effects of a cytokine.

[0115] Based on the identification of the conserved yc-box motif in cytokines which bind to the yc-subunit, Applicants have devised a novel, 19-mer custom derivative peptide which is an artificial composite peptide combining partial amino acid sequences of both the human IL-2 and IL-15 yc-box. The 19-mer peptide, herein referred to as ΒΝΖ-γ, consists of the amino acid sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), where the amino acids depicted by bold characters are conserved between IL-2 and IL-15 and the underlined amino acids represent positions where the physico-chemical properties of the amino acids are conserved.

[0116] Applicants discovered that the 19-mer ΒΝΖ-γ, suppresses IL-15 and IL-9 induced cellular proliferation, but not IL-3 or IL-4 induced cellular proliferation. See FIG. 3 A and EXAMPLE 2. Applicants further demonstrated that ΒΝΖ-γ inhibits IL-15 mediated phosphorylation of the intracellular cytokine signal transduction molecule, STAT-5. See FIG. 4A and EXAMPLE 5. These results demonstrate that custom peptide derivatives of the conserved yc-box motif can inhibit the activity of multiple yc-cytokines.

[0117] Several embodiments relate to custom derivative peptides of the 19-mer ΒΝΖ-γ amino acid sequence, I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), which can inhibit the activity of one or more yc-cytokines. Custom peptide derivatives of the 19-mer ΒΝΖ-γ amino acid sequence include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1). Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of sequence: I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1). In several embodiments, the amino acid residues of the custom derivative peptides retain similar physico-chemical properties with the amino acid residues of ΒΝΖ-γ, but exhibit different biological inhibition specificity to the 6 yc-cytokine family members from that of the original 19-mer peptide. Peptide derivatives of BNZ-y may be 19, 20, 21, 22, 23, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length. In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/peptides. The other moieties may include proteins or peptides that stabilize the composite peptide, or other moieties, including without limitation, bovine serum albumin (BSA), albumin, Keyhole Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell- specific antigen, a receptor, a ligand, a metal ion and Poly Ethylene Glycol (PEG).

[0118] In some embodiments, any of the custom peptide derivatives disclosed herein can comprise one or more intra-peptide hydrocarbon linker elements. In some embodiments, the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) comprises one or more intra-peptide hydrocarbon linker elements. In some embodiments, the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 1. In some embodiments, the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 7 residues apart on SEQ ID NO: 1. In some embodiments, the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 1 and 7 residues apart on SEQ ID NO: 1.

[0119] In some embodiments, when the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 1. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 1. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 1.

[0120] In some embodiments, when the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 1. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position C terminal to the last residue of SEQ ID NO: 1. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 1.

[0121] In some embodiments, when the 19-mer ΒΝΖ-γ (SEQ ID NO: 1) is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 1 and at a position C terminal to the last residue SEQ ID NO: 1. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 1 and at a single position C terminal to the last residue of SEQ ID NO: 1. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 1 and at a position C terminal to the last residue of SEQ ID NO: 1.

[0122] In some embodiments of the 19-mer ΒΝΖ-γ (SEQ ID NO: 1), none or one or more of the amino acids are joined through each intra-peptide hydrocarbon linker element positioned on the one or more additional moieties conjugated to one or more of the residues of SEQ ID NO: 1. In some embodiments, 0 or 1 of the amino acids joined through each intra- peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 1. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 1.

[0123] In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element may include natural or non- natural amino acids in the (D) as well as (L), or the (R) as well as (S), stereochemical configuration. In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise natural amino acids, non-natural amino acids, or a combination thereof. In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise the (D) stereochemical configuration, the (L) stereochemical configuration, or a combination thereof. In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise the (R) stereochemical configuration, the (S) stereochemical configuration, or a combination thereof.

[0124] In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon or non- substituted side chain alpha carbon. In some embodiments of the 19-mer ΒΝΖ-γ, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon, a non-substituted side chain alpha carbon or a combination thereof. The yc-box amino acid sequence comprising D/E-F-L-E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2) (where X denotes any amino acid) is the conserved sequence identified in the D-helix of each member of the yc-cytokine family that is important for interacting with the common yc-subunit of each multi-unit cytokine receptor.

[0125] In some embodiments, any of the custom peptide derivatives disclosed herein can comprise one or more intra-peptide hydrocarbon linker elements. In some embodiments, the composite peptide of SEQ ID NO: 2 comprises one or more intra-peptide hydrocarbon linker elements. In some embodiments, the composite peptide of SEQ ID NO: 2 comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 2. In some embodiments, the composite peptide of SEQ ID NO: 2 comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 7 residues apart on SEQ ID NO: 2. In some embodiments, the composite peptide of SEQ ID NO: 2 comprises one or more intra- peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 2 and 7 residues apart on SEQ ID NO: 2.

[0126] In some embodiments, when the composite peptide of SEQ ID NO: 2 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 2. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 2. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 2.

[0127] In some embodiments, when the composite peptide of SEQ ID NO: 2 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 2. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position C terminal to the last residue of SEQ ID NO: 2. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 2.

[0128] In some embodiments, when the composite peptide of SEQ ID NO: 2 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 2 and at a position C terminal to the last residue SEQ ID NO: 2. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 2 and at a single position C terminal to the last residue of SEQ ID NO: 2. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 2 and at a position C terminal to the last residue of SEQ ID NO: 2. [0129] In some embodiments of the composite peptide of SEQ ID NO: 2, none or one or more of the amino acids are joined through each intra-peptide hydrocarbon linker element positioned on the one or more additional moieties conjugated to one or more of the residues of SEQ ID NO: 2. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 2. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 2.

[0130] In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element may include natural or non-natural amino acids in the (D) as well as (L), or the (R) as well as (S), stereochemical configuration. In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise natural amino acids, non- natural amino acids, or a combination thereof. In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise the (D) stereochemical configuration, the (L) stereochemical configuration, or a combination thereof. In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra- peptide hydrocarbon linker element comprise the (R) stereochemical configuration, the (S) stereochemical configuration, or a combination thereof.

[0131] In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon or non- substituted side chain alpha carbon. In some embodiments of the composite peptide of SEQ ID NO: 2, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon, a non-substituted side chain alpha carbon or a combination thereof.

[0132] Several embodiments relate to custom peptide derivatives of the yc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7, which are depicted in FIG. 1A. Other embodiments relate to custom derivative peptides which are artificial composite peptides combining the amino acid sequence of two or more of the human IL-15, IL-2, IL-21, IL-4, IL-9, and IL-7 yc-box motifs. Several embodiments relate to custom peptide derivatives of the yc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7 having a partial amino acid sequence that shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequences of the of the yc-box motifs of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7. Custom peptide derivatives of the of the yc-box motifs of IL- 15, IL-2, IL-21, IL-4, IL-9, or IL-7 further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of the yc-box motif sequences of IL-15, IL-2, IL-21, IL-4, IL-9, or IL-7.

[0133] Several embodiments relate to custom peptide derivatives that would inhibit the function of one, all, or selective members of the yc-cytokines. In some embodiments, the custom peptide derivatives selectively target individual yc-cytokine family members. For example, a custom peptide derivative can selectively inhibit the function of IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21. In other embodiments, a custom peptide derivative can inhibit 2 or more yc-cytokine family members.

[0134] For example, the custom peptide derivatives of the present embodiments can selectively inhibit the function of IL-2 in combination with one or more of IL-4, IL-7, IL-9, IL-15, and IL-21; IL-4 in combination with one or more of IL-7, IL-9, IL-15, and IL-21; IL-7 in combination with one or more of IL-9, IL-15, and IL-21; IL-9 in combination with one or more of IL-2, IL-4, IL-7, IL-15, and IL-21 ; IL-15 in combination with one or more of IL-2, IL-4, IL-7, IL-9, and IL-21 ; or IL-21 in combination with one or more of IL-2, IL-4, IL-7, IL-9, and IL-15. In other embodiments, custom peptide derivatives can comprehensively target all yc-cytokine family members.

[0135] Not wishing to be bound by a particular theory, the custom peptide derivatives can inhibit the function of all or selective members of the yc-cytokines by diminishing the binding of yc-cytokines to the yc-subunit, for example, as a competitive inhibitor. Such custom peptide derivatives may be used in diverse applications, including as a clinical drug.

[0136] Several embodiments relate to custom peptide derivatives that would modulate (including enhance or reduce) the function of one, two, or more of selective members of the yc-cytokines. In some embodiments, the custom peptide derivatives selectively target individual yc-cytokine family members. For example, a custom peptide derivative can selectively enhance or inhibit the function of IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21. In other embodiments, a custom peptide derivative can enhance or inhibit two or more yc-cytokine family members. In certain embodiments, custom peptide derivatives may comprise P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), which can enhance or inhibit the activity of one, two or more of yc-cytokines. In certain embodiments, custom peptide derivatives may comprise P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), which can inhibit the activity of at least IL-15 and IL-21.

[0137] In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines. In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines by suppressing cell proliferation induced by the one or more yc-cytokines. In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines by inhibiting phosphorylation of the intracellular cytokine signal transduction molecule mediated by the one or more yc-cytokines. In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines by suppressing cell proliferation induced by the one or more yc-cytokines and by inhibiting phosphorylation of the intracellular cytokine signal transduction molecule mediated by the one or more yc-cytokines. In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines by one or more other mechanisms.

[0138] In some embodiments, one or more of the peptide sequences disclosed herein suppress proliferation of one or more cell types induced by one or more of the cytokines disclosed herein (e.g., IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21). In some embodiments, one or more of the peptide sequences disclosed herein suppress proliferation of one or more cell types induced by all of the cytokines disclosed herein. In some embodiments, one or more of the peptide sequences disclosed herein suppress proliferation of one or more cell types induced by some but not all of the cytokines disclosed herein. For example, the 21-mer SEQ ID NO: 3 suppressed IL-15 and IL-21 induced cellular proliferation, but not IL-2, IL-4, or IL-9 induced cellular proliferation. See FIG. 3C and EXAMPLE 2.

[0139] In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit the activity of one or more yc-cytokines by inhibiting phosphorylation of one or more intracellular cytokine signal transduction molecules mediated by the one or more yc-cytokines disclosed herein (e.g., IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21). In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit phosphorylation of one or more intracellular cytokine signal transduction molecules mediated by all of the yc-cytokines disclosed herein. In some embodiments, one or more of the custom peptide derivatives of the conserved yc-box motif disclosed herein can inhibit phosphorylation of one or more intracellular cytokine signal transduction molecules mediated by some but not all of the yc-cytokines disclosed herein. For example, SEQ ID NO: 3 inhibited IL-15 mediated phosphorylation of the intracellular cytokine signal transduction molecule, STAT-5, but not IL-2 mediated phosphorylation of STAT-5.

[0140] Also, for example, SEQ ID NO: 3 inhibited IL-21 mediated phosphorylation of the intracellular cytokine signal transduction molecule STAT-3. See FIG. 4B and EXAMPLE 5.

[0141] In some embodiments, custom peptide derivatives may include any peptide whose partial amino acid sequence shows approximately 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity to amino acid sequence: P-K-E-F-L-E-R- F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3). Custom peptide derivatives further include any peptide wherein a partial amino acid sequence of that peptide derivative comprises amino acids with similar physico-chemical properties to the amino acids of sequence: P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3).

[0142] In several embodiments, the amino acid residues of the custom derivative peptides retain similar physico-chemical properties with the amino acid residues of P-K-E-F- L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), but exhibit different biological inhibition specificity to the 6 yc-cytokine family members (i.e. IL-2, IL-4, IL-7, IL-9, IL-15, or IL-21) from that of the original peptide of P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L- S (SEQ ID NO: 3). Peptide derivatives of the sequence of P-K-E-F-L-E-R-F-V-H-L-V-Q-M- F-I-H-Q-S-L-S (SEQ ID NO: 3) may be 19, 20, 21, 22, 23, 24, 25-30, 30-35, 35-40, 40-45, 45-50, or more than 50 amino acids in length.

[0143] In some embodiments, the custom peptide derivatives may be conjugated to the N-termini, C-termini and/or to the side residues of existing biological proteins/pep tides. In some embodiments, the composite peptide of SEQ ID NO: 3 may be conjugated to other moieties through the N-terminus, C-terminus or side chains of the composite peptide. The other moieties may include proteins or peptides that stabilize the composite peptide, or other moieties, including without limitation, bovine serum albumin (BSA), albumin, Keyhole Limpet Hemocyanin (KLH), Fc region of IgG, a biological protein that functions as scaffold, an antibody against a cell-specific antigen, a receptor, a ligand, a metal ion and Poly Ethylene Glycol (PEG).

[0144] In some embodiments, any of the custom peptide derivatives disclosed herein can comprise one or more intra-peptide hydrocarbon linker elements. In some embodiments, the composite peptide of SEQ ID NO: 3 comprises one or more intra-peptide hydrocarbon linker elements. In some embodiments, the composite peptide of SEQ ID NO: 3 comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 3. In some embodiments, the composite peptide of SEQ ID NO: 3 comprises one or more intra-peptide hydrocarbon linker elements that connect two separate amino acids positioned 7 residues apart on SEQ ID NO: 3. In some embodiments, the composite peptide of SEQ ID NO: 3 comprises one or more intra- peptide hydrocarbon linker elements that connect two separate amino acids positioned 4 residues apart on SEQ ID NO: 3 and 7 residues apart on SEQ ID NO: 3.

[0145] In some embodiments, when the composite peptide of SEQ ID NO: 3 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 3. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 3. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 3.

[0146] In some embodiments, when the composite peptide of SEQ ID NO: 3 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 3. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position C terminal to the last residue of SEQ ID NO: 3. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position C terminal to the last residue of SEQ ID NO: 3.

[0147] In some embodiments, when the composite peptide of SEQ ID NO: 3 is part of a longer peptide sequence, none of the amino acids joined through each intra-peptide hydrocarbon or one or more of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 3 and at a position C terminal to the last residue SEQ ID NO: 3. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a single position N terminal to the first residue of SEQ ID NO: 3 and at a single position C terminal to the last residue of SEQ ID NO: 3. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be located at a position N terminal to the first residue of SEQ ID NO: 3 and at a position C terminal to the last residue of SEQ ID NO: 3.

[0148] In some embodiments of the composite peptide of SEQ ID NO: 3, none or one or more of the amino acids are joined through each intra-peptide hydrocarbon linker element positioned on the one or more additional moieties conjugated to one or more of the residues of SEQ ID NO: 3. In some embodiments, 0 or 1 of the amino acids joined through each intra-peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 3. In some embodiments, 0, 1, 2, 3, 4 or 5 of the amino acids joined through each intra-peptide hydrocarbon linker element may be positioned on the one or more additional moieties conjugated to SEQ ID NO: 3.

[0149] In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element may include natural or non-natural amino acids in the (D) as well as (L), or the (R) as well as (S), stereochemical configuration. In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise natural amino acids, non- natural amino acids, or a combination thereof. In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element comprise the (D) stereochemical configuration, the (L) stereochemical configuration, or a combination thereof. In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra- peptide hydrocarbon linker element comprise the (R) stereochemical configuration, the (S) stereochemical configuration, or a combination thereof.

[0150] In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, or 2 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon or non- substituted side chain alpha carbon. In some embodiments of the composite peptide of SEQ ID NO: 3, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 of the amino acids joined through each intra-peptide hydrocarbon linker element are connected through a substituted side chain alpha carbon, a non-substituted side chain alpha carbon or a combination thereof.

[0151] Peptides of the present embodiments may also contain one or more rare amino acids (such as 4-hydroxyproline or hydroxylysine), organic acids or amides and/or derivatives of common amino acids, such as amino acids having the C-terminal carboxylate esterified (e.g., benzyl, methyl or ethyl ester) or amidated and/or having modifications of the N-terminal amino group (e.g., acetylation or alkoxycarbonylamino), with or without any of a wide variety of side chain modifications and/or substitutions. Side chain modifications, substitutions or a combination thereof that may be present in the custom peptide derivatives of the present embodiments include, but are not limited to, oc-methyl, oc-alkenyl, alkylation, methylation, benzylation, t-butylation, tosylation, alkoxycarbonylamino, and the like.

[0152] Residues other than common amino acids that may be present include, but are not limited to, penicillamine, tetramethylene cysteine, pentamethylene cysteine, mercaptopropionic acid, pentamethylene-mercaptopropionic acid, 2-mercaptobenzene, 2-mercaptoaniline, 2-mercaptoproline, ornithine, diaminobutyric acid, aminoadipic acid, m-aminomethylbenzoic acid, and diaminopropionic acid.

