Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THERAPEUTIC AGENTS COMPRISING ELASTIN-LIKE PEPTIDES
Document Type and Number:
WIPO Patent Application WO/2009/158704
Kind Code:
A2
Abstract:
The present invention provides therapeutic agents and compositions comprising elastin-like peptides (ELPs) and therapeutic proteins. In some embodiments, the therapeutic protein is a GLP-1 receptor agonist, insulin, or Factor VII/VIIa, including functional analogs. The present invention further provides encoding polynucleotides, as well as methods of making and using the therapeutic agents. The therapeutic agents have improvements in relation to their use as therapeutics, including, inter alia, one or more of half-life, clearance and/or persistance in the body, solubility, and bioavailability.

Inventors:
CHILKOTI ASHUTOSH (US)
Application Number:
PCT/US2009/049059
Publication Date:
December 30, 2009
Filing Date:
June 29, 2009
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV DUKE (US)
CHILKOTI ASHUTOSH (US)
International Classes:
A61K38/26; A61K38/00; A61K38/22; C07H21/00; C07K2/00
Foreign References:
US20070009602A12007-01-11
Other References:
KREYMANN, LANCET, vol. 2, no. 8571, 1987, pages 1300 - 4
WETTERGEN, DIG DIS SCI, vol. 38, 1993, pages 665 - 73
D'ALESSIO ET AL., J. CLIN INVEST, vol. 93, 1994, pages 2293 - 6
GUTNIAK ET AL., DIABETES CARE, vol. 17, 1994, pages 1039 - 44
WILLIAMS, J. CLIN ENDO METAB, vol. 81, 1996, pages 327 - 32
HOLZ ET AL., NATURE, vol. 361, 1993, pages 362 - 5
ORSKOV, DIABETOLOGIA, vol. 35, 1992, pages 701 - 11
WEIR ET AL., DIABETES, vol. 38, 1989, pages 338 - 342
GUTNIAK, M., N. ENGL J MED, vol. 226, 1992, pages 1316 - 22
NAUCK ET AL., DIABETOLOGIA, vol. 36, 1993, pages 741 - 4
CREUTZFELDT, DIABETES CARE, vol. 19, 1996, pages 580 - 6
DEACON ET AL., DIABETES, vol. 44, 1995, pages 1126 - 31
SAHA ET AL., J. PHARM. EXP. THER, vol. 316, 2006, pages 1159 - 64
TATUSOVA ET AL.: "Blast 2 seauences - a new tool for comparing protein and nucleotide seauences", FEMS MICROBIAL LETT, vol. 174, 1999, pages 247 - 250
SIEGEL ET AL., REGUL PEPT, vol. 79, no. 2-3, 1999, pages 93 - 102
FEHMANN, ENDOCRINOLOGY, vol. 130, 1992, pages 159 - 166
BUTEAU, DIABETES, vol. 52, 2003, pages 124 - 32
BULOTTA ET AL., CELL BIOCHEM BIOPHYS, vol. 40, no. 3, 2004, pages 65 - 78
DUTTAROY ET AL., DIABETES, vol. 54, 2005, pages 251 - 8
TATUSOVA ET AL.: "Blast 2 sequences - a new tool for comparing protein and nucleotide sequences", FEMS MICROBIAL LETT., vol. 174, 1999, pages 247 - 250
JEHLE ET AL., DIABETOLOGIA, vol. 39, 1996, pages 421 - 432
CHAING ET AL., BLOOD, vol. 83, no. 12, 1994, pages 3524 - 35
NILSSON ET AL., ANN REV BIOPHYS BIO STRUCTURE, vol. 3, 2005, pages 91 - 118
MEYER; CHILKOTI, NAT. BIOTECHNOL., vol. 17, no. 11, 1999, pages 1112 - 5
"Current Protocols in Molecular Bioloav", 1995
See also references of EP 2307038A4
Attorney, Agent or Firm:
HAYMAN, Mark et al. (777 6th Street N.W.,Suite 110, Washington DC, US)
Download PDF:
Claims:
What is claimed is:

1. A therapeutic agent comprising an elastin-like peptide (ELP) component and a therapeutic component, the therapeutic component having an extended circulatory half-life and/or a lower therapeutically effective dose when compared to the therapeutic component alone.

2. The therapeutic agent of claim 1 , wherein the therpeutic component is a therapeutic protein listed in Table 1 or a functional portion or functional analog thereof.

3. The therapeutic agent of claim 2, wherein an ELP component is covalently bonded to the therapeutic protein at an N- and/or C-terminus thereof.

4. The therapeutic agent of any one of claims 1-3, wherein a therapeutic component is covalently bonded to the ELP component at the N-terminus of the ELP component, and a second therapeutic component is covalently bonded to the ELP component at the C- terminus of the ELP component.

5. The therapeutic agent of any one of claims 1-4, wherein a therapeutic component is covalently bonded to the ELP component at an N- and/or C-terminus thereof.

6. The therapeutic agent of any one of claims 1-5, wherein a first ELP component is covalently bonded to the therapeutic component at the N- terminus of the therapeutic component, and a second ELP component is covalently bonded to the therapeutic component at the C- terminus of the therapeutic component.

7. The therapeutic agent of any one of claims 1-6, further comprising a spacer moiety between the ELP component and the therapeutic component.

8. The therapeutic agent of claim 7, wherein the spacer moiety comprises one or more of a protease-resistant moiety, a non-peptide chemical moiety, and a protease cleavage site.

9. The therapeutic agent of claim 8, wherein the protease cleavage site is a thrombin cleavage site, a factor Xa cleavage site, a metalloprotease cleavage site, an enterokinase cleavage site, a Tev cleavage site, and a cathepsin cleavage site.

10. The therapeutic agent of claim 9, wherein the spacer moiety comprises a non- cleavable moiety having the formula [(Gly) n -Ser] m (SEQ ID NO: 22) where n is from 1 to 4, inclusive, and m is from 1 to 4, inclusive.

11. The therapeutic agent of any one of claims 1-10, wherein the ELP comprises at least one repeating unit selected from SEQ ID NOS: 1-12.

12. The therapeutic agent of claim 11 , wherein said repeating unit is VPGXG (SEQ ID NO: 3).

13. The therapeutic agent of claim 12, wherein X is any natural or non-natural amino acid residue, and wherein X varies among at least two units.

14. The therapeutic agent of claim 13, wherein each X is independently selected from alanine, arginine, asparagine, aspartic acid, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tryptophan, tyrosine and valine residues.

15. The therapeutic agent of any one of claims 1-14, wherein the therapeutic agent has a molecular weight of less than about 50 kDa.

16. The therapeutic agent of any one of claims 1-15, wherein the ELP component has a Tt greater than 37°C.

17. The therapeutic agent of any one of claims 1-16, wherein the therapeutic agent is a genetically encoded fusion protein.

18. A polynucleotide comprising a nucleotide sequence encoding the therapeutic agent of claim 17.

19. The polynucleotide of claim 18, further comprising an expression control element operably linked to said nucleotide sequence.

20. The polynucleotide of claim 19, wherein said expression control element comprises a promoter.

21. A vector comprising the polynucleotide of any one of claims 18-20.

22. An isolated host cell containing the vector of claim 21 or the polynucleotide of any one of claims 18-20.

23. A pharmaceutical composition comprising the therapeutic agent of any one of claims 1-18, and a pharmaceutically acceptable carrier and/or excipient.

24. A method of treating or preventing a disease, disorder, or condition in a subject, comprising administering an effective amount of the therapeutic agent of any one of claims 1-17 or the pharmaceutical composition of claim 23 to a subject in need thereof.

25. The method of claim 24, wherein the subject has an indication listed in Table 1.

26. A therapeutic agent comprising an elastin-like peptide (ELP) component and a glucagon-like peptide (GLP)-I receptor agonist.

27. The therapeutic agent of claim 26, wherein the GLP-1 receptor agonist is GLP-1 or functional analog thereof.

28. The therapeutic agent of claim 26, wherein the GLP-1 receptor agonist is an exendin- 4 or functional analog thereof.

29. The therapeutic agent of any one of claims 26-28, wherein the ELP component is covalently bonded to the GLP-1 receptor agonist at an N- and/or C-terminus thereof.

30. The therapeutic agent of any one of claims 26-29, wherein a first GLP-1 receptor agonist is covalently bonded to the ELP component at the N-terminus of the ELP component, and a second GLP-1 receptor agonist is covalently bonded to the ELP component at the C-terminus of the ELP component.

31. The therapeutic agent of any one of claims 26-30, wherein the GLP-1 receptor agonist is covalently bonded to the ELP component at an N- and/or C-terminus thereof.

32. The therapeutic agent of any one of claims 26-31 , wherein a first ELP component is covalently bonded to the GLP-1 receptor agonist at the N-terminus of the GLP-1 receptor agonist, and a second ELP component is covalently bonded to the GLP-1 receptor agonist at the C- terminus of the GLP-1 receptor agonist.

33. The therapeutic agent of any one of claims 26-32, further comprising a spacer moiety between the ELP component and GLP-receptor agonist.

34. The therapeutic agent of claim 33, wherein the spacer moiety comprises one or more of a protease-resistant moiety, a non-peptide chemical moiety, and a protease cleavage site.

35. The therapeutic agent of claim 34, wherein the protease cleavage site is a thrombin cleavage site, a factor Xa cleavage site, a metalloprotease cleavage site, an enterokinase cleavage site, a Tev cleavage site, and a cathepsin cleavage site.

36. The therapeutic agent of claim 34, wherein the spacer moiety comprises a non- cleavable moiety having the formula [(Gly) n -Ser] m (SEQ ID NO.: 22) where n is from 1 to 4, inclusive, and m is from 1 to 4, inclusive.

37. The therapeutic agent of any one of claims 26-36, wherein the ELP comprises at least one repeating unit selected from SEQ ID NOS: 1-12.

38. The therapeutic agent of claim 37, wherein said repeating unit is VPGXG (SEQ ID NO: 3).

39. The therapeutic agent of claim 38, wherein X is any natural or non-natural amino acid residue, and wherein X varies among at least two units.

40. The therapeutic agent of claim 38, wherein each X is independently selected from alanine, arginine, asparagine, aspartic acid, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tryptophan, tyrosine and valine residues.

41. The therapeutic agent of any one of claims 26-40, wherein the therapeutic agent has a molecular weight of less than about 50 kDa.

42. The therapeutic agent of any one of claims 26-40, wherein the ELP component has a Tt greater than 37°C.

43. The therapeutic agent of any one of claims 26-42, wherein the therapeutic agent is a genetically encoded fusion protein.

44. A polynucleotide comprising a nucleotide sequence encoding the therapeutic agent of claim 43.

45. The polynucleotide of claim 44, further comprising an expression control element operably linked to said nucleotide sequence.

46. The polynucleotide of claim 45, wherein said expression control element comprises a promoter.

47. A vector comprising the polynucleotide of any one of claims 44-46.

48. An isolated host cell containing the vector of claim 47 or the polynucleotide of any one of claims 44-46.

49. A pharmaceutical composition comprising the therapeutic agent of any one of claims 26-43, and a pharmaceutically acceptable carrier and/or excipient.

50. A method of treating or preventing a biological condition, disorder, or disease in a subject, comripising administering an effective amount of the therapeutic agent of any one of claims 26-43 or the pharmaceutical composition of claim 49 to a subject in need thereof.

51. The method of claim 50, wherein the subject has diabetes.

52. A therapeutic agent comprising an elastin-like peptide (ELP) component and an insulin or functional analog thereof.

53. The therapeutic agent of claim 52, wherein the ELP component is covalently bonded to the insulin or functional analog at an N- and/or C-terminus thereof.

54. The therapeutic agent of claim 52 or 53, wherein a first insulin or functional analog is covalently bonded to the ELP component at the N-terminus of the ELP component, and a second insulin or functional analog is covalently bonded to the ELP component at the C- terminus of the ELP component.

55. The therapeutic agent of claim 52, wherein the insulin or functional analog is covalently bonded to the ELP component at an N- and/or C-terminus thereof.

56. The therapeutic agent of claim 52 or 53, wherein a first ELP component is covalently bonded to the insulin or functional analog at an N- terminus thereof, and a second ELP component is covalently bonded to the insulin or functional analog at a C- terminus thereof.

57. The therapeutic agent of any one of claims 52-56, further comprising a spacer moiety between the ELP component and the insulin or functional analog.

58. The therapeutic agent of claim 57, wherein the spacer moiety comprises one or more of a protease-resistant moiety, a non-peptide chemical moiety, and a protease cleavage site.

59. The therapeutic agent of claim 58, wherein the protease cleavage site is a thrombin cleavage site, a factor Xa cleavage site, a metalloprotease cleavage site, an enterokinase cleavage site, a Tev cleavage site, or a cathepsin cleavage site.

60. The therapeutic agent of claim 58, wherein the spacer moiety comprises a non- cleavable moiety having the formula [(Gly) n -Ser] m (SEQ ID NO: 22) where n is from 1 to 4, inclusive, and m is from 1 to 4, inclusive.

61. The therapeutic agent of any one of claims 52-60, wherein the ELP component comprises at least one repeating unit selected from SEQ ID NOS: 1-12.

62. The therapeutic agent of claim 61 , wherein said repeating unit is VGPXG (SEQ ID NO: 3).

63. The therapeutic agent of claim 61 , wherein the repeating unit is Ile-Pro-Gly-X-Gly (SEQ ID NO: 5) or Leu-Pro-Gly-X-Gly (SEQ ID NO: 7), wherein X is any natural or non- natural amino acid residue, and wherein X varies among at least two units.

64. The therapeutic agent of claim 62 or 63, wherein each X is independently selected from alanine, arginine, asparagine, aspartic acid, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tryptophan, tyrosine and valine residues.

65. The therapeutic agent of any one of claims 52-64, wherein the therapeutic agent has a molecular weight of less than about 50 kDa.

66. The therapeutic agent of any one of claims 52-65, wherein the ELP component has a Tt greater than 37°C.

67. The therapeutic agent of any one of claims 52-66, wherein the therapeutic agent is a genetically encoded fusion protein.

68. A polynucleotide comprising a nucleotide sequence encoding the therapeutic agent of claim 67.

69. The polynucleotide of claim 68, further comprising an expression control element operably linked to said nucleotide sequence.

70. The polynucleotide of claim 69, wherein said expression control element comprises a promoter.

71. A vector comprising the polynucleotide of any one of claims 68-70.

72. An isolated host cell containing the vector of claim 71 or the polynucleotide of any one of claims 68-70.

73. A pharmaceutical composition comprising the therapeutic agent of any one of claims 52-67, and a pharmaceutically acceptable carrier and/or excipient.

74. A method of preventing or treating a biological condition, disorder, or disease in a subject, comprising administering an effective amount of the therapeutic agent of any one of claims 52-67 or the pharmaceutical composition of claim 73 to a subject in need thereof.

75. The method of claim 74, wherein the subject has diabetes.

76. A therapeutic agent comprising an elastin-like peptide (ELP) component and a Factor Vll/Vlla or functional analog thereof.

77. The therapeutic agent of claim 76, wherein the ELP component is covalently bonded to the Factor Vll/Vlla or functional analog at an N- and/or C-terminus of the Factor Vll/Vlla.

78. The therapeutic agent of claim 76 or 77, wherein a first Factor Vll/Vlla or functional analog is covalently bonded to the ELP at the N-terminus of the ELP component, and a second Factor Vll/Vlla or functional analog is covalently bonded to the ELP component at the C- terminus of the ELP component.

79. The therapeutic agent of claim 76, wherein the Factor Vll/Vlla or functional analog is covalently bonded to the ELP component at an N- and/or C-terminus of the ELP component.

80. The therapeutic agent of claim 76 or 77, wherein a first ELP component is covalently bonded to the Factor Vll/Vlla or functional analog at an N- terminus thereof, and a second ELP component is covalently bonded to the Factor Vll/Vlla or functional analog at a C- terminus thereof.

81. The therapeutic agent of any one of claims 76-80, further comprising a spacer moiety between the ELP component and factor Vll/Vlla or functional analog.

82. The therapeutic agent of claim 81 , wherein the spacer moiety comprises one or more of a protease-resistant moiety, a non-peptide chemical moiety, and a protease cleavage site.

83. The therapeutic agent of claim 82, wherein the protease cleavage site is a thrombin cleavage site, a factor Xa cleavage site, a metalloprotease cleavage site, an enterokinase cleavage site, a Tev cleavage site, and a cathepsin cleavage site.

84. The therapeutic agent of claim 82, wherein the spacer moiety comprises a non- cleavable moiety having the formula [(Gly) n -Ser] m (SEQ ID NO: 22) where n is from 1 to 4, inclusive, and m is from 1 to 4, inclusive.

85. The therapeutic agent of any one of claims 76-84, wherein the ELP component comprises at least one repeating unit selected from SEQ ID NOS: 1-12.

86. The therapeutic agent of claim 85, wherein said repeating unit is VPGXG (SEQ ID NO: 3).

87. The therapeutic agent of claim 86, wherein X is any natural or non-natural amino acid residue, and wherein X varies among at least two units.

88. The therapeutic agent of claim 86, wherein each X is independently selected from alanine, arginine, asparagine, aspartic acid, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tryptophan, tyrosine and valine residues.

89. The therapeutic agent of any one of claims 76-88, wherein the therapeutic agent has a molecular weight of less than about 70 kDa.

90. The therapeutic agent of any one of claims 76-89, wherein the ELP component has a Tt greater than 37°C.

91. The therapeutic agent of any one of claims 76-90, wherein the therapeutic agent is a genetically encoded fusion protein.

92. A polynucleotide comprising a nucleotide sequence encoding the therapeutic agent of claim 91.

93. The polynucleotide of claim 92, further comprising an expression control element operably linked to said nucleotide sequence.

94. The polynucleotide of claim 93, wherein said expression control element comprises a promoter.

95. A vector comprising the polynucleotide of any one of claims 92-94.

96. An isolated host cell containing the vector of claim 95 or the polynucleotide of any one of claims 92-95.

97. A pharmaceutical composition comprising the therapeutic agent of any one of claims 76-91 , and a pharmaceutically acceptable carrier and/or excipient.

98. A method of preventing or treating a subject for a biological condition, disorder, or disease, comprising administering an effective amount of the therapeutic agent of any one of claims 76-91 or the pharmaceutical composition of claim 97 to a subject in need thereof.

99. The method of claim 98, wherein the subject has a hemorrhage.

100. The method of claim 98 or 99, wherein the subject has hemophilia.

Description:

THERAPEUTIC AGENTS COMPRISING ELASTIN-LIKE PEPTIDES

PRIORITY

[01] This application claims priority to US Provisional Application No. 61/076,221 , filed June 27, 2008, which is hereby incorporated by reference in its entirety.

FIELD OF THE INVENTION

[02] This application discloses therapeutic agents comprising elastin-like-peptides, and is related to PCT/US2007/077767 (published as WO 2008/030968 on March 13, 2008) having an International Filing Date of September 6, 2007. This application is also related to PCT/US2006/048572 (published as WO 2007/073486 on June 28, 2007) having an International Filing Date of December 20, 2006. WO 2008/030968 and WO 2007/073486 are each hereby incorporated by reference in their entireties.

DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY

[03] The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: PHAS_010_01 US_SeqList_ST25.txt, date recorded: June 26, 2009, file size 50 kb).

BACKGROUND OF THE INVENTION

[04] Therapeutic proteins or peptides in their native state or when recombinantly produced can be labile molecules exhibiting, inter alia, short periods of serum stability, serum half-life (i.e., circulatory half-life), or limited persistance in the body. Such molecules can also be extremely labile when formulated, such as when formulated in aqueous solutions.

[05] In some instances, polyethylene glycol (PEG) conjugated to a proteinaceous molecule results in a longer-acting, sustained activity of the molecule. PEG attachment, however, can often substantially reduce or even destroy the protein's therapeutic activity. Therapeutic proteins and/or peptides have also been stabilized by fusion to certain proteins that are capable of extending serum half-life. For example, in some instances, therapeutic proteins fused to albumin, transferrin, and antibody fragments exhibit extended serum half-

life when compared to the therapeutic protein in the unfused state. See US Patent No. 7,238,667 (particularly with respect to albumin conjugates), US Patent No. 7,176,278 (particularly with respect to transferrin conjugates), and US Patent No. 5,766,883, which are each hereby incorporated by reference in their entireties.

[06] There remains a need in the art for more stable, longer acting, and/or effective proteinaceous molecules.

SUMMARY OF THE INVENTION

[07] The present invention provides therapeutic agents comprising an elastin-like peptide (ELP) component and a therapeutic proteinacious component. The ELP component contains structural peptide units, which may be repeating units, structurally related to, or derived from, sequences of the elastin protein. Such ELP components provide certain therapeutic advantages to the therapeutic agent, such as comparatively better stability, solubility, bioavailability, half-life, persistance, and/or biological action of the therapeutic proteinaceous component. Such properties may be determined, for example, with respect to the therapeutic component's unfused or unconjugated counterpart. In some embodiments, the ELP components may undergo a reversible inverse phase transition, which may impart additional practical and/or therapeutic advantages. The invention further provides polynucleotides encoding the therapeutic agents of the invention, as well as methods of treatment or prophylaxis for certain biological conditions.

[08] In a first aspect, the invention provides a therapeutic agent comprising an elastin-like peptide (ELP) component and a therapeutic proteinacious component, as well as pharmaceutical compositions containing the same for delivery to a subject or patient in need. The therapeutic component may be selected from active portions of the therapeutic proteins listed in Table 1 , or functional analogs thereof. In certain embodiments, the therapeutic component is a GLP-1 receptor agonist, such as GLP-1 , exendin-4, or a functional analog thereof. Such therapeutic components are generally effective for, among other things, increasing insulin secretion from the pancreas in a glucose-dependent manner. In other embodiments, the therapeutic component is an insulin or functional analog thereof, which is generally effective for promoting glucose uptake from the blood and storage within cells. In still other embodiments, the therapeutic component is a Factor VIIλ/lla or functional analog thereof, which is generally effective for promoting coagulation by activation of Factor X or Factor IX.

[09] The ELP and therapeutic components may be covalently coupled by various means, including chemical coupling (e.g., conjugation) and recombinant fusion technology. In addition, the number of ELP or therapeutic components per molecule, and their respective positions within the molecule, may vary as needed. The therapeutic agent may further include one or more spacer or linker moieties, which in addition to providing the desired functional independence of the ELP and therapeutic components, may optionally provide for additional functionalities, such as a protease-sensitive feature to allow for proteolytic release or activation of the therapeutic component. The therapeutic agent may further include one or more targeting components such as, for example, a peptide or protein to target the therapeutic agent to a particular cell type, e.g., a cancer cell, or to a particular organ.

[010] In a second aspect, the invention provides polynucleotides, such polynucleotides comprising a nucleotide sequence encoding a therapeutic agent of the invention. For example, the nucleotide sequence encodes an ELP fusion with a functional portion of at least one therapeutic protein listed in Table 1 (or functional analog thereof). In certain embodiments, the therapeutic component is a GLP-1 receptor agonist (including GLP-1 and exendin-4), insulin, Factor Vll/Vlla, or functional analog thereof. Such polynucleotides may further comprise additional control element(s) operably linked to the nucleotide sequence, such as promoter elements and/or other transcription or expression- related signals. The polynucleotide may be inserted into various vectors, which may be useful for production of the therapeutic agent in host cells, including, for example, bacterial and eukaryotic host cells.

[011] In a third aspect, the invention provides a method for treating or preventing a disease, disorder, or condition in a subject, such as in a mammalian patient, including a human patient. The method comprises administering an effective amount of the therapeutic agent of the invention (or pharmaceutical composition containing the same) to a subject or patient in need thereof. For example, the patient may be in need of an agent having a biological activity or preferred indication listed in Table 1. In certain embodiments employing a GLP-1 receptor agonist / ELP compound or employing an insulin/ELP compound, the invention provides a method for treating one or more disorders including type 1 or type 2 diabetes, hyperglycemia, and impaired glucose tolerance. In certain other embodiments employing Factor Vll/Vlla / ELP compound, the invention provides a method for treating one or more disorders including hemophilia, post-surgical bleeding, anticoagulation-induced bleeding, thrombocytopenia, factor VII deficiency, factor Xl deficiency, and intracranial hemorrhage.

[012] Various other aspects, features and embodiments of the invention will be more fully apparent from the following disclosure and appended claims.

BRIEF DESCRIPTION OF THE FIGURES

[013] Figure 1 depicts plasmid pET24d-ELP1-90, encoding an ELP component with a 10 unit VPGXG (SEQ ID NO: 3) repeat motif, where guest position X is V, G, and A in the ratio of 5:3:2. This motif is repeated eight times with a final C-terminal 10-unit repeat where X is V, G, A, and W in the ratio 4:3:2:1. This ELP component is represented generally as [(VPGXG)io] 9 .

[014] Figure 2A depicts plasmid pET24d-Ex-4 ELP1-90 encoding an ELP component with VPGXG (SEQ ID NO: 3) repeat motif (as in Fig. 1) cloned in frame with an N-terminal exendin-4 component. Figure 2B depicts the nucleotide and amino acid sequence of the exendin-4/ELP fusion (SEQ ID NOS: 23 and 24). Primer sequences are indicated (SEQ ID NOS:35-40).

[015] Figure 3A depicts the nucleotide and amino acid sequence of an exendin-4 construct having an N-terminal Tev (Tobacco Etch Virus cysteine protease) cleavage site (SEQ ID NOS: 25 and 26). Primer sequences are indicated (SEQ ID NOS:38, 41 , 42). Figure 3B also depicts the nucleotide and amino acid sequence of an exendin-4 construct having an N-terminal Tev cleavage site, but with an additional sequence N-terminal to the Tev cleavage site to provide a better target for the protease (SEQ ID NOS: 27 and 28). Primer sequences are indicated (SEQ ID NOS:38, 43,44).

[016] Figure 4A depicts the nucleotide and amino acid sequence of an exendin-

4/ELP fusion as in Figures 1-3, but with a DsbA leader sequence to direct secretion into the periplasmic space (SEQ ID NOS: 29 and 30). Primer sequences are indicated (SEQ ID NOS:38, 45, 46). Figure 4B depicts plasmid pET24d-DsbA-Ex-4 ELP1-90 encoding the fusion of Fig. 4A.

[017] Figure 5A depicts pPB0868, which encodes GLP-1(A8G,7-37)ELP1-90.

Figure 5B depicts the nucleotide and amino acid sequence of the encoded fusion protein (SEQ ID NOS: 53 and 54, respectively).

[018] Figure 6A depicts pPB1022, which encodes GLP-1(A8G,7-37)ELP1-120.

Figure 6B depicts the nucleotide and amino acid sequence of the encoded fusion protein (SEQ ID NOS: 55 and 56, respectively).

[019] Figure 7A depicts pPB0788, which encodes Factor VII-ELP1-90. Figure 7B depicts the nucleotide and amino acid sequence of the encoded fusion protein (SEQ ID NOS: 57 and 58, respectively).

[020] Figure 8A depicts the nucleotide and amino acid sequence of an insulin (B,

C, and A chains) having the ELP component cloned in frame (SEQ ID NOS: 31 and 32). Primer sequences are indicated (SEQ ID NOS: 47 and 48). Figure 8B depicts plasmid pET24d lnsulin-ELP1-90 expressing the insulin/ELP fusion of Fig. 8A.

[021] Figure 9 is a Western blot for FVII-ELP1-90 from transient transfection of

Freestyle HEK293, detected with mouse anti-human FVII monoclonal antibody. Lanes are:

(1) culture media; (2) FVII ELP1-90 after purification by phase transition; and FVII control.

[022] Figure 10 is an SDS-PAGE showing recombinant production of an Exendin-

4/ELP4-60 fusion. Lanes are: (M) Protein markers; (1 ) Exendin-4 ELP4-60 from total lysate;

(2) Exendin-4 ELP4-60 from insoluble lysate; (3) Exendin-4 ELP4-60 from soluble lysate; (4) Exendin-4 ELP4-60 from 1st transition (equal volume); (5) Exendin-4 ELP4-60 from 2nd transition (concentrated); (6) Exendin-4 ELP4-60 from 3rd transition (concentrated).

[023] Figure 11 shows the activation of Factor X by FactorVlla-ELP1-90, and by

Factor Vila as a comparison. As shown, FactorVI Ia-ELP retains full activity.

[024] Figure 12 shows that Factor Vlla-ELP1-90 has a long PK when administered by i.v. in rats. FactorVI Ia has a Ty 2 of about 690 min. as compared to about 45-60 min. for Factor Vila.

[025] Figure 13 shows the high in vitro activity of GLP1-ELP and Exendin-4-ELP, when compared to the activity of Exendin peptide.

[026] Figure 14 shows that GLP1-ELP has a Ty 2 of about 12.9 hours when administered by i.v. to rats, and a T 1/2 of about 8.6 hours when administered subcutaneously (SQ).

[027] Figure 15 shows that GLP-1 ELP has a long half-life in rabbits of about 20 hours when administered i.v., and about 24 hours when administered sub-cutaneously.

[028] Figure 16 shows sustained glycemic control in diabetic mice with GLP-1 -ELP.

DETAILED DESCRIPTION OF THE INVENTION

[029] The present invention provides therapeutic agents comprising an elastin-like peptide (ELP) ("the ELP component") and a therapeutic component. The therapeutic component may be selected from Table 1 (e.g., selected from a Therapeutic Protein, or

functional portion or functional analog thereof, listed in Table 1). In certain embodiments, the therapeutic component is a GLP-1 receptor agonist, such as GLP-1 or exendin-4, or may be insulin, Factor Vll/Vlla, or functional analog thereof. The ELP component contains structural units related to, or derived from, sequences of the elastin protein, which provides certain therapeutic advantages, such as comparatively better persistence, stability, solubility, bioavailability, half-life, and/or biological action of the therapeutic component. Such properties may be determined with respect to, for example, an unfused or unconjugated counterpart of the therapeutic component. The invention further provides polynucleotides encoding the therapeutic agents of the invention, as well as methods of treatment or prophylaxis for certain biological conditions, including the preferred indications listed in Table 1 , and including diabetes (e.g., Type I and Type II), hyperglycemia, bleeding, hemophilia, and hemorrhage, among others.

[030] For ease of reference in the ensuing discussion, set out below are definitions of some terms appearing in the discussion.

[031] As used herein, the term "therapeutic agent" or "therapeutic component" refers to an agent or component capable of inducing a biological effect in vivo and/or in vitro. The biological effect may be useful for treating and/or preventing a condition, disorder, or disease in a subject or patient.

[032] As used herein, the term "coupled" means that the specified components are either directly covalently bonded to one another (e.g., via chemical conjugation or recombinant fusion technology), or indirectly covalently joined to one another (e.g., via chemcial conjugation or recombinant fusion technology) through an intervening moiety or moieties, such as a bridge, spacer, or linker.

[033] As used herein, "half-life" (which generally refers to in vivo half-life or circulatory half-life) is the period of time that is required for a 50% diminution of bioactivity of the active agent to occur. Such term is to be contrasted with "persistence," which is the overall temporal duration of the active agent in the body, and "rate of clearance" as being a dynamically changing variable that may or may not be correlative with the numerical values of half-life and persistence.

[034] The term "functional analog" refers to a protein that is an active analog (e.g., either chemical or protein analog), derivative, fragment, truncation isoform or the like of a native protein. For example, the functional analog may be a functional analog of a therapeutic protein listed in Table 1 , or may be a functional analog of a GLP-1 receptor agonist (e.g., GLP-1 , exendin), insulin, or Factor Vll/Vlla. A polypeptide is active when it retains some or all of the biological activity of the corresponding native polypeptide, as

determined in vivo or in one or more indicative in vitro assays. Exemplary activity assays for certain therapeutic proteins, which are determinative of activity, are listed Table 1. Further, such biological activities and assays for GLP-1 receptor agonists, insulin, and Factor Vll/Vlla, which are determinative of whether a given molecule is a "functional analog," are described in detail elsewhere herein.

[035] As used herein, the term "native," as used in reference to an amino acid sequence, indicates that the amino acid sequence is found in a naturally-occurring protein.

[036] As used herein, the term "spacer" refers to any moiety, peptide or other chemical entity, that may be interposed between the ELP component and the therapeutic component. For example, the spacer may be a divalent group that is covalently bonded at one terminus to the ELP component, and covalently bonded at the other terminus to the therapeutic component. The therapeutic agents may therefore be open to the inclusion of additional chemical structure that does not preclude the efficacy of the agent for its intended purpose. The spacer may, for example, be a protease-sensitive spacer moiety that is provided to control the pharmacokinetics of the agent, or the spacer may be a protease- resistant moiety.

[037] The therapeutic component and the ELP component may be coupled with one another in any suitable covalent manner, including chemical coupling and recombinant technology, such that the therapeutic agent is efficacious for its intended purpose, and such that the presence of the ELP component enhances the therapeutic component in some functional, therapeutic or physiological aspect. For example, the ELP-coupled therapeutic component may be enhanced in, e.g., its bioavailability, bio-unavailability, therapeutically effective dose, biological action, formulation compatibility, resistance to proteolysis or other degradative modality, solubility, half-life or other measure of persistence in the body subsequent to administration, rate of clearance from the body subsequent to administration, etc. Such enhancement may be determined, for example, in relation to a corresponding unconjugated or unfused counterpart therapeutic (e.g., determined relative to native GLP-1, exendin, insulin, or Factor Vll/Vlla, or a therapeutic protein listed in Table 1 ).

[038] In some embodiments, the therapeutic agent of the invention circulates or exists in the body in a soluble form, and escapes filtration by the kidney thereby persisting in the body in an active form. In some embodiments, the therapeutic agents of the invention have a molecular weight of less than the generally recognized cut-off for filtration through the kidney, such as less than about 60 kD, or in some embodiments less than about 55, 50, 45, 40, 30, or 20 kDa, and persist in the body by at least 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-

fold, or 100-fold longer than an uncoupled (e.g., unfused or unconjugated) therapeutic counterpart.

[039] The number of ELP and/or therapeutic components per molecule, and their respective positions within the molecule, may vary among embodiments of the invention. For example, in embodiments where the agent is a recombinant fusion, at least one ELP component may be placed at one or both of the N-terminus and the C-terminus. Where the ELP component is at both the N-terminus and C-terminus of the fusion, the ELP components will flank the therapeutic component. Alternatively, the therapeutic component may be positioned at either or both of the N-terminus and C-terminus. Where the therapeutic component is at both the N-terminus and C-terminus, the therapeutic component will flank the ELP component. In a further embodiment, different therapeutic components are positioned at the N-terminus and C-terminus of the molecule. As discussed in detail herein, in certain embodiments, such therapeutic component(s) may be released by proteolysis of a spacer moiety separating the ELP and therapeutic components. In certain embodiments, the therapeutic component may be inactive in the fused state, and becoming active upon proteolytic release from the ELP component(s). Alternatively, the therapeutic component remains active in the fused state, making proteolytic processing of the therapeutic agent unnecessary for biological activity.

[040] When prepared as recombinant fusions, the therapeutic agent can be prepared by known recombinant expression techniques. For example, to recombinant^ produce the therapeutic agent, a nucleic acid sequence encoding the chimeric gene is operatively linked to a suitable promoter sequence such that the nucleic acid sequence encoding such fusion protein will be transcribed and/or translated into the desired fusion protein in the host cells. Preferred promoters are those useful for expression in E. coli, such as the T7 promoter. Any commonly used expression system may be used, including eukaryotic or prokaryotic systems. Specific examples include yeast (e.g., Saccharomyces spp., Pichia spp.), baculovirus, mammalian, and bacterial systems, such as E. coli, and Caulobacter.

[041] The various aspects and embodiments of the invention are described in greater detail in the following sections.

Elastin-Like Peptide (ELP) Component

[042] The therapeutic agent of the invention may comprise one or more ELP components. The ELP components comprise or consist of structural peptide units or

sequences that are related to, or derived from, the elastin protein. Such sequences are useful for improving the properties of therapeutic proteins, such as those listed in Table 1 , as well as GLP-1 receptor agonists (e.g., GLP-1 or exendin-4), insulin, and Factor Vll/Vlla in one or more of bioavailability, therapeutically effective dose, biological action, formulation compatibility, resistance to proteolysis, solubility, half-life or other measure of persistence in the body subsequent to administration, and/or rate of clearance from the body.

[043] The- ELP component is constructed from structural units of from three to about twenty amino acids, or in some embodiments, from four to ten amino acids, such as five or six amino acids. The length of the individual structural units, in a particular ELP component, may vary or may be uniform. In certain embodiments, the ELP component is constructed of a polytetra-, polypenta-, polyhexa-, polyhepta-, polyocta, and polynonapeptide motif of repeating structural units. Exemplary structural units include units defined by SEQ ID NOS: 1-12 (below), which may be employed as repeating structural units, including tandem- repeating units, or may be employed in some combination, to create an ELP effective for improving the properties of the therapeutic component. Thus, the ELP component may comprise or consist essentially of structural unit(s) selected from SEQ ID NOS: 1-12, as defined below.

[044] The ELP component, comprising such structural units, may be of varying sizes. For example, the ELP component may comprise or consist essentially of from about 10 to about 500 structural units, or in certain embodiments about 15 to about 150 structural units, or in certain embodiments from about 20 to about 100 structural units, or from about 50 to about 90 structural units, including one or a combination of units defined by SEQ ID NOS: 1-12. Thus, the ELP component may have a length of from about 50 to about 2000 amino acid residues, or from about 100 to about 600 amino acid residues, or from about 200 to about 500 amino acid residues, or from about 200 to about 400 amino acid residues.

