Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSION PROTEIN OF THERAPEUTIC POLYPEPTIDE WITH IMPROVED PHARMACOKINETIC PROFILE AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2014/101287
Kind Code:
A1
Abstract:
The present invention provides a fusion protein comprising a therapeutic polypeptide fused to one or more flexible unstructured polypeptides and a trimeric scaffold protein. The flexible unstructured polypeptide sequence within the fusion protein is exhibited as one or more pCloud sequences derived from human fibrinogen alpha chain, and may be flanked by a proteinous connecting moiety of human origin. Also provided are pharmaceutical compositions comprising the fusion protein, nucleic acid molecules enconding the fusion protein, vectors containing the nucleic acids, host cells transformed with the vectors, and methods of making the fusion proteins of the invention, and use thereof.

Inventors:
QIAN XINGUO (CN)
HONG WEI (CN)
MA XIAOYU (CN)
Application Number:
PCT/CN2013/001602
Publication Date:
July 03, 2014
Filing Date:
December 19, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BEIJING ANXINHUAIDE BIOTECH CO LTD (CN)
International Classes:
C07K19/00; A61K38/00; A61P3/08; A61P35/00; C12N15/62; C12N15/63
Foreign References:
EP2441776A12012-04-18
US5961973A1999-10-05
Other References:
OHASHI T. ET AL.: "An experimental study of GFP-based FRET, with the application to intrinsically unstructured proteins.", PROTEIN SCIENCE, vol. 16, no. 7, July 2007 (2007-07-01), pages 1429 - 1438, XP055270976
LIU W. ET AL.: "The mechanical properties of single fibrin fibers.", JOURNAL OF THROMBOSIS AND HAEMOSTASIS., vol. 8, no. 5, May 2010 (2010-05-01), pages 1030 - 1036, XP055135666
See also references of EP 2935338A4
Attorney, Agent or Firm:
CHINA PATENT AGENT (H.K.) LTD (Great Eagle Centre23 Harbour Road, Wanchai, Hong Kong, CN)
Download PDF:
Claims:
CLAIMS

1. A fusion protein comprising a therapeutic polypeptide fused to one or more flexible un-structured polypeptide linkers and a scaffold protein, wherein the scaffold protein forms a homo-trimer in solution, the flexible un-structured polypeptide linker contains 1 to 3000 amino acid residues, wherein the sum of G, S, E, A, P and T constitutes more than 90% of the flexible un-structured polypeptide sequence, and the flexible un-structured polypeptide sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm, wherein the fusion protein exhibits an improved pharmacokinetic profile when administered to a subject compared with the therapeutic polypeptide by itself.

2. The fusion protein of claim 1 , wherein the fusion protein is configured, from N-terminus to C-terminus, using the following formula:

(Linker)m-TP-(Linker)n -Scaffold - (Linker)k Or

(Linker)m- Scaffold - (Linker)n -TP-(Linker)k wherein:

(a) Linker is the flexible un-structured polypeptide linker characterized in claim 1 ;

(b) TP is a therapeutic polypeptide selected from, but not limited to the group consisting of human glucagon-like peptide- 1 (GLP- 1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m+ n+k

>=1 ; wherein the digits indicate the number of presence of the designated polypeptides.

3. A fusion protein comprising a therapeutic polypeptide fused to one or more flexible un-structured polypeptide linkers and a trimeric scaffold protein, wherein within the fusion protein, the therapeutic polypeptide is connected with the flexible un-structured polypeptide sequence via a proteinous connecting moiety of human origin, wherein the fusion protein exhibits an improved pharmacokinetic profile when administered to a subject compared with the therapeutic polypeptide by itself.

4. The fusion protein of claim 3, wherein the fusion protein is configured, from N-terminus to C-terminus, according to the following formula: (Linker)m-TP-Loop-PCM-(Linker)n -Scaffold-(Linker)k or

(Linker)m-Scaffold -(Linker)n -PCM-Loop-TP- (Linker)k wherein:

(a) Linker is the flexible un-structured polypeptide linker characterized in claim 1 ;

(b) TP is the therapeutic polypeptide selected from, but not limited to the group consisting of human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m+n+k >= 1 , wherein the digits indicate the number of presence of the designated polypeptides;

(e) PCM is the proteinous connecting moiety of human origin; and

(f) Loop is a flexible loop, wherein the flexible loop has variable lengths from 0 to 100 residues, the flexible loops are rich in glycine (G) and serine (S), the flexible loops may also contain glutamate(E), alanine (A), proline (P) and threonine(T), and the flexible loops have greater than 95% unstructured random coil formation as determined by GOR algorithm.

5. The fusion protein of any preceding claim, wherein the flexible un-structured polypeptide linker is selected from the group consisting of (G2S)n, (G3S)n, (G4S)n, (G5S)n, (GS)n, (G2S2)n, (GS2)n, (GS3)n, (S2G)n, (S3G)n, (S4G)n, (S5G)n, (SG)n, (S2G2)n, (SG2)n, (SG3)n where n is an integer, , GGGSGGGG, GGGSGGGGS, GSGG, GGGSGGG, GGGGSGGG, GGGSGG, GGGGSGG, GGGGSGGGGSGGGGSGGGGS,

GGGGSGGGGSASSASTGGPSGGGGSGGGGS,

GGGGSGGGGSTASSASTGGPSGGGGSGGGGSAPSSGSTSGGTAAGGG GSGGGGS and GGGSGGGSGGGSTASSASTKGPSGGGSGGGSGGGSAPSSKSTSGGTAA GGGSGGGSGGGS.

6. A fusion protein comprising a therapeutic polypeptide fused to one or more pCloud sequences and a scaffold protein, wherein the scaffold protein forms a homo-trimer in solution, the pCloud sequence is a flexible un-structured polypeptides with at least 100 to about 3000 amino acid residues, wherein the fusion protein exhibits an improved pharmacokinetic profile when administered to a subject compared with the therapeutic polypeptide by itself.

7. The fusion protein of claim 6, wherein the pCloud polypeptide is characterized in:

(a) the total pCloud amino acid residues is at least 100 to about 3000 amino acid residues; (b) the pCloud polypeptide sequence is generated by use of some or all of the fragments derived from human fibrinogen alpha chain, and in pCloud sequence, the fibrinogen fragments are flanked by flexible loops with various lengths from 1 to 100 residues, therefore the pCloud polypeptide is primarily human originated and has low immunogenicity when administered to human; (c) the pCloud sequence is rich in glycine (G), serine (S) and Glutamate (E), the pCloud also contains alanine (A), proline (P), arginine (R) and threonine (T), the sum of G, S, E, A, P and T constitutes more than 90% of the pCloud sequence; (d) the pCloud sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm; and (e) the pCloud sequence does not contain any T-cell epitopes as predicted by TEPITOPE algorithm.

8. The fusion protein sequence of claim 6 or 7, wherein in pCloud sequence, the human fibrinogen fragments are flanked by flexible loops with variable lengths from 0 to 100 residues, the flexible loops are rich in glycine (G) and serine (S), the flexible loops may also contain glutamate(E), alanine (A), proline (P) and threonine(T), the flexible loops have greater than 95% unstructured random coil formation as determined by GOR algorithm.

9. The fusion protein of any one of the preceding claims, comprising the therapeutic polypeptide, one or more pCloud sequences, and the scaffold protein, wherein the pCloud sequence can be placed at either or both of the N-terminal and the C-terminal end of the therapeutic polypeptide, and the pCloud sequences can also be placed at either or both of the N-terminal and the C-terminal end of the scaffold protein, wherein the fusion protein is configured, from N-terminus to C-terminus, using the following formula:

(pCloud)m-TP-(pCloud)n -Scaffold - (pCloud)k Or

(pCloud)m- Scaffold - (pCloud)n -TP-(pCloud)k wherein:

(a) pCloud is the pCloud polypeptide characterized in claim 7 or 8;

(b) TP is a therapeutic polypeptide selected from, but not limited to the group consisting of human glucagon-like peptide- 1 (GLP-1 ), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m + n+k >= 1 , the digits indicate the number of presence of the designated polypeptides.

10. The fusion protein of any one of the preceding claims, wherein the therapeutic polypeptide is connected with the pCloud sequence via a proteinous connecting moiety of human origin, wherein a flexible loop may be utilized to fuse the therapeutic polypeptide and the proteinous connecting moiety, the flexible loop is characterized in claim 8, the proteinous connecting moiety can be fused at either the N-terminal or the C-terminal end of the therapeutic polypeptide, wherein the fusion protein is configured, from N-terminus to C-terminus, according to the following formula: (pCloud)m-TP-Loop-PCM-(pCloud)n -Scaffold- (pCloud)k or

(pCloud)m-Scaffold -(pCloud)n -PCM-Loop-TP- (pCloud)k wherein:

(a) pCloud is the pCloud polypeptide characterized in claim 7 or 8;

(b) TP is the therapeutic polypeptide selected from, but not limited to the group consisting of human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m+ n+k >=1 , the digits indicate the number of presence of the designated polypeptides;

(e) Loop is a flexible loop which is characterized in claim 8; and

(f) PCM is the proteinous connecting moiety of human origin.

1 1. The fusion protein of any preceding claim, wherein the proteinous connecting moiety is a proteinous sequence having an elongated shape of human origin.

12. The fusion protein of any of the preceding claims, wherein the proteinous connecting moiety is a human Fibronectin type III domain.

13. The fusion protein of claim 12, wherein the human Fibronectin type III domain is selected from the group consisting of Tenascin, Usherin, Titin, tripartite motif (TRIM) family members, tissue factor, TIE1 , TIE2, SPEG, SORL1 , SDK1 , ROBOl , ROB02, SDK2, Receptor-type tyrosine-protein phosphatase, prolactin receptor, L I CAM, NCAM 1 , NCAM2, myomesin 1 , myomesin 2, Myosin-binding protein C, LIFR, Leptin receptor, Integrin, Insulin receptor, Contactin, Collagen, Cytokine receptor-like factor, Inteferon receptor, Growth hormone receptor, fibronectin, leucine rich transmembrane protein (FLRT) members, IL, ephrin type-A receptor, ephrin type-B receptor, IL-6R, gp l 30, IL1 1RA, IL12RB, IL20RB, IL23R, IL27RA and IL31RA.

14. The fusion protein of any of preceding claims, wherein the scaffold protein is selected from the group consisting of human collagen noncollagenous (NC) domains which form stable homo-trimers in solution, C lq-like molecular structures which form homo-trimers in solution, TNF-like molecular structures which form homo-trimers in solution, and proteins with C-type lectin-like domains (CTLD) which form homo-trimers in solution.

15. The fusion protein of any of the preceding claims, wherein the scaffold protein is selected from the group consisting of the NCI domain within Multiplexin type of human Collagen, NC2 domain within FACIT type of collagen, human C lq A chain, Clq B chain, Cl q C chain, cbln family members, human EMILIN-1 , multimerin, ACRP30/adiponectin, adipolin, resistin, resistin-like molecule (RELM) hormone family members, human TNFalpha, TNFbeta, TRAIL, RANK ligand, Fas ligand, CD 30 ligand, CD40 ligand, CD27 ligand, OX40L, CD137, mannan-binding lectin (MBL), surfactant protein A (SP-A), surfactant protein D (SP-D), collectin liver 1 (CL-L1 ), collectin placenta 1 (CL-P1 ), conglutinin, collectin of 43 kDa (CL-43) and collectin of 46 kDa (CL-46), Langerin, Tetranectin and functional variants thereof.

16. The fusion protein of any of the preceding claims, wherein the scaffold protein is preferably selected from NCI domain within Multiplexin type of human Collagen such as NC I domain from collagen XV and collagen XVIII.

17. The fusion protein of any of the preceding claims, wherein the therapeutic polypeptide is selected, but not limited to the group consisting of human glucagon-like peptide- 1 (GLP-1 ), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof.

18. The fusion protein of any of the preceding claims, wherein the fusion protein comprises: a therapeutic polypeptide selected from the group consisting of GLP- 1 , GLP- 1 (A8G/G22E), GLP- 1 (A8G/G22E/R36G) and GLP-1(A8G/G22E/R36S); a flexible loop; a proteinous connecting moiety selected from the group consisting of human fibronectin domain 7 (Fn7), human fibronectin domain 8 (Fn8), and human Tenascin C fibronectin type III domain 3 (TNCfn3); a pCloud polypeptide sequence defined in claim 7 or 8; and a scaffold protein selected from the group consisting of human collagen XVIII NC I domain (COL18NC 1 ), human collagen XV NC I domain (COL15NC1 ), human collagen XIV NC2 domain (COL19NC2) and ACRP30 C I q-like domain.

19. The fusion protein of any of the preceding claims, wherein the fusion protein comprises: GLP- 1 (A8G/G22E/R36S) or GLP- 1 (A8G/G22E/R36G), a flexible loop, Fn8, a pCloud polypeptide, and COL18NC 1.

20. The fusion protein of any preceding claims, wherein the fusion protein is selected from the group consisting of SEQ ID NOs: 1-31.

21. The fusion protein of any one of the preceding claims, comprises a protein sequence that has at least 90% sequence identity to a sequence selected from any one of SEQ ID NOs: 1 -31.

22. The fusion protein of any of the preceding claims, wherein the length of the flexible unstructured polypeptide and/or the pCloud sequence can be varied to adjust the apparent molecular weight (hydrodynamic radius and/or radius of gyration) and/or the in vivo half life of the fusion protein.

23. The fusion protein containing the pCloud polypeptide of any of the preceding claims, wherein the flexible unstructured polypeptide and/or the pCloud sequence enhance the thermostability and solubility of a therapeutic polypeptide when connected to the therapeutic polypeptide.

24. A polynucleotide sequence encoding the fusion protein of any of the preceding claims.

25. An expression vector comprising the polynucleotide sequence of claim 24 and expression control elements.

26. A pharmaceutical composition comprising the fusion protein of any of the preceding claims 1 -23 and a pharmaceutically acceptable carrier.

27. The fusion protein of any of the preceding claims 1-23, wherein the fusion protein can be further modified by PEGylation to extend the in vivo half life of the fusion protein and PEG conjugate, the PEG moiety may have a molecular weight of between 2kDa and 100 kDa.

28. A method of improving the pharmacokinetic property of a therapeutic polypeptide, comprising the steps of fusing the therapeutic polypeptide to one or more flexible unstructured polypeptides (and/or pCloud polypeptides) and a trimeric scaffold protein, the fusion protein may further contain a proteinous connecting moiety of human origin, the fusion protein achieves a property characterized in that (a) the terminal half-life of the fusion protein is significantly longer as compared to the terminal half- life of the therapeutic polypeptide by itself; (b) stability and solubility under physiologic conditions of the fusion protein are improved as compared to the stability and solubility of the therapeutic polypeptide by itself.

Description:
FUSION PROTEIN OF THERAPEUTIC POLYPEPTIDE

WITH IMPROVED PHARMACOKINETIC PROFILE AND USE THEREOF

Field of the Invention

The present invention relates generally to a fusion protein with therapeutic efficacy. In particular, the present invention relates to a method of improving the half life of a therapeutic polypeptide by fusing with one or more flexible un-structured polypeptide sequences and a trimeric scaffold protein and the use thereof.

Background of the Invention

Many therapeutic polypeptides suffer from short terminal in vivo half life and poor thermal stability when injected into a subject. Short plasma half life is commonly due to fast renal clearance as well as to enzymatic degradation occurring during systemic circulation. The long half life time is usually required for a therapeutic polypeptide to achieve its optimal efficacy. Increasing the in vivo residence times of the therapeutic polypeptides could decrease their dosing frequencies and make them more convenient for the patients to use.

PEGylation has been widely utilized to extend the half life of a therapeutic polypeptide (see review paper [1 -4], patents 1-9). PEGylation changes the physical and chemical properties of the biomedical molecule, such as its conformation, electrostatic binding, and hydrophobicity, and results in an improvement in the pharmacokinetic behavior of the drug. In general, PEGylation improves drug solubility and decreases immunogenicity. PEGylation also increases drug stability and the retention time of the conjugates in blood. However, PEGylation has severe consequences for the biological activities of the protein. The activity of the PEGylated protein usually reduces by 20-50 fold [2, 5](patents 1 -9). In addition, the site for PEGylation needs to be carefully decided to avoid interfering with the active site of the therapeutic polypeptide. For some short peptides such as GLP-1 , PTH and Calcitonin, it would be difficult to choose the proper site for PEGylation without disturbing the biological activity of the peptides. Moreover, PEG is a heterogeneous mixture of related polymers, its conjugation to a therapeutic polypeptide results in numerous distinct species with similar molecular sizes and chemical properties. This complicates the purification and increases the production costs of the PEGylated products.

It has been reported that fusion of a therapeutic polypeptide with human IgG Fc fragment or human serum albumin (HSA) may significantly increase the half life of the therapeutic polypeptide [6-9] (patents 10, 1 1 , 12). However, recombinant fusion protein with IgG Fc fragment or HSA needs to be produced from eukaryotic systems such as mammalian cell lines or yeast cells, which significantly increases the cost of the recombinant protein.

Summary of the invention

The present invention is directed to enhance the pharmaceutical properties, stability, solubility and safety of the therapeutic polypeptides. The present invention is particularly useful for improving the pharmacokinetic properties, such as in vivo terminal half-life, of a therapeutic polypeptide.

In one aspect, the present invention provides a fusion protein comprising a therapeutic polypeptide fused to a scaffold protein which forms a homo-trimer in solution. The fusion protein further comprises one or more flexible un-structured polypeptide sequences. In some embodiments, the fusion protein further comprises a proteinous connecting moiety (PCM) of human origin. In a particular embodiment, the proteinous connecting moiety is a proteinous sequence having an elongated shape, such as a human Fibronectin type III domain.