[0153] Peptides of the present embodiments can be produced and obtained by various methods known to those skilled in the art. For example, the peptide may be produced by genetic engineering, based on the nucleotide sequence coding for the peptide of the present embodiments, or chemically synthesized by means of peptide solid-phase synthesis and the like, or produced and obtained in their combination. One skilled in the art of solid-phase peptide synthesis can readily incorporate natural or non-natural amino acids in the (D) as well as (L), or the (R) as well as (S), stereochemical configuration. It will also be apparent to one skilled in the art of solid-phase peptide synthesis to produce and obtain peptides containing one or more intra-peptide hydrocarbon linker elements of the present embodiments utilizing oc-substituted (such as oc-alkenyl) natural or non- natural amino acids in one or more of (D), (L), (R) or (S), stereochemical configurations, or a combination thereof. In some embodiments, an intra-peptide hydrocarbon linker element linking oc-substituted amino acids (e.g., oc-alkenyl amino acids) can be generated by catalyzing one or more ring-closing metathesis. In some embodiments, one or more intra-peptide hydrocarbon linker elements can be generated by catalyzing a ring-closing metathesis using benzylidenebis(tricyclohexyl- phosphine)-dichlororuthenium (Grubb 's catalyst) on the resin-bound peptide during peptide synthesis. In some embodiments, other ring-closing synthesis reactions and/or mechanisms during one or more known peptide synthesis processes are also contemplated. In some embodiments, one or more inter-peptide hydrocarbon linker elements can be generated by catalyzing a ring-closing metathesis using benzylidenebis(tricyclohexyl-phosphine)- dichlororuthenium (Grubb's catalyst) on the resin-bound peptide during peptide synthesis. In some embodiments, other ring-closing synthesis reactions and/or mechanisms during one or more known peptide synthesis processes are also contemplated.

[0154] In some embodiments, at least two alpha-alkenyl substituted amino acids in a composite peptide are linked by the at least one intra-peptide hydrocarbon linker element. In some embodiments, the at least two alpha-alkenyl substituted amino acids linked by an intra-peptide hydrocarbon linker element are separated by n-2 amino acids, wherein n represents the number of amino acids encompassed by the intra-peptide linkage.

[0155] In some embodiments, one or more intra-peptide hydrocarbon linker elements are incorporated at amino acid positions that correlate with a single oc-helical turn in a secondary structure of the composite peptide. In some embodiments, when the composite peptide comprises one or more non-contiguous single oc-helical turns, the amino acid positions that correlate with a single oc-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the single oc-helical turn and i+4 is the last amino acid position of the single oc- helical turn, and wherein amino acid positions i and i+4 comprise alpha-alkenyl substituted amino acids, and where i and i+4 are positioned 4 residues apart (4 spaced).

[0156] In some embodiments, one skilled in the art of solid-phase peptide synthesis can readily synthesize composite peptides comprising more than one intra-peptide hydrocarbon linker elements such that the composite peptide comprises more than one single oc-helical turn. In some embodiments, the more than one single oc-helical turns are noncontiguous, i.e., the more than one single oc-helical turns do not share a substituted amino acid. For example, in some embodiments, the composite peptide can comprise one or more intra-peptide hydrocarbon linker elements of Formula 1, Formula 2, Formula 3, Formula 4, Formula 5, Formula 6, Formula 7, and/or Formula 8 (See TABLE 1) that span more than one non-contiguous single a-helical turns of the composite peptide.

[0157] Not wishing to be bound to any specific peptide containing one or more intra-peptide hydrocarbon linker elements of the present embodiments, a generic peptide example containing one intra-peptide hydrocarbon linker element connecting two separate amino acids positioned 4 residues apart, or one α-helical turn (position i and position i+4), can have S-pentenylalanine (S5Ala) incorporated at each of the positions i and i+4 during solid-phase synthesis of the peptide before catalyzing ring-closing metathesis using Grubb's catalyst while the peptide is still resin-bound on the solid support. This will result in a peptide sequence containing the intra-peptide hydrocarbon linker element depicted below (SEQ ID NO: 78) positioned 4 residues apart:

i+4

SEQ ID NO: 78

[0158] In some embodiments, one or more intra-peptide hydrocarbon linker elements are incorporated at amino acid positions that correlate with a double α-helical turn in a secondary structure of the composite peptide. In some embodiments, when the composite peptide comprises one or more non-contiguous double α-helical turns, the amino acid positions that correlate with a double α-helical turn of the composite peptide correspond to amino acid positions i and i+7 of the composite peptide, where i is the first amino acid position of the double α-helical turn and i+7 is the last amino acid position of the double a- helical turn, and wherein amino acid positions i and i+7 comprise alpha-alkenyl substituted amino acids, and where i and i+7 are positioned 7 residues apart (7 spaced).

[0159] Not wishing to be bound to any specific peptide containing one or more intra-peptide hydrocarbon linker elements of the present embodiments, a generic peptide example containing one intra-peptide hydrocarbon linker element connecting two separate amino acids positioned 7 residues apart, or two oc-helical turns (position i and position i+7), can have R-octenylalanine (R8Ala) incorporated at position i and S-pentenylalanine (S5Ala) incorporated at position i+ 7 during solid-phase synthesis of the peptide before catalyzing ring-closing metathesis using Grubb' s catalyst while the peptide is still resin-bound on the solid support. This will result in a peptide sequence containing the intra-peptide hydrocarbon

i i+7

SEQ ID NO: 79

[0160] It will be appreciated by one of ordinary skill in the art that the positions of octenylalanine (R8Ala) and S-pentenylalanine (S5Ala) in SEQ ID NO: 79 can be switched such that S-pentenylalanine (S5Ala) is incorporated at position i and R-octenylalanine (R8Ala) is incorporated at position i+7 (SEQ ID NO: 80).

[0161] Not wishing to be bound to any specific peptide containing one or more intra-peptide hydrocarbon linker elements of the present embodiments, a generic peptide example containing one intra-peptide hydrocarbon linker element connecting two separate amino acids positioned 7 residues apart, or two oc-helical turns (position i and position i+7), can have S-octenylalanine (S8Ala) incorporated at position i and R-pentenylalanine (R5Ala) incorporated at position i+ 7 during solid-phase synthesis of the peptide before catalyzing ring-closing metathesis using Grubb' s catalyst while the peptide is still resin-bound on the solid support. This will result in a peptide sequence containing the intra-peptide hydrocarbon linker elements depicted below (SEQ ID NO: 81) positioned 7 residues apart: * ΛΛ ^Χ— X sSAla X X— X— X X— 85 Ala—

i i+7

SEQ ID NO: 81

[0162] It will be appreciated by one of ordinary skill in the art that the positions of S-octenylalanine (S8Ala) and R-pentenylalanine (R5Ala) in SEQ ID NO: 81 can be switched such that R-pentenylalanine (R5Ala) is incorporated at position i and S-octenylalanine (S8Ala) is incorporated at position i+7 (SEQ ID NO: 82).

[0163] In some embodiments, one skilled in the art of solid-phase peptide synthesis can readily synthesize composite peptides comprising more than one intra-peptide hydrocarbon linker elements such that the composite peptide comprises more than one double α-helical turn. In some embodiments, the more than one double α-helical turns are noncontiguous, i.e., the more than one double α-helical turns do not share a substituted amino acid. For example, in some embodiments, the composite peptide can comprise one or more intra-peptide hydrocarbon linker elements of Formula 17, Formula 18, Formula 19, and/or Formula 20 (See TABLE 1) that span more than one non-contiguous double α-helical turns of the composite peptide.

[0164] One skilled in the art of solid-phase peptide synthesis can readily synthesize peptides containing more than one intra-peptide hydrocarbon linker element of the present embodiments by incorporating a-alkenyl substituted amino acids at paired non- overlapping amino acid positions in the peptide, with each a-alkenyl substituted amino acid in the pair positioned a single α-helical turn apart (4 residues apart) or a double α-helical turn apart (7 residues apart) during solid-phase peptide synthesis before catalyzing ring-closing metathesis using Grubb' s catalyst while the peptide is still resin-bound on the solid support. In some embodiments, single peptides can comprise more than one intra-peptide hydrocarbon linker element that span a single α-helical turn (4 residues apart), can contain hydrocarbon linker elements that span a double α-helical turns (7 residues apart), or can contain a combination of both a single α-helical turn (4 residues apart) and a double α-helical turn (7 residues apart) intra-peptide hydrocarbon linker elements. [0165] In some embodiments, when the composite peptide comprises two or more contiguous single α-helical turns, the amino acid positions that correlate with the first single α-helical turn of the composite peptide correspond to amino acid positions i and i+4 of the composite peptide, where i is the first amino acid position of the first single α-helical turn and i+4 is the last amino acid position of the first single α-helical turn, and the amino acid positions that correlate with the second single α-helical turn of the composite peptide correspond to amino acid positions i+4 and i+8 of the composite peptide, where i+4 is the first amino acid position of the second single α-helical turn and i+8 is the last amino acid position of the second single α-helical turn, and wherein amino acid positions i, i+4 and i+8 comprise alpha-alkenyl substituted amino acids.

[0166] In some embodiments, one skilled in the art of solid-phase peptide synthesis can readily synthesize composite peptides comprising more than one intra-peptide hydrocarbon linker elements such that the composite peptide comprises more than one single α-helical turn. In some embodiments, the more than one single α-helical turns are contiguous, i.e., the more than one single α-helical turns share a substituted amino acid. Thus, when two single α-helical turns are contiguous, the last position of a preceding single α-helical turn is the first positon of a subsequent contiguous single α-helical turn. In some embodiments, the shared a substituted amino acid is a di- substituted amino acid (See TABLE 1).

[0167] Thus, in some embodiments, the composite peptide can comprise one or more intra-peptide hydrocarbon linker elements of Formula 9, Formula 10, Formula 11, Formula 12, Formula 13, Formula 14, Formula 15, and/or Formula 16 (See TABLE 1) that span two contiguous single α-helical turns of the composite peptide.

[0168] For example, in some embodiments, one skilled in the art of solid-phase peptide synthesis can readily synthesize a peptide containing more than one intra-peptide hydrocarbon linker element of the present embodiments by incorporating a di-substituted a- alkenyl glycine amino acid a position that is equidistant from one a-alkenyl substituted amino acid positioned a single α-helical turn apart (4 residues apart) toward the N-terminus of the peptide, and another a-alkenyl substituted amino acid positioned a single α-helical turn (4 residues apart) toward the C-terminus of the peptide during solid-phase peptide synthesis before catalyzing ring-closing metathesis using Grubb's catalyst while the peptide is still resin-bound on the solid support. [0169] In some embodiments, when the composite peptide comprises two or more contiguous double α-helical turns, the amino acid positions that correlate with the first double α-helical turn of the composite peptide correspond to amino acid positions i and i+7 of the composite peptide, where i is the first amino acid position of the first double α-helical turn and i+7 is the last amino acid position of the first double α-helical turn, and the amino acid positions that correlate with the second double α-helical turn of the composite peptide correspond to amino acid positions i+7 and i+14 of the composite peptide, where i+7 is the first amino acid position of the second double α-helical turn and i+14 is the last amino acid position of the second double α-helical turn, and wherein amino acid positions i, i+7 and i+14 comprise alpha-alkenyl substituted amino acids.

[0170] In some embodiments, one skilled in the art of solid-phase peptide synthesis can readily synthesize composite peptides comprising more than one intra-peptide hydrocarbon linker elements such that the composite peptide comprises more than one double α-helical turn. In some embodiments, the more than one double α-helical turns are contiguous, i.e., the more than one double α-helical turns share a substituted amino acid. Thus, when two double α-helical turns are contiguous, the last position of a preceding double α-helical turn is the first positon of a subsequent contiguous double α-helical turn. In some embodiments, the shared a substituted amino acid is a di-substituted amino acid (See TABLE 1).

[0171] Thus, in some embodiments, the composite peptide can comprise one or more intra-peptide hydrocarbon linker elements of Formula 21, Formula 22, and/or Formula 23 (See TABLE 1) that span two contiguous double α-helical turns of the composite peptide. When two double α-helical turns are contiguous, the last position of a preceding double a- helical turn is the first positon of a subsequent contiguous double α-helical turn.

[0172] For example, in some embodiments,, one skilled in the art of solid-phase peptide synthesis can readily synthesize a peptide containing more than one intra-peptide hydrocarbon linker element of the present embodiments by incorporating a di-substituted a- alkenyl glycine amino acid at a position equidistant from one a-alkenyl substituted amino acid positioned a double α-helical turn (7 residues apart) toward the N-terminus of the peptide, and another α-alkenyl substituted amino acid positioned a double α-helical turn apart (7 residues apart) toward the C-terminus of the peptide during solid-phase peptide synthesis before catalyzing ring-closing metathesis using Grubb' s catalyst while the peptide is still resin-bound on the solid support.

[0173] Peptides containing one or more intra-peptide hydrocarbon linker elements of the present embodiments can be produced through solid-phase peptide synthesis utilizing commercially available Boc- or Fmoc-protected a-alkenyl substituted natural or non-natural amino acids in the (D) as well as (L), or the (R) as well as (S), stereochemical configuration. The Fmoc-protected α-alkenyl substituted amino acids and the resultant hydrocarbon linker element following ring-closing metathesis that may be used in the synthesis of the custom peptide derivatives of the present embodiments include, but are not limited to TABLE 1 :

TABLE 1

α-alkenyl Substituted Amino α-alkenyl Substituted Amino α-alkenyl Substituted

Acid Acid Amino Acid

Peptide Position i Peptide Position i+4 Peptide Position i+8

R-pentenylalanine (CAS :

R-propenylalanine (CAS :

288617-77-6; R5Ala), S- 288617-76-5; R3Ala), S- pentenylalanine (CAS:

propenylalanine (CAS:

288617-73-2; S5Ala), R- 288617-71-0; S3Ala), D- N/A

pentenylglycine (CAS :

allylglycine (CAS: 170642-28

1093645-21-6; R5Gly), or S- 1; D3Gly), or L-allylglycine

pentenylglycine (CAS :

(CAS: 146549-21-5; L3Gly)

856412-22-1 ; S5Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 1

R5Ala, S5Ala, R5Gly, or R3Ala, S3Ala, D3Gly, or

N/A

S5Gly L3Gly

Hydrocarbon Linker Element Followin Ring-Closing Metathesis Formula 2

R-butenylalanine (CAS:

1311933-82-0; R4Ala), S- butenylalanine (CAS: 288617-

72-1; S4Ala), R- R4Ala, S4Ala, R4Gly, or

N/A butenylglycine (CAS: 865352- S4Gly

21-2; R4Gly), or S- butenylglycine (CAS: 851909- 08-5; S4Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 3

R5Ala, S5Ala, R5Gly, or R5Ala, S5Ala, R5Gly, or

N/A S5Gly S5Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 4

R-hexenylalanine (CAS:

288617-78-7; R6Ala), S- hexenylalanine (CAS :

R4Ala, S4Ala, R4Gly, or 288617-74-3; S6Ala), R-

N/A S4Gly hexenylglycine (CAS:

1208226-88-3; R6Gly), or S- hexenylglycine (CAS:

1251904-51-4; S6Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 5

R6Ala, S6Ala, R6Gly, or R4Ala, S4Ala, R4Gly, or N/A S6Gly S4Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 6

R-heptenylalanine (CAS :

1311933-84-2; R7Ala), S- heptenylalanine (CAS:

R3Ala, S3Ala, D3Gly, or 1311933-83-1; S7Ala), R-

N/A

L3Gly heptenylglycine (CAS :

1262886-63-4; R7Gly), or S- heptenylglycine (CAS :

1058705-57-9; S7Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 7

R7Ala, S7Ala, R7Gly, or R3Ala, S3Ala, D3Gly, or

N/A

S7Gly L3Gly

H drocarbon Linker Element Following Ring-Closing Metathesis

Formula 8

di-substituted bis-

R5Ala, S5Ala, R5Gly, or R5Ala, S5Ala, R5Gly, or propenylglycine (CAS:

S5Gly S5Gly

1311992-97-8; bis3Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 9

R3Ala, S3Ala, D3Gly, or di-substituted bis- R3Ala, S3Ala, D3Gly, or L3Gly pentenylglycine (CAS: L3Gly

1068435-19-7; bis5Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 10

R4Ala, S4Ala, R4Gly, or di-substituted bis- R4Ala, S4Ala, R4Gly, or

S4Gly butenylglycine (bis4Gly) S4Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 11

R5Ala, S5Ala, R5Gly, or R5Ala, S5Ala, R5Gly, or bis5Gly

S5Gly S5Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

,

Formula 12

R6Ala, S6Ala, R6Gly, or R6Ala, S6Ala, R6Gly, or bis4Gly

S6Gly S6Gly Hydrocarbon Linker Element Following Ring-Closing Metathesis f÷S

Formula 13

R4Ala, S4Ala, R4Gly, or di-substituted bis- R4Ala, S4Ala, R4Gly, or

S4Gly hexenylglycine (bis6Gly) S4Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

R7Ala, S7Ala, R7Gly, or R7Ala, S7Ala, R7Gly, or bis3Gly

S7Gly S7Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 15

R3Ala, S3Ala, D3Gly, or di-substituted bis- R3Ala, S3Ala, D3Gly, or L3Gly heptenylglycine (bis7Gly) L3Gly

H drocarbon Linker Element Following Ring-Closing Metathesis

Peptide Position i Peptide Position i+7 Peptide Position i+14

R-octenylalanine (CAS:

945212-26-0; R8Ala), S- octenylalanine (CAS: 288617-

R5Ala, S5Ala, R5Gly, or 75-4; S8Ala), R-

N/A

S5Gly octenylglycine (CAS:

1191429-20-5; R8Gly), or S- octenylglycine (CAS:

1262886-64-5; S8Gly)

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 17

R8Ala, S8Ala, R8Gly, or R5Ala, S5Ala, R5Gly, or

N/A

S8Gly S5Gly

H drocarbon Linker Element Following Ring-Closing Metathesis

Formula 18

R6Ala, S6Ala, R6Gly, or R7Ala, S7Ala, R7Gly, or

N/A

S6Gly S7Gly Hydrocarbon Linker Element Following Ring-Closing Metathesis

Formula 19

R7Ala, S7Ala, R7Gly, or R6Ala, S6Ala, R6Gly, or

N/A

S7Gly S6Gly

H drocarbon Linker Element Following Ring-Closing Metathesis

Formula 20

R8Ala, S8Ala, R8Gly, or R8Ala, S8Ala, R8Gly, or bis5Gly

S8Gly S8Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis -Ιί

Formula 21

R7Ala, S7Ala, R7Gly, or R7Ala, S7Ala, R7Gly, or bis6Gly

S7Gly S7Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

"i÷ 4

Formula 22

R6Ala, S6Ala, R6Gly, or R6Ala, S6Ala, R6Gly, or bis7Gly

S6Gly S6Gly

Hydrocarbon Linker Element Following Ring-Closing Metathesis

Ί÷7

Formula 23

[0174] n some embodiments, an intra-peptide hydrocarbon linker can be further functionalized through one or more chemical reactions. In some embodiments, one or more carbon-carbon double bond(s) present in the intra-peptide hydrocarbon linker (e.g., Formula 1 - Formula 23 in TABLE 1) can be utilized for organic chemical reactions to add one or more additional chemical functionalities. For example, alkene reactions may be utilized for custom peptide derivatives that contain one or more intra-peptide hydrocarbon linker elements of the present embodiments. Non-limiting examples of alkene reactions include hydroboration, oxymercuration, hydration, chlorination, bromination, addition of HF, HBr, HC1 or HI, dihydroxylation, epoxidation, hydrogenation, and cyclopropanation. In some embodiments, one or more additional chemical functionalities of the intra-peptide hydrocarbon linker elements can be achieved subsequent to the alkene reaction. Non-limiting examples include covalent addition of one or more chemical group substituents, such as nucleophilic reactions with epoxide and hydroxyl groups, and the like. In some embodiments, alkene reactions may be utilized to attach biotin, radioisotopes, therapeutic agents (non-limiting examples include rapamycin, vinblastine, taxol, etc.), non-protein fluorescent chemical groups (non-limiting examples include FITC, hydrazide, rhodamine, maleimide, etc.), and protein fluorescent groups (non-limiting examples include GFP, YFP, mCherry, etc.) to one or more inter- and/or intra-peptide hydrocarbon linker elements of the present embodiments.

[0175] In some embodiments, custom peptide derivatives comprising an intra- peptide hydrocarbon linker element spanning a single oc-helical turn (4 spaced) are provided. In some embodiments, custom peptide derivatives comprising an intra-peptide hydrocarbon linker element spanning a double oc-helical turn (7 spaced) are provided. In some embodiments, custom peptide derivatives comprising one or more intra-peptide hydrocarbon linker elements synthesized utilizing a-alkenyl substituted amino acids by ring-closing can span any number of amino acids. In some embodiments, custom peptide derivatives comprising one or more intra-peptide hydrocarbon linker elements synthesized utilizing a- alkenyl substituted amino acids by ring-closing span 2 to about 200 amino acids. In some embodiments, custom peptide derivatives comprising one or more intra-peptide hydrocarbon linker elements synthesized utilizing α-alkenyl substituted amino acids by ring-closing span 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195 or 200 amino acids, or a number within a range defined by any two of the aforementioned values. [0176] Non-limiting examples of composite peptides comprising one or more intra-peptide hydrocarbon linker elements are provided in TABLE 2. The examples in TABLE 2 are not limiting with respect to any specific a-alkenyl substituted amino acid useful for the synthesis of single α-helical turn (4 spaced) and/or double α-helical turn (7 spaced) intra-peptide hydrocarbon linker elements of the present embodiments and/or to any specific amino acid stereochemical configuration (e.g., (D) stereochemical configuration denoted with "d" in TABLE 2) in the custom peptide derivatives of the present embodiments.

TABLE 2

SEQ ID NO:

{S5Ala}-P-K-E-{S5Ala}-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S 12

{S5Ala}-K-E-F-{S5Ala}-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S 13

P-{S5Ala}-E-F-L-{S5Ala}-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S 14

P-K- { S5Ala} -F-L-E- {S5Ala} -F-V-H-L-V-Q-M-F-I-H-Q-S-L-S 15

P-K-E- { S5 Ala} -L-E-R- { S5 Ala} - V-H-L- V-Q-M-F-I-H-Q-S-L-S 16

P-K-E-F-{S5Ala}-E-R-F-{S5Ala}-H-L-V-Q-M-F-I-H-Q-S-L-S 17

P-K-E-F-L- { S5 Ala} -R-F-V- { S5 Ala} -L- V-Q-M-F-I-H-Q-S-L-S 18

P-K-E-F-L-E-{S5Ala}-F-V-H-{S5Ala}-V-Q-M-F-I-H-Q-S-L-S 19

P-K-E-F-L-E-R-{S5Ala}-V-H-L-{S5Ala}-Q-M-F-I-H-Q-S-L-S 20

P-K-E-F-L-E-R-F- { S5 Ala} -H-L- V- { S5 Ala} -M-F-I-H-Q-S-L-S 2Ϊ

P-K-E-F-L-E-R-F- V- { S5 Ala} -L- V-Q- { S5 Ala} -F-I-H-Q-S-L-S 22

P-K-E-F-L-E-R-F- V-H-{S5Ala}-V-Q-M-{S5Ala}-I-H-Q-S-L-S 23

P-K-E-F-L-E-R-F- V-H-L- { S5 Ala} -Q-M-F- { S5 Ala} -H-Q-S-L-S 24

P-K-E-F-L-E-R-F- V-H-L- V- { S5 Ala} -M-F-I- { S5 Ala} -Q-S-L-S 25

P-K-E-F-L-E-R-F- V-H-L- V-Q- { S5 Ala} -F-I-H- { S5 Ala} -S-L-S 26

P-K-E-F-L-E-R-F- V-H-L- V-Q-M-l S5Ala} -I-H-Q- { S5Ala} -L-S 27

P-K-E-F-L-E-R-F- V-H-L- V-Q-M-F- { S5 Ala} -H-Q-S- { S5 Ala} -S 28

P-K-E-F-L-E-R-F- V-H-L- V-Q-M-F-I-{S5Ala}-Q-S-L-{S5Ala} 29

P-K-E-F-L-E-R-F- V-H-L- V-Q-M-F-I-H- { S5 Ala} -S-L-S- { S5 Ala} 30

{R8Ala}-P-K-E-F-L-E-{S5Ala}-F- V-H-L- V-Q-M-F-I-H-Q-S-L-S 37

{R8Ala}-K-E-F-L-E-R-{S5Ala}-V-H-L- V-Q-M-F-I-H-Q-S-L-S 32

P-{R8Ala}-E-F-L-E-R-F-{S5Ala}-H-L- V-Q-M-F-I-H-Q-S-L-S 33

P-K-{R8Ala}-F-L-E-R-F-V-{S5Ala}-L- V-Q-M-F-I-H-Q-S-L-S 34 P-K-E-{R8Ala}-L-E-R-F-V-H-{S5Ala}-V-Q-M-F-I-H-Q-S-L-S 35

P-K-E-F-{R8Ala}-E-R-F-V-H-L-{S5Ala}-Q-M-F-I-H-Q-S-L-S 36

P-K-E-F-L- { R8 Ala} -R-F- V-H-L- V- { S5 Ala} -M-F-I-H-Q-S-L-S 37

P-K-E-F-L-E- { R8 Ala } -F- V-H-L- V-Q- { S5 Ala } -F-I-H-Q-S -L-S 38

P-K-E-F-L-E-R- {R8Ala} -V-H-L- V-Q-M- { S5 Ala} -I-H-Q-S-L-S 39

P-K-E-F-L-E-R-F- {R8Ala} -H-L- V-Q-M-F- { S5 Ala} -H-Q-S-L-S 40

P-K-E-F-L-E-R-F-V-{R8Ala}-L-V-Q-M-F-I-{S5Ala}-Q-S-L-S 41

P-K-E-F-L-E-R-F- V-H- { R8 Ala} -V-Q-M-F-I-H- { S5 Ala} -S-L-S 42

P-K-E-F-L-E-R-F- V-H-L- { R8Ala} -Q-M-F-I-H-Q- { S5 Ala} -L-S 43

P- K-E-F-L-E-R-F- V-H-L- V- {R8Ala} -M-F-I-H-Q-S- { S5 Ala} -S 44

P-K-E-F-L-E-R-F- V-H-L- V-Q- { R8 Ala} -F-I-H-Q-S-L- { S5 Ala} 45

P-K-E-F-L-E-R-F- V-H-L- V-Q-M-{R8Ala}-I-H-Q-S-L-S-{S5Ala} 46

{dP}-{dK}-{dE}-{dF}-{dL}-E-R-{R8Ala}-V-H-L-V-Q-F-{S5Ala}- I-{dH}-

47

{dQ}-{dS}-{dL}-{dS}

{dP}-{dK}-{dE}-{dF}-{dL}-E-R-{R8Ala}-V-H-L-V-Q-F-{S5Ala}-I-{ dH}-

48

{dQ}-{dS}-{dL}-S

{ S5 Ala 1 } *-P-K-E- { S5 Ala 1 } -L-E-R- { R8 Ala 2 } -V-H-L- V-Q-F- { S5 Ala 2 } -I-

49 {dH}-{dQ}-{dS}-{dL}-{dS}

{ S5 Ala! } -P-K-E- { S5 Ala 1 } -L-E-R- {R8Ala 2 } -V-H-L- V-Q-F- { S5 Ala 2 } -I-

50 {dH}-{dQ}-{dS}-{dL}-S

{ S5 Ala ! } -P-K-E- { S5 Alai } -L-E-R- {R8Ala 2 } -V-H-L- V-Q-F- { S5 Ala 2 } -

51

{S5Ala 3 }-H-Q-S-{S5Ala 3 }-{dS }

{ S5 Aki } -P-K-E- { S5 Ala ! } -L-E-R- {R8Ala 2 } -V-H-L- V-Q-F- { S5 Ala 2 } -

52 { S5 Ala 3 } -H-Q-S- { S5 Ala 3 } -S

{ dP } - { dK } - { dE } - { dF } - { dL } -E-R- { R8 Alai } - V-H-L- V-Q-F- { S5 Alai } -

53 {S5Ala 2 }-H-Q-S-{S5Ala 2 }-{dS }

{ dP } - { dK } - { dE } - { dF } - { dL } -E-R- { R8 Alai } - V-H-L- V-Q-F- { S5 Alai } -

54 {S5Ala 2 } -H-Q-S- {S5Ala 2 }-S

{ S5 Alai } -P-K-E- { S5 Alai } -L-E-R- { S5 Ala 2 } -V-H-L- { S5 Ala 2 } -Q-F-F-I-

55 {dH}-{dQ}-{dS}-{dL}-{dS}

{ S5 Alai } -P-K-E- { S5 Alai } -L-E-R- { S5 Ala 2 } -V-H-L- { S5 Ala 2 } -Q-F-F-I-

56 {dH}-{dQ}-{dS}-{dL}-S {dRl-ldRl-ldRl-ldRl-ldPl-ldKl-ldEl-ldFl-ISSAlajl-E-R-F-IRSAl a ! }-

57 {dH}-{S5Ala 2 }-V-Q-L-{S5Ala 2 }-I-{dH}-{dQ}-{dS}-{dL}-{dS}

{ S5 Ala 1 } -P-K-E- { S5 Ala 1 } -L-E-R- { S5 Ala 2 } -V-H-L- { S5 Ala 2 } -Q-F-F-

58

{ S5 Ala 3 } -H-Q-S- { S5 Ala 3 H dS }

{ S5 Ala 1 } -P-K-E- { S5 Ala 1 } -L-E-R- { S5 Ala 2 } -V-H-L- { S5 Ala 2 } -Q-F-F-

59 { S5 Ala 3 } -H-Q-S- { S5 Ala 3 } -S

{ dP } - { dK } - { dE } - { dF } - { dL } -E-R- { R8 Ala 1 } - V-H-L- V-Q-M- { S5 Ala 1 } -I-

60 {dH}-{dQ}-{dS}-{dL}-S

{ S5 Ala ! } -P-K-E- { S5 Ala 1 } -L-E-R- {R8Ala 2 } -V-H-L- V-Q-M- { S5 Ala 2 } -I-

61 {dH}-{dQ}-{dS}-{dL}-S

{ S5 Ala 1 } -P-K-E- { S5 Ala 1 } -L-E-R- { S5 Ala 2 } -V-H-L- { S5 Ala 2 } -Q-M-F-

62 { S5 Ala 3 } -H-Q-S- { S5 Ala 3 } -S

{S5Ala}-I-K-E-{S5Ala}-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S 63

I-K-E-F-L-Q-R-{S5Ala}-I-H-I-{S5Ala}-Q-S-I-I-N-T-S 64

I-K-E-F-L-Q-R- {R8 Ala} -I-H-I- V-Q-S-l S5 Ala} -I-N-T-S 65

I-K-E-F-L-Q-R-F-I-H-I-{S5Ala}-Q-S-I-{S5Ala}-N-T-S 66

I-K-E-F-L-Q-R-F-I-H-I- { R8 Ala } -Q-S-I-I-N-T- { S 5 Ala } 67

{ S5 Alaj } -I-K-E- { S5 Ak } -L-Q-R- { S5 Ala 2 } -I-H-I- { S5 Ala 2 } -Q-S-I-I-N-T-S 68

{ S5Al ai } -I-K-E- {S5Al ai } -L-Q-R- {R8Ala 2 } -I-H-I- V-Q-S-{S5Ala 2 }-I-N-T-S 69

{ S5Alai} -I- K-E-{S5Alai} -L-Q-R- F-I-H-I-{S5Ala 2 } -Q-S-I- {S5Ala 2 }-N-T-S 70

{ S5 Aki } -I-K-E- { S5 Aki } -L-Q-R-F-I-H-I- {R8Ala 2 } -Q-S-I-I-N-T- { S5 Ala 2 } TV

{ S5 Alaj } -I-K-E- { S5 Ak } -L-Q-R- { S5 Ala 2 } -I-H-I- { S5 Ala 2 } -Q-S-I-I- { dN } -

72

{dT}-{dS}

{S5Ala 1 }-I-K-E-{S5Ala 1 }-L-Q-R-{R8Ala 2 }-I-H-I-V-Q-S-{S5Ala 2 }-I-{dN}-

73

{dT}-{dS}

{SSAla -I-K-E-iSSAla -L-Q-R-F-I-H-I-iSSAla!l-Q-S-I-iSSAla!l-idN}-

74

{dT}-{dS}

{ S5 Alaj } -I-K-E- { S5 Ak } -L-Q-R- { S5 Ala 2 } -I-H-I- {bis5Gly 2,3 } -Q-S-I-

75

{S5Ala 3 }-N-T-S

{SSAla -I-K-E-iSSAla -L-Q-R-iSSAla!l-I-H-I-ibisSGly^l-Q-S-I-I-N-

76

T-{R8Ala 3 }

{ S5 Alaj } -I-K-E- { S5 Ak } -L-Q-R- { S5 Ala 2 } -I-H-I- {bis5Gly 2,3 } -Q-S-I- 77 ~ {S5Ala 3 }-{dN}-{dT}-{dS}

{S5Ala ! } - P - K - E - {SSAla^ - L - E - R - F - V - H - L - V - Q - K -

83

{S5Ala 2 } - 1 - H - Q - {S5Ala 2 } - L - S

*Subscript denotes corresponding pairs of hydrocarbon-linked -alkenyl substituted amino acids

[0177] One skilled in the art can synthesize the custom peptide derivatives with or without one or more hydrocarbon linker elements based on the present disclosure of the conserved yc-box motif and knowledge of the biochemical properties of amino acids as described in FIG. 2. Some embodiments also relate to polynucleotides comprising nucleotide sequences encoding the peptides of the present invention. "Nucleotide sequence," "polynucleotide," or "nucleic acid" can be used interchangeably, and are understood to mean either double- stranded DNA, a single- stranded DNA or products of transcription of the said DNAs (e.g., RNA molecules). Polynucleotides can be administered to cells or subjects and expressed by the cells or subjects, rather than administering the peptides themselves. Several embodiments also relate to genetic constructs comprising a polynucleotide sequence encoding the peptides of the present invention. Genetic constructs can also contain additional regulatory elements such as promoters and enhancers and, optionally, selectable markers.