[045] In some embodiments, the ELP component, or in some cases the therapeutic agent, has a size of less than about 65 kDa, or less than about 60 kDa, or less than about 55 kDa, or less than about 50 kDa, or less than about 40 kDa, or less than about 30 or 25 kDa. Three major blood proteins, Human Serum Albumin (HSA), Transferrin (Tf) and IgG, or the Fc portion of IgGs in their glycosylated form, have been exploited to extend the half-lives of proteins and peptides for improved therapeutic use. These molecules are 585, 679 and 480 amino acids in length giving molecular weights of about 66, 77, and -75 kDa (including glycosylates), respectively. They are each globular and relatively compact. The half life of these molecules is determined by a number of factors, including charge distribution, rescue of molecules by the neonatal Fc receptor (FcRn) (HSA and Fc) or cycling of Tf through the Tf

receptor (TfR), and their size which prevents filtering through the kidney glomerulus. HSA is slightly below the generally regarded cut-off for filtration through the kidney (~70 kDa) but its charge distribution helps prevent this. It would be anticipated that, in order to achieve half- life extension of the same order as that achieved with HSA, Tf and Fc, a protein of at least this molecular weight range would be required or desirable, i.e. having over 550 amino acids and being over 65 kDa. However, an ELP with a small number of amino acids relative to HSA 1 Tf and Fc (e.g., in the range of about 300 to 400) and around 30 to 40 kDa may have a half life that matches and/or exceeds that of HSA, Tf, and Fc.

[046] In some embodiments, the ELP component in the untransitioned state may have an extended, relatively unstructured and non-globular form, and thus such molecules may have a large expanded structure in comparison to HSA, Tf and Fc, so as to escape kidney filtration. In such embodiments, the therapeutic agents of the invention have a molecular weight of less than the generally recognized cut-off for filtration through the kidney, such as less than about 60 kD, or in some embodiments less than about 55, 50, 45, 40, 30, or 25 kDa, and persist in the body by at least 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20- fold, or 100-fold longer than an uncoupled (e.g., unfused or unconjugated) therapeutic counterpart.

[047] In certain embodiments, the ELP component undergoes a reversible inverse phase transition. That is, the ELP components are structurally disordered and highly soluble in water below a transition temperature (Tt), but exhibit a sharp (2-3°C range) disorder-to- order phase transition when the temperature is raised above the Tt 1 leading to desolvation and aggregation of the ELP components. For example, the ELP forms insoluble polymers, when reaching sufficient size, which can be readily removed and isolated from solution by centrifugation. Such phase transition is reversible, and isolated insoluble ELPs can be completely resolubilized in buffer solution when the temperature is returned below the Tt of the ELPs. Thus, the therapeutic agents of the invention can, in some embodiments, be separated from other contaminating proteins to high purity using inverse transition cycling procedures, e.g., utilizing the temperature-dependent solubility of the therapeutic agent, or salt addition to the medium. Successive inverse phase transition cycles can be used to obtain a high degree of purity. In addition to temperature and ionic strength, other environmental variables useful for modulating the inverse transition of the therapeutic agents include pH, the addition of inorganic and organic solutes and solvents, side-chain ionization or chemical modification, and pressure.

[048] In certain embodiments, the ELP component does not undergo a reversible inverse phase transition, or does not undergo such a transition at a biologically relevant Tt,

and thus the improvements in the biological and/or physiological properties of the molecule (as described elsewhere herein), may be entirely or substantially independent of any phase transition properties. Nevertheless, such phase transition properties may impart additional practical advantages, for example, in relation to the recovery and purification of such molecules.

[049] In certain embodiments, the ELP component(s) may be formed of structural units, including but not limited to:

(a) the tetrapeptide Val-Pro-Gly-Gly, or VPGG (SEQ ID NO: 1 );

(b) the tetrapeptide Ile-Pro-Gly-Gly, or IPGG (SEQ ID NO: 2);

(C) the pentapeptide Val-Pro-Gly-X-Gly (SEQ ID NO: 3), or VPGXG, where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;

(d) the pentapeptide Ala-Val-Gly-Val-Pro, or AVGVP (SEQ ID NO: 4);

(e) the pentapeptide Ile-Pro-Gly-X-Gly, or IPGXG (SEQ ID NO: 5), where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;

(e) the pentapeptide Ile-Pro-Gly-Val-Gly, or IPGVG (SEQ ID NO: 6);

(f) the pentapeptide Leu-Pro-Gly-X-Gly, or LPGXG (SEQ ID NO: 7), where X is any natural or non-natural amino acid residue, and where X optionally varies among polymeric or oligomeric repeats;

(g) the pentapeptide Leu-Pro-Gly-Val-Gly, or LPGVG (SEQ ID NO: 8);

(h) the hexapeptide Val-Ala-Pro-Gly-Val-Gly, or VAPGVG (SEQ ID NO: 9);

(I) the octapeptide Gly-Val-Gly-Val-Pro-Gly-Val-Gly, or GVGVPGVG (SEQ ID NO: 10);

(J) the nonapeptide Val-Pro-Gly-Phe-Gly-Val-Gly-Ala-Gly, or VPGFGVGAG (SEQ ID NO: 11); and

(K) the nonapeptides Val-Pro-Gly-Val-Gly-Val-Pro-Gly-Gly, or VPGVGVPGG (SEQ ID NO: 12).

Such structural units defined by SEQ ID NOS:1-12 may form structural repeat units, or may be used in combination to form an ELP component in accordance with the invention. In some embodiments, the ELP component is formed entirely (or almost entirely) of one or a

I l

combination of (e.g., 2, 3 or 4) structural units selected from SEQ ID NOS: 1-12. In other embodiments, at least 75%, or at least 80%, or at least 90% of the ELP component is formed from one or a combination of structural units selected from SEQ ID NOS: 1-12, and which may be present as repeating units.

[050] In certain embodiments, the ELP component(s) contain repeat units, including tandem repeating units, of the pentapeptide Val-Pro-Gly-X-Gly (SEQ ID NO:3), where X is as defined above, and where the percentage of Val-Pro-Gly-X-Gly (SEQ ID NO:3) pentapeptide units taken with respect to the entire ELP component (which may comprise structural units other than VPGXG (SEQ ID NO:3)) is greater than about 75%, or greater than about 85%, or greater than about 95% of the ELP component. The ELP component may contain motifs having a 5 to 15-unit repeat (e.g. about 10-unit repeat) of the pentapeptide of SEQ ID NO: 3, with the guest residue X varying among at least 2 or at least 3 of the units. The guest residues may be independently selected, such as from the amino acids V, I, L, A, G, and W (and may be selected so as to retain a desired inverse phase transition property). The repeat motif itself may be repeated, for example, from about 5 to about 12 times, such as about 8 to 10 times, to create an exemplary ELP component. The ELP component as described in this paragraph may of course be constructed from any one of the structural units defined by SEQ ID NOS: 1-12, or a combination thereof.

[051] In some embodiments, the ELP component may include a β-turn structure.

Exemplary peptide sequences suitable for creating a β-turn structure are described in International Patent Application PCT/US96/05186, which is hereby incorporated by reference in its entirety. For example, the fourth residue (X) in the elastin pentapeptide sequence, VPGXG (SEQ ID NO: 3), can be altered without eliminating the formation of a β- turn. Alternatively, the ELP component may lack a β-turn, or otherwise have a different conformation and/or folding character.

[052] In certain embodiments, the ELP components include polymeric or oligomeric repeats of the pentapeptide VPGXG (SEQ ID NO: 3), where the guest residue X is any amino acid. X may be a naturally occurring or non-naturally occurring amino acid. In some embodiments, X is selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tryptophan, tyrosine and valine. In some embodiments, X is a natural amino acid other than proline or cysteine.

[053] The guest residue X (e.g., with respect to SEQ ID NO: 3, or other ELP structural unit) may be a non-classical (non-genetically encoded) amino acid. Examples of non-classical amino acids include: D- isomers of the common amino acids, 2,4-

diaminobutyric acid, α-amino isobutyric acid, A- aminobutyric acid, Abu, 2-amino butyric acid, Y-Abu, ε-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, Cα-methyl amino acids, Nα-methyl amino acids, and amino acid analogs in general.

[054] Selection of X is independent in each ELP structural unit (e.g., for each structural unit defined herein having a guest residue X). For example, X may be independently selected for each structural unit as an amino acid having a positively charged side chain, an amino acid having a negatively charged side chain, or an amino acid having a neutral side chain, including in some embodiments, a hydrophobic side chain.

[055] In still other embodiments, the ELP component(s) may include polymeric or oligomeric repeats of the pentapeptides VPGXG (SEQ ID NO:3), IPGXG (SEQ ID NO:5) or LPGXG (SEQ ID NO:7), or a combination thereof, where X is as defined above.

[056] In each embodiment, the structural units, or in some cases polymeric or oligomeric repeats, of the ELP sequences may be separated by one or more amino acid residues that do not eliminate the overall effect of the molecule, that is, in imparting certain improvements to the therapeutic component as described. In certain embodiments, such one or more amino acids also do not eliminate or substantially affect the phase transition properties of the ELP component (relative to the deletion of such one or more amino acids).

[057] In each repeat, X is independently selected. The structure of the resulting

ELP components may be described using the notation ELPk [X|Yj-n], where k designates a particular ELP repeat unit, the bracketed capital letters are single letter amino acid codes and their corresponding subscripts designate the relative ratio of each guest residue X in the structural units (where applicable), and n describes the total length of the ELP in number of the structural repeats. For example, ELP1 [V 5 A 2 G 3 -IO] designates an ELP component containing 10 repeating units of the pentapeptide VPGXG (SEQ ID NO:3), where X is valine, alanine, and glycine at a relative ratio of 5:2:3; ELP1 [K 1 V 2 F 1 ^] designates an ELP component containing 4 repeating units of the pentapeptide VPGXG (SEQ ID NO:3), where X is lysine, valine, and phenylalanine at a relative ratio of 1 :2:1 ; ELP1 [K 1 V 7 F 1 -G] designates a polypeptide containing 9 repeating units of the pentapeptide VPGXG (SEQ ID NO:3), where X is lysine, valine, and phenylalanine at a relative ratio of 1 :7:1 ; ELP1 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide VPGXG (SEQ ID NO:3), where X is exclusively valine; ELP1 [V-20] designates a polypeptide containing 20 repeating units of the pentapeptide VPGXG (SEQ ID NO:3), where X is exclusively valine;

ELP2 [5] designates a polypeptide containing 5 repeating units of the pentapeptide AVGVP (SEQ ID NO:4); ELP3 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide IPGXG (SEQ ID NO:5), where X is exclusively valine; ELP4 [V-5] designates a polypeptide containing 5 repeating units of the pentapeptide LPGXG (SEQ ID NO:7), where X is exclusively valine. Such ELP components as described in this paragraph may be used in connection with the present invention to increase the therapeutic properties of the therapeutic component.

[058] Further, the Tt is a function of the hydrophobicity of the guest residue. Thus, by varying the identity of the guest residue(s) and their mole fraction(s), ELPs can be synthesized that exhibit an inverse transition over a 0-100 0 C range. Thus, the Tt at a given ELP length may be decreased by incorporating a larger fraction of hydrophobic guest residues in the ELP sequence. Examples of suitable hydrophobic guest residues include valine, leucine, isoleucine, phenyalanine, tryptophan and methionine. Tyrosine, which is moderately hydrophobic, may also be used. Conversely, the Tt may be increased by incorporating residues, such as those selected from the group consisting of: glutamic acid, cysteine, lysine, aspartate, alanine, asparagine, serine, threonine, glycine, arginine, and glutamine; preferably selected from alanine, serine, threonine and glutamic acid.

[059] The ELP component in some embodiments is selected or designed to provide a Tt ranging from about 10 to about 8O 0 C, such as from about 35 to about 6O 0 C, or from about 38 to about 45°C. In some embodiments, the Tt is greater than about 40 0 C or greater than about 42 0 C, or greater than about 45 0 C, or greater than about 50 0 C. The transition temperature, in some embodiments, is above the body temperature of the subject or patient (e.g., >37°C) thereby remaining soluble in vivo, or in other embodiments, the Tt is below the body temperature (e.g., < 37 0 C) to provide alternative advantages, such as in vivo formation of a drug depot for sustained release of the therapeutic agent.

[060] The Tt of the ELP component can be modified by varying ELP chain length, as the Tt generally increases with decreasing MW. For polypeptides having a molecular weight > 100,000, the hydrophobicity scale developed by Urry et al. (PCT/US96/05186, which is hereby incorporated by reference in its entirety) is preferred for predicting the approximate Tt of a specific ELP sequence. However, in some embodiments, ELP component length can be kept relatively small, while maintaining a target Tt, by incorporating a larger fraction of hydrophobic guest residues (e.g., amino acid residues having hydrophobic side chains) in the ELP sequence. For polypeptides having a molecular weight <100,000, the Tt may be predicted or determined by the following quadratic function: Tt = M 0

+ M 1 X + M 2 X 2 where X is the MW of the fusion protein, and M 0 = 116.21 ; M 1 = -1.7499; M 2 = 0.010349.

[061] While the Tt of the ELP component, and therefore of the ELP component coupled to a therapeutic component, is affected by the identity and hydrophobicity of the guest residue, X, additional properties of the molecule may also be affected. Such properties include, but are not limited to solubility, bioavailability, persistence, and half-life of the molecule.

[062] As described in PCT/US2007/077767 (published as WO 2008/030968), which is hereby incorporated by reference in its entrety, the ELP-coupled therapeutic component can retain the therapeutic component's biological activity. Additionally, ELPs themselves can exhibit long half-lives. Therefore, ELP components in accordance with the present invention substantially increase (e.g. by greater than 10%, 20%, 30%, 50%, 100%, 200%, 500% or more, in specific embodiments) the half-life of the therapeutic component when conjugated thereto. Such half-life (or in some embodiments persistance or rate of clearance) is determined in comparison to the half-life of the free (unconjugated or unfused) form of the therapeutic component. Furthermore, ELPs may target high blood content organs, when administered in vivo, and thus, can partition in the body, to provide a predetermined desired corporeal distribution among various organs or regions of the body, or a desired selectivity or targeting of a therapeutic agent. In sum, the therapeutic agents contemplated by the invention are administered or generated in vivo as active compositions having extended half-lives (e.g., circulatory half-life), among other potential benefits described herein.

[063] The invention thus provides various agents for therapeutic {in vivo) application, where the therapeutic component is biologically active. Such therapeutic components include those listed in Table 1 (e.g., full length or functional portions or functional analogs thereof), as well as GLP-1 receptor agonists such as GLP-1 or exendin-4, insulin, or Factor Vll/Vlla, and functional analogs thereof. The structure and activity of such therapeutic components are described in detail below. In some forms of the therapeutic agent, the coupling of the therapeutic component to the ELP component is effected by direct covalent bonding or indirect (through appropriate spacer groups) bonding (as described elsewhere herein). Further, the therapeutic component(s) and the ELP component(s) can be structurally arranged in any suitable manner involving such direct or indirect covalent bonding, relative to one another.

Glucaqon-Like Peptide (GLPV1 Receptor Agonists

[064] In certain embodiments of the invention, the therapeutic agent comprises an

ELP component fused or conjugated to a GLP-1 receptor agonist, such as GLP-1 , exendin- 4, or functional analogs thereof.

[065] Human GLP-1 is a 37 amino acid residue peptide originating from preproglucagon which is synthesized in the L-cells in the distal ileum, in the pancreas, and in the brain. Processing of preproglucagon to give GLP-1 (7-36)amιde, GLP-1 (7-37) and GLP- 2 occurs mainly in the L-cells. A simple system is used to describe fragments and analogs of this peptide. For example, GIy 8 -GLP-1 (7-37) designates a fragment of GLP-1 formally derived from GLP-1 by deleting the amino acid residues Nos. 1 to 6 and substituting the naturally occurring amino acid residue in position 8 (Ala) by GIy. Similarly, Lys 34 (N E - tetradecanoyl)-GLP-1(7-37) designates GLP-1 (7-37) wherein the ε-amino group of the Lys residue in position 34 has been tetradecanoylated. Where reference in this text is made to C-terminally extended GLP-1 analogues, the amino acid residue in position 38 is Arg unless otherwise indicated, the optional amino acid residue in position 39 is also Arg unless otherwise indicated and the optional amino acid residue in position 40 is Asp unless otherwise indicated. Also, if a C-terminally extended analogue extends to position 41 , 42, 43, 44 or 45, the amino acid sequence of this extension is as in the corresponding sequence in human preproglucagon unless otherwise indicated.

[066] The parent peptide of GLP-1 , proglucagon (PG), has several cleavage sites that produce various peptide products dependent on the tissue of origin including glucagon (PG[32-62]) and GLP-1 [7-36]NH 2 (PG[72-107]) in the pancreas, and GLP-1 [7-37] (PG[78- 108]) and GLP-1 [7-36]NH 2 (PG [78-107]) in the L cells of the intestine where GLP-1 [7- 36]NH 2 (78-107 PG) is the major product. The GLP-1 component in accordance with the invention may be any biologically active product or deivative of proglocagon, or functional analog thereof, including: GLP-1 (1-35), GLP-1 (1-36), GLP-1 (1-36)amιde, GLP-1 (1-37), GLP-1 (1-38), GLP-1 (1-39), GLP-1 (1^0), GLP-1 (1 -41), GLP-1 (7-35), GLP-1 (7- 36), GLP-1 (7-36)amιde, GLP-1 (7-37), GLP-1 (7-38), GLP-1 (7-39), GLP-1 (7^0) and GLP- 1 (7- 41 ), or a analog of the foregoing. Generally, the GLP-1 component in some embodiments may be expressed as GLP-1 (A-B), where A is an integer from 1 to 7 and B is an integer from 38 to 45, optionally with one or more amino acid substitutions as defined below.

[067] As an overview, after processing in the intestinal L-cells, GLP-1 is released into the circulation, most notably in response to a meal. The plasma concentration of GLP-1 rises from a fasting level of approximately 15 pmol/L to a peak postprandial level of 40 pmol/L. For a given rise in plasma glucose concentration, the increase in plasma insulin is

I 6

approximately threefold greater when glucose is administered orally compared with intravenously (Kreymann ef al., 1987, Lancet 2(8571): 1300-4). This alimentary enhancement of insulin release, known as the incretin effect, is primarily humoral and GLP-1 is now thought to be the most potent physiological incretin in humans. GLP-1 mediates insulin production via binding to the GLP-1 receptor, known to be expressed in pancreatic β cells. In addition to the insulinotropic effect, GLP-1 suppresses glucagon secretion, delays gastric emptying (Wettergen ef al., 1993, Dig Dis Sci 38: 665-73) and may enhance peripheral glucose disposal (D'Alessio ef a/., 1994, J. Clin Invest 93: 2293-6).

[068] A combination of actions gives GLP-1 unique therapeutic advantages over other agents currently used to treat non-insulin-dependent diabetes mellitus (NIDDM). First, a single subcutaneous dose of GLP-1 can completely normalize post prandial glucose levels in patients with NIDDM (Gutniak et al., 1994, Diabetes Care 17: 1039-44). This effect may be mediated both by increased insulin release and by a reduction in glucagon secretion. Second, intravenous infusion of GLP-1 can delay postprandial gastric emptying in patients with NIDDM (Williams ef a/., 1996, J. CHn Endo Metab 81 : 327-32). Third, unlike sulphonylureas, the insulinotropic action of GLP-1 is dependent on plasma glucose concentration (HoIz ef al., 1993, Nature 361 :362-5). Thus, the loss of GLP-1 -mediated insulin release at low plasma glucose concentration protects against severe hypoglycemia.