The flexible un-structured polypeptide sequence contains 1 to 3000 amino acid residues, wherein the sum of G, S, E, A, P and T constitutes more than 90% of the flexible un-structured polypeptide sequence; and the flexible un-structured polypeptide sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm[10]. Fusing the therapeutic polypeptide with one or more flexible unstructured polypeptide sequences and the trimeric scaffold protein can significantly increase the apparent molecular weight of the fusion protein and improve the in vivo half life of the therapeutic polypeptide. Moreover, this method renders the therapeutic polypeptide with tri-valency, which may greatly enhance the potency and efficacy of the therapeutic polypeptide (reviewed in [1 1] ). This novel method provided by the invention, termed as "Trident technology", can efficiently increase the hydrodynamic radius and/or the radius of gyration (Rg) of the polypeptide molecule to extend its half life in vivo. The flexible unstructured polypeptide sequences and the proteinous connecting moiety (PCM) within the fusion protein contribute significantly to increasing the apparent molecular weight of the fusion proteins.

In the present invention, the therapeutic polypeptide may be fused with one or more flexible unstructured polypeptides and the trimeric scaffold protein in a number of ways. In some embodiments, the fusion protein of the present invention may be configured, from N-terminus to C-terminus, using the following formula:

(Linker) m -TP-(Linker) n -Scaffold - (Linker) k Or

(Linker) m - Scaffold - (Linker) n -TP-(Linker) k wherein:

(a) Linker is the flexible un-structured polypeptide linker above;

(b) TP is a therapeutic polypeptide;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m + n+k

>=1. These digits indicate the number of presence of the designated polypeptides.

In some embodiments of the invention, the fusion protein may further contain a proteinous connecting moiety of human origin. The therapeutic polypeptide is connected with the flexible un-structured polypeptide sequence via the proteinous connecting moiety of human origin. The fusion protein exhibits an improved pharmacokinetic profile when administered to a subject compared with the therapeutic polypeptide by itself. The fusion protein may be configured, from N-terminus to C-terminus, according to the following formula:

(Linker) m -TP-Loop-PCM-(Linker) j -Scaffold-(Linker) k or

(Linker) m -Scaffold -(Linker) n -PCM-Loop-TP- (Linker) k wherein:

(a) Linker is the flexible un-structured polypeptide linker characterized above;

(b) TP is the therapeutic polypeptide;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution;

(d) m is either 0 or 1 ; and n is either 0 or 1 , j is either 0 or 1 , k is either 0 or 1 , and m+n+j+k >=1. These digits indicate the number of presence of the designated polypeptides.

(e) PCM is the proteinous connecting moiety of human origin; and

(f) Loop is a flexible loop which refers to the protein sequence which has the variable lengths from 0 to 100 residues. These flexible loops are rich in glycine (G) and serine (S). These flexible loops may also contain glutamate(E), alanine (A), proline (P) and threonine(T). These flexible loops have greater than 95% unstructured random coil formation as determined by GOR algorithm.

In a preferred embodiment of the invention, the flexible un-structured linker is exhibited as one or more flexible un-structured pCloud polypeptides. The pCloud sequence is characterized in: (a) the total pCloud amino acid residues is at least 100 to about 3000 amino acid residues; (b) the pCloud polypeptide is generated by use of some or all of the fragments derived from human fibrinogen alpha chain (table 1). In pCloud sequence, the fibrinogen fragments are flanked by flexible loops with various lengths. Therefore the pCloud polypeptide is primarily human originated and has low immunogenicity. (c) the pCloud polypeptide is rich in glycine (G), serine (S) and Glutamate (E). The pCloud polypeptide also contains alanine (A), proline (P), arginine (R) and threonine (T).

The sum of G, S, E, A, P and T constitutes more than 90% of the pCloud sequence, (d) The pCloud sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm [10]; and (e) the pCloud sequence does not contain any T-cell epitopes as predicted by TEPITOPE algorithm [ 12].

In some embodiments, the pCloud polypeptide represents a flexible unstructured polypeptide originated from human fibrinogen alpha chain. The fragments derived from human fibrinogen alpha chain (listed in table 1) can be utilized as the building blocks to constitute the pCloud polypeptide. In the pCloud sequence, the human fibrinogen alpha chain fragments are flanked by flexible loops with variable lengths from 0 to 100 residues. Attaching one or more pCloud polypeptide to a therapeutic polypeptide may significantly increase the apparent molecular weight of the therapeutic polypeptide and improve the in vivo half life of the therapeutic polypeptide. The in vivo half life of the therapeutic polypeptide connected with the pCloud sequence can be adjusted by varying the length of the pCloud sequence. More importantly, the pCloud sequence is generated based on the human fibrinogen alpha chain, therefore, the pCloud polypeptide may not stimulate the immune responses from human patients when administrated.

In some embodiments, the scaffold protein of the invention is selected from the group consisting of human collagen noncollagenous (NC) domains which form stable homo-trimers in solution, proteins which form homo-trimers in solution with Clq-like molecular structures, proteins which form homo-trimers in solution with TNF-like molecular structures, and proteins with C-type lectin-like domains (CTLD) which form homo-trimers in solution. In some embodiments, the scaffold protein is selected from the group consisting of the NC I domain within Multiplexin type of human Collagen, NC2 domain within FACIT type of collagen, human Cl q A chain, Cl q B chain, C l q C chain, cbln family members, human EMILIN- 1 , multimerin, AC P30/adiponectin, adipolin, resistin, resistin-like molecule (RELM) hormone family members, human TNFalpha, TNFbeta, TRAIL, RANK ligand, Fas ligand, CD 30 ligand, CD40 ligand, CD27 ligand, OX40L, CD 137, mannan-binding lectin (MBL), surfactant protein A (SP-A), surfactant protein D (SP-D), collectin liver 1 (CL-L1), collectin placenta 1 (CL-P1), conglutinin, collectin of 43 kDa (CL-43) and collectin of 46 kDa (CL-46), Langerin, Tetranectin and functional variants thereof. In the preferred embodiments, the NC I domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) is selected as the scaffold protein in the present invention.

In some embodiments, the therapeutic polypeptide is selected from the group consisting of human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof.

In some embodiments, the therapeutic polypeptide is connected with the pCloud sequence via a proteinous connecting moiety of human origin. In a particular embodiment, the proteinous connecting moiety is a proteinous sequence having an elongated shape, such as human Fibronectin type III domain.

In the particular embodiments of the invention, the fusion protein of the invention comprises, from N-terminus to C-terminus, a therapeutic polypeptide selected from the group consisting of GLP- 1 , GLP- 1 (A8G/G22E), GLP- 1 (A8G/G22E/R36S) and GLP-1 (A8G/G22E/R36G); a flexible loop; a proteinous connecting moiety selected from the group consisting of Fn7, Fn8 and TNCfn3; a pCloud sequence; and a scaffold protein selected from the group consisting of COL18NC1 , COL15NC 1 , COL19NC2, and ACRP30 C l q-like domain.

In a preferred embodiment of the invention, the fusion protein of the invention comprises, from N-terminus to C-terminus, GLP- 1 (A8G/G22E/R36G), a flexible loop, Fn8, a pCloud sequence, and COL18NC1.

In another preferred embodiment of the invention, the fusion protein of the invention comprises, from N-terminus to C-terminus, a first pCloud sequence, human growth hormone, a second pCloud sequence and COL18NC 1.

The present invention also provides a polynucleotide sequence encoding the fusion protein, a pharmaceutical composition comprising the fusion protein and a pharmaceutically acceptable carrier, and an expression vector comprising the polynucleotide sequence and expression control elements.

In another aspect, the present invention provides a method of improving the pharmacokinetic property of a therapeutic polypeptide, comprising the steps of fusing the therapeutic polypeptide to one or more pCloud polypeptide and a trimeric scaffold protein. In some embodiments, the therapeutic polypeptide is connected with the pCloud sequence via a proteinous connecting moiety of human origin. The fusion protein of the present invention achieves a property characterized in that (a) the terminal half-life of the therapeutic polypeptide linked to the scaffold protein and one or more pCloud sequence is significantly longer as compared to the terminal half- life of the therapeutic polypeptide by itself; (b) stability and solubility under physiologic conditions of the therapeutic polypeptide linked to the scaffold protein and one or more pCloud sequence are improved as compared to the stability and solubility of the therapeutic polypeptide by itself.

Brief Description of the Drawings

Fig. 1 shows the schematic drawings illustrating some mechanisms of the present invention. The therapeutic polypeptide is shown as a red star in Fig. la and Fig. l c and as a red helix in Fig. lb. a) The therapeutic polypeptide is connected directly to the pCloud polypeptide and the scaffold protein, b) The therapeutic polypeptide is fused with the pCloud polypeptide and the scaffold protein via a proteinous connecting moiety of human origin, preferably a proteinous sequence with an elongated shape, c) The therapeutic polypeptide and the pCloud polypeptide can be fused at the opposite terminus of the scaffold protein.

Fig. 2. The amino acid sequence of the mature human fibrinogen alpha chain. The amino acid residue numbers of the fibrinogen alpha chain are listed. The 12 unstructured fragments containing primarily the residues glycine (G), serine (S) and Glutamate (E), proline (P), arginine (R) and threonine (T) are underlined. To further reduce the immunogenicity of these fragments, mutations Y277S, V379A and D396E were introduced. The residues Y277, V379 and D396 are in bold. Fig. 3 shows the gel filtration chromatography profiles for purified GLP- 1 (A8G/G22E/R36S)-Fn8-COL 18NC 1 ,

GLP- 1 (A8G/G22E/R36S)-Fn8-COL 18NC 1 -20,

GLP-l (A8G/G22E/R36S)-Fn8-COL18NC l-30, and GLP- l (A8G/G22E/R36S)-Fn8-COL18NCl -54 using the analytical column Superdex200 (GE Healthcare). In this figure, the profiles of these proteins are labeled as NC I , NCI -20, NCI -30 and NCI -54. The elution time for the molecular marker proteins (158Kd, 67Kd and 44Kd) are shown by arrows. The X-axis refers to elution time and the Y-axis refers to UV280 absorbance intensity.

Fig. 4 shows the pharmacokinetics profiles of the GLP-1 containing proteins (GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 ,

GLP- 1 (A8G/G22E/R36S)-Fn8-COL 18NC 1 -20,

GLP- l (A8G/G22E/R36S)-Fn8-COL18NC l-30 and GLP- 1 (A8G/G22E/R36S)-Fn8-COL 18NC 1 -54) in Sprague Dawley rats measured by use of the sandwich ELISA method. In this figure, the profiles of these proteins are labeled as NCI , NC20, NC30 and NC54, respectively. The vertical axis indicates the percentage of the measured protein concentration by use of sandwich ELISA method compared with C max .

Fig. 5 shows the gel filtration chromatography profiles for purified

GLP- 1 -Fn8, GLP- 1 -Fn8-COL 18NC 1 ,

GLP- l (A8G/G22E/R36G)-Fn8-p246-COL18NCl by use of the analytical column Superdex200 (GE Healthcare). In this figure, the profiles of these proteins are labeled. The elution time for the molecular marker proteins are shown by arrows. The X-axis refers to elution time and the Y-axis refers to UV280 absorbance intensity, respectively.

Fig. 6 shows the results of cAMP assays for GLP-1 (7-37) peptide, GLP-1 -Fn8-C0L18NC1 and GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NCl . This assay is based on competitive binding technique. A monoclonal antibody specific for cAMP becomes bound to the goat anti-mouse antibody coated onto the microplate. Following a wash to remove excess monoclonal antibody, cAMP present in a sample competes with a fixed amount of horseradish peroxidase (HRP)-labeled cAMP for sites on the monoclonal antibody. This is followed by another wash to remove excess conjugate and unbound sample. A substrate solution is added to the wells to determine the bound enzyme activity. The color development is stopped and the absorbance is read at 450nm. The intensity of the color is inversely proportional to the concentration of cAMP in the sample. The Y-axis refers to the OD 450 obtained by the plate reader and the X-axis refers to the concentration of GLP-1 (7-37) peptide, GLP-1-Fn8-C0L18NC1 and GLP 1 (A8G/G22E/R36G)-Fn8-p246-COL 18NC 1.

Fig. 7 shows the pharmacokinetics profiles of the fusion proteins GLP- 1 -Fn8-C0L18NC 1 , GLP-l (A8G/G22E/R36G)-Fn8-p246-COL18NCl , and GLP-l (A8G/G22E/R36G)-Fn8-p285-COL18NCl in Sprague Dawley rats. The protein concentration in the blood samples were measured by use of the sandwich ELISA method. In this figure, the profiles of these proteins are labeled as NC I , p246 and p285, respectively. The errors bars calculated from a group of six rats are labeled.

Fig. 8 shows the pharmacokinetics profiles of the fusion proteins GLP-l(A8G/G22E/R36G)-Fn8-p246-COL18NCl in three cynomolgus monkeys. The protein concentration in the serum samples were measured by use of the sandwich ELISA method. In this figure, the three cynomolgus monekys were labeled as #1 , 2 and 3, respectively.

Fig. 9 shows the Intraperitoneal glucose tolerance test (IPGTT) results of GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l in SD rats. In this figure, GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l is labeled as p246. In Fig. 9a, the rats were injected with saline, GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l at the dose of l Onmol/kg and 20nmol/kg, respectively. 8 hours after the administrations of the fusion protein (and control), the IPGTT experiments were conducted. The three curves indicated the glucose levels in the IPGTT for rats that received negative control (saline), GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NCl at the doses of lOnmol/kg and 20nmol/kg, respectively. In Fig. 9b, the rats were injected with saline, GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NCl at the dose of 20nmol/kg. 32 hours and 102 hours after the administrations of the fusion protein (and the control), the IPGTT experiments were conducted. The three curves indicated the glucose levels in the IPGTT for rats that 32 hours and 102 hours after administration of negative control (saline),

GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NCl respectively.

Definitions

The terms "polypeptide", "peptide", and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified, for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The terms "flexible unstructured polypeptide", "flexible unstructured polypeptide sequence", and "flexible unstructured linker" "flexible unstructured polypeptide linker" are used interchangeably in this invention. The flexible un-structured polypeptide sequence contains 1 to 3000 amino acid residues, wherein the sum of G, S, E, A, P and T constitutes more than 90% of the flexible un-structured polypeptide sequence; and the flexible un-structured polypeptide sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm[10].

The term "pCloud" polypeptide is characterized in: (a) the total pCloud amino acid residues is at least 100 to about 3000 amino acid residues; (b) the pCloud polypeptide sequence is generated by use of some or all of the fragments derived from human fibrinogen alpha chain. In pCloud sequence, the fibrinogen fragments are flanked by flexible loops with various lengths. Therefore pCloud is primarily human originated and has low immunogenicity when administered to human, (c) the pCloud sequence is rich in glycine (G), serine (S) and Glutamate (E). The pCloud also contains alanine (A), proline (P), arginine (R) and threonine (T). The sum of G, S, E, A, P and T constitutes more than 90% of the pCloud sequence, (d) The pCloud sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm; and (e) the pCloud sequence does not contain any T-cell epitopes as predicted by TEPITOPE algorithm. Within the pCloud polypeptide, the human fibrinogen fragments are flanked by flexible loops with variable lengths from 0 to 100 residues.

The term "flexible loop" in this invention refers to the protein sequence which has the variable lengths from 0 to 100 residues. These flexible loops are rich in glycine (G) and serine (S). These flexible loops may also contain glutamate(E), alanine (A), proline (P) and threonine(T). These flexible loops have greater than 95% unstructured random coil formation as determined by GOR algorithm. The flexible loops are generally the flexible unstructured polypeptide linkers with shorter lengths and more flexibility. A skilled artisan will appreciate that the flexible loop may be utilized in the fusion protein as a spacer to provide flexibility.

A "fragment" is a truncated form of a native protein. The term "variant" or "functional variant" of a protein refers to a modified version of the native protein which comprises substitutions, deletions and/or additions of one or several amino acids, and which substantially retains the biological activity of the native protein. For example, a variant protein may share at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity with the reference protein. Typically, conservative substitutions of amino acids are preferred which are well known to a skilled artisan. Deletions are preferably deletions of amino acids from regions not involved in the biological function of the protein. For example, GLP- 1 (A8G/G22E/R36G) is a functional variant of wild type GLP- 1 , which contains three substitutions of amino acids and which substantially retains or increases its biological activity as shown by the cAMP assay.

"Conjugated", "linked," "connected", "fused," and "fusion" are used interchangeably herein. These terms refer to the joining together of two more chemical elements or components, by whatever means including chemical conjugation or recombinant means. For example, two distinct proteins can be connected together by "in- frame fusion", which refers to the joining of two or more open reading frames (ORFs) to form a continuous longer ORF, in a manner that maintains the correct reading frame of the original ORFs. Thus, the resulting recombinant fusion protein is a single protein containing two or more segments that correspond to polypeptides encoded by the original ORFs (which segments are not normally so joined in nature). For another example, the two proteins can also be linked together by use of a chemical crosslinker, which results in a protein conjugate that contains two individual polypeptides connected by a crosslinker.

In the context of polypeptides, a "sequence" is an order of amino acids in a polypeptide in an amino to carboxyl terminus direction in which residues that neighbor each other in the sequence are contiguous in the primary structure of the polypeptide.

The terms "DNA", "polynucleotides", "nucleic acids", "nucleotides" and "oligonucleotides" are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Polynucleotides may have any three-dimensional structure, and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: coding or non-coding regions of a gene or gene fragment, loci (locus) defined from linkage analysis, exons, introns, messenger RNA (mPvNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.