Methods of treating yc-cytokine mediated diseases

[0178] Several embodiments relate to the use of yc-antagonist peptides in the treatment of yc-cytokine mediated diseases. Use of custom peptide derivative according to the present embodiments allows for flexibility in the design of the therapeutic agent (custom design of the peptide) and enables more comprehensive outcomes, which would not be accomplished by conventional strategies employing anti-cytokine or anti-cytokine receptor antibodies.

[0179] Described herein is a novel method of blocking the action of yc-family cytokines. Such manipulations can yield effective methods of clinical interventions in treating diseases related to the dysregulation or dysfunction of yc-cytokines. Examples of disease that may be treated by disrupting the interaction between the yc-cytokine and the yc-subunit include autoimmune diseases such as systemic lupus erythematosis, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease (CD), Hashimoto's or auto-immune thyroiditis; collagen diseases including rheumatoid arthritis, inflammatory bowel disease, diabetes mellitus (e.g., type 1 diabetes mellitus), autoimmune diseases of the skin such as psoriasis; degenerative neuronal diseases such as multiple sclerosis, uvietis or inflammation of the eye and sympathetic ophthalmia, graft-versus-host disease (GvHD), myasthenia gravis, inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease), Systemic Lupus Erythematosus, and alopecia areata.

[0180] In some embodiments, the yc-antagonist peptides described herein may be used in the treatment of Human T-cell Lymphotropic type I and II (HTLV-I and HTLV-II)- associated diseases including Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy and myositis. In some embodiments, the yc-antagonist peptides described herein may be used in the treatment of other viral diseases such as influenza, AIDS, HBV and Herpes or parasitic diseases.

[0181] In several embodiments, the yc-antagonist peptides may be administered before, during, and or after transplantation of various organs as an immunosuppressant agent.

[0182] In some embodiments, the yc-antagonist peptides described herein may be used in the treatment of immune-mediated diseases such as asthma and other inflammatory respiratory diseases, such as, but not limited to sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, lung fibrosis. In some embodiments, yc-antagonist peptides may be administered to treat or prevent allergic reactions due to exposure to allergens, chemical agents or other common causes of acute respiratory disease. In some embodiments, yc-antagonist peptides may be administered to treat or prevent inflammatory responses caused by viruses, bacteria, chemical reagents, and biochemical reagents.

[0183] In several embodiments, the yc-antagonist peptides may be administered to treat some types of malignancies such as LGL-leukemia, Intraepithelial lymphoma and leukemia in Refractory Celiac Disease, NK leukemia/lymphoma and NK-T leukemia/lymphoma

[0184] In some embodiments, custom peptide derivatives according to the embodiments described herein can be used for cosmetic purposes, such as the treatment of acne, hair loss, sunburn and nail maintenance, included to ointment as anti-aging component because of the anti-inflammatory nature of them. [0185] Several embodiments relate to therapeutic antagonist peptides that would inhibit the function of all or selective members of the yc-cytokines. In some embodiments, therapeutic antagonist peptides selectively inhibit individual yc-cytokine family members (custom peptides). In other embodiments, therapeutic antagonist peptides can comprehensively inhibit all yc-cytokine family members (Simul-Block). In some embodiments, therapeutic antagonist peptides selectively inhibit subsets of the yc-cytokines. Not wishing to be bound by a particular theory, the peptide antagonists can inhibit the function of all or selective members of the yc-cytokines by diminishing the binding of yc-cytokines to the yc-subunit, for example, as a competitive inhibitor.

[0186] Several members of the yc-cytokine family, IL-2, IL-7, and IL-15, but not IL-4 have been implicated as being involved in graft versus host disease (GvHD) in an experimental mouse model. (Miyagawa et al., 2008 J. Immunol. 181:1109-19.) One embodiment relates to the use of therapeutic antagonist peptides that selectively inhibit IL-2, IL-7, and IL-15 activity for the treatment of GvHD in humans, allowing survival of the grafted tissues or bone marrow cells. Other embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit a combination of IL-2 and IL-7, IL-2 and IL-15, or IL-7 and IL-15 to treat GvHD. Other embodiments relate to the use of a combination of therapeutic antagonist peptides that selectively inhibit IL-2, IL-7, or IL-15.

[0187] Some embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-2 function for the treatment of autoimmune disorders where T-regs have been implicated as playing a role. In some embodiments, peptide-mediated inhibition of T-regs can enhance the natural anti-cancer immunity in humans, providing a novel means of anti-cancer therapy.

[0188] Several embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-4 to treat asthma.

[0189] Some embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-7 either alone or in combination with therapeutic antagonist peptides that selectively inhibit the yc-cytokine family member, IL-15, as a therapeutic agent for LGL leukemia. In some embodiments therapeutic antagonist peptides that selectively inhibit both IL-7 and IL-15 activity can be used to treat LGL leukemia. Several embodiments relate to the use of BNZ-y to treat LGL leukemia. In some embodiments, specific yc-antagonist peptides that selectively inhibit IL-15 alone or specific yc-antagonist peptides that selectively inhibit IL-15 and IL-7 are used as a therapeutic agent for CD4/CD8 T lymphocyte- associated leukemia including that caused by the HTLV-I.

[0190] Several embodiments relate to the use of yc-antagonist peptides that selectively inhibit the activity of IL-9, either alone or in combination with the other yc-cytokine family members, as a therapeutic agent for human diseases that involve the abnormal development of Thl7 cells.

[0191] Several embodiments relate to the use of therapeutic antagonist peptides that selectively inhibit IL-15 activity as a therapeutic agent for treating CD. One publication suggested that IL-21, in addition to IL-15, may play a role in CD pathogenesis. (See Bodd et al., 2010, Mucosal Immunol. 3:594-601.) Furthermore, a recent study also identified synergistic effects of IL-2, IL-15 and IL-21 contribute greatly to the pathogenesis of refractory CD (Kooy-Winkelaar, et al., 2017 Proc Natl Acad Sci U S A 114: E980-9.). This suggests that optimum treatment of CD by conventional anti-cytokine or cytokine-receptor antibodies would benefit from a combination of at least two antibodies recognizing one or more components that belong to the IL-2 system, IL-15 system, IL-21 system, or a combination thereof. In some embodiments, custom derivative antagonist peptides that selectively inhibit IL-2, IL-15, IL-21, a combination of IL-2 and IL-15, a combination of IL-2 and IL-21, and/or a combination of IL-15 and IL-21 activities are used as a therapeutic agent for treating CD. In some embodiments, the effect of custom derivative antagonist peptides that selectively inhibit a combination of IL-2 and IL-15, a combination of IL-2 and IL-21, and/or a combination of IL-15 and IL-21 can be additive or synergistic.

[0192] An additive effect is observed when the effect of a combination is equal to the sum of the effects of the individuals in the combination (e.g., the effect of a combination of two or more peptides is equal to the sum of the effects of peptides individually). A synergistic effect is observed when the effect of a combination is greater than the sum of the effects of the individuals in the combination (e.g., the effect of a combination of two or more peptides is greater than the sum of the effects of peptides individually). A synergistic effect is greater than an additive effect. Additive effect, synergistic effect, or both can occur in human patients, non-human patients, non-patient human volunteers, in vivo models, ex vivo models, in vitro models, etc.

[0193] In some embodiments, two or more peptides disclosed herein can be used in combination. In some embodiments, two or more peptides disclosed herein when used in combination yield an additive effect. In some embodiments, two or more peptides disclosed herein when used in combination yield a synergistic effect. Synergistic effect can range from about >1 to about 100-fold. In some embodiments, the synergistic effect is about 2 to about 20-fold. In some embodiments, the synergistic effect is about 20 to about 100 fold. In some embodiments, the synergistic effect is from >1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100-fold, or within a range defined by any two of the aforementioned values.

[0194] In addition to having therapeutic applications, yc-antagonist peptides have applications in consumer products as well. Several embodiments relate to the use of yc-antagonist peptides in skin care products such as anti-aging, anti-inflammatory, anti-acne, and other related applications. Some embodiments relate to the use of yc-antagonist peptides in hair products as anti-hair loss ingredient to treat hair loss caused by autoimmune disorders.

[0195] Another embodiment relates to the development of chemical compounds (non-peptide, non-protein) that have a spatial structure which resembles the 19-mer amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), or the 21-mer amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), and can fit into the pocket of the yc-subunit to structurally hinder the access of a yc-cytokine to the yc-subunit for binding. Some embodiments relate to the use of structurally similar chemical compounds as inhibitors of yc-cytokine activity. Such molecular mimicry strategy to further refine the development of synthetic compounds resembling in structure to existing biological peptide/proteins is described in Orzaez et al., 2009 Chem. Med. Chem. 4: 146-160. Another embodiment relates to administration of chemical compounds (non-peptide, non-protein) that have a resembling 3D structure as the 19-mer amino acids sequence I-K-E-F-L-Q-R-F-I-H-I- V-Q-S-I-I-N-T-S (SEQ ID NO: 1), or the 21-mer amino acid sequence P-K-E-F-L-E-R-F-V- H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), to treat yc-cytokine-mediated diseases.

[0196] Several embodiments relate to the administration of a peptide of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) to treat yc-cytokine- mediated diseases. Another embodiment relates to the administration of peptide derivatives of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), wherein the amino acid sequence of the derivative peptide has similar physico-chemical properties as a peptide of the amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), but has distinct biological activity, to treat yc-cytokine-mediated diseases. Another embodiment relates to administration of a peptide of amino acid sequence I-K-E-F-L-Q-R-F- I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) conjugated to the N- and C-termini or to the side residues of existing biological proteins/peptides into patients to treat yc-cytokine-mediated diseases. Another embodiment relates to administration of a peptide of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1) that comprises one or more intra- peptide hydrocarbon linker elements to treat yc-cytokine-mediated diseases.

[0197] Several embodiments relate to the administration of a peptide of amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3) to treat yc-cytokine-mediated diseases. Another embodiment relates to the administration of peptide derivatives of amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), wherein the amino acid sequence of the derivative peptide has similar physico- chemical properties as a peptide of the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q- M-F-I-H-Q-S-L-S (SEQ ID NO: 3), but has distinct biological activity, to treat yc-cytokine- mediated diseases. Another embodiment relates to administration of a peptide of amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3) conjugated to the N- and C-termini or to the side residues of existing biological proteins/peptides into patients to treat yc-cytokine-mediated diseases. Another embodiment relates to administration of a peptide of amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H- Q-S-L-S (SEQ ID NO: 3) that comprises one or more intra-peptide hydrocarbon linker elements to treat yc-cytokine-mediated diseases.

[0198] Several embodiments relate to administration of polyclonal and monoclonal antibodies raised against a peptide comprising of amino acid sequence I-K-E-F- L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), or raised against a peptide comprising of amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), into patients as an immunogen to treat yc-cytokine-mediated diseases. Another embodiment relates to administration of polyclonal and monoclonal antibodies that were raised against derivative peptides of amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), or raised against derivative peptides of amino acid sequence P-K-E-F-L-E-R-F-V- H-L-V-Q-M-F-I-H-Q-S-L-S (SEQ ID NO: 3), wherein the amino acid sequence of the derivative peptide has similar physico-chemical properties as a peptide of the amino acid sequence I-K-E-F-L-Q-R-F-I-H-I-V-Q-S-I-I-N-T-S (SEQ ID NO: 1), or has similar physico- chemical properties as a peptide of the amino acid sequence P-K-E-F-L-E-R-F-V-H-L-V-Q- M-F-I-H-Q-S-L-S (SEQ ID NO: 3), but has distinct biological activity, into patients as an immunogen to treat yc-cytokine-mediated diseases. Administration of yc-antagonist peptides

[0199] The present embodiments also encompass the use of yc-antagonist peptides for the manufacture of a medicament for the treatment of a disease. The present embodiments also encompass a pharmaceutical composition that includes yc-antagonist peptides in combination with a pharmaceutically acceptable carrier. The pharmaceutical composition can include a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of yc-antagonist peptides, or other compositions of the present embodiments.

[0200] The present embodiments provide methods of using pharmaceutical compositions comprising an effective amount of antagonists for yc-cytokines in a suitable diluent or carrier. A yc-antagonist of the present embodiments can be formulated according to known methods used to prepare pharmaceutically useful compositions. A yc-antagonist can be combined in admixture, either as the sole active material or with other known active materials, with pharmaceutically- suitable diluents (e.g., phosphate, acetate, Tris-HCl), preservatives (e.g., thimerosal, benzyl alcohol, parabens), emulsifying compounds, solubilizers, adjuvants, and/or carriers such as bovine serum albumin.

[0201] In some embodiments, one or more compositions and kits comprising one or more of the composite peptides or derivatives thereof disclosed herein are contemplated. In some embodiments, one or more compositions and kits are used for preventing and/or treating one or more diseases. In some embodiments, one or more compositions and kits are used for preventing and/or treating a yc cytokine-mediated disease. In some embodiments, one or more compositions and kits are used for preventing and/or treating an HTLV-1- associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease. In some embodiments, one or more compositions and kits are used for preventing and/or treating an inflammatory respiratory disease. In some embodiments, one or more compositions and kits are used for preventing and/or treating a cosmetic condition.

[0202] Some embodiments, the one or more compositions and kits comprising one or more of the composite peptides are administered to a subject in need thereof via any of the routes of administration provided herein. In some embodiments, the one or more compositions and kits comprises one or more of the composite peptides or derivatives thereof at a therapeutically effective amount to modulate the activity of one or more yc-cytokines selected from the group consisting of IL 2, IL 4, IL 7, IL 9, IL 15, and IL 21. In some embodiments, the one or more compositions and kits comprises one or more of the composite peptides or derivatives thereof at a therapeutically effective amount to prevent and/or treat one or more diseases. In some embodiments, the one or more compositions and kits comprising one or more of the composite peptides additionally comprise one or more pharmaceutically acceptable carriers, diluents, excipients or combinations thereof.

[0203] In some embodiments, one or more composite peptides in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating one or more diseases. In some embodiments, one or more composite peptides in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a yc cytokine-mediated disease. In some embodiments, one or more composite peptides in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease. In some embodiments, one or more composite peptides in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an inflammatory respiratory disease. In some embodiments, one or more composite peptides in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a cosmetic condition.

[0204] In some embodiments, one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating one or more diseases. In some embodiments, one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a yc cytokine-mediated disease. In some embodiments, one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an HTLV-l-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease. In some embodiments, one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an inflammatory respiratory disease. In some embodiments, one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a cosmetic condition.

[0205] In some embodiments, one or more derivatives of the one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating one or more diseases. In some embodiments, one or more derivatives of the one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a yc cytokine-mediated disease. In some embodiments, one or more derivatives of the one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an HTLV- 1-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) associated disease. In some embodiments, one or more derivatives of the one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating an inflammatory respiratory disease. In some embodiments, one or more derivatives of the one or more composite peptides selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 in the one or more compositions and kits are formulated as suitable for administration to a subject for preventing and/or treating a cosmetic condition. In some embodiments, the one or more derivatives of the one or more composite peptides comprise amino acid sequences that shares about 50% to about 99% identity with the one or more composite peptides. In some embodiments, the one or more derivatives of the one or more composite peptides comprise amino acid sequences that shares 50%, 50-60%, 60-70%, 70-80%, 80%, 90%, 95%, 97%, 98%, 99% or 99.8% identity with the one or more composite peptides, or within a range defined by any two of the aforementioned values.

[0206] In some embodiments, one or more yc-cytokine-mediated disease is selected from the group consisting of CD4-leukemia, CD8-leukemia, LGL-leukemia, systemic lupus erythematosis, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, rheumatoid arthritis, diabetes mellitus, psoriasis, multiple sclerosis, uvietis, inflammation of the eye, and graft-versus-host disease (GvHD). In some embodiments, one or more HAM/TSP associated disease is selected from the group consisting of Adult T-cell Leukemia (ATL), HTLV-associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), and other non-neeoplastic inflammatory diseases associated with HTLV such as uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, and myositis. In some embodiments, one or more inflammatory respiratory disease is selected from the group consisting of asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, and lung fibrosis). In some embodiments, one or more cosmetic disease is selected from the group consisting of acne, hair loss, sunburn, nail maintenance, and appearance of aging.

[0207] Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16 th ed. 1980 Mack Publishing CO. Additionally, such compositions can contain a yc-antagonist complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts, or spheroblasts. Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance of a yc-antagonist. A yc-antagonist can be conjugated to antibodies against cell-specific antigens, receptors, ligands, or coupled to ligands for tissue-specific receptors.

[0208] Methods of administrating yc- antagonists of the present embodiments may be selected as appropriate, depending on factors, such as the type of diseases, the condition of subjects, and/or the site to be targeted. The yc- antagonists can be administered topically, orally, parenterally, rectally, or by inhalation. The term "parenteral" includes subcutaneous injections, intravenous, intramuscular, intraperitoneal, intracisternal injection, or infusion techniques. These compositions will typically include an effective amount of a yc-antagonist, alone or in combination with an effective amount of any other active material. Several non- limiting routes of administrations are possible including parenteral, subcutaneous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, intralesional, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal.