[069] When given to healthy subjects, GLP-1 potently influences glycemic levels as well as insulin and glucagon concentrations (Orskov, 1992, Diabetologia 35:701-11 ), effects which are glucose dependent (Weir et al., 1989, Diabetes 38: 338-342). Moreover, it is also effective in patients with diabetes (Gutniak, M., 1992, N. Engl J Med 226: 1316-22), normalizing blood glucose levels in type 2 diabetic subjects and improving glycemic control in type 1 patients (Nauck et al., 1993, Diabetologia 36: 741-4, Creutzfeldt ef al., 1996, Diabetes Care 19:580-6).

[070] GLP-1 is, however, metabolically unstable, having a plasma half-life (ti /2 ) of only 1-2 minutes in vivo. Moreover, exogenously administered GLP-1 is also rapidly degraded (Deacon et al., 1995, Diabetes 44: 1126-31 ). This metabolic instability has limited the therapeutic potential of native GLP-1.

[071] GLP-1 [7-36]NH 2 has the following amino acid sequence:

HAEGTFTSDVSSYLEGQAAKEFIAWLVKGR (SEQ ID NO: 13), which may be employed as the GLP-1 component in accordance with the invention. Alternatively, the GLP-1 component may contain glycine (G) at the second position, giving, for example, the sequence HGEGTFTSDVSSYLEGQAAKEFIAWLVKGR (SEQ ID NO: 17). The GLP-1 component may be a biologically active fragment of GLP-1 , for example, as disclosed in US

2007/0041951 , which is hereby incorporated by reference in its entirety. Other fragments and modified sequences of GLP-1 are known in the art (U.S. Pat. No. 5,614,492; U.S. Pat. No. 5,545,618; European Patent Application, Publication No. EP 0658568 A1; WO 93/25579, which are hereby incorporated by reference in their entireties). Such fragments and modified sequences may be used in connection with the present invention, as well as those described below.

[072] Certain structural and functional analogs of GLP-1 have been isolated from the venom of the GiIa monster lizards (Heloderma suspectum and Heloderma horridum) and have shown clinical utility. Such molecules find use in accordance with the present invention. In particular, exendin-4 is a 39 amino acid residue peptide isolated from the venom of Heloderma suspectum and shares approximately 52% homology with human GLP- 1. Exendin-4 is a potent GLP-1 receptor agonist that stimulates insulin release, thereby lowering blood glucose levels. Exendin-4 has the following amino acid sequence: HGEGTFTSDLSKQMEEEAVRLFEWLKNGGPSSGAPPPS (SEQ ID NO: 14). A synthetic version of exendin-4 known as exenatide (marketed as Byetta®) has been approved for the treatment of Type-2 Diabetes. Although exenatide is structurally analogous to native GLP-1 , it has a longer half-life after injection.

[073] While exenatide has the ability to lower blood glucose levels on its own, it can also be combined with other medications such as metformin, a thiozolidinedione, a sulfonylureas, and/or insulin to improve glucose control. Exenatide is administered by injection subcutaneously twice per day using a pre-filled pen device. Typical human responses to exenatide include improvements in the initial rapid release of endogenous insulin, an increase in β-cell growth and replication, suppression of pancreatic glucagon release, delayed gastric emptying, and reduced appetite - all of which function to lower blood glucose. Unlike sulfonylureas and meglitinides, exenatide increases insulin synthesis and secretion in the presence of glucose only, thus lessening the risk of hypoglycemia. Despite the therapeutic utility of exenatide, it has certain undesirable traits, including the requirement of twice daily injections, gastrointestional side effects, and similar to native GLP-1 , a relatively short half-life (i.e. approximately 2 hr).

[074] Various functional analogs of GLP-1 and exendin-4 are known, and which find use in accordance with the invention. These include liraglutide (Novo Nordisk, WO98/008871 ), R1583/taspoglutide (Roche, WO00/034331 ), CJC-1131 (ConjuChem, WO00/069911), ZP-10/AVE0010 (Zealand Pharma, Sanofi-Aventis, WO01/004156), and LY548806 (EIi Lilly, WO03/018516).

[075] Liraglutide, also known as NN2211 , is a GLP-1 receptor agonist analog that has been designed for once-daily injection (Harder et al., 2004, Diabetes Care 27: 1915-21). Liraglutide has been tested in patients with type-2 diabetes in a number of studies and has been shown to be effective over a variety of durations. In one study, treatment with liraglutide improved glycemic control, improved β-cell function, and reduced endogenous glucose release in patients with type-2 diabetes after one week of treatment (Degn et al., 2004, Diabetes 53: 1187-94). In a similar study, eight weeks of 0.6-mg liraglutide therapy significantly improved glycemic control without increasing weight in subjects with type 2 diabetes compared with those on placebo (Harder et al., 2004, Diabetes Care 27: 1915-21 ).

[076] Thus, in certain embodiments, the GLP-1 receptor agonist in accordance with the invention is as described in WO98/008871 , which is hereby incorporated by reference in its entirety. The GLP-1 receptor agonist may have at least one lipophilic substituent, in addition to one, two, or more amino acid substitutions with respect to native GLP-1. For example, the lipophilic substituent may be an acyl group selected from CH 3 (CH 2 ) n CO-, wherein n is an integer from 4 to 38, such as an integer from 4 to 24. The lipophilic substituent may be an acyl group of a straight-chain or branched alkyl or fatty acid (for example, as described in WO98/008871 , which description is hereby incorporated by reference).

[077] In certain embodiments, the GLP-1 component is Arg 26 -GLP-1 (7-37), Arg 34 -

GLP-1(7-37), Lys 36 -GLP-1 (7-37), Arg 2634 Lys 36 -GLP-l (7-37), Arg 2634 Lys 38 -GLP-l (7-38), Arg 28 34 Lys 39 -GLP-1 (7-39), Arg 26|34 Lys 40 -GLP-1 (7-40), Arg 26 Lys 36 -GLP-1(7-37), Arg 34 Lys 36 - GLP-1(7-37), Arg 26 Lys 39 -GLP-1(7-39), Arg 34 Lys 40 -GLP-1 (7^0), Arg 2634 Lys 36|39 -GLP-l (7-39), Arg 26 ' 34 Lys 3M0 -GLP-1(7-40), Gly 8 Arg 26 -GLP-1(7-37); Gly 8 Arg 34 -GLP-1(7-37); Gly 8 Lys 38 -GLP- 1(7-37); GIy 8 Arg 26 34 Lys 36 -GLP-1 (7-37), Gly 8 Arg 2634 Lys 39 -GLP-1(7-39), Gly 8 Arg 26 ' 34 Lys 40 - GLP-1(7^0), Gly 8 Arg 26 Lys 36 -GLP-1(7-37), Gly 8 Arg 34 Lys 36 -GLP-1 (7-37), Gly 8 Arg 26 Lys 39 -GLP- 1(7-39); Gly 8 Arg 34 Lys 40 -GLP-1(7-40), Gly 8 Arg 28 ' 34 Lys 3639 -GLP-1(7-39) and

Gly 8 Arg 2634 Lys 3540 -GLP-1(7-40), each optionally having a lipophilic substituent. For example, the GLP-1 receptor agonist may have the sequence/structure Arg 34 Lys 26 -(N-ε-(γ- Glu(N-α-hexadecanoyl)))-GLP-l(7-37).

[078] Taspoglutide, also known as R1583 or BIM 51077, is a GLP-1 receptor agonist that has been shown to improve glycemic control and lower body weight in subjects with type 2 diabetes mellitus treated with metformin (Abstract No. A-1604, June 7, 2008, 68th American Diabetes Association Meeting, San Francisco, CA).

[079] Thus, in certain embodiments, the GLP-1 receptor agonist is as described in

WO00/034331 , which is hereby incorporated by reference in its entirety. In certain

exemplary embodiments, the GLP-1 receptor agonist has the sequence [Aib 8 35 ]hGLP-1(7- 36)NH 2 (e.g. taspoglutide), wherein Aib is alpha-aminoisobutyric acid.

[080] CJC-1131 is a GLP-1 analog that consists of a DPP-IV-resistant form of GLP-

1 joined to a reactive chemical linker group that allows GLP-1 to form a covalent and irreversible bond with serum albumin following subcutaneous injection (Kim et al., 2003, Diabetes 52: 751-9). In a 12-week, randomized, double-blind, placebo-controlled multicenter study, CJC-1131 and metformin treatment was effective in reducing fasting blood glucose levels in type 2 diabetes patients (Ratner et al., Abstract No. 10-OR, June 10-14th, 2005, 65th American Diabetes Association Meeting, San Francisco, CA).

[081] Thus, in certain embodiments, the GLP-1 receptor agonist is as described in

WO00/069911 , which is hereby incorporated by reference in its entirety. In some embodiments, the GLP-1 receptor agonist is modified with a reactive group which reacts with amino groups, hydroxyl groups or thiol groups on blood components to form a stable covalent bond. In certain embodiments, the GLP-1 receptor agonist is modified with a reactive group selected from the group consisting of succinimidyl and maleimido groups. In certain exemplary embodiments, the GLP-1 receptor agonist has the sequence/structure: D- Ala 8 Lys 37 -(2-(2-(2-maleimidopropionamido(ethoxy)ethoxy)acetamide))-GL P-1(7-37) (e.g. CJC-1 131 ).

[082] AVE0010, also known as ZP-10, is a GLP-1 receptor agonist that may be employed in connection with the invention. In a recent double-blind study, patients treated with once daily dosing of AVE0010 demonstrated significant reductions in HbAIc levels (Ratner et al., Abstract No. 433-P, 68th American Diabetes Association Meeting, San Francisco, CA.). At the conclusion of the study, the percentages of patients with HbAIc <7% ranged from 47-69% for once daily dosing compared to 32% for placebo. In addition, AVE0010 treated patients showed dose-dependent reductions in weight and post-prandial plasma glucose.

[083] Thus, in certain embodiments, the GLP-1 receptor agonist is as described in

WO01/004156, which is hereby incorporated by reference in its entirety. For example, the GLP-1 receptor agonist may have the sequence:

HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPSKKKKKK-NH2 (SEQ ID NO: 18) (e.g. AVE0010).

[084] LY548806 is a GLP-1 derivative designed to be resistant to proteolysis by dipeptidase-peptidyl IV (DPP-IV) (Jackson et al., Abstract No. 562, June 10-14th, 2005, 65th American Diabetes Association Meeting, San Francisco, CA). In an animal model of hyperglycemia, LY548806 has been shown to produce a significant lowering of blood

glucose levels during the hyperglycemic phase (Saha et al., 2006, J. Pharm. Exp. Ther. 316: 1159-64). Moreover, LY548806 was shown to produce a significant increase in insulin levels consistent with its known mechanism of action, namely stimulation of insulin release in the presence of hyperglycemia.

[085] Thus, in certain embodiments, the GLP-1 receptor agonist is as described in

WO03/018516, which is hereby incorporated by reference in its entirety. In some embodiments, the therapeutic agents of the present invention comprise GLP-1 analogs wherein the backbone for such analogs or fragments contains an amino acid other than alanine at position 8 (position 8 analogs). The backbone may also include L-histidine, D- histidine, or modified forms of histidine such as desamino-histidine, 2-amino-histidine, β- hydroxy-histidine, homohistidine, α-fluoromethyl-histidine, or α-methyl-histidine at position 7. In some embodiments, these position 8 analogs may contain one or more additional changes at positions 12, 16, 18, 19, 20, 22, 25, 27, 30, 33, and 37 compared to the corresponding amino acid of native GLP-1. In other embodiments, these position 8 analogs may contain one or more additional changes at positions 16, 18, 22, 25 and 33 compared to the corresponding amino acid of native GLP-1. In certain exemplary embodiments, the GLP- 1 receptor agonist has the sequence: HVEGTFTSDVSSYLEEQAAKEFIAWLIKGRG-OH (SEQ ID NO: 19) (e.g. LY548806).

[086] Thus, the present invention provides therapeutic agents comprising an elastin-like peptide (ELP) and a GLP-1 receptor agonist. For example, in certain embodiments, the GLP-1 receptor agonist is GLP-1 (SEQ ID NO: 13, 17, or 59) or a functional analog thereof. In other embodiments, the GLP-1 receptor agonist is exendin-4 (SEQ ID NO:14) or a functional analog thereof. Such functional analogs of GLP-1 or exendin-4 include functional fragments truncated at the C-terminus by from 1 to 10 amino acids, including by 1 , 2, 3, or up to about 5 amino acids (with respect to SEQ ID NOS: 13, 14, 17, or 59). Such functional analogs may contain from 1 to 10 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the native sequence (e.g., SEQ ID NOS 13, 14, and 59), and in each case retaining the activity of the peptide. For example, the functional analog of GLP-1 or exendin-4 may have from 1 to about 3, 4, or 5 insertions, deletions and/or substitutions (collectively) with respect to SEQ ID NOS: 13, 59 and 14, and in each case retaining the activity of the peptide. Such activity may be confirmed or assayed using any available assay, including those described herein. In these or other embodiments, the GLP-1 receptor agonist component has at least about 50%, 75%, 80%, 85%, 90%, or 95% identity with the native sequence (SEQ ID NOS: 13, 59, and 14). The determination of sequence identity between two sequences (e.g., between a native sequence and a functional analog) can be accomplished using any alignment tool, including Tatusova et al.,

Blast 2 sequences - a new tool for comparing protein and nucleotide sequences. FEMS Microbiol Lett. 174:247-250 (1999). Such functional analogs may further comprise additional chemical modifications, such as those described in this section and/or others known in the art.

[087] In certain embodiments, the GLP1-ELP fusion has a sequence exemplified herein as SEQ ID NOS: 54 and 56. When processed, the mature form of such fusion protein will begin with the His 7 of GLP.

[088] In another aspect, the present invention provides methods for the treatment or prevention of type 2 diabetes, impaired glucose tolerance, type 1 diabetes, hyperglycemia, obesity, binge eating, bulimia, hypertension, syndrome X, dyslipidemia, cognitive disorders, atheroschlerosis, non-fatty liver disease, myocardial infarction, coronary heart disease and other cardiovascular disorders. The method comprises administering the therapeutic agent comprising the elastin-like peptide (ELP) and the GLP-1 receptor agonist (as described above) to a patient in need of such treatment. In these or other embodiments, the present invention provides methods for decreasing food intake, decreasing β-cell apoptosis, increasing β-cell function and β-cell mass, and/or for restoring glucose sensitivity to β-cells. Generally, the patient may be a human or non-human animal patient (e.g., dog, cat, cow, or horse). Peferably, the patient is human.

[089] The treatment with a ELP/GLP-1 receptor agonist compound according to the present invention may also be combined with one or more pharmacologically active substances, e.g. selected from antidiabetic agents, antiobesity agents, appetite regulating agents, antihypertensive agents, agents for the treatment and/or prevention of complications resulting from or associated with diabetes and agents for the treatment and/or prevention of complications and disorders resulting from or associated with obesity. In the present context, the expression "antidiabetic agent" includes compounds for the treatment and/or prophylaxis of insulin resistance and diseases wherein insulin resistance is the pathophysiological mechanism.

[090] The ability of a GLP-1 or exendin-4 analog, or an GLP-1 receptor agonist/ELP compound, to bind the GLP-1 receptor may be determined by standard methods, for example, by receptor-binding activity screening procedures which involve providing appropriate cells that express the GLP-1 receptor on their surface, for example, insulinoma cell lines such as RINmSF cells or INS-1 cells. In addition to measuring specific binding of tracer to membrane using radioimmunoassay methods, cAMP activity or glucose dependent insulin production can also be measured. In one method, a polynucleotide encoding the GLP-1 receptor is employed to transfect cells to thereby express the GLP-1 receptor protein.

Thus, these methods may be employed for testing or confirming whether a suspected GLP receptor agonist is active. An exemplary assay is described in greater detail herein.

[091] In addition, known methods can be used to measure or predict the level biologically activity of a GLP-1 receptor agonist or GLP-1 receptor agonist/ELP in vivo (Se e.g. Siegel, et ai, 1999, Regul Pept 79(2-3): 93-102). In particular, GLP-1 receptor agonis or GLP-1 receptor agonist/ELP compounds can be assessed for their ability to induce U production of insulin in vivo using a variety of known assays for measuring GLP-1 activil For example, an ELP/GLP-1 receptor agonist compound can be introduced into a cell, sui as an immortalized β-cell, and the resulting cell can be contacted with glucose. If the o produces insulin in response to the glucose, then the modified GLP-1 is genera considered biologically active in vivo (Fehmann ef a/., 1992, Endocrinology 130: 159-16* An exemplary assay is described in greater detail herein.

[092] The ability of an GLP-1 receptor agonist/ELP compound to enhance β-c proliferation, inhibit β-cell apoptosis, and regulate islet growth may also be measured usii known assays. Pancreatic β-cell proliferation may be assessed by 3 H-tymidine or Brc incorporation assays (See e.g. Buteau et al., 2003, Diabetes 52: 124-32), wherein pancrea β-cells such as INS(832/13) cells are contacted with an ELP/ GLP-1 receptor agon compound and analyzed for increases in 3 H-thymidine or BrdU incorporation. Tl antiapoptotic activity of an ELP/GLP-1 receptor agonist compound can be measured cultured insulin-secreting cells and/or in animal models where diabetes occurs as consequence of an excessive rate of beta-cell apoptosis (See e.g. Bulotta ef a/., 2004, C Biochem Biophys 40(3 suppl): 65-78).

[093] In addition to GLP-1 , other peptides of this family, such as those derived frc processing of the pro-glucagon gene, such as GLP-2, GIP, and oxyntomodulin, could conjugated or fused to the ELP component (as described herein) to enhance the therapeu potential.

Insulin

[094] In other embodiments, the present invention provides a therapeutic age comprising an ELP component coupled to insulin (e.g., via fusion or conjugation), lnsu injections, e.g. of human insulin, can be used to treat diabetes. The insulin-making cells the body are called β-cells, and they are found in the pancreas gland. These cells clui together to form the "islets of Langerhans", named for the German medical student w described them.

[095] The synthesis of insulin begins at the translation of the insulin gene, which resides on chromosome 11. During translation, two introns are spliced out of the mRNA product, which encodes a protein of 110 amino acids in length. This primary translation product is called preproinsulin and is inactive. It contains a signal peptide of 24 amino acids in length, which is required for the protein to cross the cell membrane.