The term "functional variant" of a protein refers to a modified version of the native protein which comprises substitutions, deletions and/or additions of one or several amino acids, e.g., less than 15 amino acids, or preferably less than 10 or 5 amino acids, and which substantially retains the biological activity of the native protein. Typically, conservative substitutions of amino acids are preferred which are well known to a skilled artisan. Deletions are preferably deletions of amino acids from regions not involved in the biological function of the protein. For example, GLP- 1 (A8G/G22E) and GLP- 1 (A8G/G22E/R36G) are the functional variants of wild type GLP- 1 , which contains two or three substitutions of amino acids and which substantially retains its biological activity such as increasing cAMP level. Detailed description of the invention

In one aspect of the present invention, there is provided a method to increase the half life of a therapeutic polypeptide by fusing the therapeutic polypeptide to one or more flexible un-structured polypeptide sequences and a scaffold protein. The scaffold protein can form a stable homo-trimer in solution. This method can efficiently increase the hydrodynamic radius and/or the radius of gyration (Rg) of the polypeptide molecule to extend its half life in vivo. Moreover, changing the length of the flexible unstructured polypeptide linker within the fusion protein can adjust the in vivo half life of the fusion protein in a tunable manner. In some embodiments, the fusion protein of the invention may further comprise a proteinous connecting moiety of human origin, preferably a proteinous sequence with an elongated shape. The proteinous connecting moiety can be connected to the therapeutic polypeptide via a flexible loop. The proteinous connecting moiety can be linked to the scaffold protein via a flexible, un-structured linker whose length is adjustable. This novel method provided by the invention, termed as "Trident technology", can efficiently improve the pharmacokinetics profile of the therapeutic polypeptide.

In a preferred embodiment of the invention, the flexible un-structured linker is exhibited as one or more un-structured pCloud polypeptides. Within the fusion protein, the therapeutic polypeptide, the pCloud polypeptides and the scaffold protein can be arranged in a number of manners. For example, in some embodiments, the therapeutic polypeptide is connected directly to the pCloud polypeptide and the scaffold protein (Fig. la). In some embodiments, the therapeutic polypeptide is fused with the pCloud polypeptide and the scaffold protein via a proteinous connecting moiety of human origin, preferably a proteinous sequence with an elongated shape (Fig. lb). In some embodiments, the therapeutic polypeptide and the pCloud polypeptide can be fused at the opposite terminus of the scaffold protein (Fig. lc).

In preferred embodiments of the present invention, the method of the invention may have several major advantages over the traditional PEGylation method or Fc/HSA fusion method. 1. In the method of the invention, PEGylation on the polypeptide molecule is not essential, therefore the biological activity of the therapeutic polypeptide is fully retained. 2. Because the scaffold protein forms a homo-trimer, the fusion protein of the therapeutic polypeptide with the scaffold protein may greatly increase the apparent size of the fusion protein to slow down renal filtration. Moreover, the trimer formation also renders the fusion protein tri-valency. This may greatly increase the activity of the therapeutic protein. 3. The length of the flexible unstructured polypeptide linker within the fusion protein plays an important role in determining the in vivo half life of the fusion protein. The method of the invention provides a platform to fine tune the in vivo half life of a therapeutic polypeptide by varying the length of the flexible unstructured polypeptide linker within the fusion protein. 4. The scaffold protein and pCloud polypeptide is preferably generated from human proteins, usually from human extracellular proteins, therefore, no foreign protein sequences are introduced into the fusion protein. The immunogenicity of the fusion proteins generated using the method is low. 5. In many cases, the recombinant fusion protein of the present invention can be generated using E.coli expression system, which eliminates the need of the expensive chemical synthesis process for some therapeutic polypeptides or the need of using the eukaryotic expression systems.

The flexible unstructured polypeptide sequences

In this invention, the flexible unstructured polypeptide sequences within the fusion protein play critical roles in extending the half-life of the therapeutic polypeptide. The lengths of the flexible unstructured polypeptide sequences play important role in determining the hydrodynamic radius and/or the radius of gyration of the fusion protein. The primary sequences of the flexible unstructured polypeptide heavily affect the stability and solubility of the fusion protein.

The term "flexible un-structured polypeptide" refers to an amino acid sequence which is flexible in movement and which does not form any regular stable secondary and tertiary protein structures. The flexible un-structured polypeptide sequence contains 1 to 3000 amino acid residues, wherein the sum of G, S, E, A, P and T constitutes more than 90% of the flexible un-structured polypeptide sequence; and the flexible un-structured polypeptide sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm[10].

If the therapeutic polypeptide is a relatively large protein (such as Interferon, Growth hormone, Erythropoietin, G-CSF, or TNFR2, usually a protein with more than 100 amino acid residues), it may be directly fused to the scaffold protein through a flexible, un-structured polypeptide linker. In some other cases, the therapeutic polypeptide might be a short peptide (such as GLP-1 , Exenatide, GLP-2, C-peptide, Calcitonin or PTH, usually a peptide not more than 100 residues). To efficiently utilize our method, the fusion protein may further contain a proteinous connecting moiety of human origin, preferably a proteinous sequence with an elongated shape, such as the human Fibronectin type III domain. The therapeutic polypeptide and flexible unstructured polypeptide linker is connected by use of the proteinous connecting moiety. The proteinous connecting moiety can further increase the hydrodynamic radius and/or the radius of gyration (Rg) of the fusion protein. Moreover, the proteinous connecting moiety can stabilize the therapeutic polypeptide. In some embodiments, the proteinous connecting moiety may comprise a whole protein, a truncated version of a protein, a protein domian or domains in tandem, or protein fragments. A skilled artisan will appreciate that the proteinous connecting moiety may comprise some non-proteinous modifications which are not formed by amino acids, such as PEG.

The length of the flexible, unstructured linker may play an important role in determining the hydrodynamic radius and/or the radius of gyration (Rg) and the in vivo half life of the fusion protein. The flexible, unstructured polypeptide linker may contain sequences such as (G5S)n, (G4S)n, (G3S)n, (GS)n, (G2S2)n, (G3S3)n, (GS3)n where n is an integer, or other sequences that are rich in G, S, A, T or P. The length of the flexible linker may vary from 1 to 3000 amino acid residues, and particularly within the range of 5 to 500 amino acid residues. It has been reported that the un-structured stretches of polypeptides may act like PEG molecule and increase the hydrodynamic radius and/or the Rg of the protein molecule[13]. By use of our method, a relatively shorter flexible linker is needed to reach the desired Rg due to the trimer formation compared with the monomer. This may have great advantages for therapeutic proteins by reducing the immunogenicity.

In some embodiments of the invention, the fusion protein may consist of one or more flexible, un-structured polypeptide linkers. It will be appreciated by a skilled artisan that these flexible, un-structured linkers within the fusion protein may be the same or different.

Our data clearly showed that varying the length of the flexible, unstructured linker or linkers can efficiently change the hydrodynamic radius and/or Rg of the molecule and control the in vivo half life of the engineered protein molecule. Therefore, our method can generate a recombinant protein with tunable in vivo half life by varying the flexible, unstructured linker length within the fusion protein. This is advantageous compared with the traditional therapeutic IgG with fixed in vivo half life. In addition, our "Trident technology" may offer the fusion protein tri-valency for the ligand, in contrast, IgG only has bi-valency.

The pCloud polypeptide

The present invention provides compositions comprising the "pCloud" polypeptide. In a preferred embodiment of the invention, the flexible un-structured linker is exhibited as one or more un-structured pCloud polypeptides. In some embodiments, pCloud polypeptides are generally extended polypeptides that have low degree or no secondary or tertiary structures under physiologic conditions.

The pCloud polypeptide is characterized in: (a) the total pCloud amino acid residues is at least 100 to about 3000 amino acid residues; (b) the pCloud polypeptide sequence is generated by use of some or all of the fragments derived from human fibrinogen alpha chain. In pCloud sequence, the fibrinogen fragments are flanked by flexible loops with various lengths. Therefore pCloud is primarily human originated and has low immunogenicity when administered to human, (c) the pCloud sequence is rich in glycine (G), serine (S) and Glutamate (E). The pCloud also contains alanine (A), proline (P), arginine (R) and threonine (T). The sum of G, S, E, A, P and T constitutes more than 90% of the pCloud sequence, (d) The pCloud sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm; and (e) the pCloud sequence does not contain any T-cell epitopes as predicted by TEPITOPE algorithm.

It has been reported that fusing an unstructured polypeptide to the therapeutic polypeptide can significantly extend the in vivo half life of the therapeutic polypeptide (XTEN technology)[13]. However, in XTEN technology, the unstructured polypeptide is generated using artificial peptide fragments and it is not fused with a trimeric scaffold protein. These artificial peptides in XTEN technology represent foreign peptides to human body and it is likely that these foreign peptides may elicit immune responses within the patients when administrated. Because many therapeutic polypeptide, such as human Growth hormone and GLP-1 analogues, needs to be applied to the patients for an extended period, the foreign peptides introduced by the XTEN technology may represent a potential threat for the patients. In the present invention, a flexible unstructured "pCloud" polypeptide generated by use of human fibrinogen alpha chain sequences was demonstrated to efficiently extend the in vivo half life of therapeutic polypeptides. In addition, in the present invention, the pCloud polypeptide is further fused with a trimeric scaffold protein, which further enhances the pharmacokinetic profile of the therapeutic polypeptide.

In some embodiments of this invention, to constitute an unstructured pCloud polypeptide with low immunogenicity, we took advantage of the human fibrinogen alpha chain sequence. Human Fibrinogen (factor Γ) is a soluble, 340 kDa plasma glycoprotein, that is converted by thrombin into fibrin during blood clot formation [14-16]. Fibrinogen is synthesized in the liver by the hepatocytes. The normal concentration of fibrinogen in the human blood plasma is quite high (1.5-3 mg/ml), which strongly suggests that the human fibrinogen sequence may exhibit very low immunogenicity. Human fibrinogen is a hetero-hexamer that contains two sets of three different chains (α, β, and γ), linked to each other by disulfide bonds. Within the fibrinogen alpha (a) chain, an intrinsic unstructured region (residues 262-455) is present (Fig. 2). This unstructured region of fibrinogen contains minimum secondary structures as determined by GOR algorithm [10, 17]. 12 fragments within this fibrinogen unstructured region (residues 262-455) have been identified to contain primarily the residues glycine (G), serine (S) and Glutamate (E), proline (P), arginine (R) and threonine (T). In some embodiments of this invention, to further reduce the immunogenicity of these fragments, mutations Y277S, V379A and D396E were introduced in fragment 1 , 9 and 1 1 , respectively (Fig. 2). In some embodiments, the resultant 12 fragments derived from human fibrinogen alpha chain sequence are utilized as the building blocks to generate the pCloud polypeptides (Table. 1). In some embodiments, the variants of these fragments that share at least 70%, 75%, 80%, 85%o or 90% amino acid sequence identity with the fragments listed in table 1 may be utilized as the building blocks for pCloud polypeptides. Table 1. the protein sequences of the 12 fragments derived from human fibrinogen alpha chain

In some embodiments of this invention, in order to generate the pCloud polypeptides, the fragments listed in table 1 derived from human fibrinogen alpha chain are flanked by flexible loops with variable lengths from 0 to 100 residues. These flexible loops are rich in glycine (G) and serine (S). These loops also contain glutamate(E), alanine (A), proline (P) and threonine(T). The flexible loops have greater than 95% unstructured random coil formation as determined by GOR algorithm. The flexible loop sequences can be selected, but not limited, from the table 2.

The flexible loops are generally the flexible unstructured polypeptide linkers with shorter lengths and more flexibility. In the present invention, the flexible loops are utilized to connect the human fibrinogen alpha chain fragments to constitute the pCloud polypeptide. In addition, the flexible loops are also utilized to connect therapeutic polypeptide and the proteinous connecting moiety of human origin in the fusion protein. The flexible loops may also be utilized to link the therapeutic polypeptide with the scaffold protein, the scaffold protein with the flexible unstructured polypeptide (or the pCloud polypeptide), and the proteinous connecting moiety with the flexible unstructured polypeptide (or the pCloud polypeptide). A skilled artisan will appreciate that the flexible loop may be utilized in the fusion protein as a spacer to provide flexibility.

Table 2: the protein sequences of the flexible loops utilized in the pCloud sequence to connect the fibrinogen alpha chain fragments. In the first line of the table, several G/S rich linkers are listed, n is an integer that can be adjusted based on needs.

(G2S)n, (G3S)n, (G4S)n, (G5S)n, (GS)n, (G2S2)n, (GS2)n, (GS3)n, (S2G)n,

(S3G)n, (S4G)n, (S5G)n, (SG)n, (S2G2)n, (SG2)n, (SG3)n

GSESG

GSGSG

GSPSG

GSTSG

GSGSESG

GSESGSG

GSTSESG

GSESTSG

GSPSESG

GSESPSG

GSGEGSG

GSGTGSG

GSGPGSG

GSPSESGSG

GSPSGSESG

GSESPSGSG

GSESGSPSG

GSGSPSESG

GSGSESPSG

GSTSESGSG

GSTSGSESG

GSESTSGSG

GSESGSTSG

GSGSTSESG

GSGSESTSG

GSGSESGSG

GGSGEGSGG

GSGGEGGSG

GGSGESGSG

GSGSEGSGG

GGSGSESGSGG

GGSGGGSGG

GGGGSGG

GGGSGGGG In some embodiments, all of the fragments listed in table 1 are utilized to generate the pCloud sequence, or alternatively at least 10 fragments listed in table 1 , or alternatively at least 8 fragments in table 1 , or alternatively at least 6 fragments in table 1 , or alternatively at least 5 fragments, or alternatively at least 3 fragments in table 1 , or at least one fragment in table 1 are utilized to generate the pCloud sequence. In some embodiments, the fragments listed in table 1 can be connected in the order as they appear in the human fibrinogen alpha chain sequence to constitute the pCloud sequence. In some embodiments, the fragments listed in table 1 can be connected in the order that is distinct from they appear in the human fibrinogen alpha chain sequence to generate the pCloud sequence. In one aspect of the present invention, the pCloud polypeptide contains 100 to 3000 amino acid residues generated by use of the human fibrinogen derived fragments listed in table 1. The pCloud sequence has greater than 90% unstructured random coil formation as determined by GOR algorithm.

In some cases, a pCloud sequence may comprise charged residues separated by other residues such as serine or glycine, which may lead to better expression or purification behavior. The charged residues, such as D, E, K and R, may also prevent the aggregations of the pCloud polypeptide. In some preferred embodiments of this invention, the pCloud polypeptide may carry a net negative charge under physiologic conditions that may contribute to the unstructured conformation and reduced binding of the pCloud polypeptide component to the mammalian proteins and tissues. Based on the net charge, pCloud polypeptide may have an isoelectric point (pi) of 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, or even 6.5. In preferred embodiments, the pCloud polypeptide will have an isoelectric point between 2.0 and 5.0.

Because large hydrophobic amino acid residues (such as I, L, V, M, F, W, Y) can induce protein aggregations and may form a core structure for a polypeptide, in some embodiments of the present invention, the content of the hydrophobic amino acids (I, L, V, M, F, W, Y) in the pCloud polypeptide will typically be less than 5%, or less than 2%, or less than 1 % of the total amino acid residues.

In some cases, the invention provides compositions in which the pCloud sequences have a low degree of immunogenicity or are essentially non- immunogenic. Several facts can contribute to the low immunogenicity of pCloud, such as the unstructured conformation, the high degree of solubility, the low degree or lack of residues with large side chains, the low degree or lack of self-aggregation, the low degree or lack of proteolytic sites within the sequence, the low degree or lack of hydrophobic residues, and the lack of epitopes in the pCloud sequence.

In some embodiments of the invention, the pCloud polypeptides are generated by use of human fibrinogen alpha chain fragment sequences, therefore, the pCloud polypeptides may not stimulate any immune responses from human body. Moreover, the pCloud polypeptide primarily consists of unstructured sequence and contains low degree of secondary structures, which will prevent the pCloud polypeptide to activate the B-cells. To be efficiently recognized by the host humoral immune system, the foreign polypeptide needs to form a stable conformation. The precise folding of the polypeptide may allow it to form the epitope that can be recognized as "foreign" by the host humoral immune system, resulting in the production of antibodies to the polypeptide or triggering a cell-mediated immune response. In addition, the pCloud polypeptides lack a predicted T-cell epitope when analyzed by TEPITOPE algorithm [12], wherein the TEPITOPE algorithm prediction for epitopes within the pCloud sequence is based on a score of -7 or greater, or -8 or greater, or -9 or greater. The data strongly suggest that the pCloud polypeptide may not be recognized by MHC molecules and T-cell receptors to trigger the T-cell activations which may lead to the release of cytokines further activating other lymphocytes such as B cells to produce antibodies or activating T killer cells as a full cellular immune response. The construction of the fusion protein of the therapeutic polypeptide, the pCloud polypeptide and the scaffold protein

In this invention, the therapeutic polypeptide is connected to one or more pCloud sequences and the scaffold protein to extend the in vivo half life of the therapeutic polypeptide. The pCloud sequence can be placed at either or both of the N-terminal and the C-terminal end of the therapeutic polypeptide. The pCloud sequences can also be placed at either or both of the N-terminal and the C-terminal end of the scaffold protein. The pCloud sequences within the fusion protein could be the same or be different from each other. In some embodiments, the fusion protein of the present invention is configured using the following formula:

(pCloud) m -TP-(pCloud) n -Scaffold - (pCloud) k Or

(pCloud) m - Scaffold - (pCloud) n -TP-(pCloud) k wherein:

(a) pCloud is the pCloud polypeptide characterized above, they could be different from each other.

(b) TP is a therapeutic polypeptide selected, but not limited from the group consisting of human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof.

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution.

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m + n+k

>=1.

In some cases, the therapeutic polypeptide might be a short peptide (such as

GLP- 1 or PTH, usually a peptide not more than 100 residues). To efficiently utilize our method, the therapeutic polypeptide is connected with the pCloud sequence via a proteinous connecting moiety of human origin. A flexible loop may be utilized to fuse the therapeutic polypeptide and the proteinous connecting moiety. The flexible loop has been characterized above. The proteinous connecting moiety can stabilize the therapeutic polypeptide and further increase the hydrodynamic radius and/or the Rg of the fusion protein. In some embodiments, the proteinous connecting moiety may comprise a whole protein, a truncated version of a protein, a protein domain or domains in tandem, or protein fragments. A skilled artisan will appreciate that the proteinous connecting moiety may comprise some non-proteinous modifications which are not formed by amino acids, such as PEG.