[0209] The one or more composite peptides disclosed herein can be administered at any dose, via any of the routes of administration, and at any frequency of administration as determined by one of ordinary skill in the art based on various parameters non-limiting examples of which include the condition being treated, the severity of the condition, patient compliance, efficacy of treatment, side effects, etc.

[0210] The amount of the peptide contained in pharmaceutical compositions of the present embodiments, dosage form of the pharmaceutical compositions, frequency of administration, and the like may be selected as appropriate, depending on factors, such as the type of diseases, the condition of subjects, and/or the site to be targeted. Such dosages and desired drug concentrations contained in the compositions may vary affected by many parameters, including the intended use, patient's body weight and age, and the route of administration. Pilot studies will first be conducted using animal studies and the scaling to human administration will be performed according to art- accepted practice.

[0211] In one embodiment, host cells that have been genetically modified with a polynucleotide encoding at least one yc-antagonist peptide are administered to a subject to treat a proliferation disorder and/or to reduce the growth of malignant cells. The polynucleotide is expressed by the host cells, thereby producing the peptides within the subject. Preferably, the host cells are allogeneic or autogeneic to the subject.

[0212] In a further aspect, yc-antagonist peptides can be used in combination with other therapies, for example, therapies inhibiting cancer cell proliferation and growth. The phrase "combination therapy" embraces the administration of yc-antagonist peptides and an additional therapeutic agent as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of these therapeutic agents. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually minutes, hours, days or weeks depending upon the combination selected).

[0213] A combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by an appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. There therapeutic agents can be administered by the same route or by different routes. The sequence in which the therapeutic agents are administered is not narrowly critical.

[0214] Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients (such as, but not limited to, a second and different therapeutic agent) and non- drug therapies (such as, but not limited to, surgery or radiation treatment). Where the combination therapy further comprises radiation treatment, the radiation treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and radiation treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the radiation treatment is temporarily removed from the administration of the therapeutic agents, perhaps by days or even weeks.

[0215] In certain embodiments, yc-antagonist peptides can be administered in combination with at least one anti-proliferative agent selected from the group consisting of chemotherapeutic agent, an antimetabolite, and antitumorgenic agent, and antimitotic agent, and antiviral agent, and antineoplastic agent, an immunotherapeutic agent, and a radiotherapeutic agent.

[0216] In certain embodiments, yc-antagonist peptides can be administered in combination with at least one anti-inflammatory agent selected from the group consisting of steroids, corticosteroids, and nonsteroidal anti-inflammatory drugs.

[0217] Also provided are kits for performing any of the methods provided herein. In some embodiments, kits may include one or more yc-antagonist according to any of the embodiments provided herein. In some embodiments, the kit may include instructions. Instructions may be in written or pictograph form, or may be on recorded media including audio tape, audio CD, video tape, DVD, CD-ROM, or the like. The kits may comprise packaging. Definitions

[0218] As used herein, the term "patient" or "subject" refers to the recipient of a any of the embodiments of the composite peptides disclosed herein and includes all organisms within the kingdom animalia. In some embodiments, any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, etc.) are included. In preferred embodiments, the animal is within the family of mammals, such as humans, bovine, ovine, porcine, feline, buffalo, canine, goat, equine, donkey, deer, and primates. The most preferred animal is human. In some embodiments, the patient is a male or a female.

[0219] As used herein, the term "treat" or any variation thereof (e.g., , treatment, treating, etc.), refers to any treatment of a patient diagnosed with a biological condition, such as CD4-, CD8-, and LGL-leukemia, an autoimmune disease, systemic lupus erythematosis, Sjogren's syndrome, Wegener's granulomatosis, Celiac disease, Hashimoto's thyroiditis, a collagen disease, rheumatoid arthritis, inflammatory bowel disease, diabetes mellitus, psoriasis, a degenerative neuronal disease, multiple sclerosis, uvietis, inflammation of the eye, graft-versus-host disease (GvHD), myasthenia gravis, Human T-cell Lymphotropic type I and II (HTLV-I and HTLV-II)-associated diseases, Adult T-cell Leukemia (ATL), HTLV- associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), uveitis (HU), arthropathy, pneumopathy, dermatitis, exocrinopathy, myositis, influenza, AIDS, HBV, Herpes, asthma, sinusitis, hay fever, bronchitis, chronic obstructive pulmonary disease (COPD), allergic rhinitis, acute and chronic otitis, lung fibrosis, NK leukemia/lymphoma and NK-T leukemia/lymphoma.

[0220] The term treat, as used herein, includes: (i) preventing or delaying the presentation of symptoms associated with the biological condition of interest in an at-risk patient who has yet to display symptoms associated with the biological condition; (ii) ameliorating the symptoms associated with the biological condition of interest in a patient diagnosed with the biological condition; (iii) preventing, delaying, or ameliorating the presentation of symptoms associated with complications, conditions, or diseases associated with the biological condition of interest in either an at-risk patient or a patient diagnosed with the biological condition; (iv) slowing, delaying or halting the progression of the biological condition; and/or (v) preventing, delaying, slowing, halting or ameliorating the cellular events of inflammation; and/or (vi) preventing, delaying, slowing, halting or ameliorating the histological abnormalities and/or other clinical measurements of the biological condition.

[0221] The term "symptom(s)" as used herein, refers to common signs or indications that a patient is suffering from a specific condition or disease.

[0222] The term "effective amount," as used herein, refers to the amount necessary to elicit the desired biological response. In accordance with the present embodiments, an effective amount of a yc-antagonist is the amount necessary to provide an observable effect in at least one biological factor for use in treating a biological condition.

[0223] "Recombinant DNA technology" or "recombinant" refers to the use of techniques and processes for producing specific polypeptides from microbial (e.g., bacterial, yeast), invertebrate (insect), mammalian cells or organisms (e.g., transgenic animals or plants) that have been transformed or transfected with cloned or synthetic DNA sequences to enable biosynthesis of heterologous peptides. Native glycosylation pattern will only be achieved with mammalian cell expression system. Prokaryotic expression systems lack the ability to add glycosylation to the synthesized proteins. Yeast and insect cells provide a unique glycosylation pattern that may be different from the native pattern.

[0224] A "nucleotide sequence" refers to a polynucleotide in the form of a separate fragment or as a component of a larger DNA construct that has been derived from DNA or RNA isolated at least once in substantially pure form, free of contaminating endogenous materials and in a quantity or concentration enabling identification, manipulation, and recovery of its component nucleotide sequences by standard molecular biology methods (as outlined in Current Protocols in Molecular Biology).

[0225] "Recombinant expression vector" refers to a plasmid comprising a transcriptional unit containing an assembly of (1) a genetic element or elements that have a regulatory role in gene expression including promoters and enhances, (2) a structure or coding sequence that encodes the polypeptide according to the present embodiments, and (3) appropriate transcription and translation initiation sequence and, if desired, termination sequences. Structural elements intended for use in yeast and mammalian system preferably include a signal sequence enabling extracellular secretion of translated polypeptides by yeast or mammalian host cells.

[0226] "Recombinant microbial expression system" refers to a substantially homogenous monoculture of suitable microorganisms, for example, bacteria such as E. coli, or yeast such as S. cerevisiae, that have stably integrated a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit as a component of a residual plasmid. Generally, host cells constituting a recombinant microbial expression system are the progeny of a single ancestral transformed cell. Recombinant microbial expression systems will express heterologous polypeptides upon induction of the regulatory elements linked to a structural nucleotide sequence to be expressed.

[0227] As used herein, the section headings are for organizational purposes only and are not to be construed as limiting the described subject matter in any way. All literature and similar materials cited in this application, including but not limited to, patents, patent applications, articles, books, treatises, and internet web pages are expressly incorporated by reference in their entirety for any purpose. When definitions of terms in incorporated references appear to differ from the definitions provided in the present teachings, the definition provided in the present teachings shall control. It will be appreciated that there is an implied "about" prior to the temperatures, concentrations, times, etc discussed in the present teachings, such that slight and insubstantial deviations are within the scope of the present teachings herein.

[0228] Although this invention has been disclosed in the context of certain embodiments and examples, those skilled in the art will understand that the present invention extends beyond the specifically disclosed embodiments to other alternative embodiments and/or uses of the invention and obvious modifications and equivalents thereof. In addition, while several variations of the invention have been shown and described in detail, other modifications, which are within the scope of this invention, will be readily apparent to those of skill in the art based upon this disclosure.

[0229] It is also contemplated that various combinations or sub-combinations of the specific features and aspects of the embodiments may be made and still fall within the scope of the invention. It should be understood that various features and aspects of the disclosed embodiments can be combined with, or substituted for, one another in order to form varying modes or embodiments of the disclosed invention. Thus, it is intended that the scope of the present invention herein disclosed should not be limited by the particular disclosed embodiments described above.

[0230] It should be understood, however, that this detailed description, while indicating preferred embodiments of the invention, is given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art.

EXAMPLES

[0231] The following Examples are presented for the purposes of illustration and should not be construed as limitations.

EXAMPLE 1 - Method for Assessing the Inhibitory Activity of yc-Antagonist Peptide

[0232] The capacity of any custom derivative peptide prepared according to the present embodiments for inhibiting the action of one yc-cytokine family member is determined using mammalian cellular assays to measure their proliferative response to the yc-cytokine family member.

[0233] For each of the six yc-cytokines, indicator cell lines: NK92, a human NK cell line NK92 available by American Type Culture Collection (ATCC) (catalog # CRL- 2407), CTLL-2, a murine CD8 T cells line available from ATCC, and PT-18, a murine mast cell line and its subclone ΡΤ-18β, is transfected with human IL-2R gene to make the cells responsive to IL-2 and IL-15 (Tagaya et al., 1996, EMBO J. 15:4928-39), and is used to quantitatively determine the yc-cytokine's growth-promoting activity (See Current protocols in Immunology from Wiley and Sons for a methodological reference). The indicator cells demonstrate semi-linear dose-dependent response when measured by a colorimetric WST-1 assay over a range of concentrations (See Clontech PT3946-1 and associated user manual, incorporated herein by reference, for a detailed description of the reagents and methods).

[0234] Once the appropriate doses of the cytokine that yield the 50-70% and 95% maximum response from the indicator cell line is determined, various concentrations (ranging from 1 pM to 10 μΜ) of the purified or synthesized custom derivative peptide is added to each well containing the cytokine and indicator cells. The reduction in light absorbance at 450nm is used as an indicator of inhibition of cytokine- stimulated cellular proliferation. EXAMPLE 2 - The Selective Inhibition of the Growth-Promoting Activities of Certain Yc-Cvtokines by ΒΝΖ-γ and SEP ID NO: 3

[0235] Using ΡΤ-18β cells as described above, the ability of the ΒΝΖ-γ peptide to specifically inhibit the growth-promoting activity of select yc-cytokines was determined (FIG. 3A). IL-3, a non-yc-cytokine that supports the growth of ΡΤ-18β cells, was used as a negative control. Briefly, ΡΤ-18β cells were incubated either with two different dilutions of ΒΝΖ-γ peptide produced by HEK293T cells (1 :20 or 1 :50 dilution of the original supernatant of HEK293T cells transfected with a ΒΝΖ-γ expression construct) or without ΒΝΖ-γ peptide in the presence of IL-3, IL-9, IL-15, or IL-4 (1 nM of each cytokine in the culture).

[0236] The growth-responses of the cells were determined 2 days after the introduction of ΒΝΖ-γ peptide and the cytokine using the WST-1 assay. The growth- promoting activity of IL-3 (a non yc-cytokine) was not inhibited by ΒΝΖ-γ. In contrast, the activity of IL-15 and IL-9 were significantly (p<0.01 Student's T test) reduced by the ΒΝΖ-γ peptide. Cellular proliferation stimulated by IL-4, another yc-cytokine, was not affected by the by the addition of ΒΝΖ-γ peptide. Results for IL-3, IL-9, IL-15, and IL-4 are shown at FIG. 3A.

[0237] In a similar assay, the murine cell line CTTL2 was used. In this assay the cells were cultured with 0.5 nM of recombinant IL-2 in RPMI 10% fetal Calf Serum. To set up the proliferation assay, cells were washed from the cytokines 3 times. Cells were seeded at 1 x 10(5) cells per well of a 96-well plate with final concentration of 50 pM of IL-2 or IL-15. Various concentration of ΒΝΖ-γ peptide (0.1, 1, and 10 ug/ml) was added to each well. Cells were cultured for 20 hours and in the last 4 hours, 3 H-thymidine was added to the plates. Cells were harvested using a plate reader. The data are shown in FIG. 3B.

[0238] The human NK cell line NK92 was used for a dose response of SEQ ID NO: 3 against IL-2, IL-15, and IL-21. Cells were cultured with 0.5 nM of recombinant IL-2 in RPMI 10% fetal Calf Serum. To set up the proliferation assay, cells were washed from the cytokines 3 times, and subsequently seeded at 2.5 x 10(4) cells per well of a 96-well plate with cytokine concentrations that correspond to 70% NK92 cell proliferation levels. Commercially purchased SEQ ID NO: 3 (at >99% purity) was added to each well at a final concentration of 0-10 μΜ for IL-2, at 0-0.1 μΜ for IL-15, and at 0-0.1 μΜ for IL-21. The growth-responses of the cells were determined 2 days after the introduction of SEQ ID NO: 3 and the cytokine using the WST-1 assay. Cellular proliferation stimulated by the yc-cytokine IL-2 was not affected by the by the addition of SEQ ID NO: 3 up to 10 μΜ. In contrast, the activity of IL-15 and IL-21 were potently inhibited by SEQ ID NO: 3. The data are shown in FIG. 3D.

EXAMPLE 3 - Method for Measuring Inhibition yc-Cytokine Activity by Assaying 3H- thymidine Incorporation of as a Marker of Cellular Proliferation

[0239] Inhibition of yc-cytokine-induced proliferation of an indicator cell population by antagonist custom derivative peptides is measured by the 3H-thymidine incorporation assay. Briefly, radiolabeled thymidine (1 microCi) is given to 20-50,000 cells undergoing proliferation in the presence of cytokines. The cell-incorporated radioactivity is measured by trapping cell-bound radioactivity to a glass-fiber filter using a conventional harvester machines (for example, Filtermate Universal Harvester from Perkin-Elmer), after which the radioactivity is measured using a b-counter (for example, 1450 Trilux microplate scintillation counter).

EXAMPLE 4 - Method for Measuring Inhibition yc-Cytokine Activity by Assaying Incorporation of a Cell-Tracker Dye as a Marker of Cellular Proliferation

[0240] Indicator cells are incubated in the presence of a selected yc-cytokine or in the presence of a selected yc-cytokine and a selected custom derivative peptide. The cell population is then labeled in vitro using a cell-tracker dye, for example, CMFDA, C2925 from Invitrogen, and the decay of cellular green fluorescence at each cellular division is monitored using a flow-cytometer (for example, Beckton-Dickinson FACScalibur). Typically, in response to yc-cytokine stimulation 7-10 different peaks corresponding to the number of divisions that the cells have undergone will appear on the green fluorescence channel. Incubation of the cells with the selected yc-cytokine and antagonist custom derivative peptide reduces the number of peaks to only 1 to 3, depending on the degree of the inhibition. EXAMPLE 5 - Inhibition of Intracellular Signaling by Custom Peptide Derivative Antagonists

[0241] In addition to stimulating cellular proliferation, binding of the yc-cytokines to their receptors causes a diverse array of intracellular events. (Rochman et al. 2009 Nat. Rev. Immunol. 9:480-90, Pesu et al. 2005 Immunol. Rev. 203: 127-142.) Immediately after the cytokine binds to its receptor, a tyrosine kinase called Jak3 (Janus-kinase 3) is recruited to the receptor at the plasma membrane. This kinase phosphorylates the tyrosine residues of multiple proteins including the yc-subunit, STAT5 (Signal Transducer and Activator of Transcription 5) and subunits of the PI3 (Phosphatidylinositol 3) kinase. Among these, the phosphorylation of STAT5 has been implicated in many studies as being linked to the proliferation of cells initiated by the yc-cytokine. (Reviewed in Hennighausen and Robinson, 2008 Genes Dev. 22:711-21.) In accordance with these published data, whether or not the ΒΝΖ-γ peptide inhibits the tyrosine phosphorylation of STAT5 molecule in ΡΤ-18β cells stimulated by IL-15 was examined (results shown in FIG. 4A).

[0242] ΡΤ-18β cells were stimulated by IL-15 in the presence or absence of ΒΝΖ-γ peptide. Cytoplasmic proteins were extracted from the cells according to a conventional method as described in Tagaya et al. 1996 EMBO J. 15:4928-39. The extracted cytoplasmic proteins were resolved using a standard SDS-PAGE (Sodium Dodecyl-Sulfate PolyAcrylamide Gel Electrophoresis) and the phorphorylation status was confirmed by an anti-phospho-STAT5 antibody (Cell Signaling Technology, Catalog # 9354, Danvers MA) using immunoblotting (See FIG. 4A, top panel). To confirm that each lane represented a similar total protein load, the membrane was then stripped, and re-probed with an anti- STAT5 antibody (Cell Signaling Technology, Catalog # 9358) (See FIG. 4A, bottom panel).