[096] Once the preproinsulin reaches the endoplasmic reticulum, a protease cleaves off the signal peptide to create proinsulin. Proinsulin consists of three domains: an amino-terminal B chain, a carboxyl-terminal A chain, and a connecting peptide in the middle known as the C-peptide. Insulin is composed of two chains of amino acids named chain A (21 amino acids - GIVEQCCASVCSLYQLENYCN) (SEQ ID NO: 15) and chain B (30 amino acids FVNQHLCGSHLVEALYLVCGERGFFYTPKA) (SEQ ID NO: 16) that are linked together by two disulfide bridges. There is a 3rd disulfide bridge within the A chain that links the 6th and 11th residues of the A chain together. In most species, the length and amino acid compositions of chains A and B are similar, and the positions of the three disulfide bonds are highly conserved. For this reason, pig insulin can replace deficient human insulin levels in diabetes patients. Today, porcine insulin has largely been replaced by the mass production of human proinsulin by bacteria (recombinant insulin).

[097] J Insulin molecules have a tendency to form dimers in solution, and in the presence of zinc ions, insulin dimers associate into hexamers. Whereas monomers of insulin readily diffuse through the blood and have a rapid effect, hexamers diffuse slowly and have a delayed onset of action. In the design of recombinant insulin, the structure of insulin can be modified in a way that reduces the tendency of the insulin molecule to form dimers and hexamers but that does not interrupt binding to the insulin receptor. In this way, a range of preparations are made, varying from short acting to long acting.

[098] Within the endoplasmic reticulum, proinsulin is exposed to several specific peptidases that remove the C-peptide and generate the mature and active form of insulin. In the Golgi apparatus, insulin and free C-peptide are packaged into secretory granules, which accumulate in the cytoplasm of the β-cells. Exocytosis of the granules is triggered by the entry of glucose into the beta cells. The secretion of insulin has a broad impact on metabolism.

[099] There are two phases of insulin release in response to a rise in glucose. The first is an immediate release of insulin. This is attributable to the release of preformed insulin, which is stored in secretory granules. After a short delay, there is a second, more prolonged release of newly synthesized insulin.

[0100] Once released, insulin is active for a only a brief time before it is degraded by enzymes, lnsulinase found in the liver and kidneys breaks down insulin circulating in the plasma, and as a result, insulin has a half-life of only about 6 minutes. This short duration of action results in rapid changes in the circulating levels of insulin.

[0101] Insulin analogs have been developed with improved therapeutic properties

(Owens et a/., 2001 , Lancet 358: 739-46; Vajo ef a/., 2001 , Endocr Rev 22: 706-17), and such analogs may be employed in connection with the present invention. Various strategies, including elongation of the COOH-terminal end of the insulin B-chain and engineering of fatty acid-acylated insulins with substantial affinity for albumin are used to generate longer-acting insulin analogs. However, in vivo treatments with available longer-acting insulin compounds still result in a high frequency of hypo- and hyperglycemic excursions and modest reduction in HbAic. Accordingly, development of a truly long-acting and stable human insulin analog still remains an important task.

[0102] Functional analogs of insulin that may be employed in accordance with the invention include rapid acting analogs such as lispro, aspart and glulisine, which are absorbed rapidly (< 30 minutes) after subcutaneous injection, peak at one hour, and have a relatively short duration of action (3 to 4 hours). In addition, two long acting insulin analogs have been developed: glargine and detemir, and which may be employed in connection with the invention. The long acting insulin analogs have an onset of action of approximately two hours and reach a plateau of biological action at 4 to 6 hours, and may last up to 24 hours.

[0103] Thus, in one embodiment, the insulin component may contain the A and/or B chain of lispro (also known as Humalog, EIi Lilly). Insulin lispro differs from human insulin by the substitution of proline with lysine at position 28 and the substitution of lysine with proline at position 29 of the insulin B chain. Although these modifications do not alter receptor binding, they help to block the formation of insulin dimers and hexamers, allowing for larger amounts of active monomeric insulin to be available for postprandial injections.

[0104] In another embodiment, the insulin may contain an A and/or B chain of aspart

(also known as Novolog, Novo Nordisk). Insulin aspart is designed with the single replacement of the amino acid proline by aspartic acid at position 28 of the human insulin B chain. This modification helps block the formation for insulin hexamers, creating a faster acting insulin.

[0105] In yet another embodiment, the insulin may contain an A and/or B chain of glulisine (also known as Apidra, Sanofi-Aventis). Insulin glulisine is a short acting analog created by substitution of asparagine at position 3 by lysine and lysine at position 29 by

glutamine of human insulin B chain. Insulin glulisine has more rapid onset of action and shorter duration of action compared to regular human insulin.

[0106] In another embodiment, the insulin may contain an A and/or B chain of glargine (also known as Lantus, Sanofi-Aventis). Insulin glargine differs from human insulin in that the amino acid asparagine at position 21 of the A chain is replaced by glycine and two arginines are added to the C-terminus of the B-chain. Compared with bedtime neutral protamine Hagedorn (NPH) insulin (an intermediate acting insulin), insulin glargine is associated with less nocturnal hypoglycemia in patients with type 2 diabetes.

[0107] In yet another embodiment, the insulin may contain an A and/or B chain from detemir (also known as Levemir, Novo Nordisk). Insulin detemir is a soluble (at neutral pH) long-acting insulin analog, in which the amino acid threonine at B30 is removed and a 14- carbon, myristoyl fatty acid is acetylated to the epsilon-amino group of LysB29. After subcutaneous injection, detemir dissociates, thereby exposing the free fatty acid which enables reversible binding to albumin molecules. So at steady state, the concentration of free unbound insulin is greatly reduced resulting in stable plasma glucose levels.

[0108] In some embodiments, the insulin may be a single-chain insulin analog (SIA)

(e.g. as described in 6,630,438 and WO08/019368, which are hereby incorporated by reference in their entirety). Single-chain insulin analogs encompass a group of structurally- related proteins wherein the A and B chains are covalently linked by a polypeptide linker. The polypeptide linker connects the C-terminus of the B chain to the N-terminus of the A chain. The linker may be of any length so long as the linker provides the structural conformation necessary for the SIA to have a glucose uptake and insulin receptor binding effect. In some embodiments, the linker is about 5-18 amino acids in length. In other embodiments, the linker is about 9-15 amino acids in length. In certain embodiments, the linker is about 12 amino acids long. In certain exemplary embodiments, the linker has the sequence KDDNPNLPRLVR (SEQ ID NO.: 20) or GAGSSSRRAPQT (SEQ ID NO.: 21 ). However, it should be understood that many variations of this sequence are possible such as in the length (both addition and deletion) and substitutions of amino acids without substantially compromising the effectiveness of the produced SIA in glucose .uptake and insulin receptor binding activities. For example, several different amino acid residues may be added or removed from either end without substantially decreasing the activity of the produced SIA.

[0109] An exemplary single-chain insulin analog currently in clinical development is albulin (Duttaroy et al., 2005, Diabetes 54: 251-8). Albulin can be produced in yeast or in mammalian cells. It consists of the B and A chain of human insulin (100% identity to native

human insulin) linked together by a dodecapeptide linker and fused to the NH 2 terminals of the native human serum albumin. For expression and purification of albulin, Duttaroy et al. constructed a synthetic gene construct encoding a single-chain insulin containing the B- and A- chain of mature human insulin linked together by a dodecapeptide linker using four overlapping primers and PCR amplification. The resulting PCR product was ligated in-frame between the signal peptide of human serum albumin (HSA) and the NH 2 terminus of mature HSA, contained within a pSAC35 vector for expression in yeast. In accordance with the present invention, the HSA component of abulin may be replaced with an ELP component as described herein.

[0110] Thus, in one aspect, the present invention provides therapeutic agents comprising an elastin-like peptide (ELP) and an insulin or functional analog thereof. For example, in certain embodiments, the insulin is a mammalian insulin, such as human insulin or porcine insulin. In accordance with the invention, the ELP component may be coupled (e.g., via recombinant fusion or chemical conjugation) to the insulin A chain, or B chain, or both. The insulin may comprise each of chains A, B, and C (SEQ ID NOS: 51 and 52), or may contain a processed form, containing only chains A and B. In some embodiments, chains A and B are connected by a short linking peptide, to create a single chain insulin. The insulin may be a functional analog of human insulin, including functional fragments truncated at the N-terminus and/or C-terminus (of either or both of chains A and B) by from 1 to 10 amino acids, including by 1 , 2, 3, or about 5 amino acids. Functional analogs may contain from 1 to 10 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the native sequence (e.g., SEQ ID NOS 15 and 16), and in each case retaining the activity of the peptide. For example, functional analogs may have 1 , 2, 3, 4, or 5 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the native sequence (which may contain chains A and B, or chains A, B, and C). Such activity may be confirmed or assayed using any available assay, including those described herein. In these or other embodiments, the insulin component has at least about 75%, 80%, 85%, 90%, 95%, or 98% identity with each of the native sequences for chains A and B (SEQ ID NOS:15 and 16). The determination of sequence identity between two sequences (e.g., between a native sequence and a functional analog) can be accomplished using any alignment tool, including Tatusova et al., Blast 2 sequences - a new tool for comparing protein and nucleotide sequences. FEMS Microbiol Lett. 174:247-250 (1999). The insulin component may contain additional chemical modifications known in the art.

[0111] In another aspect, the present invention provides methods for the treatment or prevention of diabetes, including type I and Il diabetes. The method comprises administering an effective amount of the therapeutic agent comprising an elastin-like peptide

(ELP) component and an insulin (or functional analog thereof) component to a patient in need thereof. Generally, the patient may be a human or non-human animal (e.g., dog, cat, cow, or horse) patient. Preferably, the patient is human.

[0112] To characterize the in vitro binding properties of an insulin analog or an ELP- containing insulin analog, competition binding assays may be performed in various cell lines that express the insulin receptor (Jehle ef a/., 1996, Diabetologia 39: 421-432). For example, competition binding assays using CHO cells overexpressing the human insulin receptor may be employed. Insulin can also bind to the IGF-1 receptor with a lower affinity than the insulin receptor. To determine the binding affinity of an ELP-containing insulin analog, a competition binding assay can be performed using 125 l-labeled IGF-1 in L6 cells.

[0113] The activities of insulin include stimulation of peripheral glucose disposal and inhibition of hepatic glucose production. The ability of an ELP-containing insulin analog to mediate these biological activities can be assayed in vitro using known methodologies. For example, the effect of an ELP-containing analog on glucose uptake in 3T3-L1 adipocytes can be measured and compared with that of insulin. Pretreatment of the cells with a biologically active analog will generally produce a dose-dependent increase in 2- deoxyglucose uptake. The ability of an ELP-containing insulin analog to regulate glucose production may be measured in any number of cells types, for example, H4lle hepatoma cells. In this assay, pretreatment with a biologically active analog will generally result in a dose-dependent inhibition of the amount of glucose released.

Factor Vl I (Vila)

[0114] In certain embodiments, the invention provides therapeutic agents conprising an ELP component coupled (e.g., via fusion or conjugation) to a Factor Vll/Vlla. Coagulation is the biological process of blood clot formation involving many different serine proteases as well as their essential cofactors and inhibitors. It is initiated by exposure of Factor VII (FVII) and Factor Vila (FVIIa) to its membrane bound cofactor, tissue factor (TF), resulting in production of Factor Xa (FXa) and more FVIIa. The process is propagated upon production of Factor IXa (FIXa) and additional FXa that, upon binding with their respective cofactors FVIIIa and FVa, form platelet bound complexes, ultimately resulting in the formation of thrombin and a fibrin clot. Thrombin also serves to further amplify coagulation by activation of cofactors such as FV and FVII and zymogens such as Factor Xl. Moreover, thrombin activates platelets leading to platelet aggregation, which is necessary for the formation of a hemostatic plug.

[0115] Factor VII circulates in the blood in a zymogen form, and is converted to its active form, Factor Vila, by either factor IXa, factor Xa, factor XIIa, or thrombin by minor proteolysis. Factor Vila is a two-chain, 50 kilodalton (kDa) plasma serine protease. The active form of the enzyme comprises a heavy chain (254 amino acid residues) containing a catalytic domain and a light chain (152 residues) containing 2 epidermal growth factor (EGF)-like domains. The mature factor Vll/Vlla that circulates in plasma is composed of 406 amino acid residues (SEQ ID NO: 33). The light and heavy chains are held together by a disulfide bond.

[0116] As noted above, Factor Vila is generated by proteolysis of a single peptide bond from its single chain zymogen, Factor VII, which is present at approximately 0.5 μg/ml in plasma. The conversion of zymogen Factor VII into the activated two-chain molecule occurs by cleavage of an internal peptide bond. In human Factor VII, the cleavage site is at Arg152-lle153 (Hagen ef a/., 1986, PNAS USA 83: 2412-6).

[0117] "Factor Vll/Vlla" as used in this application means a product consisting of either the unactivated form (factor VII) or the activated form (factor Vila) or mixtures thereof. "Factor Vll/Vlla" within the above definition includes proteins that have an amino acid sequence of native human factor Vll/Vlla. It also includes proteins with a slightly modified amino acid sequence, for instance, a modified N-terminal end including N-terminal amino acid deletions or additions so long as those proteins substantially retain the activity of factor Vila. "Factor VII" within the above definition also includes natural allelic variations that may exist and occur from one individual to another. Also, degree and location of glycosylation or other post-translation modifications may vary depending on the chosen host cells and the nature of the host cellular environment.

[0118] In the presence of calcium ions, Factor Vila binds with high affinity to TF. TF is a 263 amino acid residue glycoprotein composed of a 219 residue extracellular domain, a single transmembrane domain, and a short cytoplasmic domain (Morrissey ef a/., 1987, Ce// 50: 129-35). The TF extracellular domain is composed of two fibronectin type III domains of about 105 amino acids each. The binding of FVIIa is mediated entirely by the TF extracellular domain (Muller et a/., 1994, Biochem. 33:10864-70). Residues in the area of amino acids 16-26 and 129-147 contribute to the binding of FVIIa as well as the coagulant function of the molecule. Residues Lys20, Trp45, Asp58, Tyr94, and Phe140 make a large contribution (1 kcal/mol) to the free energy (δG) of binding to FVIIa.

[0119] TF is expressed constitutively on cells separated from plasma by the vascular endothelium. Its expression on endothelial cells and monocytes is induced by exposure to inflammatory cytokines or bacterial lipopolysaccharides (Drake et a/., 1989, J. Cell Biol. 109:

389). Upon tissue injury, the exposed extracellular domain of TF forms a high affinity, calcium dependent complex with FVII. Once bound to TF, FVII can be activated by peptide bond cleavage to yield serine protease FVIIa. The enzyme that catalyzes this step in vivo has not been elucidated, but in vitro FXa, thrombin, TF:FVIIa and FIXa can catalyze this cleavage. FVIIa has only weak activity upon its physiological substrates FX and FIX whereas the TF: FVI Ia complex rapidly activates FX and FIX.

[0120] The TF: FVI Ia complex constitutes the primary initiator of the extrinsic pathway of blood coagulation. The complex initiates the extrinsic pathway by activation of FX to Factor Xa (FXa), FIX to Factor IXa (FIXa), and additional FVII to FVIIa. The action of TF:FVIIa leads ultimately to the conversion of prothrombin to thrombin, which carries out many biological functions. Among the most important activities of thrombin is the conversion of fibrinogen to fibrin, which polymerizes to form a clot. The TF:FVIIa complex also participates as a secondary factor in extending the physiological effects of the contact activation system.

[0121] The initiation and subsequent regulation of coagulation is complex, since maintenance of hemostasis is crucial for survival. There is an exquisite balance between hemostasis (normal clot formation and dissolution) and thrombosis (pathogenic clot formation). Serious clinical conditions involving aberrations in coagulation include deep vein thrombosis, myocardial infarction, pulmonary embolism, s stroke and disseminated intravascular coagulation (in sepsis). There are also many bleeding coagulopathies where there is insufficient clot formation. These include hemophilia A (FVIII deficiency) or hemophilia B (FIX deficiency), where procoagulant therapy is required. The challenge in this therapeutic area is to operate in the narrow window between too much and too little coagulation.

[0122] The use of exogenous FVIIa as a therapeutic agent has been shown to induce hemostasis in patients with hemophilia A and B (Hedner, 2001, Seminars Hematol. 38 (suppl. 12): 43-7; Hedner, 2004, Seminars Hematol. 41 (suppl. 1 ): 35-9). It also has been used to treat bleeding in patients with liver disease, anticoagulation-induced bleeding, surgery, thrombocytopenia, thrombasthenia, Bemard-Soulier syndrome, von Willebrand disease, and other bleeding disorders (See e.g. Roberts et a/., 2004, Blood 104: 3858-64).

[0123] Commercial preparations of human recombinant FVIIa are sold as

NovoSeven.™ NovoSeven™ is indicated for the treatment of bleeding episodes in hemophilia A or B patients and is the only recombinant FVIIa effective for bleeding episodes currently available. A circulating recombinant FVIIa half-life of 2.3 hours was reported in "Summary Basis for Approval for NovoSeven™" FDA reference number 96-0597. Moreover,

the half-life of recombinant FVIIa is shorter in pediatric patients (~ 1.3 hours), suggesting that higher doses of recombinaint FVIIa may be required in this population (Roberts et a/., 2004, Blood 104: 3858-64). Accordingly, relatively high doses and frequent administration are necessary to reach and sustain the desired therapeutic or prophylactic effect. As a consequence, adequate dose regulation is difficult to obtain and the need of frequent intravenous administrations imposes restrictions on the patient's way of living.

[0124] A molecule with a longer circulation half-life would decrease the number of necessary administrations. Given the frequent injections associated with currently available FVIIa therapy and the potential for obtaining more optimal therapeutic FVIIa levels with concomitant enhanced therapeutic effect, there is a clear need for improved FVII or FVIIa- like molecules with a longer half-life in vivo.

[0125] Recombinant human coagulation factor Vila (rFVIIa, NovoSeven; Novo

Nordisk A/S, Copenhagen, Denmark) has proven to be efficacious for the treatment of bleeding episodes in hemophilia patients with inhibitors. A small fraction of patients may be refractory to rFVIIa treatment and could potentially benefit from genetically modified FVIIa molecules with increased potencies. To this end, FVIIa analogs with increased intrinsic activity have been investigated that exhibit superior hemostatic profiles in vitro (see e.g. WO02/077218 or WO05/074975, which are hereby incorporated by reference in their entirety, and Tranholm et al., 2003, Blood 102(10): 3615-20, which is also incorporated by reference). These analogs may also be used as more efficacious hemostatic agents in other indications where efficacy of rFVIIa has been observed, including in thrombocytopenia and trauma.

[0126] Thus, in some embodiments, the Factor Vila analog that may be used in accordance with the invention is as described in WO02/077218 or WO05/074975. For example, the FVIIa analog may have a glutamine substituted for methionine at position 298 (i.e. M298Q-FVIIa). In certain exemplary embodiments, the FVIIa analog contains two additional mutations, valine at position 158 replaced by aspartic acid and glutamic acid at position 296 replaced by valine (i.e. V158D/E296V/M298Q-FVIIa). Additionally or alternatively, the Factor Vila analog may have an alanine residue substitution for lysine at position 337 (i.e. V158D/E296V/M298Q/K337A-FVIIa). In still other embodiments, the Factor Vila analog has a substitution or insertion selected from Q250C; P406C; and 407C, wherein a cysteine has also been introduced in the C-terminal sequence (see, e.g. US 7,235,638, which is hereby incorporated by reference in its entirety). The Factor Vila analog may further comprise a substitution or insertion at one or more of positions 247, 260, 393, 396, and/or 405.