In some embodiments, the fusion protein containing the therapeutic polypeptide, the proteinous connecting moiety, the pCloud polypeptides, and the scaffold protein of the present invention is configured according to the following formula: (pCloud) m -TP-Loop-PCM-(pCloud) n -Scaffold- (pCloud) k or

(pCloud) m -Scaffold -(pCloud) n -PCM-Loop-TP- (pCloud) k wherein:

(a) pCloud is the pCloud polypeptide characterized above;

(b) TP is the therapeutic polypeptide selected, but not limited from the group consisting of human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof;

(c) Scaffold indicates the scaffold protein which forms a homo-trimer in solution.

(d) m is either 0 or 1 ; and n is either 0 or 1 , k is either 0 or 1 , and m + n+k

>=1.

(e) Loop is a flexible loop characterized above; and

(f) PCM is the proteinous connecting moiety of human origin.

In some embodiments, the proteinous connecting moiety within the fusion protein is a proteinous sequence having an elongated shape of human origin. In some embodiments, the proteinous connecting moiety within the fusion protein is a human Fibronectin type III domain. In a particular embodiment of this invention, the proteinous connecting moiety within the fusion protein contains a human Fibronectin type III domain 8 (Fn8). Using the wild type human Fibronectin type III domain as the proteinous connecting moiety in the present invention is because Fibronectin type III domain has an elongated molecular shape and it may greatly extend the hydrodynamic radius and/or the radius of gyration of the fusion protein. This mechanism is totally different from using mutant Fibronectin type III domain as a binder to human Albumin to extend half life as shown before (patent 13). Fibronectin(Fn) is a high-molecular weight (~440kDa) glycoprotein of the extracellular matrix that binds to a number of proteins including integrins, collagen, fibrin and heparan sulfate proteoglycans (e.g. syndecans) [18] . Fibronectin exists as a protein dimer, consisting of two nearly identical polypeptide chains linked by a pair of C-terminal disulfide bonds. Each fibronectin monomer has a molecular weight of 230-250 kDa and contains three types of domains: type I, II, and III. Type I and type II are stabilized by intra-chain disulfide bonds, while fibronectin type III domains do not contain any disulfide bonds [19] . The Fibronectin type III domain is an evolutionary conserved protein domain that is widely found in animal proteins. The human fibronectin protein in which this domain was first identified contains 16 copies of this domain (Fnl to Fnl6). The fibronectin type III domain family (pfam ID: PF00041) member contains about 95 amino acids long and possesses a beta sandwich structure. Fibronection type III domain forms a very stable domain structure with the melting temperature of -70 °C as measured by DSC [20 ' 21] . Fibronectin type III domains are found in a wide variety of extracellular proteins. In human genome, fibronection type III domain exists in many proteins including Tenascin, Usherin, Titin, tripartite motif (TRIM) family members, tissue factor, TIE1 , TIE2, SPEG, SORL1 , SDK1 , ROBOl , ROB02, SDK2, Receptor-type tyrosine-protein phosphatase, prolactin receptor, LI CAM, NCAM1 , NCAM2, myomesin 1 , myomesin 2, Myosin-binding protein C, LIFR, Leptin receptor, Integrin, Insulin receptor, Contactin, Collagen, Cytokine receptor-like factor, Inteferon receptor, Growth hormone receptor, fibronectin, leucine rich transmembrane protein (FLRT) members, IL, ephrin type-A receptor, ephrin type-B receptor, IL-6R, gp l 30, IL1 I RA, IL12RB, IL20RB, IL23R, IL27RA and IL31RA etc. Therefore, using Fibronectin type III domain as the proteinous connecting moiety in our method may have low immunogenicity. The Fibronectin type III domains can also be used in tandem fashion in the proteinous connecting moiety. In addition, Fibronectin type III domain can be expressed in recombinant form using a number of expression systems including E. coli, using Fibronectin type III domain as the proteinous connecting moiety in the method of the invention may greatly increase the expression yield of the fusion protein. The scaffold protein

Collagens are a diverse family of proteins that constitute the major structural component of the extracellular matrix [22-24]. Collagen is composed of a triple helix, which generally consists of two identical chains (oil) and an additional chain that differs slightly in its chemical composition (a2). Classification according to supramolecular structure assigns collagens to fibril, fibril-associated containing interrupted triple helicies (FACIT), beaded filament, anchoring fibril, network-forming, transmembrane or multiple triple helicies with interruptions (Multiplexin) families[25]. To date, 43 unique -chains that belong to 28 types of collagens (types I-XXVIII) have been discovered in vertebrates. The alpha chains of collagens consist of at least one triple helical collagenous domain of varying length and two noncollagenous (NC) domains of variable sequence, size, and shape that are positioned at the N and C terminus. The collagenous domains contain the G-X-Y repeats and form the typical triple helix within the collagen molecule while some of the NC domains form homo-trimers to stabilize the collagen triple helix. Studies on classic fibril-forming collagens found that the extreme carboxy-terminal NC (NC I) domains were essential for trimerization[26]. The Multiplexin family members also utilize NC I domains for trimerization. On the other hand, Fibril associated-collagens (FACIT) have recently been shown to trimerize via their NC2 domains (the second NC domain from the carboxy-terminal end)[27, 28]. The crystal structures of NC I domains from the network-forming collagens IV, VIII and X indicated these NC I domains constituted a stable homo-trimer with the cl q-like molecular structure [29-31]. About 130 a. a. residues are present in each monomer. In contrast, the NCI domains from the multiplexin family members such as Collagen XV and XVIII formed the much smaller homo-trimer with the length of about 55 residues in each monomer[26, 32]. Both types of the collagen NC domains form very stable homo-trimers in solution (Tm>60°C) and are suitable as the scaffold protein as described in the invention.

Many other human proteins or domains thereof which can form homo-trimers in solution may also serve as the scaffold proteins in the method of the invention. The Clq family is characterized by a C-terminal conserved globular Cl q domain (pfam ID: PF00386), which can form a stable homo-trimer [33-35]. The C l q-like protein family includes, but not limited to, human C l q A chain, C l q B chain, C lq C chain, cbln family members, human EMILI -1 , multimerin, ACRP30/adiponectin, adipolin, resistin and resistin-like molecule (RELM) hormone family members. Tumor necrosis factor (TNF) refers to a cytokine that can induce cell apoptosis and inflammation[36, 37]. TNF family (Pfam ID: PF00229) members include, but not limited, human TNFalpha, TNFbeta, TRAIL, RANK ligand, Fas ligand, CD 30 ligand, CD40 ligand, CD27 ligand, OX40L and CD137. TNF family members form homo-trimers in solution and demonstrated the similar molecular structure as the C l q family members. Therefore, these two family members are also named as Cl q/TNF-related proteins (CTRP)[34].

The superfamily of proteins containing C-type lectin-like domains (CTLDs, pfam ID: PF00059) is a large group of extracellular proteins with diverse functions including cell-cell adhesion, immune response to pathogens and apoptosis [38]. A number of CTLD proteins contain a neck and a C-terminal C-type carbohydrate-recognition domain (CRD) and they form homo-trimer in solution. This type of CTLD includes mannan-binding lectin (MBL), surfactant protein A (SP-A), surfactant protein D (SP-D), collectin liver 1 (CL-L1), collectin placenta 1 (CL-P1), conglutinin, collectin of 43 kDa (CL-43) and collectin of 46 kDa (CL-46), Langerin and Tetranectin [39-42].

In this invention, the CTRP family members, including the Clq-like domains and TNF family members, can be utilized to fuse with the therapeutic polypeptide to extend the in vivo half life of the fusion protein. Alternatively, the CTLD family members can be employed as the scaffold protein to drive the trimerization of the therapeutic polypeptides.

In the preferred embodiments, the NCI domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) and NC2 domain within FACIT type of collagen (such as collagen IX, XII, XIV, XVI, XIX, XX, XXI, and XXII) may serve as the scaffold proteins in the method of this invention. The therapeutic polypeptide and the pCloud polypeptides can be fused to either the N-terminus or the C-terminus of the scaffold protein. The trimer formation of the fusion protein can efficiently increase the hydrodynamic radius of the protein molecule. The fusion protein may demonstrate a much larger apparent size than a compact molecule with the same molecular weight. Therefore the fusion protein will show a much reduced clearing rate by renal filtration and will exhibit an extended half life in vivo.

In the preferred embodiments, the NCI domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) were selected as the scaffold protein in the present invention. NC I domain does not utilize the G-X-Y repeats and does not form the typical triple helix. In addition, no disulfide bridges between the NC I domains are required to form the stable homo-trimer. Moreover, NCI domain contains only about 55 amino acid residues and represents a small protein, which makes it less likely to interfere with the proper functions of the therapeutic polypeptide in the fusion protein. All the features of NC 1 domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) make it an ideal scaffold protein in the present invention. Using NCI domain as the scaffold protein is preferred in the present invention, which is quite different from using other collagen domains as the scaffold protein as described elsewhere (patent 14, 15).

It will be appreciated by a skilled artisan that the term "scaffold protein" as used herein does not necessarily mean an entire wild type protein; a domain or functional variant thereof which can form a stable homo-trimer in solution and can therefore serve the purpose of the invention may also be used in our method. In some embodiments of the invention, for example, the NCI domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) was used as a scaffold protein.

The therapeutic polypeptide

In some embodiments of this invention, the therapeutic polypeptide may be selected from, but not limited to, human glucagon-like peptide- 1 (GLP-1), Exenatide, GLP-2, C-peptide, Calcitonin, human Parathyroid hormone (PTH), glucagon, G-CSF, GM-CSF, Interferon, interleukin factors, VEGF receptors, TNF alpha receptors, RANK, Growth hormone, Erythropoietin, blood-coagulation factors, single-chain Fv, single domain antibodies and functional variants thereof. In a preferred embodiment of this invention, we use GLP- 1 as one of the examples to illustrate how the method of the invention can significantly improve the pharmacokinetics property of GLP- 1 and its mutants while retaining its biological activity. The natural incretin hormone glucagon-like peptide- 1 (GLP- 1) supports glucose homeostasis by enhancing glucose-dependent insulin secretion from ?-cells and suppressing inappropriately elevated postprandial glucagon secretion from ct-cells. In addition, GLP- 1 has been demonstrated to reduce appetite and food intake and inhibit gastric emptying, which may facilitate weight management [43, 44]. Therefore, GLP-1 remains to be a very promising therapeutic polypeptide for type 2 diabetes and weight loss. However, GLP-1 is a 30 residue polypeptide with a very short half life in vivo, which severely limits its applications. In the present invention, we demonstrated data to show that the half life of GLP-1 can be significantly extended by use of the method of the invention.

In some other preferred embodiments of this invention, we could fuse pCloud polypeptides and the scaffold protein with the human growth hormone, human Interferon alpha-2b and human G-CSF. These therapeutic polypeptides (human growth hormone, Interferon alpha-2b and G-CSF) suffer significantly from their short in vivo half life. Fusing with pCloud polypeptide and the scaffold protein may greatly improve the pharmacokinetic profiles of the therapeutic polypeptides in vivo.

One advantage of the method of our "Trident technology" is that it can render tri-valency for the therapeutic polypeptide. It has been well documented that multivalency of protein can greatly enhance its affinity and avidity to binding partner[l l , 45-47]. Antibody IgG is a Y-shaped molecule with bi-valency and utilizes two identical variable domains to interact with its ligand. The fusion protein generated using the method of the invention has tri-valency and therefore might behave better than the traditional human monoclonal antibody IgG in interacting with the ligand. For example, TNF alpha forms a homo-trimer in solution and interact with three TNF receptors simultaneously. To inhibit the TNF alpha function, TNF receptor 2 (TNFR2, p75) have been fused to IgG Fc fragment to constitute Etanercept (Enbrel) to treat severe rheumatoid arthritis. However, one Enbrel molecule can only block two out of three possible binding sites located on TNF alpha homo-trimer. In contrast, our fusion protein of TNFR2 and collagen XVIII NC 1 domain generated (described below in examples) can form a homo-trimer and block all three binding sites of TNFalpha while retaining a long half life in vivo.

The scaffold protein utilized in this invention can form homo-trimers by simultaneous self assembly. In the preferred embodiments, the NCI domain within Multiplexin type of human Collagen (such as collagen XV and XVIII) were selected as the scaffold protein. In NC 1 domain, no inter-chain disulfide bonds are needed to drive the trimerization, which makes it more convenient for protein expression. Many expression systems such as E. coli, yeast, insect cell and mammalian cell systems can be utilized to express the fusion proteins generated by the invention. In the sharp contrast, therapeutic monoclonal antibodies rely purely on the mammalian systems for mass productions. Summary for Trident technology

Improving the pharmacokinetic property of a therapeutic polypeptide may have major impacts on its clinical application. In the case of GLP-1 , extending its in vivo half life transformed it into a practical drug with great efficacy and broad markets. It is well known that increasing the apparent molecular weight (hydrodynamic radius and/or radius of gyration) of a therapeutic polypeptide can result in an improvement in the pharmacokinetic behavior of the therapeutic polypeptide possibly due to the slower renal clearance. The apparent molecular weight (the hydrodynamic radius and/or Rg) of a protein is determined by its molecular weight as well as by its structure, including shape and compactness. The flexible unstructured polypeptides (in preferred embodiments, pCloud polypeptides) can adopt unstructured conformations due to electrostatic repulsion between individual charges of the polypeptide and/or the inherent flexibility imparted by the particular amino acids in the sequence that lack potential to confer secondary structures. The extended and unstructured conformation of the flexible unstructured polypeptides (in preferred embodiments, pCloud polypeptides) may have a greater proportional hydrodynamic radius and/or Rg compared to polypeptides of a comparable sequence length and/or molecular weight that have tight secondary and/or tertiary structures, such as typical globular proteins. Methods for determining the hydrodynamic radius and/or Rg are well known in the art, such as by the use of size exclusion chromatography (SEC), as described in U.S. Patent Nos. 6,406,632 and 7,294,513.

In one aspect, the present invention provides a novel technique termed as

"Trident technology" which allows the therapeutic polypeptide to fuse with one or more flexible unstructured polypeptides (in preferred embodiments, pCloud polypeptides) and a trimeric scaffold protein. The trimer formation may greatly increase the hydrodynamic radius of the fusion molecule and improve the in vivo half life of the fusion protein. In addition, the flexible unstructured polypeptides (in preferred embodiments, pCloud polypeptides) may act like PEG molecules and increase the apparent size of the fusion protein. As the result, fusing the therapeutic polypeptide with flexible unstructured polypeptides (in preferred embodiments, pCloud polypeptides) and the trimeric scaffold protein may render the fusion protein a much larger apparent molecular size compared to a compactly folded globular protein with the same molecular weight. This will greatly improve the pharmacokinetic profile of the therapeutic polypeptide. In some embodiments, fragments derived from human fibrinogen alpha chain sequence were utilized as the building blocks to generate the pCloud polypeptides which rendered low immunogenicity when administered to human. Moreover, the method of the invention can provide the therapeutic polypeptide with tri-valency, which may greatly increase the affinity and avidity of the fusion protein toward the ligand.

To further extend the in vivo half life of the fusion protein generated by the method of the invention, the fusion protein can be further modified by PEGylation. The PEG moiety may have a molecular weight of between 2 kDa and 100 kDa. For specific PEGylation, the Cys residue may need to be generated in the fusion protein using site-directed mutagenesis. Methods of Preparing fusion proteins generated by the Present

Invention

The fusion proteins of the present invention can be produced through the application of recombinant DNA technology. Recombinant polynucleotide constructs encoding a fusion polypeptide of the present invention typically include an expression control sequence operably-linked to the coding sequences of the fusion polypeptide, including naturally-associated or heterologous promoter regions. As such, another aspect of the invention includes vectors containing one or more nucleic acid sequences encoding a fusion polypeptide of the present invention. For recombinant expression of one or more polypeptides of the invention, the nucleic acid containing all or a portion of the nucleotide sequence encoding the fusion polypeptide is inserted into an appropriate cloning vector, or an expression vector (i.e. , a vector that contains the necessary elements for the transcription and translation of the inserted polypeptide coding sequence) by recombinant DNA techniques well known in the art and as detailed below. Methods for producing diverse populations of vectors have been described by Lerner et al , U.S. Pat. No. 6,291 ,160; 6,680,192.

In general, expression vectors useful in recombinant DNA techniques are often in the form of plasmids. In some embodiments of the present invention, "plasmid" and "vector" can be used interchangeably as plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors that are not technically plasmids, such as viral vectors (e.g. , replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. Preferably, the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences encoding the fusion polypeptide. These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers, e.g. , ampicillin-resistance or kanamycin-resistance, to permit detection of those cells transformed with the desired DNA sequences. Vectors can also encode a signal peptide, e.g. , pectate lyase, useful to direct the secretion of extracellular antibody fragments. See U.S. Pat. No. 5,576, 195. The recombinant expression vectors of the invention may comprise a nucleic acid encoding a fusion polypeptide in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors may include one or more regulatory sequences selected on the basis of the host cells to be used for expression that are operatively-linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g. , in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g. , polyadenylation signals). Such regulatory sequences are described, e.g. , in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g. , tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of polypeptide desired, etc.

Another aspect of the invention pertains to the fusion polypeptide-expressing host cells, which contain a nucleic acid encoding one or more fusion polypeptides. The recombinant expression vectors of the invention can be designed for expression of a fusion polypeptide in prokaryotic or eukaryotic cells. For example, a fusion polypeptide can be expressed in bacterial cells such as Escherichia coli, insect cells, fungal cells, e.g. , yeast, or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. ( 1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, e.g. using T7 promoter regulatory sequences and T7 polymerase.