[0243] These results demonstrated that tyrosine phosphorylation of STAT5, a marker of signal transduction, was induced by IL-15 in ΡΤ-18β cells, and tyrosine phosphorylation of STAT5 was markedly reduced by the ΒΝΖ-γ peptide.

[0244] Similar to the IL-15 induced tyrosine phosphorylation of STAT5 via Jak3, it is well known in the art that Jak-STAT signaling mediated via IL-21 receptor binding preferentially induces Jak-mediated tyrosine phosphorylation of STAT3 (Habib et al. 2003 J Allergy Clin Immunol. 112: 1033-45). In accordance with these published data, the ability of SEQ ID NO: 3 to inhibit the tyrosine phosphorylation of the STAT5 molecule by IL-2 or IL- 15 in CTLL-2 cells, and the ability of SEQ ID NO: 3 to inhibit the tyrosine phosphorylation of the STAT3 molecule by IL-21 in NK92 cells was examined (results shown in FIG. 4B).

[0245] CTLL-2 cells were stimulated by IL-2 or IL-15 in the presence or absence of SEQ ID NO: 3. CTLL-2 cells were also stimulated by IL-2 or IL-15 in the presence of anti-IL-2 antibody (R & D Systems, Catalog # MAB202, Minneapolis, MN) or anti-IL-15 antibody (R & D Systems, Catalog # MAB247, Minneapolis, MN) as positive controls. NK92 cells were stimulated by IL-21 in the presence or absence of SEQ ID NO: 3. NK92 cells were also stimulated by IL-21 in the presence of anti-IL-21 antibody (Mabtech, Catalog # 3540-1-250, Cincinnati, OH) as a positive control. Cytoplasmic proteins were extracted from the cells according to a conventional method as described in Tagaya et al. 1996 EMBO J. 15:4928-39. The extracted cytoplasmic proteins were resolved using a standard SDS-PAGE (Sodium Dodecyl-Sulfate PolyAcrylamide Gel Electrophoresis) and the phosphorylation status was confirmed by an anti-phospho-STAT5 antibody (Cell Signaling Technology, Catalog # 9354, Danvers MA) or an anti-phospho-STAT3 antibody (Cell Signaling Technology, Catalog # 9145, Danvers MA) using immunoblotting (See FIG. 4B, top panel). To confirm that each lane represented a similar total protein load, the membrane was then stripped, and re-probed with an anti-STAT5 antibody (Cell Signaling Technology, Catalog # 9358) or an anti-STAT3 antibody (Cell Signaling Technology, Catalog # 9139) (See FIG. 4B, bottom panel).

EXAMPLE 6 - Rational Design for ΒΝΖ-γ Derivative Antagonistic Peptides

[0246] Derivative peptides are prepared based from the core sequence D/E-F-L- E/Q/N-S/R-X-I/K-X-L/I-X-Q (SEQ ID NO: 2) (where X denotes any amino acid) by substituting the defined amino acids of the core sequence with amino acids having identical physico-chemical properties as designated in FIG. 2.

[0247] Alternatively, custom peptides or their derivative peptides can be prepared based on the sequence alignment of the D-helix regions of different yc-cytokine family members. For example, as shown in FIG. 6, one or more sequences conserved in yc-cytokine family (SEQ ID NO: 4 - SEQ ID NO: 9) members can be combined to form a peptide such as SEQ ID NO: 3. EXAMPLE 7 - Method of Identifying the Inhibitory Specificity of Antagonistic Custom Derivative Peptides

[0248] The yc-cytokine inhibitory specificity of antagonistic custom derivative peptides is determined by assaying the ability of a custom derivative peptide to inhibit the proliferative response of a cytokine-responsive cell line to each of the 6 yc-cytokines. For example, a mouse cell line, CTLL-2, is used to determine if a candidate peptide inhibits the function of IL-2 and IL-15. ΡΤ-18(β) cells are used to determine if a candidate peptide inhibits the function of IL-4 and IL-9. PT-18 (7a) cells are used to determine if a candidate peptide inhibits the function of IL-7, and PT- 18(2 la) cells are used to determine if a candidate peptide inhibits the function of IL-21. ΡΤ-18(β) denotes a subclone of PT-18 cells that exogenously express human IL-2R by gene transfection (See Tagaya et al. 1996), PT-18(7a) denotes a subclone that expresses human IL-7Ra by gene transfection and PT-18(21Ra) cells express human IL-21Ra.

[0249] Another alternative is to use other cell lines that respond to an array of cytokines. An example of this cell line in a human NK cell line NK92 that is commercially available by ATCC (catalog # CRL-2407). This cell line is an IL-2 dependent cell line that responds to other cytokines including IL-9, IL-7, IL-15, IL-12, IL-18, IL-21 (Gong et al. 1994 Leukemia 8: 652-658,, Kingemann et al., 1996, Biol Blood Marrow Transplant 2:68;75, Hodge DL et al., 2002 J. Immunol. 168:9090-8).

[0250] The human NK cell line NK92 was used to determine the yc-cytokine inhibitory specificity of SEQ ID NO: 3. In this assay the cells were cultured with 0.5 nM of recombinant IL-2 in RPMI 10% fetal Calf Serum. To set up the inhibitory specificity assay, cells were washed from the cytokines 3 times. Cells were seeded at 2.5 x 10(4) cells per well of a 96-well plate with cytokine concentrations that correspond to 70% NK92 cell proliferation levels. Commercially purchased SEQ ID NO: 3 (at 70% purity levels) was added to each well at a final concentration of 2.5 μΜ. The growth-responses of the cells were determined 2 days after the introduction of SEQ ID NO: 3 and the cytokine using the WST-1 assay. Cellular proliferation stimulated by the yc-cytokines IL-2, IL-4, and IL-9 was not affected by the by the addition of SEQ ID NO: 3. In contrast, the activity of IL-15 and IL-21 were significantly reduced by SEQ ID NO: 3. Results for IL-2, IL-4, IL-9, IL-15, and IL-21 are shown at FIG. 3C.

EXAMPLE 8 - Enhanced Inhibitory Activity of Custom Peptide Derivatives Containing Intra- Peptide Hydrocarbon Linker Element

[0251] The human NK cell line NK92 was used to determine the enhanced inhibitory activity of custom peptide derivatives containing intra-peptide hydrocarbon linker element. Cells were cultured with 0.5 nM of recombinant IL-2 in RPMI 10% fetal Calf Serum. To set up the proliferation assay, cells were washed from the cytokines 3 times. Cells were seeded at 2.5 x 10(4) cells per well of a 96-well plate with a final IL-15 concentration of 0.25 ng/mL. Commercially purchased unmodified SEQ ID NO: 3 and custom peptide derivatives of SEQ ID NO: 3 containing an intra-peptide hydrocarbon linker element were added to each well at equal concentrations. The growth-responses of the cells were determined 2 days after the introduction of the peptides and the cytokine using the WST-1 assay. Multiple custom peptide derivatives of SEQ ID NO: 3 containing an intra-peptide hydrocarbon linker element showed enhanced inhibitory activity as compared to the unmodified SEQ ID NO: 3. The data are shown in FIG. 7.

EXAMPLE 9 - Time-Course Protease Stability Measurement of Custom Peptide Derivatives Containing Intra-Peptide Hydrocarbon Linker Element in Simulated Intestinal Fluid

[0252] To evaluate the gastric stability of the custom peptide derivatives containing one or more intra-peptide hydrocarbon linker elements of the present embodiments a time-course protease stability measurement of unmodified SEQ ID NO: 3 in comparison with a representative custom peptide derivative of SEQ ID NO: 3 containing one hydrocarbon linker element (SEQ ID NO: 39), and another representative custom peptide derivative of SEQ ID NO: 3 containing two hydrocarbon linker elements and certain amino acid positions in the (D) stereochemical configuration (SEQ ID NO: 57) was carried out in simulated intestinal fluid over 60 minutes. All peptides were commercially synthesized and purchased. Simulated intestinal fluid was made according to USP specifications (Test Solutions, United States Pharmacopeia 36). 50 of 10 mg/mL peptide was mixed with 450 pre-warmed (37°C) simulated intestinal fluid, and aliquots of the reaction mixture were removed at prescribed time intervals over a time-period of 60 minutes and stopped with 0.1 M HC1. Peptide stability was measured by reversed phase HPLC on a Phenomenex Aeris Peptide column (4.6 x 250 mm) with settings: 3.6 μιη particle size, non-porous; 5.0 μL· injection, 0.5mL/min; mobile phase A: 25% Acetonitrile with 0.1% TFA; mobile phase B: 100% Acetonitrile with 0.1% TFA. The multi-step buffer gradient goes from 100% A to 100% B over 26 minutes. Time point 0 minutes was set to 100% undigested peptide, and data at later time-points were graphed and connected via a smooth curve out to the end-point of 60 minutes (FIG. 8 A). Results show the percentage of undigested peptide present in the simulated intestinal fluid is greatly enhanced with one or more hydrocarbon linker elements and certain amino acid positions in the (D) stereochemical configuration, compared to the unmodified peptide. The data are shown in FIG. 8A.

EXAMPLE 10 - Preparation of jc- Antagonist Peptides

[0253] Custom derivative yc-antagonist peptides are synthesized chemically by manual and automated processes.

[0254] Manual synthesis: Classical liquid-phase synthesis is employed, which involves coupling the carboxyl group or C-terminus of one amino acid to the amino group or N-terminus of another. Alternatively, solid-phase peptide synthesis (SPPS) is utilized.

[0255] Automated synthesis: Many commercial companies provide automated peptide synthesis for a cost. These companies use various commercial peptide synthesizers, including synthesizers provided by Applied Biosystems (ABI). Custom derivative yc-antagonist peptides are synthesized by automated peptide synthesizers.

EXAMPLE 11 - Biological Production of Custom Derivative jc- Antagonist Peptides Using Recombinant Technology

[0256] A custom derivative yc-antagonist peptide is synthesized biologically as a pro-peptide that consists of an appropriate tagging peptide, a signal peptide, or a peptide derived from a known human protein that enhances or stabilizes the structure of the ΒΝΖ-γ peptide or a peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof, and improves their biological activities. If desired, an appropriate enzyme-cleavage sequence proceeding to the N-terminus of the peptide shall be designed to remove the tag or any part of the peptide from the final protein.

[0257] A nucleotide sequence encoding the custom derivative peptide with a stop codon at the 3' end is inserted into a commercial vector with a tag portion derived from thioredoxin of E. coli and a special peptide sequence that is recognized and digested by an appropriate proteolytic enzyme (for example, enterokinase) intervening between the tag portion and the nucleotide sequence encoding the custom derivative peptide and stop codon. One example of a suitable vector is the pThioHis plasmid available from Invitrogen, CA. Other expression vectors may be used.

EXAMPLE 12 - Conjugation of Custom Peptides and Derivative to Carrier Proteins for Immunization

Purposes and Generation of Antibody against the Custom Peptides

[0258] ΒΝΖ-γ and other custom derivative peptides, such as a peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof are used to immunize animals to obtain polyclonal and monoclonal antibodies. Peptides are conjugated to the N- or the C-terminus of appropriate carrier proteins (for example, bovine serum albumin, Keyhole Limpet Hemocyanin (KLH), etc.) by conventional methods using Glutaraldehyde or m-Maleimidobenzoyl-N- Hydroxysuccinimide Ester. The conjugated peptides in conjunction with an appropriate adjuvant are then used to immunize animals such as rabbits, rodents, or donkeys. The resultant antibodies are examined for specificity using conventional methods. If the resultant antibodies react with the immunogenic peptide, they are then tested for the ability to inhibit individual yc-cytokine activity according to the cellular proliferation assays described in Examples 1-3. Due to the composite nature of the derivative peptides it is possible to generate a single antibody that recognizes two different cytokines simultaneously, because of the composite nature of these peptides.

EXAMPLE 13 - Method for Large Scale Production of Custom Derivative jc- Antagonist Peptides

[0259] Recombinant proteins are produced in large scale by the use of cell-free system as described elsewhere. (See Takai et al., 2010 Curr. Pharm. Biotechnol. l l(3):272-8.) Briefly, cDNAs encoding the yc-antagonist peptide and a tag are subcloned into an appropriate vector (See Takai et al., 2010 Curr. Pharm. Biotechnol. l l(3):272-8), which is subjected to in vitro transcription, followed immediately by an in vitro translation to produce the tagged peptide. The pro-polypeptide is then purified using an immobilized antibody recognizing the tagged epitope, treated by the proteolytic enzyme and the eluate (which mostly contains the custom derivative peptide of interest) is tested for purity using conventional 18% Tricine-SDS-PAGE (Invitrogen) and conventional comassie staining. Should the desired purity of the peptide not be met (>98%), the mixture is subjected to conventional HPLC (high-performance liquid chromatography) for further purification.

EXAMPLE 14 - Use of Custom Derivative yc-Antagonist Peptides to Block Cytokine Function in HAM/TSP

[0260] HTLV- 1-associated myelopathy (HAM)/ tropical spastic paraparesis (TSP) is a chronic progressive myelopathy seen in some people infected with Human T- Lymphotropic Virus Type I (HTLV- 1). Infiltration of lymphocytes in the spinal cord is associated with the immune response to HTLV-I and results in the release of certain cytokines. Some of these cytokines may also damage nerves.

[0261] Patients with HAM/TSP show an elevated state of the immune system that is similar to that observed in autoimmune diseases (Oh et al. 2008 Neurol Clin. 26:781-785). This elevated state is demonstrated by the ability of HAM/TSP patient's T-cells to undergo spontaneous proliferation in an ex vivo culture for about a week in the absence of exogenously added cytokines. The spontaneous proliferation of T-cells in HAM/TSP patients is attributed, at least partly, to autocrine/paracrine loops of IL-2, IL-9, and IL-15. It has been shown that adding blocking antibody against the IL-2 or IL-15 receptors can inhibit spontaneous T-cell proliferation in a HAM/TSP ex vivo culture system.

[0262] These observations, along with other data derived from ex vivo studies, have provided the rationale for taking two monoclonal antibodies (an anti-IL-2 receptor alpha or anti-Tac and an anti-IL-15 receptor beta chain) into the clinic for treatment of HAM/TSP (Azimi et al. 2001 Proc. Natl. Acad. Sci. 98: 14559-64., Azimi et al., 1999 J. Immunol 163:4064-72). Anti-cytokine receptor antagonists according to the embodiments described herein, would not only be valuable as a therapeutic immuno-modulatory agent for treatment of HAM/TSP, but modulation of immune response in HAM/TSP by anti-cytokine receptor antagonists according to the present embodiments acts proof-of-concept for the use of the anti-cytokine receptor antagonists according to the present embodiments in the treatment of other auto-immune diseases.

[0263] To demonstrate the efficacy of custom derivative yc-antagonist peptides according to the embodiments described herein, we tested the ability of ΒΝΖ-γ peptide to block immune response to HTLV-I in a spontaneous T-cell proliferation assay using a HAM/TSP ex vivo culture system. Proliferation assays were performed on HAM/TSP patient blood samples with and without the addition of ΒΝΖ-γ. These assays evaluated the ability of ΒΝΖ-γ to block the function of cytokines, such as IL-2 and IL-15, present in the ex vivo HAM/TSP patient blood culture and prevent spontaneous T-cell proliferation in these samples.

[0264] In an ex vivo spontaneous T-cell proliferation assay, PBMC from HAM/TSP patient was cultured at 1 x 10(6) cells per well of a 96 well plate in RPMI-10% FCS. Increasing concentrations of ΒΝΖ-γ peptide were added to each well. As a control, an irrelevant peptide was used in similar fashion. The cells were incubated in a 37°C C02 incubator for 3, 4, and 6 days. The amount of 1 uCi of 3 H-thymidine was added to the cells. After an additional 6 hour incubation, cells were harvested their proliferation rate was measured. The data for a representative HAM/TSP patient is shown in FIG. 5A - FIG. 5D. As indicated in FIG. 5A - FIG. 5D, ΒΝΖ-γ peptide inhibits the spontaneous proliferation of T- cells in HAM/TSP culture at a concentration of about 1 ug/ml.

[0265] Other immunological markers were additionally measured in this assay. The percentage of the viral specific CD8 cells was measured during the ex vivo culture using viral protein tetramers. The population of CD4+CD25+ cells, a marker of T-cell activation, as well as Ki67 staining, a marker of T-cell proliferation, was monitored in a flow cytometry assay.

[0266] Other forms of the conjugated ΒΝΖ-γ peptide derivative or a custom peptide comprising the sequence of SEQ ID NO: 3, and a derivative thereof can be used in a similar future assay. They include albumin, BSA, PEG that can be conjugated to the peptide after chemical synthesis. Other biological forms of custom peptides such as the ΒΝΖ-γ peptide conjugate or a custom peptide comprising the sequence of SEQ ID NO: 3, and a derivative thereof may include regions of known protein entities (including but not limited to Fc region of human IgG) that are fused to the custom peptides.

EXAMPLE 15 - Method of treating Adult T-cell Leukemia (ATL) in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0267] A human patient suffering from Adult T-cell Leukemia is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, ΒΝΖ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient enters remission.