[0127] In these or other embodiments, the Factor Vila analog comprises a substitution relative to the sequence of native Factor Vila selected from: (a) a substitution of Lys157 with an amino acid selected from the group consisting of GIy, VaI, Ser, Thr, Asp, and GIu; (b) a substitution of Lys337 with an amino acid selected from the group consisting of Ala, GIy, VaI, Ser, Thr, GIn, Asp, and GIu; (c) a substitution of Asp334 with any amino acid other than Ala or Asn; and (d) a substitution of Ser336 with any amino acid other than Ala or Cys (see e.g. US 7,176,288, which is hereby incorporated by reference in its entirety). Additionally or alternatively, the Factor Vila analog comprises a substitution of the Leu at position 305 of Factor VII with an amino acid residue selected from the group consisting of VaI, lie, Met, Phe, Trp, Pro, GIy, Ser, Thr, Cys, Tyr, Asn, GIu, Lys, Arg, His, Asp and GIn (see e.g. US 6,905,683, which is hereby incorporated by reference in its entirety).

[0128] Thus, in one aspect, the present invention provides therapeutic agents comprising an elastin-like peptide (ELP) and a Factor Vll/Vlla, or functional analog thereof. For example, in certain embodiments, the Factor Vll/Vlla is human Factor Vll/Vlla (e.g., SEQ ID NO: 33). The Factor Vll/Vlla may be a functional analog of human Factor Vll/Vlla, including functional fragments truncated at the N-terminus and/or C-terminus by from 1 to 10 amino acids, including by 1 , 2, 3, or about 5 amino acids. Functional analogs may contain from 1 to 10 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the native sequence (e.g., SEQ ID NO: 33), and in each case retaining the activity of the peptide. For example, such analogs may have from 1 to about 5 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the native full length sequence, or with respect to one or both of the heavy and light chains. Such activity may be confirmed or assayed using any available assay, including those described herein. In these or other embodiments, the Factor Vll/Vlla component has at least about 75%, 80%, 85%, 90%, 95%, or 98% identity with the native sequence (SEQ ID NO:33). The determination of sequence identity between two sequences (e.g., between a native sequence and a functional analog) can be accomplished using any alignment tool, including Tatusova et al., Blast 2 sequences - a new tool for comparing protein and nucleotide sequences. FEMS Microbiol Lett. 174:247- 250 (1999).

[0129] In exemplary embodiments, the FactorVII-ELP fusion has the amino acid sequence of SEQ ID NO:58. SEQ ID NO:58 further comprises a TEV protease cleavage site between the FactorVII and ELP sequences, which may be beneficial for removing the ELP sequence post expression where desired. However, in accordance with the invention, the tev sequence may be entirely removed, or replaced with another linking sequence as disclosed herein.

[0130] In another aspect, the present invention provides methods for the treatment or prevention of bleeding-related disorders. The method comprises administering an effective amount of the therapeutic agent comprising an elastin-like peptide (ELP) and a Factor VIIλ/lla or functional analog thereof to a patient in need. In certain embodiments, the bleeding-related disorder is one or more of hemophilia (A or B), post-surgical bleeding, anticoagulation-induced bleeding, thrombocytopenia, Factor VII deficiency, Factor Xl deficiency, bleeding in patients with liver disease, thrombasthenia, Bemard-Soulier syndrome, von Willebrand disease, and intracranial hemorrhage. Generally, the patient is a human or non-human animal (e.g., dog, cat, cow, or horse) patient. Perferably, the patient is human.

[0131] To characterize the in vitro binding properties of a suspected Factor Vll/Vlla analog, or an ELP-containing Factor Vila analog, TF binding assays can be performed as described previously (See, e.g., Chaing et al., 1994, Blood 83(12): 3524-35). Briefly, recombinant human TF can be coated onto lmmulon Il plates in carbonate antigen buffer overnight at 4 0 C. BSA is also coated onto the plates for use as a control. ELP-containing Factor Vila analogs may be added at various concentrations in TBS-T buffer. After several washes, monospecific polyclonal rabbit anti-human FVIIa sera is added and incubated for approximately an hour at room temperature. Next, goat anti-rabbit IgG conjugated to alkaline phosphatase is added, followed by the alkaline phosphatase substrate PNPP, which is used for detection. After subtraction of background, the absorbance at - 405 nm is taken to be directly proportional to the degree of Factor Vila binding to the immobilized TF. These values can then be compared to control plasma containing Factor Vila.

[0132] The clotting ability of a Factor Vll/Vlla analog or an ELP-containing Factor

Vila analog can be measured in human FVII deficient plasma. In this assay, the ELP- containing Factor Vila analog diluted to varying concentrations directly into FVII deficient plasma. In a coagulometer, one part plasma ± a FVIIa analog can be mixed with 2 parts Innovin™ (Dade, Miami, FIa.) prothrombin time reagent (recombinant human tissue factor with phospholipids and CaCI 2 ). Clot formation is detected optically and time to clotting measured. Clotting time (seconds) is compared to the mean clotting time of FVII-deficient plasma alone and plotted as the fractional clotting time versus FVIIa analog concentration.

Therapeutic Proteins

[0133] The present invention further provides therapeutic agents comprising an ELP component and at least one therapeutic protein selected from Table 1. The ELP component and therapeutic protein may be coupled by recombinant fusion or chemical conjugation as

described herein. Such therapeutic proteins are listed in Table 1 by protein name and GeneSeq Accession No. The amino acid sequence of each Therapeutic Protein, which is known in the art, is hereby incorporated by reference for each Therapeutic Protein listed in Table 1. Such therapeutic proteins are further described in US patent or PCT publications that are also listed in Table 1 , and such US patent and PCT publications are hereby incorporated by reference, especially with respect to the structure of such therapeutic proteins and described functional analogs.

[0134] Table 1 further describes the biological activity of each listed Therapeutic

Protein, as well as an exemplary assay for determining the activity of functional analogs or agents of the invention (e.g., fusion with an ELP component). Generally, functional analogs of therapeutic proteins listed in Table 1 may include functional fragments truncated at the N- terminus and/or C-terminus by from 1 to 10 amino acids, including by 1 , 2, 3, 4 or about 5 amino acids. Functional analogs may contain from 1 to 10 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the base sequence (e.g., as listed in Table 1), and in each case retaining the full or partial biological activity (as listed in Table 1) of the therapeutic protein. For example, functional analogs may have 1 , 2, 3, 4, or 5 amino acid insertions, deletions, and/or substitutions (collectively) with respect to the base sequence. Such activity may be confirmed or assayed using any available assay, including those described in the Table. In these or other embodiments, the therapeutic protein has at least about 75%, 80%, 85%, 90%, 95%, or 98% identity with the corresponding base sequence. The molecules may further comprise additional chemical modifications known for each in the art.

[0135] In some embodiments, the therapeutic protein (e.g., as selected from Table 1) has a size of less than about 25 kDa, or less than about 10 kDa, or less than about 5 kDa, and the corresponding therapeutic agent of the invention (e.g., comprising the ELP component) has a molecular weight of less than about 60 kDa, 55 kDa, 50 kDa, or 40 kDa.

[0136] Table 1 further lists preferred indications for each therapeutic protein, for which the corresponding therapeutic agent finds use, such as in a method for treatment or prevention related to such indication.

Table 1

I38

Conjugation and Coupling

[0132] The present invention provides therapeutic agents comprising an ELP component and a therapeutic component, such as therapeutic proteins listed in Table 1 , as well as a GLP-1 receptor agonists, insulin, Factor Vll/Vlla, and functional analogs as described. Such agents may be prepared by recombinant technology and/or chemical coupling (e.g., conjugation).

[0133] A recombinantly-produced ELP fusion protein, in accordance with certain embodiments of the invention, includes the ELP component and the therapeutic component associated with one another by genetic fusion. For example, the fusion protein may be generated by translation of a polynucleotide encoding the therapeutic component cloned in- frame with the ELP component (or vice versa). Such an ELP fusion protein may contain one or more copies of the therapeutic component attached to the N-terminus and/or the C- terminus of the ELP component. In some embodiments, the therapeutic proteinacious component is attached to both the N- and C-terminus of the ELP component and the fusion protein may contain one or more equivalents of the therapeutic component on either or both ends of the ELP component.

[0134] In certain embodiments, the ELP component and the therapeutic components can be fused using a linker peptide of various lengths to provide greater physical separation and allow more spatial mobility between the fused portions, and thus maximize the accessibility of the therapeutic component, for instance, for binding to its cognate receptor. The linker peptide may consist of amino acids that are flexible or more rigid. For example, a flexible linker may include amino acids having relatively small side chains, and which may be hydrophilic. Without limitation, the flexible linker may contain a stretch of glycine and/or serine residues. More rigid linkers may contain, for example, more sterically hindering amino acid side chains, such as (without limitation) tyrosine or histidine. The linker may be less than about 50, 40, 30, 20, 10, or 5 amino acid residues. The linker can be covalently linked to and between an ELP component and a therapeutic component, for example, via recombinant fusion.

[0135] The linker or peptide spacer may be protease-cleavable or non-cleavable. By way of example, cleavable peptide spacers include, without limitation, a peptide sequence recognized by proteases {in vitro or in vivo) of varying type, such as Tev, thrombin, factor Xa, plasmin (blood proteases), metalloproteases, cathepsins (e.g., GFLG, etc.), and proteases found in other corporeal compartments. In some embodiments employing cleavable linkers, the fusion protein ("the therapeutic agent") may be inactive, less active, or

less potent as a fusion, which is then activated upon cleavage of the spacer in vivo. Alternatively, where the therapeutic agent is sufficiently active as a fusion, a non-cleavable spacer may be employed. The non-cleavable spacer may be of any suitable type, including, for example, non-cleavable spacer moieties having the formula [(Gly)n-Serjm (SEQ ID NO.: 22) where n is from 1 to 4, inclusive, and m is from 1 to 4, inclusive. Alternatively, a short ELP sequence different than the backbone ELP could be employed instead of a linker or spacer, while accomplishing the necessary effect.

[0136] In still other embodiments, the therapeutic agent is a recombinant fusion having a therapeutic component flanked on each terminus by an ELP component. At least one of said ELP components may be attached via a cleavable spacer, such that the therapeutic component is inactive, but activated in vivo by proteolytic removal of a single ELP component. The resulting single ELP fusion being active, and having an enhanced half-life (or other property described herein) in vivo.

[0137] In other embodiments, the present invention provides chemical conjugates of the ELP component and the therapeutic component. The conjugates can be made by chemically coupling an ELP component to a therapeutic component by any number of methods well known in the art (See e.g. Nilsson et al., 2005, Ann Rev Biophys Bio Structure 34: 91-118). In some embodiments, the chemical conjugate can be formed by covalently linking the therapeutic component to the ELP component, directly or through a short or long linker moiety, through one or more functional groups on the therapeutic proteinacious component, e. g., amine, carboxyl, phenyl, thiol or hydroxyl groups, to form a covalent conjugate. Various conventional linkers can be used, e. g., diisocyanates, diisothiocyanates, carbodiimides, bis (hydroxysuccinimide) esters, maleimide- hydroxysuccinimide esters, glutaraldehyde and the like.

[0138] Non-peptide chemical spacers can additionally be of any suitable type, including for example, by functional linkers described in Bioconjugate Techniques, Greg T. Hermanson, published by Academic Press, Inc., 1995, and those specified in the Cross- Linking Reagents Technical Handbook, available from Pierce Biotechnology, Inc. (Rockford, Illinois), the disclosures of which are hereby incorporated by reference, in their respective entireties. Illustrative chemical spacers include homobifunctional linkers that can attach to amine groups of Lys, as well as heterobifunctional linkers that can attach to Cys at one terminus, and to Lys at the other terminus.

[0139] In certain embodiments, relatively small ELP components (e.g., ELP components of less than about 30 kDa, 25 kDa, 20 kDa, 15 kDa, or 10 kDa), that do not transition at room temperature (or human body temperature, e.g., Tt >37 C C), are chemically

coupled or crosslinked. For example, two relatively small ELP components, having the same or different properties, may be chemically coupled. Such coupling, in some embodiments, may take place in vivo, by the addition of a single cysteine residue at or around the C-terminus of the ELP. Such ELP components may each be fused to one or more therapeutic components, so as to increase activity or avidity at the target.

Polynucleotides. Vectors, and Host Cells

[0140] In another aspect, the invention provides polynucleotides comprising a nucleotide sequence encoding the therapeutic agent of the invention. Such polynucleotides further comprise, in addition to sequences encoding the ELP and therapeutic components, one or more expression control elements. For example, the polynucleotide, may comprise one or more promoters or transcriptional enhancers, ribosomal binding sites, transcription termination signals-, and polyadenylation signals, as expression control elements. The polynucleotide may be inserted within any suitable vector, which may be contained within any suitable host cell for expression.

[0141] A vector comprising the polynucleotide can be introduced into a cell for expression of the therapeutic agent. The vector can remain episomal or become chromosomally integrated, as long as the insert encoding the therapeutic agent can be transcribed. Vectors can be constructed by standard recombinant DNA technology. Vectors can be plasmids, phages, cosmids, phagemids, viruses, or any other types known in the art, which are used for replication and expression in prokaryotic or eukaryotic cells. It will be appreciated by one of skill in the art that a wide variety of components known in the art (such as expression control elements) may be included in such vectors, including a wide variety of transcription signals, such as promoters and other sequences that regulate the binding of RNA polymerase onto the promoter. Any promoter known to be effective in the cells in which the vector will be expressed can be used to initiate expression of the therapeutic agent. Suitable promoters may be inducible or constitutive. Examples of suitable promoters include the SV40 early promoter region, the promoter contained in the 3' long terminal repeat of Rous sarcoma virus, the HSV-1 (herpes simplex virus-1 ) thymidine kinase promoter, the regulatory sequences of the metallothionein gene, etc., as well as the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells; insulin gene control region which is active in pancreatic beta cells, immunoglobulin gene control region which is active in lymphoid cells, mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells, albumin gene control

region which is active in liver, alpha-fetoprotein gene control region which is active in liver, alpha 1 -antitrypsin gene control region which is active in the liver, beta-globin gene control region which is active in erythroid cells, myelin basic protein gene control region which is active in oligodendrocyte cells in the brain, myosin light chain-2 gene control region which is active in skeletal muscle, and gonadotropin releasing hormone gene control region which is active in the hypothalamus.

Pharmaceutical Compositions

[0142] The present invention further provides pharmaceutical compositions comprising the therapeutic agents of the invention (as described above) together with a pharmaceutically acceptable carrier or excipient. Such pharmaceutical compositions may be employed in the methods of treatment as described above, for each of the therapeutic proteins, e.g., the therapeutic proteins listed in Table 1 , GLP-1 receptor agonists, insulin, and Factor Vl I/VI Ia embodiments.

[0143] The therapeutic agents of the invention may overcome certain deficiencies of peptide agents when administered (e.g., parenterally), including in some embodiments, the limitation that such peptides may be easily metabolized by plasma proteases or cleared from circulation by kidney filtration. Traditionally, the oral route of administration of peptide agents may also be problematic, because in addition to proteolysis in the stomach, the high acidity of the stomach destroys such peptide agents before they reach their intended target tissue. Peptides and peptide fragments produced by the action of gastric and pancreatic enzymes are cleaved by exo and endopeptidases in the intestinal brush border membrane to yield di- and tripeptides, and even if proteolysis by pancreatic enzymes is avoided, polypeptides are subject to degradation by brush border peptidases. Any of the peptide agents that survive passage through the stomach are further subjected to metabolism in the intestinal mucosa where a penetration barrier prevents entry into the cells. In certain embodiments, the therapeutic agents of the invention may overcome such deficiencies, and provide compositional forms having enhanced efficacy, bioavailability, therapeutic half-life, persistence, degradation assistance, etc. The therapeutic agents of the invention thus include oral and parenteral dose forms, as well as various other dose forms, by which peptide agents can be utilized in a highly effective manner. For example, in some embodiments, such agents may achieve high mucosal absorption, and the concomitant ability to use lower doses to elicit an optimum therapeutic effect.

[0144] The therapeutic agents of the present invention may be administered in smaller doses and/or less frequently than unfused or unconjugated counterparts. While one

of skill in the art can determine the desirable dose in each case, a suitable dose of the therapeutic agent for achievement of therapeutic benefit, may, for example, be in a range of about 1 microgram (μg) to about 100 milligrams (mg) per kilogram body weight of the recipient per day, preferably in a range of about 10 μg to about 50 mg per kilogram body weight per day and most preferably in a range of about 10 μg to about 50 mg per kilogram body weight per day. The desired dose may be presented as one dose or two or more sub- doses administered at appropriate intervals throughout the day. These sub-doses can be administered in unit dosage forms, for example, containing from about 10 μg to about 1000 mg, preferably from about 50 μg to about 500 mg, and most preferably from about 50 μg to about 250 mg of active ingredient per unit dosage form. Alternatively, if the condition of the recipient so requires, the doses may be administered as a continuous infusion.

[0145] The mode of administration and dosage forms will of course affect the therapeutic amount of the peptide active therapeutic agent that is desirable and efficacious for a given treatment application. For example, orally administered dosages can be at least twice, e.g., 2-10 times, the dosage levels used in parenteral administration methods.

[0146] The therapeutic agents of the invention may be administered per se as well as in various forms including pharmaceutically acceptable esters, salts, and other physiologically functional derivatives thereof. The present invention also contemplates pharmaceutical formulations, both for veterinary and for human medical use, which include therapeutic agents of the invention. In such pharmaceutical and medicament formulations, the therapeutic agents can be used together with one or more pharmaceutically acceptable carrier(s) therefore and optionally any other therapeutic ingredients. The carrier(s) must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the formulation and not unduly deleterious to the recipient thereof. The therapeutic agents are provided in an amount effective to achieve the desired pharmacological effect, as described above, and in a quantity appropriate to achieve the desired daily dose.

[0147] The formulations of the therapeutic agent include those suitable for parenteral as well as non-parenteral administration, and specific administration modalities include oral, rectal, buccal, topical, nasal, ophthalmic, subcutaneous, intramuscular, intravenous, transdermal, intrathecal, intra-articular, intra-arterial, sub-arachnoid, bronchial, lymphatic, vaginal, and intra-uterine administration. Formulations suitable for oral and parenteral administration are preferred.

[0148] When the therapeutic agent is used in a formulation including a liquid solution, the formulation advantageously can be administered orally or parenterally. When the therapeutic agent is employed in a liquid suspension formulation or as a powder in a

biocompatible carrier formulation, the formulation may be advantageously administered orally, rectally, or bronchially.