Expression of polypeptides in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of the recombinant polypeptides. The vectors may add a number of amino acids to a polypeptide encoded therein, usually to the amino terminus of the recombinant polypeptide. Such vectors with extra amino acid residues typically serve three purposes: (i) to increase expression of recombinant polypeptide; (ii) to direct the recombinant protein to periplasmic space; and (iii) to aid in the purification of the recombinant polypeptide by acting as a ligand in affinity purification. Typical expression vectors serving this purpose include pGEX (GE Healthcare), pMAL (New England Biolabs), pET20b(Novagen), pET43b (Novagen), pET32b(Novagen) and pRIT5 (GE Healthcare).

Examples of suitable inducible E. coli expression vectors include pTrc vectors (Invitrogen), pQE (Qiagen) and pET vectors (Novagen). One strategy to maximize recombinant polypeptide expression is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in the expression host, e.g. , E. coli 48 . Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.

In another embodiment, the fusion polypeptide expression vector is a yeast expression vector. Examples of vectors for expression in yeast Saccharomyces cerivisae include pYES2 (Invitrogen), pMFa 49 and pJRY88 50 . The fusion protein may also be expressed in Pichia system using the vectors pPICZ pGAPZ and pPIC9(InVitrogen). Alternatively, a fusion polypeptide can be expressed in insect cells using baculovirus expression vectors or using the stable insect cell lines. Baculovirus systems available for expression of polypeptides in cultured insect cells {e.g. , SF9 cells) include the BaculoGold system (BD Biosciences), BaculoDirect system (Invitrogen) and BacVector system (Novagen). The stable insect expression systems include, but not limited to, DES system (Invitrogen) and InsectDirect (Novagen).

In another embodiment, a nucleic acid encoding a fusion polypeptide of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include, e.g., but are not limited to, pcDNA3.1 (Invitrogen), pSecTag (invitrogen), and pTriEx series vectors (Novagen). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells useful for expression of the fusion polypeptide of the present invention, please see, e.g., Chapters 16 and 17 of Sambrook, et al , MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.

In the eukaryotic expression systems, the recombinant fusion protein can be expressed in the cytoplasm. Or alternatively, the fusion protein can be secreted into the medium by adding an N-terminal secretion signal.

Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention is to be introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.

A host cell can be any prokaryotic or eukaryotic cell. For example, a fusion polypeptide can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells. Mammalian cells are a preferred host for expressing nucleotide segments encoding immunoglobulins or fragments thereof. See Winnacker, From Genes To Clones, (VCH Publishers, NY, 1987). A number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include Chinese hamster ovary (CHO) cell lines, 293 cells, various COS cell lines, HeLa cells, L cells and myeloma cell lines. Preferably, the cells are nonhuman. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer, and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from endogenous genes, cytomegalovirus, SV40, adenovirus, bovine papillomavirus, and the like. Other suitable host cells are known to those skilled in the art.

For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate a foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g. , resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding the fusion polypeptide or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).

Once expressed, the fusion polypeptides are purified from culture media and/or host cells. Purification of recombinant polypeptides is well known in the art and includes ammonium sulfate precipitation, affinity chromatography purification technique, column chromatography, ion exchange purification technique, gel filtration and the like (see generally Scopes, Protein Purification (Springer- Verlag, N.Y., 1982).

Formulation of Pharmaceutical Compositions

The present invention envisions treating a disease, for example, type II diabetes, in a mammal by the administration of the fusion protein compositions of the present invention. Administration of the fusion protein in accordance with the present invention may be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners. The administration of the vaccines of the invention may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated.

The pharmaceutical composition of the present invention may be delivered via various routes and to various sites in a mammal body to achieve a particular effect. One skilled in the art will recognize that although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. Local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, peritoneal, subcutaneous, intradermal, as well as topical administration.

The amount of the administered fusion protein of the present invention will vary depending on various factors including, but not limited to, the particular disease, the weight, the physical condition, and the age of the mammal, and whether prevention or treatment is to be achieved. Such factors can be readily determined by the clinician employing animal models or other test systems which are well known to the art. Generally, the amount of the fusion protein of the present invention to be administered to a mammal subject may vary in the range of lng/kg to l OOmg/kg of the subject body weight. In an embodiment of the invention, the amount of administration was from lug/kg to l .Omg/kg.

When the fusion proteins of the invention are prepared for administration, they are preferably combined with a pharmaceutically acceptable carrier to form a pharmaceutical formulation, or unit dosage form. Commonly used pharmaceutically acceptable carriers are well known to a skilled artisan in the field of pharmacy. The total active ingredients in such formulations include from 0.1 to 99.9% by weight of the formulation. The active ingredient for administration may be present as a powder or as granules; as a solution, a suspension or an emulsion. Thus, the therapeutic agent may be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and may be presented in unit dosage form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers with an added preservative. The active ingredients may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for re-constitution with a suitable vehicle, e.g. , sterile, pyrogen-free water, before use.

In general, water, suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions. Solutions for parenteral administration contain the active ingredient, suitable stabilizing agents and, if necessary, buffer substances. Antioxidizing agents such as sodium bisulfate, sodium sulfite or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium ethylenediaminetetraacetic acid (EDTA). In addition, parenteral solutions can contain preservatives such as benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, a standard reference text in this field.

Additionally, standard pharmaceutical methods can be employed to control the duration of action. These are well known in the art and include control release preparations and can include appropriate macromolecules, for example polymers, polyesters, polyamino acids, polyvinyl, pyrolidone, ethylenevinylacetate, methyl cellulose, carboxymethyl cellulose or protamine sulfate. The concentration of macromolecules as well as the methods of incorporation can be adjusted in order to control release. Additionally, the agent can be incorporated into particles of polymeric materials such as polyesters, polyamino acids, hydrogels, poly (lactic acid) or ethylenevinylacetate copolymers. In addition to being incorporated, these agents can also be used to trap the compound in microcapsules.

The terms "pharmaceutically-acceptable," "physiologically-tolerable," and grammatical variations thereof, as they refer to compositions, carriers, and reagents, are used interchangeably and represent that the materials are capable of administration to or upon a subject without the production of undesirable physiological effects to a degree that would prohibit administration of the composition.

Preferred examples of such carriers include, but are not limited to, water, saline, Ringer's solutions, and dextrose solution. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and compounds for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or compound is incompatible with the fusion polypeptides, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.

A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. The fusion polypeptides compositions of the present invention can be administered by parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal; intramuscular route or as inhalants. The fusion polypeptides can optionally be administered in combination with other agents that are at least partly effective in treating various diseases including various actin- or microfilament-related diseases.

Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial compounds such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating compounds such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and compounds for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.

Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, e.g., water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, e.g. , by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal compounds, e.g. , parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic compounds, e.g. , sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition a compound which delays absorption, e.g. , aluminum monostearate and gelatin.

Sterile injectable solutions can be prepared by incorporating the fusion polypeptides in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the binding agent into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.

Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the binding agent can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding compounds, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating compound such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening compound such as sucrose or saccharin; or a flavoring compound such as peppermint, methyl salicylate, or orange flavoring.

For administration by inhalation, the fusion polypeptides are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g. , a gas such as carbon dioxide, or a nebulizer.

Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, e.g., for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the fusion polypeptides are formulated into ointments, salves, gels, or creams as generally known in the art.

The fusion polypeptides can also be prepared as pharmaceutical compositions in the form of suppositories (e.g. , with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery. Examples

The present invention is further illustrated by the following examples, which should not be construed as limiting in any way.

Example 1. Construction of an expression vector of GLP- 1 fused with human Fibronectin type III domain 7 (Fn7) and human collagen XVIII NC I domain (COL 18NC 1 )

In this example, GLP- 1 polypeptide was fused to the N-terminus of the human collagen XVIII NC I domain (COL 18NC 1 ) which forms a stable homo-trimer. To further extend the Rg of the molecule, the human Fibronectin type III domain 7 (Fn7) was utilized as the proteinous connecting moiety. Fn7 was connected to the GLP- 1 polypeptide and COL 18NC 1 through a flexible loop (GGGSGGGG) and a flexible, unstructured linker (GGGSGG). The pET29b vector (Novagen) was used to construct a recombinant plasmid containing the GLP- 1 -Fn7-C0L 18NC 1 fusion gene. First, human COL 18NC 1 was cloned into pET29b by BamUl and Xhol to result in the pET29b-COL18NCl vector. PCR reaction was carried out using human collagen XVIII cDNA as the template by using the following primers:

Col 18NC 1 -forward:

CGGGATCCGGTGGCGGCGCCTCCTCAGGGGTGAGG

Col 18NC 1 -reverse:

C C GC T C GAGT T ACCCTCGTGGGAGTGGTGTCCGGGCCTCC The PCR product was digested by restrictive enzyme BamUl and Xhol (Fermentas) and ligated into the pET29b vector by use of T4 ligase (Fermentas). The sequence of resulted pET29b-COL18NC l vector was confirmed by DNA sequencing.

PCR reaction was carried out using human Fibronectin cDNA as the template by using the following primers:

Glp 1 -Fn7-Forward:

GGAATTCCATATGCATGCCGAAGGGACTTTTACCAGTGATGTAAGTTCTTAT TTGGAAGGTCAAGCTGCAAAAGAATTCATTGCTTGGCTGGTGAAAGGCCGTG GTGGTGGCGGCTCTGGTGGCGGTGGCACACCATTGTCTCCACCAACAAACTT GCATCTG

Glpl-Fn7-Reverse:

CGGGATCCACCACCAGCTGGGATGATGGTATCAGAGATAGGGACACTTTCC

The PCR product was digested by restrictive enzyme Ndel and BamUl (Fermentas) and ligated into the digested pET29b-COL18NCl vector. The optimized DNA sequence of human GLP- 1 (7-37) was included in the primer named as Glp 1 -Fn7-Forward. The cloned GLP-l-Fn7-COL18NCl fusion gene was confirmed by DNA sequencing. The protein sequence of GLP- 1 -Fn7-C0L18NC 1 was listed as SEQ ID NO: 1.

SEQ ID NO: 1 , GLP-l-Fn7-COL18NC l protein sequence. Fn7 is connected to GLP- 1 and COL18NC 1 by use of a flexible loop and a flexible unstructured linker. The flexible loop between GLP-1 and Fn7 (GGGSGGGG) is underlined. The flexible, unstructured linker (GGGSGG) between Fn7 and COL18NC 1 is also underlined. GLP- 1 sequence is in italic.

HAEGTFTSD VSSYLEGQAAKEFIA WL VKGRG GGGSGGGG

TPLSPPTNLHLEANPDTGVLTVSWERSTTPDITGYRITTTPTNGQQGNSLEEVVHAD QSSCTFDNLSPGLEYNVSVYTV D

DKESVPISDTIIPAGGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELY VRVQNGFRKVQLEARTPLPRG

Example 2. Cloning of GLP- 1 fused with Fibronectin type III domain 8 (Fn8) and human collagen XVIII NC I domain (COL18NC1 )

Other human Fibronectin type III domains may also act as the proteinous connecting moiety between the therapeutic polypeptide and the scaffold protein in our method. In this example, we showed that Fibronectin type III domain 8 (Fn8) can be utilized as the proteinous connecting moiety between GLP- 1 and collagen XVIII NCI domain. The gene encoding GLP- 1 and human Fibronectin type III domain 8 (Fn8) was amplified by PCR using the following primers and human Fibronectin cDNA as the template:

Fn8-Forward:

GGAATTCCATATGCATGCCGAAGGGACTTTTACCAGTGATGTAAGTTCTTAT TTGGAAGGTCAAGCTGCAAAAGAATTCATTGCTTGGCTGGTGAAAGGCCGTG GTGGTGGCGGCTCTGGTGGCGGTGGCTCTGCTGTTCCTCCTCCCACTGACCT GCGATTC

Fn8-Reverse:

CGGGATCCACCACCACCTGTTTTCTGTCTTCCTCTAAGAGGTGTGC

The PCR product was digested by Ndel and BamR\. The digested insert was ligated into the digested vector pET29b-COL 18NC l (generated in example 1 ). The resulted vector was named as pET29b-GLP- l -Fn8-COL18NC l . The protein sequence of the fusion protein GLP- 1 -Fn8-C0L 18NC 1 was listed as SEQ ID NO: 2.

SEQ ID NO: 2, GLP- 1-Fn8-C0L18NC1 protein sequence, GLP-1 sequence is in italic. Fn8 is connected to GLP-1 and COL18NC 1 by use of a flexible loop and a flexible unstructured linker. The flexible loop between GLP- 1 and Fn8 (GGGSGGGGS) is underlined. The flexible unstructured linker (GGGSGG) between Fn8 and COL18NC 1 is also underlined.

HAEGTFTSD SSVLEGQAAKEF/A WLyKGRGGGGSGGGGSAVPPPTOLRFTN GPOTMRVTVJAPPPSlOLTWLVKY SPV

NEEDVAELS1SPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQ TG

GGGSGGGASSGVRLWATROA LGOVHEVPEGWLIFVAEOEELYVRVQNGFR VQLEARTPLPRG Example 3. Expression and Purification of fusion protein GLP- 1 -Fn8-C0L 18NC 1

The constructed expression vector pET29b-Glp l -Fn8-COL18NC l , after sequencing confirmation, was used to transform Escherichia coli BL21 (DE3) for protein expression (for detailed protocols of the transformation, see Molecular Coining: A Laboratory Manual). A single colony was selected from the culture dish, and placed into a 10 ml LB liquid medium with kanamycin (final concentration, 50 iglm\), then shaken at 37 °C at 220 rpm overnight. 1 L LB culture was inoculated and allowed to grown until OD 60 o reached 0.4- 1.0. Isopropyl thiogalactoside (IPTG) was added to a final concentration of 0.2mM. After a successive culture at 30 °C for overnight, cells were collected by centrifugation. The cells were diluted 1 : 20 with 20mM Tris, NaCl 50mM, 2mM EDTA, pH 8.0, and, after a thorough mix, disrupted by sonication. Insoluble precipitates were removed by centrifugation at 13 ,000 RCF for 30 min. The proteins of interest were present in the supernatant, with the expressed product comprising 20% of soluble proteins. 50ml of the supernatant was loaded on a HiTrap Q column (5ml) (GE healthcare). The fusion protein of GLP 1 -Fn8-C0L 18NC 1 was eluted with about 0.3 M NaCl in the buffer. The eluted protein was further purified by use of a gel filtration column S-200 (GE Healthcare), and the buffer was replaced with PBS (pH 7.5). The final product was confirmed by SDS-PAGE electrophoresis.

Example 4: GLP-1 mutants fused with Fn8-C0L18NC 1

The mutations of A8G/G22E, A8V/G22E, A8S/G22E,

A8G/G22E/R36S and A8G/G22E/R36G within the GLP- 1 sequence may increase its resistance to protease digestion, reduce immunogenicity and boost its biological activity [6, 48]. In this example, we constructed a vector to fuse GLP-1 (A8G/G22E) with Fibronectin type III domain 8 and collagen XVIII NC I domain. The PCR reaction was carried out using vector pET29b-GLP l -Fn8-COL 18NC l prepared in example 2 as the template with the following primers: Glp- l (A8G/G22E)-Fn8-forward:

GGAATTCCATATGCATGGCGAAGGGACTTTTACCAGTGATGTAAGTTCTTAT TT GGAAGAGCAAGCTGCAAAAGAATTCATTGC

Col 18NC 1 -reverse:

CCGCTCGAGTTACCCTCGTGGGAGTGGTGTCCGGGCCTCC

The PCR product was digested by Ndel and Xhol (Fermentas). The digested insert was ligated into the digested vector of pET29b. The resulted vector was named as pET29b-GLP l (A8G/G22E)-Fn8-COL18NC l . The expression and purification protocol of the fusion protein of GLP- l (A8G/G22E)-Fn8-COL 18NCl was the same as described in example 3. Other mutations within the GLP- 1 , such as A8V/G22E, A8S/G22E, A8G/G22E/R36S and A8G/G22E/R36G, can be generated using the Quikchange II site-directed mutagenesis kit (Agilent) using the GLP- 1 (A8G/G22E)-Fn8-C0L18NC 1 gene as the template. The protein sequences of these GLP 1 mutants fused with Fn8-C0L 18NC 1 were listed as SEQ ID NO: 3-7. The expression and purification protocol of these fusion proteins can be carried out using similar protocols described in example 3.

SEQ ID NO: 3, GLP-1 (A8G/G22E)-Fn8-C0L18NC 1 protein sequence, the GLP- 1 mutation sites (A8G/G22E) are underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGRGGGGSGGGGSAVPPPTDLRFTNIGPDTMRV TWAPPPSIDLTNFLVRYSPV K

NEEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFR VQLEARTPLPRG SEQ ID NO: 4, GLP- 1 (A8V/G22E)-Fn8-COL18NC l protein sequence, the GLP- 1 mutation sites

(A8V/G22E) are underlined.

HVEGTFTSDVSSYLEEQAAKEFIAWLVKGRGGGGSGGGGSAVPPPTDLRFTNIGPDTMRV TWAPPPSIDLTN FLVRYSPV

NEEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQ TG

GGGSGGGASSGVRLWATRQA LGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 5, GLP- 1 (A8S/G22E)-Fn8-COL18NC l protein sequence, the GLP- 1 mutation sites (A8S/G22E) are underlined.

HSEGTFTSDVSSYLEEQAAKEFIAWLV GRGGGGSGGGGSAVPPPTDLRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPV

NEEDVAELSISPSDNAVVLT LLPGTEYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEA RTPLPRG

SEQ ID NO: 6, GLP- 1 (A8G/G22E/R36S)-Fn8-COL18NC l protein sequence, the GLP- 1 mutation sites (A8G/G22E/R36S) are underlined.