EXAMPLE 16 - Method of treating HAM/TSP in a Human Patient by Administration of Custom Derivative jc- Antagonist Peptide

[0268] A human patient suffering from HAM/TSP is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, ΒΝΖ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 17 - Use of Custom Derivative yc-Antagonist Peptides to Block Cytokine Function

[0269] A human patient suffering a disease state who is in need of reducing the function of at least IL-15 and IL-21 is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 18 - Method of treating Celiac disease in a Human Patient by Administration of Custom Derivative jc- Antagonist Peptide

[0270] In celiac disease IL-15 is chronically up-regulated in the lamina propria and epithelium of the intestine and directly correlates with the severity of mucosal damage (Jabri et al. 2000 Gastroenterology 118:867-879., Maiuri et al. 2000 Gastroenterology 119:996-1006., Mention et al. 2003 Gastroenterology 125:730-45.; Di Sabatino et al. 2006 Gut 55:469-77.), and IL-21 production and function plays a positive role for disease progression (De Nitto et al. 2009 World J Gastroenterol 15:4609-14.). IL-15 has also been shown to directly correlate, and positively regulate, the increased expression of IL-21 observed in the CD mucosal environment (Sarra et al. 2013 Mucosal Immunol 6: 244-55.)· A recent study also identified synergistic effects of IL-2, IL-15 and IL-21 contribute greatly to the pathogenesis of refractory CD (Kooy-Winkelaar, et al., 2017 Proc Natl Acad Sci U S A 114: E980-9.).

[0271] A human patient suffering from Celiac disease is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, ΒΝΖ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 19 - Method of treating Rheumatoid Arthritis in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0272] In rheumatoid arthritis (RA) IL-15 expression is elevated in the synovial fluid and rheumatoid nodules of patients, increases the trans-endothelial migration of CD4+ and CD8+ T cells in the RA environment, and serum levels of IL-15 are positively correlated with disease progression (Oppenheimer-Marks et al. 1998 J Clin Invest 101: 1261-72., Harada et al. 1999 Arthritis Rheum 42: 1508-16., Gonzalez- Alvaro et al. 2003 Clin Exp Rheumatol 21:639-42., Hessian et al. 2003 Arthritis Rheum 48:334-8., Ruckert et al. 2009 Immunology 126:63-73.). In RA IL-21 positively impacts the expression of pro-inflammatory cytokines TNF-oc and IL-6, and the invasion and migration of fibroblast-like synoviocytes (FLS). FLS play a key role in RA pathogenesis through aggressive proliferation and invasion in patients (Xing et al. 2016 Clin Exp Immunol 184: 147-58., Xing et al. 2016 Scand J Immunol 83:64- 71.).

[0273] A human patient suffering from rheumatoid arthritis is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, ΒΝΖ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down. EXAMPLE 19 - Method of treating Multiple Sclerosis in a Human Patient by Administration of Custom Derivative jc- Antagonist Peptide

[0274] In multiple sclerosis (MS) IL-15 expression is elevated in the serum and cerebrospinal fluid of patients (Kivisakk et al. 1998 Clin Exp Immunol 111: 193-7., Pashenkov et al. 1999 Scand J Immunol 50:302-8., Vaknin-Dembinsky et al. 2008 J Neuroimmunol 195: 135-9.). Brain lesions of MS patients also contain astrocytes and B cells with increased levels of IL-15 (Saikali et al. 2010 J Immunol 185:5693-703., Schneider et al. 2011 J Immunol 187:4119-28.). In MS IL-21 production plays a positive role for disease progression and its expression is strongly elevated in CD4+ cells of acute and chronic MS lesions (Tzartos et al. 2011 Am J Pathol 178:794-802., Ghalamfarsa et al. 2016 J Immunotoxicol 13:274-85.).

[0275] A human patient suffering from multiple sclerosis is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 20 - Method of treating Type 1 Diabetes Mellitus in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0276] In type 1 diabetes mellitus (TID) IL-15 expression is elevated in the serum of patients (Kuczynski et al. 2005 Diabetes Res Clin Pract 69:231-6.). TID could be completely prevented by the inhibition of IL-15 signaling at the onset of insulitis in the non- obese diabetic mouse model (Bobbala et al. 2012 Diabetologia 55: 3010-3020.). The human pancreatic islet expression of IL-15 is also observed in TID patients (Chen et al. 2013 Proc Natl Acad Sci U S A 110: 13534-9.). Furthermore, high IL-21 production is correlated with TID disease progression (Ferreira et al. 2015 Diabetologia 58: 781-90.).

[0277] A human patient suffering from type 1 diabetes mellitus is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 21 - Method of treating Psoriasis in a Human Patient by Administration of Custom Derivative yc- Antagonist Peptide

[0278] The expression of IL-15 is elevated in skin lesions in psoriasis patients (Waldmann 2013 J Investig Dermatol Symp Proc 16:S28-30.). IL-21 production and function plays a positive role for psoriasis disease progression (Caruso et al. 2009 Cell Cycle 8: 3629- 30., Botti et al. 2012 Curr Pharm Biotechnol 13: 1861-7.), and expression of the yc-cytokine is elevated in the serum of patients and is associated with disease severity (He et al. 2012 Br J Dermatol 167: 191-3.).

[0279] A human patient suffering from psoriasis is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 22 - Method of treating Inflammatory Bowel Diseases in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0280] In inflammatory bowel diseases (IBD, including ulcerative colitis and Crohn's disease) increased levels of IL-15 is observed in inflamed mucosa (Liu et al. 2000 J Immunol 164:3608-15., Vainer et al. 2000 Cytokine 12: 1531-6.). IBD patients also experience increased levels of IL-21 in the gut (Monteleone et al. 2005 Gastroenterology 128: 687-94.), which is positively correlated with increased gut mucosa inflammation (De Nitto et al. 2010 World J Gastroenterol 16: 3638-41.).

[0281] A human patient suffering from an inflammatory bowel disease is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down. EXAMPLE 23 - Method of treating Systemic Lupus Erythematosus in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0282] In systemic lupus erythematosus (SLE) IL-15 expression is elevated in the serum of patients (Aringer et al. 2001 Rheumatology (Oxford) 40:876-81.)· Dysfunction of IL-15 signaling is positively associated with SLE pathogenesis (Baranda et al. 2005 Rheumatology (Oxford) 44:1507-13.). In SLE IL-21 expression is elevated in the serum of patients (Nakou et al. 2013 Clin Exp Rheumatol 31: 172-9.). Genetic polymorphisms in IL-21 are also positively associated with SLE (Sawalha et al. 2008 Ann Rheum Dis 67:458-61.). IL-21 production in SLE is positively correlated to T cell and B cell alterations observed in SLE pathogenesis (Terrier et al. 2012 J Rheumatol 39:1819-28.), and IL-21 signaling is critical for SLE pathogenic progression in the BXSB-Yaa murine disease model (Bubier et al. 2009 Proc Natl Acad Sci U S A 106: 1518-23.).

[0283] A human patient suffering from systemic lupus erythematosus is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, a composite peptide comprising the sequence of SEQ ID NO: 3 or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 24 - Method of treating Alopecia Areata Disease in a Human Patient by Administration of Custom Derivative yc-Antagonist Peptide

[0284] In alopecia areata disease (AAD) IL-15 expression is elevated in the lesional scalp biopsies of patients (Fuentes-Duculan et al. 2016 Exp Dermatol 4:282-6., Waldmann 2013 J Investig Dermatol Symp Proc 16:S28-30.), and antibodies targeting the yc-cytokines IL-2 and IL-15 each showed inhibitory activity in an AAD mouse model, but none of the blocking antibodies alone could reverse established AAD (Xing et al. 2014 Nat Med 9:1043-9.). In AAD IL-21 expression is elevated in the serum of patients versus healthy controls (Atwa et al. 2016 Int J Dermatol 55:666-72.). Genome-wide association studies have also positively correlated IL-2 and IL-21 with AAD (Jagielska et al. 2012 J Invest Dermatol 132:2192-7, Petukhova et al. 2010 Nature 466:113-7.). [0285] A human patient suffering from alopecia areata disease is identified. An effective dose, as determined by the physician, of custom derivative yc-antagonist peptide, for example, ΒΝΖ-γ, a custom peptide comprising the sequence of SEQ ID NO: 3, or a derivative thereof is administered to the patient for a period of time determined by the physician. Treatment is determined to be effective if patient' s symptoms improve or if the progression of the disease has been stopped or slowed down.

EXAMPLE 25 - Time-Course Protease Stability Measurement of Custom Peptide Derivatives Containing Intra-Peptide Hydrocarbon Linker Element in Simulated Intestinal Fluid

[0286] To evaluate the gastric stability of the custom peptide derivatives containing one or more intra-peptide hydrocarbon linker elements of the present embodiments a time-course protease stability measurement of unmodified SEQ ID NO: 3 in comparison with a representative custom peptide derivative of SEQ ID NO: 3 containing one hydrocarbon linker element (SEQ ID NO: 39), and another representative custom peptide derivative of SEQ ID NO: 3 containing two hydrocarbon linker elements and certain amino acid positions in the (D) stereochemical configuration (SEQ ID NO: 57) was carried out in simulated intestinal fluid over 60 minutes. An additional time-course protease stability experiment was conducted in simulated intestinal fluid over 120 minutes to compare two representative custom peptide derivatives of SEQ ID NO: 3, the first containing one hydrocarbon linker element (SEQ ID NO: 39), and the second containing two hydrocarbon linker elements (SEQ ID NO: 83). All peptides were commercially synthesized and purchased. Simulated intestinal fluid was made according to USP specifications (Test Solutions, United States Pharmacopeia 36). 50 of 10 mg/mL peptide was mixed with 450 pre-warmed (37°C) simulated intestinal fluid, and aliquots of the reaction mixture were removed at prescribed time intervals over a time-period of 60 minutes and stopped with 0.1 M HC1. Peptide stability was measured by reversed phase HPLC on a Phenomenex Aeris Peptide column (4.6 x 250 mm) with settings: 3.6 μιη particle size, non-porous; 5.0 μΐ, injection, 0.5mL/min; mobile phase A: 25% Acetonitrile with 0.1% TFA; mobile phase B: 100% Acetonitrile with 0.1% TFA. The multi-step buffer gradient goes from 100% A to 100% B over 26 minutes. Time point 0 minutes was set to 100% undigested peptide, and data at later time-points were graphed and connected via a smooth curve out to the end-point of 60 minutes (FIG. 8 A) or 120 minutes (FIG. 8B). Results show the percentage of undigested peptide present in the simulated intestinal fluid is greatly enhanced with one or more hydrocarbon linker elements and certain amino acid positions in the (D) stereochemical configuration. The data are shown in FIG. 8A and FIG. 8B.

EXAMPLE 26 - Inhibition of Cytokine-Induced Gene Transcription by Representative Custom Peptide Derivative

[0287] The cytokine interferon gamma (IFNy) represents a useful marker for inflammation in multiple human immune- inflammatory disease states. Transcription of the IFNy gene is inducible by IL-15, IL-21, the non-yc cytokine IL-12, and other select cytokines. The ability to block cytokine-induced IFNy transcription therefore represents another useful approach to determine the biological activity of custom peptide derivatives.

[0288] NK92 cells were brought to a quiescent state by PBS washing and culturing in the absence of cytokine for 48 hours. Cells were treated with SEQ ID NO: 83 at 10 μΜ for 10 minutes, and then stimulated with human IL-15, IL-21, or IL-12 at 1 ng/mL for 2 hours. A cytokine only treatment group, and another treatment group where the NK92 cells were left untreated by peptide, and not stimulated by cytokine were also included as control conditions. Following incubation with peptide and cytokine, cells were harvested, RNA was isolated for cDNA generation and used as template for qPCR analysis to quantify IFNy transcripts. SEQ ID NO: 83 showed inhibition of IL-15 and IL-21 induction of IFNy gene transcription, but did not block the non-yc cytokine IL-12 induction of IFNy gene transcription. The data are shown in FIG. 9.

References

[0289] All references cited in this disclosure are incorporated herein by reference in their entireties.

[0290] Antony, P.A., Paulos, CM., Ahmadzadeh, M., Akpinarli, A., Palmer, D.C., Sato, N., Kaiser A., Heinrichs, C.S., Klebanoff, C.A., Tagaya, Y., and Restifo, NP., Interleukin-2-dependent mechanisms of tolerance and immunity in vivo. 2006 J. Immunol. 176:5255-66.

[0291] Aringer M, Stummvoll GH, Steiner G, Roller M, Steiner CW, Hofler E, Hiesberger H, Smolen JS, Graninger WB. 2001. Serum interleukin-15 is elevated in systemic lupus erythematosus. Rheumatology (Oxford) 40: 876-81.

[0292] Atwa MA, Youssef N, Bayoumy NM. 2016 T-helper cytokines (interleukins 17, 21, 22, and 6, and tumor necrosis factor-a) in patients with alopecia areata: association with clinical type and severity. Int J Dermatol 55:666-72.

[0293] Azimi, N., Nagai, M., Jacobson, S., Waldmann, T.A., IL-15 plays a major role in the persistence of Tax-specific CD8 cells in HAM/TSP patients. 2001 Proc. Natl. Acad. Sci. 98:14559-64.

[0294] Azimi, N., Mariner J., Jacobson S., Waldmann T.A., How does interleukin 15 contribute to the pathogenesis of HTLV type-1 associated myelopathy/tropical spastic paraparesis? 2000 AIDS Res. Hum. Retroviruses 16: 1717-22.

[0295] Azimi, N., Jacobson, S., Leist, T., Waldmann, T.A., Involvement of IL-15 in the pathogenesis of human T lymphotropic virus type-I-associated myelopathy/tropical spastic paraparesis: implications for therapy with a monoclonal antibody directed to the IL- 2/15R beta receptor. 1999 J. Immunol. 163:4064-72.

[0296] Azimi, N., Brown, K., Bamford, R.N., Tagaya, Y., Siebenlist, U., Waldmann, T.A., Human T cell lymphotropic virus type I Tax protein trans-activates interleukin 15 gene transcription through an NF-kappaB site. 1998 Proc. Natl. Acad. Sci. USA 95:2452-7.

[0297] Baranda L, de la Fuente H, Layseca-Espinosa E, Portales -Perez D, Nino- Moreno P, Valencia-Pacheco G, Abud-Mendoza C, Alcocer-Varela J, Gonzalez-Amaro R. 2005. IL-15 and IL-15R in leucocytes from patients with systemic lupus erythematosus. Rheumatology (Oxford) 44: 1507-13.

[0298] Bazan, J.F., Hematopoietic receptors and helical cytokines. 1990 Immunol. Today 11:350-354. [0299] Bettini, M., and Vignali, D.A., Regulatory T cells and inhibitory cytokines in autoimmunity. 2009 Curr. Opin. Immunol. 21:612-8.

[0300] Bobbala D, Chen XL, Leblanc C, Mayhue M, Stankova J, Tanaka T, Chen YG, Ilangumaran S, Ramanathan S. 2012. Interleukin-15 plays an essential role in the pathogenesis of autoimmune diabetes in the NOD mouse. Diabetologia 55: 3010-3020.

[0301] Bodd, M., Raki, M., Tollefsen, S., Fallang, L.E., Bergseng, E., Lundin, K.E., Sollid, L.M., HLA-DQ2-restricted gluten-reactive T cells produce IL-21 but not IL-17 or IL-22. 2010 Mucosal Immunol. 3:594-601.

[0302] Botti E, Spallone G, Caruso R, Monteleone G, Chimenti S, Costanzo A. 2012. Psoriasis, from pathogenesis to therapeutic strategies: IL-21 as a novel potential therapeutic target. Curr Pharm Biotechnol 13: 1861-7.

[0303] Bubier JA, Sproule TJ, Foreman O, Spolski R, Shaffer DJ, Morse HC, 3rd, Leonard WJ, Roopenian DC. 2009. A critical role for IL-21 receptor signaling in the pathogenesis of systemic lupus erythematosus in BXSB-Yaa mice. Proc Natl Acad Sci U S A 106: 1518-23.

[0304] Caruso R, Costanzo A, Monteleone G. 2009. Pathogenic role of interleukin-21 in psoriasis. Cell Cycle 8: 3629-30.

[0305] Chen J, Feigenbaum L, Awasthi P, Butcher DO, Anver MR, Golubeva YG, Bamford R, Zhang X, St Claire MB, Thomas CJ et al. 2013. Insulin-dependent diabetes induced by pancreatic beta cell expression of IL-15 and IL-15Ralpha. Proc Natl Acad Sci U S A 110: 13534-9.

[0306] De Nitto D, Monteleone I, Franze E, Pallone F, Monteleone G. 2009. Involvement of interleukin-15 and interleukin-21, two gamma-chain-related cytokines, in celiac disease. World J Gastroenterol 15: 4609-14.

[0307] De Nitto D, Sarra M, Pallone F, Monteleone G. 2010. Interleukin-21 triggers effector cell responses in the gut. World J Gastroenterol 16: 3638-41.