[0149] When the therapeutic agent is used directly in the form of a powdered solid, the active agent can be advantageously administered orally. Alternatively, it may be administered bronchially, via nebulization of the powder in a carrier gas, to form a gaseous dispersion of the powder which is inspired by the patient from a breathing circuit comprising a suitable nebulizer device.

[0150] The formulations comprising the therapeutic agent of the present invention may conveniently be presented in unit dosage forms and may be prepared by any of the methods well known in the art of pharmacy. Such methods generally include the step of bringing the therapeutic agents into association with a carrier which constitutes one or more accessory ingredients. Typically, the formulations are prepared by uniformly and intimately bringing the therapeutic agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into dosage forms of the desired formulation.

[0151] Formulations suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active ingredient as a powder or granules; or a suspension in an aqueous liquor or a non-aqueous liquid, such as a syrup, an elixir, an emulsion, or a draught.

[0152] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, with the therapeutic agent being in a free-flowing form such as a powder or granules which optionally is mixed with a binder, disintegrant, lubricant, inert diluent, surface active agent, or discharging agent. Molded tablets comprised of a mixture of the powdered peptide active therapeutic agent-ELP construct(s) with a suitable carrier may be made by molding in a suitable machine.

[0153] A syrup may be made by adding the peptide active therapeutic agent-ELP construct(s) to a concentrated aqueous solution of a sugar, for example sucrose, to which may also be added any accessory ingredient(s). Such accessory ingredient(s) may include flavorings, suitable preservative, agents to retard crystallization of the sugar, and agents to increase the solubility of any other ingredient, such as a polyhydroxy alcohol, for example glycerol or sorbitol.

[0154] Formulations suitable for parenteral administration conveniently comprise a sterile aqueous preparation of the therapeutic agent, which preferably is isotonic with the

blood of the recipient (e.g., physiological saline solution). Such formulations may include suspending agents and thickening agents or other microparticulate systems which are designed to target the peptide active therapeutic agent to blood components or one or more organs. The formulations may be presented in unit-dose or multi-dose form.

[0155] Nasal spray formulations comprise purified aqueous solutions of the therapeutic agent with preservative agents and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucus membranes.

[0156] Formulations for rectal administration may be presented as a suppository with a suitable carrier such as cocoa butter, hydrogenated fats, or hydrogenated fatty carboxylic acid.

[0157] - Topical formulations comprise the therapeutic agent dissolved or suspended in one or more media, such as mineral oil, petroleum, polyhydroxy alcohols, or other bases used for topical pharmaceutical formulations.

[0158] In addition to the aforementioned ingredients, the formulations of this invention may further include one or more accessory ingredient(s) selected from diluents, buffers, flavoring agents, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants), and the like.

[0159] The features and advantages of the present invention are more fully shown with respect to the following non-limiting examples.

EXAMPLES EXAMPLE 1 : CONSTRUCTION OF VARIOUS ELP COMPONENT CONSTRUCTS

[0160] Cloning steps were conducted in Escherichia coli strain XL1-Blue (rec A1 , endM , gyrA96, tf)/-1 , hsdRM (r k " , m k +), supE44, re/A1 , /ac[F', proAB, /σcl q ZδM15, Tn10 (Tet r )] (Stratagene La JoIIa, CA). pUC19 (NEB, Beverly, MA) was used as the cloning vector for the ELP construction (Meyer and Chilkoti, Nat. Biotechnol., 17(11):1112-5, 1999). Modified forms of pET15b and pET24d vectors (Novagen) were used to express ELP and ELP-fusion proteins in BL21 Star (DE3) strain (F " , ompT, hsdS B (r B " m B ' ), gal, dcm, rne131 , (DE3)) (Invitrogen Carlsbed, CA) or BLR(DE3) (F " , ompT, hsdS B (r B ' m B ~ ), gal, dcm, δ(srf- recA) 306::Tn10(TcR)(DE3)) (Novagen Madison, Wl). Synthetic DNA oligos were purchased from Integrated DNA Technologies, Coralville, IA. All vector constructs were made using standard molecular biology protocols (e.g., Current Protocols in Molecular Biology, ed. Ausubel, et al., 1995).

Construction of ELP1 FVSAPG 3 I Gene Series

[0161] The ELP1 [V 5 A 2 G 3 ] series designate polypeptides containing multiple repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is valine, alanine, and glycine at a relative ratio of 5:2:3.

[0162] The ELP1 [V 5 A 2 G 3 ] series monomer, ELP1 [V 5 A 2 G 3 -IO], was created by annealing four 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with EcoRI and Hind\\\ compatible ends (Meyer and Chilkoti, Nat. Biotechnol., 17(11 ):1112-5, 1999). The oligos were annealed in a 1 μM mixture of the four oligos in 50 μl IX ligase buffer (Invitrogen) to 95°C in a heating block than the block was allowed to cool slowly to room temperature. The ELP1 [V 5 A 2 G 3 -10]/EcoRI-H/πcflll DNA segment was ligated into a pUC19 vector digested with EcoRI and Hind\\\ and CIAP dephosphorylated (Invitrogen) to form pUC19-ELP1 [V 5 A 2 G 3 -IO]. Building of the ELP1 [V 5 A 2 G 3 ] series library began by inserting ELP1 [V 5 A 2 G 3 -IO] Pfflvil/Bgll fragment from pUC19-ELP1 [V 5 A 2 G 3 -IO] into pUC19- ELP1 [V 5 A 2 G 3 -IO] linearized with PfIW and dephosphorylated with CIAP to create pUC19- ELP1 [V 5 A 2 G 3 ^O]. pUC19-EL.P1 [V 5 A 2 G 3 ^O] was then built up to pUC19-ELP1 [V 5 A 2 G 3 -SO] and pUC19-EL.P1 [VsA 2 G 3 -40] by ligating ELP1 [V 5 A 2 G 3 -IO] or ELP1 [V 5 A 2 G 3 -20] PfIUMBgW fragments respectively into Pfflvll digested pUC 19-ELP1 [V 5 A 2 G 3 ^O]. This procedure was used to expand the ELP1 [V 5 A 2 G 3 ] series to create pUC19-ELP1 [V 5 A 2 G 3 ^O], pUC19-ELP1 [V 5 A 2 G 3 ^O] and pUC19-ELP1 [V 5 A 2 G 3 -IeO] genes.

Construction of ELP1 [K 1 V 2 F 1 I Gene Series

[0163] The ELP1 [K 1 V 2 F 1 ] series designate polypeptides containing multiple repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of 1:2:1.

[0164] The ELP1 [K 1 V 2 F 1 ] series monomer, ELP1 [K 1 V 2 F 1 ^], was created by annealing two 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with EcoRI and Hindlll compatible ends (Meyer and Chilkoti, 1999). The oligos were annealed in a 1 μM mixture of the four oligos in 50 μl 1X ligase buffer (Invitrogen) to 95°C in a heating block then the block was allowed to cool slowly to room temperature. The ELP1 [K 1 V 2 F 1 ^]ZECoRI- Hind\\\ DNA segment was ligated into a pUC19 vector digested with EcoRI and Hind\\\ and CIAP dephosphorylated (Invitrogen) to form pUC19-ELP1 [K 1 V 2 F 1 ^]. Building of the ELP1 [K 1 V 2 F 1 ] series library began by inserting ELP1 [K 1 V 2 FM] PflMMBgh fragment from pUC19- ELP1 [K 1 V 2 F 1 ^] into pUC19-ELP1 [K 1 V 2 F 1 ^] linearized with P/7M1

and dephosphorylated with CIAP to create pUC19-ELP1 [K 1 V 2 F 1 -S]. Using the same procedure the ELP1 [K 1 V 2 F 1 ] series was doubled at each ligation to form pUC19-ELP1 [K 1 V 2 F 1 -Ie], pUC19-ELP1 [K 1 V 2 F 1 ^] 1 pUC19- ELP1 [K 1 V 2 Fi-64] and pUC19-ELP1 [K 1 V 2 F 1 - 128].

Construction of ELP1 [K 1 V 1 F 1 I Gene Series

[0165] The ELP1 [K 1 V 7 F 1 ] series designate polypeptides containing multiple repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is lysine, valine, and phenylalanine at a relative ratio of 1 :7:1.

[0166] The ELP1 [K 1 V 7 F 1 ] series monomer, ELP1 [K 1 V 7 F 1 -S], was created by annealing four 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with P/7MI and Hind\\\ compatible ends. The ELP1 [K 1 V 7 F 1 ^] DNA segment was than ligated into PffMI/Hindlll dephosphorylated PUC19-ELP1 [V 5 A 2 G 3 -IeO] vector thereby substituting ELP1 [V 5 A 2 G 3 -IeO] for ELP1 [K 1 V 7 F 1 ^] to create the pUC19-ELP1 [K 1 V 7 F 1 ^] monomer. The ELP1 [K 1 V 7 F 1 ] series was expanded in the same manner as the ELP1 [K 1 V 2 F 1 ] series to create pUC19- ELP1 [K 1 V 7 F 1 -Ie], PUC19-ELP1 [K 1 V 7 F 1 -Se], pUC19-ELP1 [KiV 7 Fi-72] and pUC19-ELP1 [K,V 7 F 1 -144].

Construction of ELP1 M Gene Series

[0167] The ELP1 [V] series designate polypeptides containing multiple repeating units of the pentapeptide VPGXG (SEQ ID NO: 3), where X is exclusively valine.

[0168] The ELP1 [V] series monomer, ELP1 [V-5], was created by annealing two 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with EcoRI and Hind\\\ compatible ends. The ELP1 [V-5] DNA segment was than ligated into EcoRI//-//ncflll dephosphorylated pUC19 vector to create the pUC19-ELP1 [V-5] monomer. The ELP1 [V] series was created in the same manner as the ELP1 [V 5 A 2 G 3 ] series, ultimately expanding pUC19-EL.P1 [V-5] to pUC19-ELP1 [V-60] and pUC19-ELP1 [V-120].

Construction of ELP2 Gene Series

[0169] The ELP2 series designate polypeptides containing multiple repeating units of the pentapeptide AVGVP.

[0170] The ELP2 series monomer; ELP2 [5], was created by annealing two 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with EcoRI

I 58

and Hind\\\ compatible ends. The ELP2 [5] DNA segment was than ligated into EcoR\IHiπd\\\ dephosphorylated pUC19 vector to create the pUC19-ELP2[5] monomer. The ELP2 series was expanded in the same manner as the ELP1 [KiV 2 Fi] series to create pUC19-ELP2[10], pUC19- ELP2 [30], pUC 19-ELP2 [60] and pUC 19-ELP2 [ 120].

Construction of ELP3 M Gene Series

[0171] The ELP3 [V] series designate polypeptides containing multiple repeating units of the pentapeptide IPGXG (SEQ ID NO: 5), where X is exclusively valine.

[0172] The ELP3 [V] series monomer, ELP3 [V-5], was created by annealing two 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with PfLMI amino terminal and GGC carboxyl terminal compatible ends due to the lack of a convenient carboxyl terminal restriction site but still enable seamless addition of the monomer. The ELP3 [V- 5] DNA segment was then ligated into PflM1/Bgll dephosphorylated pUC19- ELP4[V-5], thereby substituting ELP4 [V-5] for ELP3 [V-5] to create the pUC19-ELP3 [V-5] monomer. The ELP3 [V] series was expanded by ligating the annealed ELP3 oligos into pUC19-ELP3[V-5] digested with PfIMI. Each ligation expands the ELP3 [V] series by 5 to create ELP3 [V-10], ELP3 [V-15], etc.

Construction of the ELP4 IVl Gene Series

[0173] The ELP4 [V] series designate polypeptides containing multiple repeating units of the pentapeptide LPGXG (SEQ ID NO: 7), where X is exclusively valine.

[0174] The ELP4 [V] series monomer, ELP4 [V-5], was created by annealing two 5' phosphorylated, PAGE purified synthetic oligos to form double stranded DNA with EcoRI and Hind\\\ compatible ends. The ELP4 [V-5] DNA segment was than ligated into EcoR\IHind\\\ dephosphorylated pUC19 vector to create the pUC19-ELP4[V-5] monomer. The ELP4 [V] series was expanded in the same manner as the ELP1 [K 1 V 2 F 1 ] series to create pUC19- ELP4[V-10], pUC19-ELP4[V-30], pUC19-ELP4[V-60] and pUC19-ELP4[V-120].

[0175] The ELP genes were also inserted into other vectors such as pET15b-SD0, pET15b-SD3, pET15b-SD5, pET15b-SD6, and pET24d-SD21. The pET vector series are available from Novagen, San Diego, CA.

[0176] The pET15b-SD0 vector was formed by modifying the pET15b vector using

SDO double-stranded DNA segment containing the multicloning restriction site (Sacl-λ/del- Nco\-Xho\-SnaB\-Bamh\). The SDO double-stranded DNA segment had Xba\ and SamHI

compatible ends and was ligated into Xbal/SamHI linearized and 5'-dephosphorylated pET15b to form the pet15b-SD0 vector.

[0177] The pET15b-SD3 vector was formed by modifying the pET15b-SD0 vector using SD3 double-stranded DNA segment containing a Sffl restriction site upstream of a hinge region- thrombin cleavage site followed by the multicloning site (Nde\-Nco\-Xho\- SnaBI-SarnHI). The SD3 double-stranded DNA segment had Sacl and Ndel compatible ends and was ligated into Sacl/Ndel linearized and 5'-dephosphorylated pET15b-SD0 to form the pET15b-SD3 vector.

[0178] The pET15b-SD5 vector was formed by modifying the pET15b-SD3 vector using the SD5 double-stranded DNA segment containing a Sfi\ restriction site upstream of a thrombin cleavage site followed by a hinge and the multicloning site (Nde\-Nco\-Xho\-SnaB\- SamHI). The SD5 double-stranded DNA segment had Sffl and Nde\ compatible ends and was ligated into Sfil/Ndel linearized and 5'-dephosphorylated pET15b-SD3 to form the pET15b-SD5 vector.

[0179] The pET15b-SD6 vector was formed by modifying the pET15b-SD3 vector using the SD6 double-stranded DNA segment containing a Sffl restriction site upstream of a linker region-TEV cleavage site followed by the multicloning site (Nde\-Nco\-Xho\-SnaB\- BamHi). The SD6 double-stranded DNA segment had Sffl and Nhe\ compatible ends and was ligated into Sfil/Ndel linearized and 5'-dephosphorylated pET15b-SD3 to form the pET15b-SD6 vector.

[0180] The pET24d-SD21 vector was formed by modifying the pET24d vector using the SD21 double-stranded DNA segment with λ/col and Nhe\ compatible ends. The SD21 double-stranded DNA segment was ligated into Nco\INhe\ linearized and 5' dephosphorylated pET24d to create the pET24d-SD21 vector, which contained a new multicloning site λ/col-Sf//-λ//7el-βamHI-EcoRI-Sacl-Sa/l-H/ncflll-λ/o/1-X λol with two stop codons directly after the Sfil site for insertion and expression of ELP with the minimum number of extra amino acids.

[0181] The pUC19-EL.P1 [V 5 A 2 G 3 ^O], pUC19-ELP1 [V 5 A 2 G 3 -QO], and pUC19-ELP1

[V 5 A 2 G 3 -IeO] plasmids produced in XL1-Blue were digested with PfIW and Bgl\, and the ELP-containing fragments were ligated into the Sffl site of the pET15b-SD3 expression vector as described hereinabove to create pET15b-SD3-ELP1 [V 5 A 2 G 3 -SO], pET15b-SD5- ELP1 [V 5 A 2 G 3 ^O] and pET15b-SD5- ELP1 [VsA 2 G 3 -180], respectively.

[0182] The pUC19-ELP1 [VsA 2 G 3 -90], pUC19-ELP1 [V 5 A 2 G 3 -IeO], pUC19-ELP1 [V-

60] and pUC19-ELP1 [V-120] plasmids produced in XL1-Blue were digested with PfIW and

Bgl\, and the ELP-containing fragments were ligated into the Sffl site of the pET15b-SD5 expression vector as described hereinabove to create pET15b-SD5-ELP1 [V 5 A 2 G 3 -QO], pET15b-SD5-ELP1 [V 5 A 2 G 3 -180], pET15b-SD5-ELP1 [V-60] and pET15b-SD5-ELP1 [V- 120], respectively.

[0183] The pUC19-ELP1 [V 5 A 2 G 3 -QO] plasmid produced in XL1-Blue was digested with PffMI and Bgl\, and the ELP-containing fragment was ligated into the Sffl site of the pET15b-SD6 expression vector as described hereinabove to create pET15b-SD6-ELP1 [V 5 A 2 G 3 -QO].

[0184] The pUC19-ELP1 [K 1 V 2 F 1 -64], and pUC19-ELP1 [K 1 V 2 F 1 -^e] plasmids produced in XL1-Blue were digested with PffMI and Bgl\, and the ELP-containing fragments were ligated into the Sffl site of the pET24d-SD21 expression vector as described hereinabove to create pET24d-SD21- ELP1 [K 1 V 2 F 1 -64] and pET24d-SD21-ELP1 [K 1 V 2 F 1 - 128], respectively.

[0185] The pUC19-ELP1 [K 1 V 7 F 1 -72] and pUC19-ELP1 [K 1 V 7 F 1 -144] plasmids produced in XL1-Blue were digested with PffMI and Bgl\, and the ELP-containing fragments were ligated into the Sffl site of the pET24d-SD21 expression vector as described hereinabove to create pET24d-SD21- ELP1 [K 1 V 7 F 1 -72], pET24d-SD21-ELP1 [K 1 V 7 F 1 -144], respectively.

[0186] The pUC19-ELP2[60] and pUC1Q-ELP2[120] plasmids produced in XL1-Blue were digested with λ/col and HincλW, and the ELP-containing fragments were ligated into the λ/col and Hind\\\ sites of the pET24d-SD21 expression vector as described hereinabove to create pET24d- SD21-ELP2[60], pET24d-SD21-ELP2[120], respectively.

[0187] The pUC1Q-ELP4[V-60] and pUC1Q-ELP4[V-120] plasmids produced in XL1-

Blue were digested with Ncol and Hindlll, and the ELP-containing fragments were ligated into the λ/col and Hind\\\ sites of the pET24d-SD21 expression vector as described hereinabove to create pET24d- SD21-ELP4[V-60], pET24d-SD21-ELP4[V-120], respectively.

EXAMPLE 2: ISOLATION AND PURIFICATION OF FUSION PROTEINS CONTAINING INSULIN A PEPTIDE (InsA)

[0188] ELP-lnsA fusion proteins included the following:

[0189] Insulin A peptide and ELP1 [V-60] polypeptide with an enterokinase protease cleavage site therebetween.

[0190] Insulin A peptide and ELP1 [V 5 A 2 G 3 -QO] polypeptide with an enterokinase protease cleavage site therebetween.

[0191] Insulin A peptide and ELP1 [V-120] polypeptide with an enterokinase protease cleavage site therebetween.

[0192] Insulin A peptide and ELP1 [V 5 A 2 G 3 -180] polypeptide with an enterokinase protease cleavage site therebetween.