HGEGTFTSDVSSYLEEQAA EFIAWLVKGSGGGGSGGGGSAVPPPTDLRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPV

NEEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEA RTPLPRG

SEQ ID NO: 7, GLP- 1 (A8G/G22E/R36G)-Fn8-COL 18NC l protein sequence, the GLP-1 mutation sites

(A8G/G22E/R36G) are underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGGGGSGGGGSAVPPPTDLRFTNIGPDTMRV TWAPPPSIDLTNFLVRYSPV

NEEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQ TG

GGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEA RTPLPRG

Example 5. Between the therapeutic polypeptides and the scaffold proteins, various lengths of the flexible un-structured linkers can be used to generate the fusion proteins with the desired hydrodynamic radius and/or radius of gyration (Rg)

In this example, we presented data to demonstrate that the flexible unstructured linkers with various lengths can be utilized in our method to adjust the hydrodynamic radius and/or Rg of the fusion protein. It is well established that a protein with a larger hydrodynamic radius and/or Rg may exhibit a longer half life in vivo. Therefore, the method of the invention may adjust the in vivo half life of the therapeutic polypeptide in a tunable fashion.

In the fusion protein of GLP- 1(A8G/G22E)-Fn8-C0L18NC 1 described in example 4, the flexible unstructured linker between Fn8 and COL18NC 1 contains six residues (GGGSGG). To generate the flexible un-structured linkers with different lengths, we have synthesized the genes GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -20,

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -30,

GLP-l(A8G/G22E/R36S)-Fn8-COL18NC l-54 and GLP- l(A8G/G22E/R36S)-Fn8-COL18NC l-60. In these genes, the length of the flexible unstructured polypeptide linker between Fn8 and COL18NC1 contained 20, 30, 54 and 60 residues, respectively. The protein sequences of

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -20,

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -30,

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -54 and

GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl-60 are listed as SEQ ID NO: 8-1 1.

The synthetic genes of GLP- l(A8G/G22E/R36S)-Fn8-COL18NCl-20,

GLP- l(A8G/G22E/R36S)-Fn8-COL18NC l-30,

GLP-l(A8G/G22E/R36S)-Fn8-COL18NC l-54 and GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl-60 were grafted to the pET29b by Ndel and Xhol for protein expressions. The expression and purification of these fusion proteins were carried out using similar protocols described in example 3. To estimate the hydrodynamic radius and/or the Rg of the fusion proteins, the purified proteins were loaded on an analytical gel filtration column Superdex200 (GE Healthcare).

The gel filtration data clearly showed that varying the length of the flexible, unstructured linker between Fn8 and COL18NC 1 can significantly change the apparent molecular size of the fusion proteins in solution (Fig. 3). GLP-l (A8G/G22E/R36S)-Fn8-COL18NC l trimer exhibited an apparent molecular weight of ~100Kd while its genuine molecular weight is ~68Kd. GLP-l (A8G/G22E/R36S)-Fn8-COL18NCl -60 trimer (data not shown) and GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl -54 trimer exhibited an apparent molecular weight of ~200Kd while its genuine molecular weight is ~80Kd. GLP-l(A8G/G22E/R36S)-Fn8-COL18NC l-20, GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl-30 exhibited larger apparent molecular weight than their genuine molecular weight as well. Therefore, our method can provide the therapeutic polypeptide with a larger hydrodynamic Radius and/or Rg which exhibited increased apparent molecular size on gel filtration profile. Moreover, the flexible unstructured linker between the therapeutic polypeptide and the scaffold protein may adjust the hydrodynamic radius and/or the Rg of the fusion molecule in a tunable manner.

SEQ ID NO: 8, protein sequence of GLP- l (A8G/G22E/R36S)-Fn8-COL18NC l -20, the flexible unstructured linker (20 residues) between Fn8 and COL18NC 1 is underlined. The flexible loop between GLP-1 and Fn8 (GGGSGG) is also underlined.

HGEGTFTSDVSSYLEEQAAK.EFIAWLVKGSGGGGSGGAVPPPTDLRFTNIGPDTMRVTW APPPSIDLTNFLVRYSPVKN

EEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEOHESTPLRGRO TGGGGGSGGGGSGGGGSGGGGS

GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPR G

SEQ ID NO: 9, protein sequence of GLP- l (A8G/G22E/R36S)-Fn8-COL18NC l -30, the flexible unstructured linker (30 residues) between Fn8 and COL18NC1 is underlined. The flexible loop between GLP-1 and Fn8 (GGGSGG) is also underlined.

HGEGTFTSDVSSYLEEOAAKEFIAWLV GSGGGGSGGAVPPPTDLRFTNIGPDT RVTWAPPPSIDLTNFLVRYSPVK

EEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQKTG

GGGGSGGGGSASSASTGGPSGGGGSGGGGS

GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPR G SEQ ID NO: 10, protein sequence of GLP-l (A8G/G22E/R36S)-Fn8-COL18NC l-54, the flexible unstructured linker (54 residues) between Fn8 and COL 18NC1 is underlined. The flexible loop between GLP-1 and Fn8 (GGGSGG) is also underlined.

HGEGTFTSDVSSYLEEOAAKEFIAWLVKGSGGGGSGGAVPPPTDLRFTNIGPDTMRVTWA PPPSIDLTNFLVRYSPVKN EEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQ TG GGGGSGGGGSTASSASTGGPSGGGGSGGGGSAPSSGSTSGGTAAGGGGSGGGGS GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 1 1 , protein sequence of GLP-l (A8G/G22E/R36S)-Fn8-COL18NC l-60, the flexible unstructured linker (60 residues) between Fn8 and COL18NC1 is underlined. The flexible loop between GLP- 1 and Fn8 (GGGSGG) is also underlined.

HGEGTFTSDVSSYLEEQAA £FIAWLV GSGGGGSGGAVPPPTDLRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKN EEDVAELSISPSDNAVVLTNLLPGTEYVVSVSSVYEQHESTPLRGRQKTG GGGSGGGSGGGSTASSASTKGPSGGGSGGGSGGGSAPSS STSGGTAAGGGSGGGSGGGS GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFR VQLEARTPLPRG

Example 6 : The pharmacokinetics studies for GLP-l (A8G/G22E/R36S)-Fn8-COL18NCl ,

GLP- l(A8G/G22E/R36S)-Fn8-COL18NC l-20,

GLP-l (A8G/G22E/R36S)-Fn8-COL18NCl-30,

GLP- 1 ( A8G/G22E/R36S)-Fn8-COL 18NC 1-54.

To evaluate the pharmacokinetics profiles for the GLP-1 containing fusion proteins generated using the method of the invention, we purified GLP- 1 (A8 G/G22E/R36S)-Fn8-COL 18NC 1 ,

GLP- l(A8G/G22E/R36S)-Fn8-COL18NCl-20,

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -30,

GLP- 1 (A8G/G22E/R36S)-Fn8-COLl 8NC 1 -54 in PBS buffer, pH 7.2. These fusion proteins were administered on Sprague-Dawley (SD) rats by intraperitoneal injections at the doses of 0.66mg/kg, 0.72mg/kg, 0.75mg/kg,0.78mg/kg animal respectively. Blood samples were taken at various time points after injections such as 0-min, 30-min, 1-hour, 2-hour, 4-hour, 8-hour, 24-hour, 48-hour, 3-day, 4-day, 5-day. 7-day, 10-day. The serum samples were centrifuged and kept at -80°C freezer.

The GLP-1 concentrations within the serum samples were examined by use of the sandwich ELISA method. The rabbit polyclonal antibody against human Fibronectin at the concentration of 3ug/ml (Ab299, Abeam company) was coated on ELISA plate for 1 hour at room temperature. Then the plate was washed by PBST buffer three times and the wells were blocked by PBS with 10% FBS for 1 hour at room temperature. The plate was washed three times before the serum samples containing GLP-1 fusion proteins were added. The serum samples could be diluted to 20-10000 folds before use. The ELISA plate was incubated with the serum samples at room temperature for 1 hour and then washed by PBST buffer five times. Then mouse monoclonal against human GLP-1 peptide antibody (sc57510, Santa Cruz Biotich) at the concentration of lug/ml in PBST buffer was added to the wells. The plate was washed extensively after incubation of 1 hour at room temperature. The secondary antibody, Goat anti-rabbit IgG HRP conjugated antibody (Beijing ZSGB-Bio company, ZB 5301), was added into the wells and the color was developed using TMB (3,3 ',5,5 '-tetramethylbenzidine, BD biosciences, Cat 555214 ). The plate reader (Bio-Rad microplate reader Model 680) was utilized to obtain the OD450 readings. This method has been calibrated using purified proteins first.

Fig. 4 showed the pharmacokinetics profiles of the GLP- 1 containing proteins by use of the sandwich ELISA method described above. The pharmacokinetics parameters were obtained by using the WinNonlin software (Table 3). The data clearly showed that the GLP- 1 containing fusion proteins generated by use of the method of the invention exhibited much extended in vivo half life possibly due to their enlarged Rg. The data also showed that the flexible, unstructured linker between the therapeutic polypeptide and the scaffold protein may adjust the in vivo half life of the fusion molecules in a tunable manner.

Table 3. Pharmacokinetics parameters for the GLP-1 containing fusion proteins in Sprague-Dawley rat. GLP- 1 (A8G/G22E/R36S)-Fn8-COL18NCl , GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl-20,

GLP- 1 (A8G/G22E/R36S)-Fn8-COL 18NC 1 -30,

GLP-l(A8G/G22E/R36S)-Fn8-COL18NCl-54 were shown in abbreviation as NCI, NCI -20, NCI -30, and NCI -54 respectively.

Example 7: construction of the fusion protein containing GLP-1 mutants, Fn8, pCloud sequence and COL18NC1

In this example, we introduced pCloud polypeptide into the GLP-1 containing fusion protein. We constructed the vector to fuse GLP-1 wild type or its mutants with Fibronectin type III domain 8(Fn8), the pCloud polypeptide and collagen XVIII NCI domain.

The pCloud sequence in this example (p246) comprises all the 12 fibrinogen fragments listed in table 1. In the protein sequence of p246, the 12 fragments listed in table 1 were placed in the order as they appear in the human fibrinogen alpha chain sequence. The flexible loops that were utilized to connect these fibrinogen-derived fragments in p246 sequence are GSGSESGSG, GGGSGGGS and GGSGGGSGG. The optimized gene encoding p246 andCOL18NCl was synthesized, digested by BamHI and Xhol and ligated into the digested vector of pET29b. The resulted vector was named as pET29b-p246-COL18NCl. The optimized gene encoding GLP-1(A8G/G22E) and Fn8 was synthesized, digested by Ndel and BamHI and ligated into the digested pET29b-p246-COL18NCl. The resulted vector was named as pET29b-GLP-l(A8G/G22E)-Fn8-p246-COL18NCl. The protein sequence of the fusion protein GLP-l(A8G/G22E)-Fn8-p246-COL18NCl was listed as SEQ ID NO: 12. From N-terminus to C-terminus, GLP- l (A8G/G22E)-Fn8-p246-COL 18NCl contains GLP- 1 (A8G/G22E), a flexible loop, Fn8, pCloud sequence p246 and the scaffold protein COL18NC 1 .

Other mutations within the GLP- 1 , such as A8V/G22E, A8S/G22E,

A8G/G22E/R36S and A8G/G22E/R36G, can be generated using the Quikchange II site-directed mutagenesis kit (Agilent) using the GLP- l (A8G/G22E)-Fn8-p246-COL 18NCl gene as the template. The resulted vectors of the fusion proteins were named as P ET29b-GLP- l (A8V/G22E)-Fn8-p246-COL 18NC l ,

pET29b-GLP- 1 ( A8 S/G22E)-Fn8-p246-COL 18NC 1 ,

P ET29b-GLP- l (A8G/G22E/R36S)-Fn8-p246-COL 18NC l and pET29b-GLP- 1 ( A8G/G22E/R36G)-Fn8 -p246-COL 18NC 1. The protein sequences of these fusion proteins were listed as SEQ ID NO: 13- 16. The expression and purification protocol of these fusion proteins can be carried out using similar protocols described in example 3.

SEQ ID NO: 12, GLP-l(A8G/G22E)-Fn8-p246-COL18NCl protein sequence, the GLP-1 mutation sites (A8G/G22E) are in Italic and bold. The flexible loop between GLP1(A8G/G22E) and Fn8 is in bold. The pCloud sequence p246 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGRGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSESGSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGG5GSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEG LIF VAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 13, GLP-1 (A8V/G22E)-Fn8-p246-COLl 8NC1 protein sequence, the GLP-1 mutation sites (A8V/G22E) are in Italic and bold. The pCloud sequence p246 is underlined.

HVEGTFTSDVSSYLEEQAAKEFIAWLVKGRGSGGGSGGGSGGGSGGGSGSGGAVPPP TD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESG SGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPG SGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRL ATRQAMLGQVHEVPEGWLIF

VAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 14, GLP-1 (A8S/G22E)-Fn8-p246-COL18NCl protein sequence, the GLP-1 mutation sites (A8S/G22E) are in Italic and bold. The pCloud sequence p246 is underlined.

HSEGTFTSDVSSYLEEQAAKEFIAWLVKGRGSGGGSGGGSGGGSGGGSGSGGAVPPP TD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNA VLTNLLPGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESG SGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPG SGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGW LIF

VAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 15, GLP-l(A8G/G22E/R36S)-Fn8-p246-COL18NCl protein sequence, the GLP-1 mutation sites (A8G/G22E/R36S) are in Italic and bold. The pCloud sequence p246 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGSGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAEPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSC-SSGPGSTGGSGSESGSGPGSSGTGGTATGSGSE SGSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSE5GSGPGSPRPGSTGTGSGSESGSGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGW LIF

VAEQEELYVRVQNGFRKVQLEARTPLPRG SEQ ID NO: 16, GLP-l(A8G/G22E/R36G)-Fn8-p246-COL18NCl protein sequence, the GLP-1 mutation sites (A8G/G22E/R36G) are in Italic and bold. The flexible loop between GLP1(A8G/G22E/R36G) and Fn8 is in bold. The pCloud sequence p246 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSESGSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIF VAEQEELYVRVQNGFRKVQLEARTPLPRG

Example 8 : Cloning and expression of GLP- 1 (A8G/G22E/R36G) fused with Tenascin C fibronectin type III domain 3 (TNCfn3), pCloud sequence p246 and human collagen XVIII NC 1 (COL 18NC 1 )

In this example, we demonstrated that the fibronectin type III domain that can be used as the proteinous connecting moiety for our method is not limited within human fibronectin. Other suitable fibronectin type III domain may alternatively be utilized as the proteinous connecting moiety in the method of the invention. Here we showed that a fibronectin type III domain from human Tenascin C can be utilized to connect the therapeutic polypeptide and the pCloud sequence as well.

In this example, the human Tenascin C fibronectin type III domain 3 (TNCfn3) was connected to the GLP- 1 (A8G/G22E/R36G) and the pCloud sequence as the proteinous connecting moiety. The gene encoding GLP-1 (A8G/G22E/R36G), the flexible loop and TNCfn3 was synthesized, digested by Ndel and BamHI and ligated into the digested pET29b-GLP-l(A8G/G22E/R36G)-p246-COL 18NCl vector. The resulted vector was named as pET29b-GLP l (A8G/G22E/R36G)-TNCfn3-p246-COL18NC l . The expression and purification protocol of the fusion protein GLP l (A8G/G22E/R36G)-TNCfn3-p246-COL 18NC l (SEQ ID NO: 17) was the same as described in example 3. SEQ ID NO: 17, GLP-1 (A8G/G22E/R36G)- TNCfh3-p246-COL18NCl protein sequence, The flexible unstructured linker between GLP1(A8G/G22E/R36G) and TNCfh3 is in bold. The TNCfn3 sequence is in italic. The pCloud sequence p246 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGG

RLDAPSQIEVKDVTDTTALI TWFKPLAEIDGIEL TYGIKDVPGDR TTIDL TEDENQ YSIG

NLKPD TE YEVSLISRR GDMS SNPAKE TF TG GGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSES5ASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIF

VAEQEELYVRVQNGFRKVQLEARTPLPRG

Example 9: Cloning and expression of GLP- 1 (A8G/G22E/R36G) fused with Fn8, pCloud sequence p246 and human collagen XV NC I (COL 15NC 1 )

In this example, we presented data to show that the NC I domain from collagen XV can alternatively be utilized as the scaffold protein in our method. It has been reported that human collagen XV NC I domain, like the human collagen XVIII NC I domain, forms a stable homo-trimer[26, 32] . To generate the expression vector encoding the fusion protein of GLP l (A8G/G22E/R36G)-Fn8-p246-COL15NCl , the gene encoding pCloud sequence p246 and COL15NC 1 domain was synthesized, digested by BamHI and Xhol and ligated into the digested pET29b-GLP- l (A8G/G22E/R36G)-Fn8-p246-COL 18NC l vector generated before. The resulted vector was named as pET29b-GLP l (A8G/G22E/R36G)-Fn8-p246-COL15NC l and the sequence of the vector was confirmed by DNA sequencing. The protein sequence of GLP 1 (A8G/G22E/R36G)-Fn8-C0L15NC 1 was listed as SEQ ID NO: 18. The expression and purification protocol of the fusion protein GLP 1 (A8G/G22E/R36G)-Fn8-C0L 15NC 1 was the same as described in example 3. SEQ ID NO: 18, GLP-l (A8G/G22E/R36G)-Fn8-p246-COL15NCl protein sequence, the sequence of COL15NC1 is underlined. The GLP-1 mutation sites (A8G/G22E/R36G) are in Italic.

HOTGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPP TD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESG SGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPG SGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGG

NLVTAFSNMDDMLQKAHLVIEGTFIYLRDSTEFFIRVRDG KKLQLGELIPIPA

Example 10: Cloning and expression of GLP- 1 (A8G/G22E/R36G) fused with Fn8, pCloud sequence and human collagen XIX NC2 domain (COL19NC2)

In this example, we demonstrated that the NC2 domain from collagen XIX can be utilized as the scaffold protein in our method. It has been shown that human collagen XIX NC2 domain (COL 19NC2) forms a highly stable homo-trimer[28]. To generate the expression vector encoding the fusion protein of GLP l (A8G/G22E/R36G)-Fn8-p246-COL19NC2, the gene encoding pCloud sequence p246 and COL19NC2 domain was synthesized, digested by BamHI and Xhol and ligated into the digested pET29b-GLP- 1 (A8G/G22E/R36G)-Fn8 -p246-COL 15NC 1 vector generated before. The resulted vector was named as pET29b-GLP l (A8G/G22E/R36G)-Fn8-p246-COL19NC2 and the sequence of the vector was confirmed by DNA sequencing. The protein sequence of GLP l (A8G/G22E/R36G)-Fn8-p246-COL19NC2 was listed as SEQ ID NO: 19. The expression and purification protocol of the fusion protein GLP l (A8G/G22E/R36G)-Fn8-p246-COL19NC2 was the same as described in example 3. SEQ ID NO: 19, GLP-l(A8G/G22E/R36G)-Fn8-p246-COL19NC2 protein sequence, the sequence of COL19NC2 is underlined. The GLP-1 mutation sites (A8G/G22E/R36G) are in Italic.

HOTGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESG SGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPG SGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGG

GIPADAVSFEEIKKYINQEVLRIFEERMAVFLSQLKLPAAMLAAQAYGRP

Example 1 1 : Cloning and expression of GLP- 1 (A8G/G22E/R36G) fused with Fn8, pCloud sequence and human ACRP30 C-terminal Cl q-like domain

ACRP30 (also referred to as Adiponectin, GBP-28, apMl and AdipoQ) is a protein hormone that modulates a number of metabolic processes, including glucose regulation and fatty acid catabolism[49]. ACRP30 contains a C-terminal globular domain that forms a homo-trimer with typical Clq-like structure [50]. In this example, we demonstrated that the C l q-like domain, such as the ACRP30 C l q-like domain, can be utilized as the scaffold protein in our method. To generate the expression vector encoding the fusion protein of GLP l (A8G/G22E/R36G)-Fn8-p246-ACRP30 C l q-like domain, the gene encoding pCloud sequence p246 and ACRP30 C l q-like domain was synthesized, digested by BamHI and Xhol and ligated into the digested pET29b-GLP- l (A8G/G22E/R36G)-Fn8-p246-COL15NC l vector generated before. The resulted vector was named as pET29b-GLP l (A8G/G22E/R36G)-Fn8-p246-ACRP30 and the sequence of the vector was confirmed by DNA sequencing. The protein sequence of GLPl (A8G/G22E/R36G)-Fn8-p246-ACRP30 was listed as SEQ ID NO:20. SEQ ID NO: 20, GLP l (A8G/G22E/R36G)-Fn8-p246-ACRP30 protein sequence, the sequence of ACRP30 C l q-like domain is underlined. The GLP-1 mutation sites (A8G/G22E/R36G) are in Italic.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESP GSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG

PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESG SGP

GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPG SGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGG

VYRSAFSVGLETRVTVPNVPIRFTKIFYNQQNHYDGSTGKFYCNIPGLYYFSYHITV Y KD

VKV

SLFKKDKAVLFTYDQYQEKNVDQASGSVLLHLEVGDQVWLQVYGDGDHNGLYADNVN DSTF TGFLLY

HDTN

Example 12: Constructions of the fusion proteins of GLP- 1 (A8G/G22E/R36G), Fn8, pCloud sequences with various lengths and COL18NC l

Numerous pCloud sequences can be generated by use of the human Fribrinogen fragments listed in Table 1 and the flexible loops listed in table 2. In the pCloud sequences, the fibrinogen fragments are flanked by flexible loops. In this example, we generated three pCloud sequences p245, p271 and p285 using the described method. In the pCloud sequences of p245 and p271 , the 12 human fibrinogen alpha chain fragments listed in table 1 were connected in the order as they appear in the human fibrinogen alpha chain sequence to constitute the pCloud sequences. In the case of p285, the fragments listed in table 1 were connected in the order that is distinct from they appear in the human fibrinogen alpha chain sequence to generate the pCloud sequence. In p285, some fibrinogen alpha chain fragments were utilized more than once. To construct p245, the flexible loops GGGSGGGSGS, GSGSESTSG, GSTSESGSG, GSTSGSESG, GSESGSTSG, GSESTSGSG, GSGSTSESG and GGSGGGSGG listed in table 2 were utilized to connect the human fribrinogen alpha chain fragments. To construct p271 , the flexible loops GGGSGGGS, GGSGGGSGG and GGSGSESGSGG were utilized to connect the human fibrinogen alpha chain fragments. To construct p285, the flexible loops GGGSGGGS, GGSGGGSGG and GSGSESGSG were utilized to connect the fibrinogen alpha chain fragments.

The genes encoding pCloud sequence (p245, p271 and p285, respectively) and the scaffold protein COL18NC 1 was synthesized, digested by BamHI and Xhol and ligated into the digested vector pET29b-GLP- 1 (A8G/G22E/R36G)-Fn8 -p246-COL 18NC 1 generated before. The resulted vector were named as pET29b-GLP- l (A8G/G22E/R36G)-Fn8-p245-COL18NC l ,

pET29b-GLP- 1 (A8G/G22E/R36G)-Fn8-p271 -COL 18NC 1 and pET29b-GLP- l (A8G/G22E/R36G)-Fn8-p285-COL18NC l . The resulted fusion proteins were named as

GLP- l (A8G/G22E/R36G)-Fn8-p245-COL18NC l ,

GLP- 1 ( A8 G/G22E/R36G)-Fn8 -p271 -COL 18NC 1 and GLP- l (A8G/G22E/R36G)-Fn8-p285-COL18NC l , respectively. The protein sequences of these fusion proteins were listed as SEQ ID NO:21 -23. These fusion proteins were expressed and purified using the protocols described in example 3.

SEQ ID NO: 21 , GLP-l (A8G/G22E/R36G)-Fn8-p245-COL18NCl protein sequence, the GLP-1 mutation sites (A8G/G22E/R36G) are in Italic and bold. The flexible loop between GLP1(A8G/G22E/R36G) and Fn8 is in bold. The pCloud sequence p245 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT EYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGSGSGSTSESGGSTSSGTGSETESPGSGSESTSGPSSAGSGSTSESGSGSG SSGPG

STGGSTSGSESGPGSSGTGGTATGSESGSTSGPGSSGPGSTGSGSTSESGSGSGSSG TGS

TGGSESGSTSGPGSPRPGSTGTGSTSGSESGPGSSERGSAGGSESTSGSGTSESSAS GST

GGSTSGSESGSESGSGSTSESGSGPESPGSGGSGSTSESGTSGSTGSESTSGSGGGS GGG

SGG

GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPR G SEQ ID NO: 22, GLP- l(A8G/G22E/R36G)-Fn8-p271 -COL18NCl protein sequence, the GLP-1 mutation sites (A8G/G22E/R36G) are in Italic and bold. The flexible loop between GLP1 (A8G/G22E/R36G) and Fn8 is in bold. The pCloud sequence p271 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT

EYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGSGGSGSESGSGGGSTSSGTGSETESPGGSG

SESGSGGPSSAGSGGSGSESGSGGSGSSGPGSTGGGSGSESGSGGPGSSGTGGTATG GSGSESGSGG PGSSGPGSTGSGGSGSESGSGGSGSSGTGSTGGGSGSESGSGGPGSPRPGSTGTGGSGSE SGSGGP GSSERGSAGGGSGSESGSGGTSESSASGSTGGGSGSESGSGGSESGSGGSGSESGSGGPE SPGSGGG SGSESGSGGTSGSTGGSGSESGSGGGGSGGGSGG

GASSGVRLWATRQAMLGQVHEVPEGWLI FVAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 23, GLP-1 (A8G/G22E/R36G)-Fn8-p285-COL18NCl protein sequence, the GLP- 1 mutation sites (A8G/G22E/R36G) are in Italic and bold. The flexible loop between GLP1(A8G/G22E/R36G) and Fn8 is in bold. The pCloud sequence p285 is underlined.

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGSGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT EYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSGSESGSGPSSAGSGSGSESGSGS GSSGP

GSTGGSGSESGSGPGSSGTGGTATGSGSESGSGPGSSGPGSTGSGSGSESGSGSGSS GTG

STGGSGSESGSGPGSPRPGSTGTGSGSESGSGPGSSERGSAGGSGSESGSGTSESSA SGS

TGGSGSESGSGSESGSGSGSESGSGPESPGSGGSGSESGSGTSGSTGSGSESGSGPG SSG

PGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGGGSGGGSGG

GASSGVRLWATRQAMLGQVHEVPEGWLI FVAEQEELYVRVQNGFRKVQLEARTPLPRG

Example 13. The therapeutic polypeptide fused with pCloud polypeptides and the scaffold protein exhibits a much larger hydrodynamic radius and/or Rg for its molecular weight

In this example, we presented data to demonstrate that connecting the therapeutic polypeptide with the unstructured pCloud polypeptide and the trimeric scaffold protein can render the therapeutic polypeptide a much larger hydrodynamic radius and/or Rg compared with that for a tightly folded protein with the same molecular weight. The apparent molecular weights of the fusion proteins were estimated by use of an analytical gel filtration column Superdex-200 (GE Healthcare) mounted on the AKTA FPLC system (GE Healthcare). The purified fusion proteins GLP-1 -Fn8, GLP- 1 -Fn8 -COL 18NC 1 , GLP- 1 (A8G/G22E/R36G)-Fn8-p246-COL 18NC 1 were loaded on the Superdex200 column. GLP-1 -Fn8 is a fusion protein of GLP- 1 and Fn8 and contains the first 144 amino acid residues of the fusion protein GLP- 1 -Fn8-C0L18NC 1 (SEQ ID NO:2). GLP- 1 -Fn8 does not contain the scaffold protein COL 18NC 1 , so it forms a monomer in solution. Fig. 5 showed the chromatography profiles for these fusion proteins. The apparent molecular weights for GLP-1 -Fn8, GLP- 1 -Fn8-C0L18NC 1 , GLP- 1 (A8G/G22E/R36G)-Fn8-p246-COL18NCl showed by gel filtration analysis were ~20Kd, ~100Kd and ~550Kd, respectively. The genuine molecular weights for the fusion proteins GLP- 1 -Fn8 monomer, GLP- 1 -Fn8-C0L18NC 1 trimer, and

GLP-l (A8G/G22E/R36G)-Fn8-p246-COL18NCl trimer were ~15Kd, 65Kd and 123Kd, respectively. The data clearly showed that the trimer formation caused by the scaffold protein COL18NC1 domain provided GLP- 1 -Fn8-C0L18NC 1 a larger apparent molecular weight compared with its genuine molecular weight ( l OOKd vs 65Kd). More importantly, the data further indicated that including the unstructured pCloud polypeptide into the fusion protein GLP- 1 (A8G/G22E/R36G)-Fn8-p246-COL18NCl dramatically enlarged the apparent molecular weight of the fusion protein compared with its genuine molecular weight (550Kd vs 123Kd). Therefore, our "Trident Technology" method could provide the therapeutic polypeptide with a large hydrodynamic radius which exhibited the increased apparent molecular size as shown by gel filtration profile for the extended in vivo half life.

Example 14. cAMP assay for measuring GLP- 1 activity

Through binding and activating a specific G protein-coupled receptor

(GLP-1 receptor), GLP- 1 stimulates the signaling pathway to increase cAMP level in cells. Therefore, measuring the cytoplasmic cAMP level can be an accurate method to evaluate the biological activity of GLP-1. Chinese Hamster Ovary (CHO) cells stably transfected with human GLP-1 receptor (GLP-1 R) were generated and named as S-CHO cells. S-CHO cells were propagated in DMEM medium with 10% FCS containing 0.05mg/ml G418. Before analysis, SCHO cells were grown to 70-80% confluence in 6-well plates at 37°C. The cells were treated 0.2mM 3-isobutyl-l -methylxanthine (IBMX). Cells were incubated with GLP- 1 fusion proteins at various concentrations of InM, 3nM, l OnM, 33nM, lOOnM for 15 min at 37°C. The cells were then lysed by use of cold lysis buffer. The supernatants of the cell extracts were used for cAMP level determinations. The Parameter cAMP ELISA kit from R&D Systems was utilized to measure the cAMP concentrations in the cell lysates. The EC50 values of the GLP- 1 fusion proteins were generated by using the software Origin. The GLP- 1 (7-37) peptide (Anaspec) and BSA were used as positive and negative controls. Fig. 6 showed the results of cAMP assays for GLP1 (7-37) peptide and some other GLP- 1 fusion proteins. The EC50 values of a number of GLP-1 containing fusion proteins are listed in Table 4. The data indicated that the fusion of GLP 1 with the flexible unstructured polypeptide and the scaffold protein did not affect GLP-1 activity. As a matter of fact, if the lengths of flexible unstructured linker sequences are increased, the activities of fusion protein improved. We reasoned that a longer flexible, unstructured linker and/or the pCloud polypeptide may provide the GLP-1 peptide with more freedom to interact with the GLP-1 receptor.

Table 4. The activities of GLP- 1 and GLP- 1 containing fusion proteins measured by cAMP assay

Example 15 : The pharmacokinetics studies for GLP- 1 -Fn8-COL 18NC 1 ,

GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NCl and GLP l (A8G/G22E/R36G)-Fn8-p285-COL 18NC l

To evaluate the pharmacokinetics profiles for the GLP-1 containing fusion proteins generated using the method of the invention, we purified the recombinant fusion proteins of GLP- 1 -Fn8 -COL 18NC 1 , GLP l (A8G/G22E/R36G)-Fn8-p246-COL 18NCl and GLP l (A8G/G22E/R36G)-Fn8-p285-COL 18NCl in PBS buffer, pH 7.2. These fusion proteins were administered into SD(Sprague-Dawley) rats by intraperitoneal injections at the dose of 5 nmol/kg for animals, respectively. Blood samples were taken at various time points after injections such as 0-min, 30-min, 1 -hour, 2-hour, 4-hour,6-hour, 8-hour, 24-hour, 48-hour, 3-day, 4-day, -day, 6-day and 7-day. The serum samples were centrifuged and kept at -80°C freezer.

The GLP- 1 concentrations within the samples were examined by use of the sandwich ELISA method. The rabbit polyclonal antibody against human Fibronectin at the concentration of 3ug/ml (Ab299, Abeam company) was coated on ELISA plate for 1 hour at room temperature. Then the plate was washed by PBST buffer three times and the wells were blocked by PBS with 10% FBS for 1 hour at room temperature. The plate was washed three times before the serum samples containing GLP- 1 fusion proteins were added. The serum samples could be diluted to 20-10000 folds before use. The ELISA plate was incubated with the serum samples at room temperature for 1 hour and then washed by PBST buffer five times. Then mouse monoclonal against human GLP- 1 peptide antibody (sc57510, Santa Cruz Biotich) at the concentration of lug/ml in PBST buffer was added to the wells. The plate was washed extensively after incubation of 1 hour at room temperature. The secondary antibody, Goat anti-rabbit IgG HRP conjugated antibody (Beijing ZSGB-Bio company, ZB 5301), was added into the wells and the color was developed using TMB (3,3',5,5 '-tetramethylbenzidine, BD biosciences, Cat 555214 ). The plate reader (Bio-Rad microplate reader Model 680) was utilized to obtain the OD450 readings. This method has been calibrated using purified proteins first.

Fig. 7 showed the pharmacokinetics profiles of the GLP-1 containing proteins in SD rats by use of the sandwich ELISA method described above. The pharmacokinetics parameters were obtained by using the WinNonlin software (Table 5). The data clearly showed that the GLP- 1 containing fusion proteins generated by use of the method of the invention exhibited much extended in vivo half life possibly due to their enlarged hydrodynamic radius and/or Rg. Particularly, including the pCloud polypeptide into the fusion protein dramatically improved the pharmacokinetic profile of fusion protein. The half life of GLP- 1 -Fn8-C0L 18NC 1 reached 5.3 hours compared with the half life of a couple of minutes for GLP- 1 peptide. The half life of GLP 1 (A8G/G22E/R36G)-Fn8-p246-COLl 8NC 1 and GLP l (A8G/G22E/R36G)-Fn8-p285-COL 18NC l were further extended to 30.8 and 31.2 hours, respectively, due to the applications of the flexible unstructured pCloud sequences within the fusion proteins.

The toxicity of GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l in SD rats was also examined. A single dose of 30nmol/kg and repeated doses of 5nmol/kg every three days for four weeks did not induce unacceptable adverse effects such as significant weight loss and fever in rats.

To further investigate the pharmacokinetics profiles for the GLP-1 containing fusion proteins generated using the method of the invention, the fusion protein GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l was administrated into the cynomolgus monkeys by subcutaneous injection. The fusion protein was administered on three cynomolgus monkeys at the dose of 5 nmol/kg. Blood samples were taken at various time points after injections such as 0-min, , 2-hour, 4-hour, 6-hour, 8-hour, 24-hour, 48-hour, and daily until the 15th day. The serum samples were centrifuged and kept at -80°C freezer. The concentrations of the fusion protein within the serum samples were examined by using the sandwich ELISA method described above. The pharmacokinetics profile of the fusion protein GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NCl in cynomolgus monkeys was shown in Fig. 8. The pharmacokinetics parameters were obtained by using the WinNonlin software (Table 5). The half life of the fusion protein GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NCl in in cynomolgus monkeys was estimated to be -67 hours, which is significantly longer than the half life of GLP- 1 -Fc fusion protein (half life -51 hours) in monkeys [6]. The pharmacokinetics data in rats and monkeys strongly suggested that a weekly dose or even once every ten day of GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NCl in human may be effective.