[0308] De Rezende, L.C., Silva I.V., Rangel, L.B., Guimaraes, M.C., Regulatory T cells as a target for cancer therapy. 2010 Arch. Immunol. Ther. Exp. 58: 179-90.

[0309] Di Sabatino A, Ciccocioppo R, Cupelli F, Cinque B, Millimaggi D, Clarkson MM, Paulli M, Cifone MG, Corazza GR. 2006. Epithelium derived interleukin 15 regulates intraepithelial lymphocyte Thl cytokine production, cytotoxicity, and survival in coeliac disease. Gut 55: 469-77. [0310] Dubois, S., Mariner, J., Waldmann, T.A., Tagaya, Y., IL-15Ralpha recycles and presents IL-15 In trans to neighboring cells. 2002 Immunity 17:537-47.

[0311] Dodge DL. Et al., IL-2 and IL-12 alter NK cell responsiveness to WN- gamma-inducible protein 10 by down-regulating CXCR3 expression.J. Immun. 168:6090-8.

[0312] Fehniger, T.A., Suzuki, K., Ponnappan, A., VanDeusen, J.B., Cooper, M.A., Florea, S.M., Freud, A.G., Robinson, M.L., Durbin, J., Caligiuri, M.A., Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. 2001 J. Exp. Med. 193:219-31.

[0313] Ferreira RC, Simons HZ, Thompson WS, Cutler AJ, Dopico XC, Smyth DJ, Mashar M, Schuilenburg H, Walker NM, Dunger DB et al. 2015. IL-21 production by CD4+ effector T cells and frequency of circulating follicular helper T cells are increased in type 1 diabetes patients. Diabetologia 58: 781-90.

[0314] Fisher, A.G., Burdet, C, LeMeur, M., Haasner, D., Gerber, P., Cerediq, R., Lymphoproliferative disorders in an IL-7 transgenic mouse line. 1993 Leukemia 2:S66-68.

[0315] Fuentes-Duculan J, Gulati N, Bonifacio KM, Kunjravia N, Zheng X, Suarez-Farinas M, Shemer A, Guttman-Yassky E, Krueger JG, Biomarkers of alopecia areata disease activity and response to corticosteroid treatment. 2016 Exp Dermatol 4:282-6.

[0316] Ghalamfarsa G, Mahmoudi M, Mohammadnia-Afrouzi M, Yazdani Y, Anvari E, Hadinia A, Ghanbari A, Setayesh M, Yousefi M, Jadidi-Niaragh F. 2016. IL-21 and IL-21 receptor in the immunopathogenesis of multiple sclerosis. J Immunotoxicol 13:274-85.

[0317] Gong JH, et al. Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia 8: 652- 658, 1994.

[0318] Gonzalez- Alvaro I, Ortiz AM, Garcia- Vicuna R, Balsa A, Pascual-Salcedo D, Laffon A. 2003. Increased serum levels of interleukin- 15 in rheumatoid arthritis with long- term disease. Clin Exp Rheumatol 21: 639-42.

[0319] Habib, T., Nelson, A., Kaushansky, K., IL-21 : a novel IL-2-family lymphokine that modulates B, T, and natural killer cell responses. 2003 J Allergy Clin. Immunol. 112: 1033-45.

[0320] Harada S, Yamamura M, Okamoto H, Morita Y, Kawashima M, Aita T, Makino H. 1999. Production of interleukin-7 and interleukin- 15 by fibroblast- like synoviocytes from patients with rheumatoid arthritis. Arthritis Rheum 42: 1508-16. [0321] He Z, Jin L, Liu ZF, Hu L, Dang EL, Feng ZZ, Li QJ, Wang G. 2012. Elevated serum levels of interleukin 21 are associated with disease severity in patients with psoriasis. Br J Dermatol 167: 191-3.

[0322] Hennighausen, L., Robinson, G.W., Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B. 2008 Genes Dev. 22:711-21.

[0323] Hessian PA, Highton J, Kean A, Sun CK, Chin M. 2003. Cytokine profile of the rheumatoid nodule suggests that it is a Thl granuloma. Arthritis Rheum 48: 334-8.

[0324] Jabri B, de Serre NP, Cellier C, Evans K, Gache C, Carvalho C, Mougenot JF, Allez M, Jian R, Desreumaux P et al. 2000. Selective expansion of intraepithelial lymphocytes expressing the HLA-E-specific natural killer receptor CD94 in celiac disease. Gastroenterology 118: 867-79.

[0325] Jagielska D, Redler S, Brockschmidt FF, Herold C, Pasternack SM, Garcia Bartels N, Hanneken S, Eigelshoven S, Refke M, Barth S, Giehl KA, Kruse R, Lutz G, Wolff H, Blaumeiser B, Bohm M, Blume-Peytavi U, Becker T, Nothen MM, Betz RC. 2012 J Invest Dermatol 132:2192-7.

[0326] Kivisakk P, Matusevicius D, He B, Soderstrom M, Fredrikson S, Link H. 1998. IL-15 mRNA expression is up-regulated in blood and cerebrospinal fluid mononuclear cells in multiple sclerosis (MS). Clin Exp Immunol 111 : 193-7.

[0327] Klingemann HG, et al. A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood. Biol. Blood Marrow Transplant. 2: 68-75, 1996.

[0328] Kooy-Winkelaar, Y.M, Bouwer, D., Janssen, G.M., Thompson, A., Brugman, M.H., Schmitz, F., de Ru, A.H., van Gils, T., Bouma, G., van Rood, J.J. et al. 2017 CD4 T-cell cytokines synergize to induce proliferation of malignant and nonmalignant intraepithelial lymphocytes. Proc Natl Acad Sci U S A 114: E980-9.

[0329] Krause, CD. and Pestka, S., Evolution of the Class 2 cytokines and receptors, and discovery of new friends and relatives. 2005 Pharmacol, and Therapeutics 106:299-346.

[0330] Kuczynski S, Winiarska H, Abramczyk M, Szczawinska K, Wierusz- Wysocka B, Dworacka M. 2005. IL-15 is elevated in serum patients with type 1 diabetes mellitus. Diabetes Res Clin Pract 69: 231-6.

[0331] Kundig, T.M., Schorle, H., Bachmann, M.F., Hengartener, H., Zinkernagel, R.M., Horak, I., Immune Responses of the interleukin-2-deficient mice. 1993 Science 262: 1059-61. [0332] Le Buanec, H., Paturance, S., Couillin, I., Schnyder-Candrian, S., Larcier, P., Ryffel, B., Bizzini, B., Bensussan, A., Burny, A., Gallo, R., Zagury, D., Peltre, G., Control of allergic reactions in mice by an active anti-murine IL-4 immunization. 2007 Vaccine 25:7206-16.

[0333] Littman, D.R., Rudensky, AY., Thl7 and regulatory T cells in mediating and restraining inflammation. 2010 Cell 140(6):845-58.

[0334] Liu Z, Geboes K, Colpaert S, D'Haens GR, Rutgeerts P, Ceuppens JL. 2000. IL-15 is highly expressed in inflammatory bowel disease and regulates local T cell- dependent cytokine production. J Immunol 164: 3608-15.

[0335] Maiuri L, Ciacci C, Auricchio S, Brown V, Quaratino S, Londei M. 2000. Interleukin 15 mediates epithelial changes in celiac disease. Gastroenterology 119: 996-1006.

[0336] Mention JJ, Ben Ahmed M, Begue B, Barbe U, Verkarre V, Asnafi V, Colombel JF, Cugnenc PH, Ruemmele FM, Mclntyre E et al. 2003. Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease. Gastroenterology 125: 730-45.

[0337] Miyagawa, F., Tagaya, Y., Kim, B.S., Patel, H.J., Ishida, K., Ohteki, T., Waldmann, T.A., Katz, S.I., IL-15 serves as a costimulator in determining the activity of autoreactive CD8 T cells in an experimental mouse model of graft-versus-host-like disease. 2008 J. Immunol. 181: 1109-19.

[0338] Monteleone G, Monteleone I, Fina D, Vavassori P, Del Vecchio Blanco G, Caruso R, Tersigni R, Alessandroni L, Biancone L, Naccari GC et al. 2005. Interleukin-21 enhances T-helper cell type I signaling and interferon-gamma production in Crohn's disease. Gastroenterology 128: 687-94.

[0339] Nakou M, Papadimitraki ED, Fanouriakis A, Bertsias GK, Choulaki C, Goulidaki N, Sidiropoulos P, Boumpas DT. 2013. Interleukin-21 is increased in active systemic lupus erythematosus patients and contributes to the generation of plasma B cells. Clin Exp Rheumatol 31 : 172-9.

[0340] Noguchi, M., Yi, H., Rosenblatt, H.M., Filipovich, A.H., Adelstein, S., Modi, W.S., McBride, O.W., Leonard, W.J., Interleukin 2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. 1993 Cell 73: 147-157.

[0341] Oh, U., Jacobson S., Treatment of HTLV-I-Associated Myelopathy/Tropical Spastic Paraparesis: Towards Rational Targeted Therapy 2008 Neurol Clin. 2008 26: 781-785. [0342] Oppenheimer-Marks N, Brezinschek RI, Mohamadzadeh M, Vita R, Lipsky PE. 1998. Interleukin 15 is produced by endothelial cells and increases the transendothelial migration of T cells in vitro and in the SCID mouse-human rheumatoid arthritis model In vivo. J Clin Invest 101 : 1261-72.

[0343] Orzaez, M., Gortat, A., Mondragon, L., Perez-Paya, E., Peptides and Peptide Mimics as Modulators of Apototic Pathways. 2009 Chem. Med. Chem. 4: 146-160.

[0344] O'Shea, J.J., Targeting the Jak/STAT pathway for immunosuppression.

2004 Ann. Rheum. Dis. 63:(suppl II): U67-71.

[0345] Pashenkov M, Mustafa M, Kivisakk P, Link H. 1999. Levels of interleukin- 15 -expressing blood mononuclear cells are elevated in multiple sclerosis. Scand J Immunol 50: 302-8.

[0346] Paul, W.E., Pleiotropy and redundancy: T cell-derived lymphokines in the immune response. 1989 Cell 57:521-4.

[0347] Pesu M, Candotti F, Husa M, Hofmann SR, Notarangelo LD, and O'Shea JJ. Jak3, severe combined immunodeficiency, and a new class of immunosuppressive drugs.

2005 Immunol. Rev. 203: 127-142.

[0348] Pesu, M., Laurence, A., Kishore, N., Zwillich, S., Chan, G., O'Shea, J.J., Therapeutic targeting of Janus kinases. Immunol. 2008 Rev. 223: 132-142.

[0349] Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y, Kim H, Singh P, Lee A, Chen WV, Meyer KC, Paus R, Jahoda CA, Amos CI, Gregersen PK, Christiano AM. 2010 Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature 466: 113-7.

[0350] Rochman, Y., Spolski, R., Leonard, W.J., New Insights into the regulation of T cells by gamma c family cytokines. 2009 Nat. Rev. Immunol. 9:480-90.

[0351] Ruckert R, Brandt K, Ernst M, Marienfeld K, Csernok E, Metzler C, Budagian V, Bulanova E, Paus R, Bulfone-Paus S. 2009. Interleukin- 15 stimulates macrophages to activate CD4+ T cells: a role in the pathogenesis of rheumatoid arthritis? Immunology 126: 63-73.

[0352] Saikali P, Antel JP, Pittet CL, Newcombe J, Arbour N. 2010. Contribution of astrocyte-derived IL-15 to CD8 T cell effector functions in multiple sclerosis. J Immunol 185: 5693-703.

[0353] Sakaguchi, S., Yamaguchi, T., Nomura, T., Ono, M., Regulatory T cells and immune tolerance. 2008 Cell 133: 775-87. [0354] Sarra M, Cupi ML, Monteleone I, Franze E, Ronchetti G, Di Sabatino A, Gentileschi P, Franceschilli L, Sileri P, Sica G et al. 2013. IL-15 positively regulates IL-21 production in celiac disease mucosa. Mucosal Immunol 6: 244-55.

[0355] Sato, N., Sabzevari, H., Fu, S., Ju, W., Bamford, R.N., Waldmann, T.A., and Tagaya, Y. 2011 Development of an IL-15-Autocrine CD8 T-cell Leukemia in IL-15 Transgenic mice requires the cis-expression of IL-15R alpha. Blood 117:4032-40.

[0356] Sawalha AH, Kaufman KM, Kelly JA, Adler AJ, Aberle T, Kilpatrick J, Wakeland EK, Li QZ, Wandstrat AE, Karp DR et al. 2008. Genetic association of interleukin- 21 polymorphisms with systemic lupus erythematosus. Ann Rheum Dis 67: 458-61.

[0357] Schneider R, Mohebiany AN, Ifergan I, Beauseigle D, Duquette P, Prat A, Arbour N. 2011. B cell-derived IL-15 enhances CD8 T cell cytotoxicity and is increased in multiple sclerosis patients. J Immunol 187: 4119-28.

[0358] Sugamura, K., Asao, H., Kondo, M., Tanaka, N., Ishii, N., Nakamura, M., Takeshita, T., The common gamma-chain for multiple cytokine receptors. 1995 Adv. Immunol. 59: 225-277.

[0359] Sugamura, K., Asao, H., Kondo, M., Tanaka, N., Ishii, N., Ohbo, K., Nakamura, M., Takeshita, T., The interleukin-2 receptor gamma chain: its role in the multiple cytokine receptor complexes and T cell development in XSCID. 1996 Annu. Rev. Immunol. 14: 179-205.

[0360] Tagaya, Y., Burton, J.D., Miyamoto, Y., Waldmann, TA., Identification of a novel receptor/signal transduction pathway for IL-15/T in mast cells. 1996 EMBO J. 15:4928-39.

[0361] Tagaya, Y., Memory CD8 T cells now join "Club 21". 2010 J. Leuk. Biol. 87: 13-15.

[0362] Takai, K., Sawasaki, T., and Endo. Y. The Wheat-Germ Cell-Free Expression System, 2010 Curr. Pharm. Biotechnol. 11 :272-8.

[0363] Takeshita, T., Asao, H., Ohtani, K., Ishii, N., Kumaki, S., Tanaka, N., Manukata, H., Nakamura, M., Sugamura, K., Cloning of the Gamma chain of the Human IL2 receptor. 1992 Science 257:379-382.

[0364] Tanaka, T., et al., A novel monoclonal antibody against murine IL-2 receptor beta-chain. Characterization of receptor expression in normal lymphoid cells and EL- 4 cells. 1991 J. Immunol. 147:2222-28. [0365] Terrier B, Costedoat-Chalumeau N, Garrido M, Geri G, Rosenzwajg M, Musset L, Klatzmann D, Saadoun D, Cacoub P. 2012. Interleukin 21 correlates with T cell and B cell subset alterations in systemic lupus erythematosus. J Rheumatol 39: 1819-28.

[0366] Tzartos JS, Craner MJ, Friese MA, Jakobsen KB, Newcombe J, Esiri MM, Fugger L. 2011. IL-21 and IL-21 receptor expression in lymphocytes and neurons in multiple sclerosis brain. Am J Pathol 178: 794-802.

[0367] Vainer B, Nielsen OH, Hendel J, Horn T, Kirman I. 2000. Colonic expression and synthesis of interleukin 13 and interleukin 15 in inflammatory bowel disease. Cytokine 12: 1531-6.

[0368] Vaknin-Dembinsky A, Brass SD, Gandhi R, Weiner HL. 2008. Membrane bound IL-15 is increased on CD14 monocytes in early stages of MS. J Neuroimmunol 195: 135-9.

[0369] Waldmann, T.A., Anti-Tac (daclizumab, Zenapax) in the treatment of leukemia, autoimmune diseases, and in the prevention of allograft rejection: a 25-year personal odyssey. 2007 J. Clin. Immunol. 27: 1-18.

[0370] Waldmann TA. 2013. The biology of IL-15: implications for cancer therapy and the treatment of autoimmune disorders. J Investig Dermatol Symp Proc 16: S28- 30.

[0371] Walensky, L.D., Bird, G.H., Hydrocarbon-Stapled Peptides: Principles, Practice, and Progress. 2014 J. Med. Chem. 57: 6275-88.

[0372] Xing L, Dai Z, Jabbari A, Cerise JE, Higgins CA, Gong W, de Jong A, Harel S, DeStefano GM, Rothman L, Singh P, Petukhova L, Mackay-Wiggan J, Christiano AM, Clynes R. 2014 Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat Med 9: 1043-9.

[0373] Xing R, Jin Y, Sun L, Yang L, Li C, Li Z, Liu X, Zhao J. 2016 Interleukin- 21 induces migration and invasion of fibroblast-like synoviocytes from patients with rheumatoid arthritis. Clin Exp Immunol 184: 147-58.

[0374] Xing R, Yang L, Jin Y, Sun L, Li C, Li Z, Zhao J, Liu X. 2016 Interleukin- 21 Induces Proliferation and Proinflammatory Cytokine Profile of Fibroblast-like Synoviocytes of Patients with Rheumatoid Arthritis. Scand J Immunol 83: 64-71.