[0193] A single colony of E. coli strain BLR (DE3) (Novagen) containing the respective ELP-lnsA fusion protein was inoculated into 5 ml CircleGrow (Q-BIOgene, San Diego, CA) supplemented with 100 μg/ml ampicillin (Sigma) and grown at 37°C with shaking at 250 rpm for 5 hours. The 5 ml culture was then inoculated into a 500 ml culture and allowed to grow at 25°C for 16 hours before inducing with 1 mM IPTG for 4 hours at 25°C. The culture was harvested and suspended in 40 ml 20 mM Tris-HCI pH 7.4, 50 mM NaCI, 1 mM DTT and 1 Complete EDTA free Protease inhibitor pellet (Roche, Indianapolis, IN). Cells were lysed by ultrasonic disruption on ice for 3 minutes, which consisted of 10 seconds bursts at 35% power separated by 30 second cooling down intervals. Cell debris was removed by centrifugation at 20,00Og, 4 0 C for 30 minutes.

[0194] Inverse phase transition was induced by adding NaCI to the cell lysate at room temperature to achieve a final concentration of 1.0 M therein, followed by centrifugation at 20,00Og for 15 minutes at room temperature. The resulting pellet contained the respective ELP-lnsA fusion protein and non-specifically NaCI precipitated proteins.

[0195] The pellet was re-suspended in 40 ml ice-cold ml 20 mM Tris-HCI pH 7.4, 50 mM NaCI, 1 mM DTT and re-centrifuged at 20,000 g, 4°C for 15 minutes to remove the non- specifically NaCI precipitated proteins. The inverse transition cycle was repeated two additional times to increase the purity of the respective ELP-lnsA fusion protein and reduce the final volume to 0.5 ml.

EXAMPLE 3: HALF-LIFE OF ELP1

[0196] The pharmacokinetics of ELP1 were determined by intravenously administering [ 14 C]ELPI to nude mice (Balb/c nu/nu) bearing a leg/flank FaDu xenograft and collecting blood samples at various time intervals after administration. The blood pharmacokinetics exhibited a characteristic distribution and elimination response for large macromolecules, which was well described by a bi-exponential process.

[0197] The plasma concentration time-course curve was fit to the analytical solution of a two-compartment model to approximate both an elimination and distribution response. Certain pharmakinetic parameters are shown in Table 1 below. The distribution volume of the ELP (1.338 μl) was nearly identical to the hypothetical plasma volume of 1.363 μl (Barbee, R.W., et al., Am. J. Physio. 263(3) (1992) R728-R733), indicating that the ELP did not rapidly distribute or bind to specific organs and tissues directly after administration. The AUC is a measure of the cumulative exposure to ELP in the central compartment or the blood plasma. The body clearance is defined as the rate of ELP elimination in the body relative to its plasma concentration and is the summation of clearance through all organs including the kidney, liver and others.

Table 1 : Pharmacokinetic parameters calculated for [ 14 C]ELPI

[0198] The mass transfer rate constants are from a standard two-compartment model (ki; from central to peripheral compartment; k 2 , from peripheral to central compartment; and k e , elimination from central compartment). The distribution volume (V d ), central compartment concentration time-course area under the curve (AUC) and body clearance (CI B ) are displayed. Data are shown as the mean values (n = 5, except V d and initial plasma concentration (C 0 ) was calculated from a similar cohort with n = 3).

EXAMPLE 4: BIODISTRIBUTION OF ELPs IN NUDE MICE 1 4 C labeled ELP1-150 and/or 14 C labeled ELP2-160

[0199] 14 C labeled ELP1-150 and/or 14 C labeled ELP2-160 were administered to nude mice with a FaDu tumor (mean +/- SD, n=6). The tumor was heated post administration of the ELP in a water bath at 41.5 0 C. The distribution was highest to the organs with the highest blood content: liver, kidneys, spleen, and lungs.

14 C labeled ELP2-fVi AaG 7 -1601

[0200] 14 C labeled ELP2-[\λ A 8 G 7 -160] (T,>60°C) was administered to nude mice for a plasma concentration of 15μM. ELP concentrations were determined following 1 hour of heating (41°C) of an implanted FaDu tumor, located in the right hind leg of the nude mouse. Data are shown as the mean, plus the 95% confidence interval. N=6.

[0201] ELP concentration was measured 1.5 hours following systemic administration of 14 C labeled ELP2-[V 1 A 8 G 7 -160]. The highest distribution is seen in organs with the highest blood content: liver, kidneys, spleen, and lungs.

EXAMPLE 5: EXENDIN-4 ELP FUSION

[0202] The DNA sequence for Exendin-4 (Ex-4) (SEQ ID NO: 14) was reverse translated from the amino acid sequence using codons optimized for E. coli expression. The DNA sequence encoding Exendin-4 was constructed by annealing together synthetic oligonucleotides with overhanging 5' and 3' ends compatible with the restriction sites Nde\ and Xho\ in the plasmid pET24d-ELP1-90 (Figure 1). This plasmid was digested with the restriction enzymes Nde\ and Xho\ and the annealed DNA sequence was ligated into the cut vector. Insertion was confirmed by restriction digest and DNA sequencing. The resulting plasmid was designated as pET24d-Ex-4 ELP1-90 (Figure 2A), and the sequence of the resulting Exendin-4-ELP fusion shown in Figure 2B. Primers for construction of the fusion are also indicated.

[0203] pET24d-Ex-4 ELP1-90 was used to transform the E. coli strain BRL

(Invitrogen) and selected transformants were grown in media 3 (1.2% Tryptone Peptone, 2.4% yeast extract, 5g/L casamino acids, 2% glycerol, 2.313g Potassium phosphate dibasic/L, 12.541 g Potassium phosphate monobasic/L) in shake flasks. Production proceeded by autoinduction by inoculating 1 OD cells into 1 L of media 3 and allowing growth to proceed for 17 hr at 37°C without addition of inducer. The product was recovered by collection of the cell pellet, sonicated to disrupt the cells and recovered by thermal and/or salt induced transition modulated by the ELP moiety (Improved Non-chromatoqraphic Purification of a Recombinant Protein by Cationic Elastin-like Polypeptides. Dong Woo Lim, Kimberly Trabbic-Carlson, J. Andrew MacKay, and Ashutosh Chilkoti. Biomacromolecules 2007, 8, 1417-1424).

[0204] This example is with the ELP designated 1-90. This is based on the VPGXG

(SEQ ID NO: 3) motif where X is a V, G or A in the ratio 5:3:2 in a 10 unit repeat, repeated 8x with a final (C-terminal) 10-unit repeat where X is a V, G, A and W in the ratio 4:3:2:1.

[0205] [(VPGXG)IO] 9 where the X residue in the ten sequential iterations of the repeat unit (numerical subscript) can be described as [(V 1 , 4 , 5, β, ioG 2 , 7, 9A 3 , β)β (Vi, 4 .5. βG 2 , 7.

[0206] The ELP may be any combination of VPGXG (SEQ ID NO: 3) units where X is any of the 20 natural amino, acids, except proline, in any combination of repeat units of

any length. In addition, the amino acid may be an unnatural amino acid for which the host strain has been engineered to accept an engineered tRNA for incorporation at specific codon (Wang L, Brock A, Herberich B, Schultz PG. Expanding the genetic code of Escherichia coli. [2001] Science 292, 498-500).

[0207] This construct was produced in the cytosol with an N-terminal methionine, which is normally removed by methionine aminopeptidase. Complete and accurate processing of the methionine, however, cannot be assumed; this enzyme may also remove the N-terminal histidine of the Exendin-4 moiety. This could result in a mixture of, unprocessed, processed and incorrectly processed products. Consequently, further constructs were developed to generate products with correctly processed N-termini.

[0208] Primers were designed to add a Tev protease (Tobacco Etch Virus cysteine protease) cleavage site between the N-terminal methionine and the histidine at the N- terminus of Exendin-4. This allows for removal of the methionine and the Tev recognition sequence to give the mature N-terminus of Exendin-4 (histidine). This can be done post- production or the Tev protease can be co-expressed to cleave the recognition sequence during production, for instance, as an intein (Ge, X., Yang, D. S. C, Trabbic-Carlson, K., Kim, B., Chilkoti, A. and Filipe, C. D. M. Self-Cleavable Stimulus Responsive Tags for Protein Purification without Chromatography. J. Am. Chem. Soc. 127, 11228-11229, 2005). The Tev Exendin-4 sequence is shown in Figure 3A. Figure 3B shows additional sequences added, labeled as "Linker Tev," provide a better target for the Tev protease.

[0209] An alternative route to obtaining a correctly processed N-terminus for Ex-4 is to use a leader or signal sequence that directs the product to the periplasm and which is cleaved by a signal peptidase in the process. In this instance, a signal sequence, DsbA, that directs the transcript to the signal recognition particle for direct secretion of the polypeptide into the periplasm is given. (See Figure 4A). The plasmid pET24d-DsbA-Ex-4 ELP1-90 is shown in Figure 4B.

[0210] While this example illustrates the preparation of therapeutic agents with

Exendin-4 sequences, such sequences can be replaced with GLP-1 , insulin, Factor VIIλ/lla, or other therapeutic protein listed in Table 1 , generated in exactly or a similar manner as detailed for Exendin-4.

EXAMPLE 6: GLP1-ELP FUSION PROTEIN

[0211] The ELP plasmid constructs were used to prepare two GLP1-ELP fusion proteins, GLP1 (A8G,7-37)ELP1-90 and GLP1(A8G,7-37)ELP1-120. The plasmid contructs,

fusion-encoding nucleotide sequence, as well as the amino acid sequence of the resulting fusion proteins are shown in Figures S and 6.

[0212] Both constructs contain an N-terminal Tev protease site to allow processing to the mature form where His 7 of GLP1 is at the N-terminus. The processed fusion proteins have calculated molecular weights of about 39,536 and about 50,828, respectively.

EXAMPLE 7: FVII ELP FUSION PROTEIN

[0213] The coagulation factor VII (FVII) gene was modified by PCR from a cDNA clone (Oragene) to add restriction sites at the 5' and 3' ends for cloning into the ELP- containing vector. At the 5 1 end an Nhe\ site was added and at the 3' end a λ/ofl site was added. . The DNA and amino acid sequences of the Factor VII gene are shown in the accompanying Sequence Listing as SEQ ID NOS: 34 and 33, respectively. The DNA sequences of the 5' and 3' primers used to PCR amplify the factor VII (FVII) gene were:

P13: CTAGCTAGCATGGTCTCCCAGGCCCTC (SEQ ID NO.: 49)

P14: TATTCTTGCGGCCGCGGGAAATGGGGCTCGCAG (SEQ ID NO.: 50)

[0214] The resulting PCR fragment was digested with the restriction enzymes Nhe\ and λ/ofl and ligated into the plasmid pcDNA3.1+ ELP1-90 previously digested with the restriction enzymes Nhe\ and λ/ofl (Figure 7A).

[0215] The resulting plasmid, pcDNA3.1+ FVII-ELP1-90, was transiently transfected into HEK293 cells and culture media harvested. The ELP fusion was purified by phase transition (Figures 9 and 10 ).

[0216] The nucleotide and amino acid sequences of the FactorVII-ELP fusion is shown in Figure 7B. As shown, the FactorVII-ELP fusion protein contains a Tev protease linker between the FactorVII component and the ELP component. This linker is optional.

EXAMPLE 8: INSULIN ELP FUSION PROTEIN

[0217] The cDNA for the human insulin gene is modified at the 5' and 3' ends for insertion in to pET24d-ELP1-90. The 5' primer adds an N-terminal methionine for bacterial expression and an λ/cfel restriction enzyme site. The 3' primer adds an Xho\ restriction enzyme site. The PCR product and the plasmid are both digested with the restriction enzymes λ/del and Xho\ and ligated together. The sequence of the insulin (Chains B, C, and A fused to ELP1 is shown in Figure 8A.

[0218] Correct insertion is determined by restriction digest and DNA sequencing.

The resulting plasmid, designated pET24d lnsulin-ELP1-90, is shown in Figure 8B.

[0219] The native insulin form is generated after recovery from E. coli by treatment with trypsin and carboxypeptidase B to remove the C-peptide chain.

[0220] For correct processing of the N-terminus of the B-chain similar modifications to those made for the Exendin-4 fusion (protease cleavage site, signal sequence) can be implemented (see Example 4). Alternatively, the first two residues can be Met - Arg, which can also be removed by trypsin digestion in production of the final material (R. M. Belagaje, S. G. Reams, S. C. Ly and W. F. Proutv, Increased production of low molecular weight recombinant proteins in Escherichia coli. Protein Sci. 6, 1953-1962, 1997).

[0221] Additional constructs would place the insulin cDNA at the 3' end of the ELP for a C-terminal fusion, add linkers between the Insulin and ELP sequences, and/or use modified forms of insulin which have no C-peptide (single chain insulins as described) removing the need for additional processing.

EXAMPLE 9: SYNTHESIS OF THE ELP GENE FOR CONJUGATION

[0222] A gene encoding a 50 amino acid sequence was constructed from chemically-synthesized oligonucleotides using standard molecular biology protocols. The 50 amino acid sequence contained 10 repeats of the pentapeptide VPGXG (SEQ ID NO: 3), where the guest residues (V, G, and A in a 5:3:2 molar ratio) were selected to provide a Tt of 40 0 C. The gene was oligomerized end-to-end by standard molecular biology techniques, to produce an oligomeric ELP gene. Additionally a single 50 amino acid sequence was constructed containing the 10 repeat pentapeptide VPGXG (SEQ ID NO: 3) polypeptide where the guest residues were V, G, A and C in a 4:3:2:1 molar ratio. This sequence could be added at any cycle of the oligomerization process to introduce a single cysteine residue into the final construct at a chosen point along the length of the construct.

[0223] The example given here is with the ELP designated 1-90. This is based on the VPGXG (SEQ ID NO: 3) motif where X is a V, G or A in the ratio 5:3:2 in a 10-unit repeat, repeated 8x with a final (C-terminal) 10-unit repeat where X is a V, G, A and C in the ratio 4:3:2:1 , i.e., [(VPGXG)10]9 (SEQ ID NO.: 3).

[0224] Alternatively, the residue could be one of either arginine, lysine, aspartic acid or glutamic acid. The purpose of these amino acids is to provide a reactive side chain for the chemical conjugation of, for example, insulin. In this particular case the use of an ELP would be to extend the circulating half-life of the therapeutic protein (e.g., insulin) to provide

prolonged basal glucose control. Conjugated to an ELP that transitions at body temperature, the insulin would form a precipitated depot at the site of injection in a similar manner to Lantus® (Sanofi Aventis) but without the requirement for formulation in acidic (pH 4.0) conditions with m-cresol for a more tolerable injection.

EXAMPLE 10: POTENCY AND HALF-LIFE OF FACTOR VII-ELP

[039] Figure 11 shows the activation of Factor X by FactorVI Ia-ELP 1-90, and by

Factor Vila as a comparison. Factor VII-ELP was produced in HEK cells. Factor Vila was derived from human plasma. As shown, FactorVlla-ELP retains full activity.

[040] When administered to rats by i.v., Factor VII-ELP demonstrated a half-life of about 690 minutes. In contrast, Factor VII demonstrated a half-life of 45-60 minutes. Half- life in this example was measured by sandwich ELISA for FactorVII. Figure 12.

[041] Also in contrast, the reported half-life for NovoSeven™ is 45 minutes, the reported half-life for FactorVlla-albumin. fusion is 263 minutes, and the reported half-life for Factor VIIa-PEG is 300 minutes in mice and 600 minutes in dog.

EXAMPLE 11 : GLP-1 ( OR EXENDIN-4) IN VITRO BIOASSAY

[0132] Activation of the GLP-1 receptor (GLP1 R) results in production of cAMP secondary messenger within the cell. Therefore, GLP-1 or Exendin-4 analogs and corresponding therapeutic agents may be tested by their ability to activate GLP 1 R on the cell surface and produce cAMP.

[0133] For this bioassay CHO cells transfected with cDNA coding for GLP1 R are used. These cells respond to stimulation by GLP-1 and produce high levels of cAMP. Log phase growing cells are plated and increasing concentrations of test compounds (e.g., therapeutic agent of the invention, or GLP-1 or exendin-4 functional analog) are added to the cells. After an appropriate incubation period (usually 15-60 min) in physiological buffer at 37°C the cAMP produced is measured using a CatchPoint cAMP assay kit from Molecular Devices (Sunnyvale, CA). The EC 50 of each test compound as compared to GLP-1 peptide or Exendin-4 peptide (or as compared to an unfused or unconjugated counterpart of a therapeutic agent of the invention) is indicative of the changes in activity due to a specific modifications introduced into the peptide, or due to particular chemical or recombinant coupling to an ELP component.

[0134] As shown in Figure 13, both GLP1-ELP (PB0868) and Exendin-4-ELP (PB

0859) maintain high activity in vitro, shown in comparison to Exendin alone. It is of note that the specific activity of Albugon® and Liraglutide® run 50-100 fold less than the exendin peptide.

EXAMPLE 12: GLP-1 i OR EXENDIN-4) IN VIVO BIQASSAY

[0135] The activity of GLP-1 or Exendin analogues or corresponding therapeutic agents may be tested in animals. For this assay, normal or diabetic animals may be used. Diabetic animals with blood glucose concentration 300-500 mg/dl are injected with different doses of GLP-1 or Exendin analogues or corresponding therapeutic agent, and changes in blood glucose monitored with a glucometer. The drop in glucose at different times points post administration is compared to that resulting with standard amounts of GLP-1 or Exendin-4 peptide, or compared to an unfused or unconjugated counterpart of a therapeutic agent of the invention. Alternatively, the blood glucose excursion in normal or diabetic animals during specific time period after administration of exogenous glucose is compared to GLP-1 or Exendin-4 (or to unfused or unconjugated counterparts of therapeutic agents). In this way the activity of the analogues and fusion proteins can be compared to the natural peptides.

[0136] Figure 14 shows the pharmacokinetics of GLP1-ELP1-120 in rats administered both by i.v. and subcutaneously. Three rats were used for each time point. The dose was ~10mg/kg. The T 1/2 when administered by i.v. was about 12.9 hours. The l m when administered subcutaneously was about 8.6 hours.

[0137] Figure 15 shows the pharmacokinetics of GLP1-ELP1-120 in rabbits administered both by i.v. and subcutaneously. Three rabbits were used for each time point. The dose was ~1 mg/kg. The T 1/2 when administered by i.v. was about 20 hours. The Ti /2 when administered subcutaneously was about 24 hours.

[0138] Figure 16 shows the sustained glycemic control in diabetic mice with GLP1-

ELP1-90.

[0139] All reference cited herein are hereby incorporated by reference in their entireties. While the invention has been has been described herein in reference to specific aspects, features and illustrative embodiments of the invention, it will be appreciated that the utility of the invention is not thus limited, but rather extends to and encompasses numerous other variations, modifications and alternative embodiments, as will suggest themselves to those of ordinary skill in the field of the present invention, based on the disclosure herein