The serum samples of the cynomolgus monkeys receiving the fusion protein GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l were withdrawn one month after dosing to examine whether specific antibody against GLPl (A8G/G22E/R36G)-Fn8-p246-COL 18NC l has been generated. The analysis of the serum samples of the three monkeys receiving the fusion protein by use of ELISA method indicated that no specific antibody was induced against GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l in any of the cynomolgus monkeys.

Table 5. Pharmacokinetics parameters for the GLP-1 containing fusion proteins in Sprague-Dawley rats and cynomolgus monkeys. In the table, the fusion protein GLP- 1 -Fn8-C0L18NC 1 ,

GLP 1 ( A8 G/G22E/R36G)-Fn8 -p246-COL 18NC 1 and GLP l (A8G/G22E/R36G)-Fn8-p285-COL18NCl were shown in abbreviations as NC I , p246-NC l and p285-NC l .

Example 16: the Intraperitoneal glucose tolerance test (IPGTT) of GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l in SD rats.

To evaluate the efficacy of GLPl (A8G/G22E/R36G)-Fn8-p246-COL 18NC l to reduce the glucose level in animal models, we performed the Intraperitoneal glucose tolerance test (IPGTT) in SD rats. The fusion protein GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l was administered on SD(Sprague-Dawley) rats by intraperitoneal injections at the dose of 10 nmol/kg and 20 nmol/kg for animals, respectively. Glucose was injected into the animals with the dose of 2g/kg at 8hours, 32 hours and 100 hours after the injection of the fusion protein

GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l . Blood samples were taken at various time points after injections such as 0-min, 10-min, 20-min,30-min, 60-min and 120-min. The glucose levels within the blood samples were measured using a Accu-Chek Performa blood glucose meter (Roche) immediately. In Fig. 9a, the rats were injected with GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l at the dose of l Onmol/kg and 20nmol/kg about 8 hours before the IPGTT experiments were conducted. The data showed in Fig. 9a indicated that the fusion protein GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l at the dose of l Onmol/kg and 20nmol/kg can efficiently reduce the glucose level. In Fig. 9b, about 32 hours and 100 hours after the rats were injected with GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l at the dose of 20nmol/kg , the IPGTT experiments were conducted. The data in Fig. 9b clearly showed that a single dose of GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l can maintain its glucose-reducing activity in vivo for an extended period of time. Even at 100 hours after the injection of the fusion protein GLP l (A8G/G22E/R36G)-Fn8-p246-COL18NC l into the animals, the fusion protein can still reduce the glucose level significantly. The IPGTT experiments in rats strongly suggested that the fusion protein GLPl (A8G/G22E/R36G)-Fn8-p246-COL18NC l can be utilized as a long acting GLP- 1 analogue to treat diabetes.

Example 17: Construction of the fusion protein of Interferon, pCloud polypeptide and the scaffold protein COL 18NC 1

In this example, we demonstrate that Interferon can be fused with the pCloud polypeptide and the scaffold protein to increase the in vivo half life of Interferon. The gene encoding human Interferon alpha-2b was synthesized, digested by Ndel and BamHI and ligated into the digested vectors pET29b-GLP- l (A8G/G22E/R36G)-Fn8-p246-COL18NC l and pET29b-GLP- 1 ( A8G/G22E/R36G)-Fn8 -p271 -COL 18NC 1 generated before. The resultant fusion protein was named as IFN-p246-COL18NC l and IFN-p271 -COL 18NC l , respectively. The protein sequences of these fusion proteins were listed as SEQ ID NO: 24 and 25. The Interferon containing fusion proteins were expressed by use of the Pichia expression system (Life Technologies). The genes encoding IFN-p246-COL18NCl and IFN-p271 -COL 18NC l were amplified by PCR and cloned into the expression vector pPICZalphaA(Life Technologies) by use of Xhol and Notl. The protein expression was carried out by following the protocols from Life Technologies. The secreted recombinant proteins were purified using the similar protocol described in example 3. Alternatively, the fusion protein IFN-p246-COL18NC l and IFN-p271 -COL 18NC l can be expressed by use of the E.coli expression system as described in example 3.

To measure the biological activity of the fusion protein IFN-p246-COL18NC l and IFN-p271 -COL 18NC l to activate the JAK/STAT pathway, the Cignal ISRE Luciferase Reporter Assay Kit (Qiagen) was utilized. Hela cells were transfected with vector that contains the interferon stimulated response element (ISRE) reporter. After 16 hours of transfection, medium was changed to assay medium (Opti-MEM + 10% heat inactivated FBS + 0.1 mM NEAA + ImM Sodium pyruvate + 100 U/ml penicillin + 100 μg/ml streptomycin). After 24 hours of transfection, cells were treated with IFN-p246-COL18NC l and IFN-p271 -COL 18NC l fusion proteins at various concentrations for 18 hours. Dual Luciferase assay was performed, and promoter activity values are expressed as arbitrary units using a Renilla reporter for internal normalization. The activity of commercially available Interferon alpha-2b was measured as 2.20x10 5 IU/ug and the activities of IFN-p246-COL18NC l and IFN-p271 -COL 18NCl were measured as 1.85xl0 5 IU/ug and 1.72x10 s IU/ug. The data clearly showed that fusing with pCloud sequence and the scaffold protein did not diminish the biological activity of Interferon alpha 2b.

SEQ ID NO: 24, protein sequence of IFN-p246-COL18NCl , The pCloud sequence p246 is underlined.

CDLPQTHSLGSRRTLMLLAQMRRISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHEMI QQIFNLFSTKDSSAAWDETLLDKFYTELYQQLNDLEACVIQGVGVTETPLMNEDSILAVR KYFQRITLYLKEKKYSPCAWEVVRAEIMRSFSLSTNLQESLRSKE GGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIF VAEQEELYVRVQNGFRKVQLEARTPLPRG SEQ ID NO: 25, protein sequence of IFN-p271-COL18NCl , The pCloud sequence p271 is underlined.

CDLPQTHSLGSRRTLMLLAQMRRISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHEMI QQIFNLFSTKDSSAAWDETLLDKFYTELYQQLNDLEACVIQGVGVTETPLMNEDSILAVR KYFQRITLYLKEKKYSPCAWEVVRAEIMRSFSLSTNLQESLRSKE GGGSGGGSGGSGSESGSGGGSTSSGTGSETESPGGSG

SESC-SGGPSSAGSGGSGSESGSGGSGSSGPGSTGGGSGSESGSGGPGSSGTGGTAT GGSGSESGSGG

PGSSGPGSTGSGGSGSESGSGGSGSSGTG5TGGGSGSESGSGGPGSPRPGSTGTGGSGSE SGSGGP

GSSERGSAGGGSGSESGSGGTSESSASGSTGGGSGSESGSGGSESGSGGSGSESGSGGPE SPGSGGG SGSESGSGGTSGSTGGSGSESGSGGGGSGGGSGG

GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPR G

Example 18 : Construction of the fusion protein of TNFR2, COL 18NC1 and pCloud polypeptide

TNF Receptor (TNFR2, or p75) has been fused to IgGl Fc fragment to constitute a fusion protein Etanercept (Enbrel). Etanercept has been successfully utilized to treat severe active rheumatoid arthritis by blocking the TNF alpha functions[5 1 ] . In this example, we applied the method of the present invention on TNFR2 to generate the fusion protein of TNFR2, COL18NC1 and the pCloud polypeptide. In this example, the TNFR2 has been placed at the N-terminus of the scaffold protein COL18NC 1 while the pCloud polypeptide has been positioned at the C-terminus of the scaffold protein COL18NC 1 . The rationale for this design is to make sure that three TNFR2 can properly interact with and block the function of the TNF alpha trimer simultaneously. The resultant TNFR2-COL 18NC 1 -pCloud fusion protein is tri-valent and can block all three binding sites of TNFalpha while retaining a long half life in vivo. On the other hand, one Etanercept molecule can only block two out of three possible binding sites located on TNF alpha homo-trimer.

To construct the fusion protein, the optimized gene encoding TNFR2 was synthesized and cloned into the pPICZalphaA(Life Technologies) by Xhol and BamHI. The resultant vector is named as pPICZalphaA-TNFR2. The optimized gene encoding COL18NC 1 and p246 was synthesized and cloned into the vector pPICZalphaA-TNFR2 by use of BamHI and Notl. The protein sequence of the resultant fusion protein TNFR2-COL 18NC l -p246 is listed as SEQ ID NO: 26. The optimized gene encoding COL 18NC 1 and p271 was synthesized and cloned into the vector pPICZalphaA-TNFR2 by use of BamHI and Notl. The protein sequence of the resultant fusion protein TNFR2-COL 18NCl -p271 is listed as SEQ ID NO: 27.

SEQ ID NO: 26, TNFR2-COL18NC l -p246 protein sequence. The COL18NC1 protein sequence is in italic and bold. The pCloud sequence p246 is underlined.

LPAQVAFTPYAPEPGSTCRLREYYDQTAQMCCSKCSPGQHAKVFCTKTSDTVCDSCEDST YTQLWNWVPECLSCGSRCSSDQVETQACTREQNRICTCRPGWYCALSKQEGCRLCAPLRK CRPGFGVARPGTETSDVVCKPCAPGTFSNTTSSTDICRPHQICNVVAIPGNASMDAVCTS TSPTRSMAPGAVHLPQPVSTRSQHTQPTPEPSTAPSTSFLLPMGPSPPAEGSTGD GGGSGGGSGGG

GASSGVRLWA RQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPRG

GGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG

SGSESGSGTSGSTGSGSESGSGGGSGGGSGG

SEQ ID NO: 27, TNFR2-COL18NCl -p271 protein sequence. The COL18NC1 protein sequence is in italic and bold. The pCloud sequence p271 is underlined.

LPAQVAFTPYAPEPGSTCRLREYYDQTAQMCCSKCSPGQHAKVFCTKTSDTVCDSCE DST YTQLWNWVPECLSCGSRCSSDQVETQACTREQNRICTCRPGWYCALSKQEGCRLCAPLRK CRPGFGVARPGTETSDVVCKPCAPGTFS TTSSTDICRPHQICNVVAIPGNASMDAVCTS TSPTRSMAPGAVHLPQPVSTRSQHTQPTPEPSTAPSTSFLLPMGPSPPAEGS GD GGGSGGGSGGG

GASSGVRLWATRQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFKKVQLEARTPLPR G

GGGSGGGSGGSGSESGSGGGSTSSGTGSETESPGGSG

SESGSGGPSSAGSGGSGSESGSGGSGSSGPGSTGGGSGSESGSGGPGSSGTGGTATG GSGSESGSGG PGSSGPGSTGSGGSGSESGSGGSGSSGTGSTGGGSGSESGSGGPGSPRPGSTGTGGSGSE SGSGGP GSSERGSAGGGSGSESGSGGTSESSASGSTGGGSGSESGSGGSESGSGGSGSESGSGGPE SPGSGGG SGSESGSGGTSGSTGGSGSESGSGGGGSGGGSGG

The protein expressions of TNFR2-COL18NC l -p246 and TNFR2-COL 18NC l -p271 were carried out by use of the pichia system as described before. The secreted recombinant proteins were purified using the similar protocol described in example 3. The purified protein was kept in 20mM Hepes buffer (pH 7.5), NaCl 150mM. The purity of the fusion protein was examined by SDS-PAGE electrophoresis (purity >95%). The biological activity of TNFR2-COL18NC l -p246 and TNFR2-COL 18NC l -p271 was measured by its ability to block the TNF-alpha signaling. The positive control TNFR2-Fc fusion protein (R&D systems) can efficiently block the apoptosis of L-929 mouse fibroblast cells induced by TNF-alpha (0.25 ng/mL) in the presence of actinomycin D. The ED50 of TNFR2-Fc fusion protein was shown to be ~5ng/ml using the abovementioned assay. Our data indicated that TNFR2-COL 18NC l -p246 and TNFR2-COL 18NC l -p271 can inhibit the cell killing activity of TNF alpha for L929 cells with the ED50 of l -5ng/ml using the same assay. This suggested that TNFR2-COL18NC l -p246 and TNFR2-COL18NC l -p271 can function as efficiently as TNFR2-IgGl Fc fusion protein in the in vitro studies.

Example 19: Construction of the fusion protein containing VEGFRl R2, COL18NC 1 and pCloud sequences

Vascular endothelial growth factor (VEGF) plays a critical role during normal embryonic angiogenesis and also in the pathological angiogenesis such as cancer. Numerous studies suggested that inhibiting VEGF functions may be an efficient treatment for cancer patients. VEGF-Trap was created by fusing the second Ig domain of VEGF receptor 1 (VEGFRl ) with the third Ig domain of VEGF receptor 2 (VEGFR2) and the Fc fragment of IgG [52] . VEGF-trap has high affinity to VEGF and has shown promising anti-cancer efficacy in clinical trials. In this example, we constructed a novel fusion protein VEGFRl R2 that consists of human VEGFRl second Ig domain and human VEGFR2 third Ig domain. Then we applied the method of the present invention to VEGFRl R2 to generate the tri-valent VEGFRl R2-COL 18NC l -p246 and VEGFRl R2-COL 18NC l -p271 fusion proteins.

The synthetic gene that encodes the human VEGFRl R2 was grafted into the digested vectors pPICZalphaA-TNFR2-COL 18NC l -p246 and pPICZalphaA-TNFR2-COL18NC l -p271 generated in example 14 by use of Xhol and BamHI. The resultant fusion proteins were named as VEGFRl R2-COL18NC l -p246 and VEGFRlR2-COL18NC l -p271 and their protein sequences were listed as SEQ ID NO: 28 and 29.

The protein expressions of VEGFRlR2-COL18NCl-p246 and VEGFRlR2-COL18NCl-p271 were carried out by use of the pichia system as described in example 13. The secreted recombinant proteins were purified using the similar protocol described in example 3. The purified protein was kept in 20mM Hepes buffer (pH 7.5), NaCl 150mM. The purity of the fusion protein was examined by SDS-PAGE electrophoresis (purity >95%). The biological activities of VEGFRlR2-COL18NCl-p246 and VEGFRlR2-COL18NCl-p271 were shown by its ability to interact with VEGF. Our data from SPR by use of Biacore 2000 (GE healthcare) indicated that VEGFRlR2-COL18NCl-p246 and VEGFRlR2-COL18NCl-p271 fusion proteins can bind VEGF with the similar affinity as the VEGFRlR2-IgG Fc fusion protein.

SEQ ID NO: 28, VEGFRlR2-COL18NCl-p246 fusion protein sequence. The COL18NC1 protein sequence is in italic and bold. The pCloud sequence p271 is underlined.

SDTGRPFVEMYSEIPEI IHMTEGRELVIPCRVTSP ITVTLKKFPLDTLI PDGKRIIWDS

RKGFI ISNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTIIDWLSPSHGIELSVGEKL

VLNCTARTELNVGIDFNWEYPSSKHQH KLVNRDL TQSGSEM KFLSTLTIDGVTRSDQ

GLYTCAASSGLMTKKNSTFVRVHE

GGGSGGGSGG

GASSGVRLWAmQAMLGQVHEVPEGWLIFVAEQEELYVRVQNGFRKVQLEARTPLPRG

GGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG

SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSES GSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG SGSESGSGTSGSTGSGSESGSGGGSGGGSGG SEQ ID NO: 29, VEGFRlR2-COL18NCl -p271 fusion protein sequence. The COL18NC1 protein sequence is in italic and bold. The pCloud sequence p271 is underlined.

SDTGRPFVEMYSEIPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRII DS

RKGFI ISNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTIIDVVLSPSHGIELSVGEKL

VLNCTARTELNVGIDFNWEYPSSKHQHKKLVNRDLKTQSGSE KKFLSTLTIDGVTRSDQ

GLYTCAASSGL TKKNSTFVRVHE

GGGSGGGSGG

GASSGVRLWA RQAML GQVHEVPEGWL IFVAEQEEL YVRVQNGFRKVQLEAR TPLPRG

GGGSGGGSGGSGSESGSGGGSTSSGTGSETESPGGSG

SESGSGGPSSAGSGGSGSESGSGGSGSSGPGSTGGGSGSESGSGGPGSSGTGGTATG GSGSESGSGG PGSSGPGSTGSGGSGSESGSGGSGSSGTGSTGGGSGSESGSGGPGSPRPGSTGTGGSGSE SGSGGP GSSERGSAGGGSGSESGSGGTSESSASGSTGGGSGSESGSGGSESGSGGSGSESGSGGPE SPGSGGG SGSESGSGGTSGSTGGSGSESGSGGGGSGGGSGG

Example 20: Cloning and expression of Exenatide (EX) fused with Fn8, pCloud sequence and human collagen XVIII NC I (COL 18NC 1 )

Exenatide (INN, marketed as Byetta, Bydureon) is a glucagon-like peptide- 1 agonist (GLP-1 agonist) medication, belonging to the group of incretin mimetics, approved in April 2005 for the treatment of type II diabetes mellitus. To efficiently extend the in vivo half life of Exenatide, in this example, we have constructed the fusion proteins of Exenatide, Fn8, pCloud polypeptide and the scaffold protein COL18NC1 by use of the method of the present invention. The optimized gene encoding Exenatide and Fn8 was synthesized, digested by Ndel and BamHI and ligated into the digested vectors pET29b-GLP- 1 (A8G/G22E/R36G)-Fn8-p246-COL 18NC 1 and pET29b-GLP- 1 (A8G/G22E/R36G)-Fn8-p271 -COL 18NC 1 generated in examples 4 and 9. The resulted fusion proteins were named as EX-Fn8-p246-COL18NC l and EX-Fn8-p271 -COL18NCl and their protein sequences were listed as SEQ ID NO: 30 and 31.

The protein expressions and purifications of EX-Fn8-p246-COL 18NC l and EX-Fn8-p271 -COL 18NC l were carried out as described in example 3. The biological activities of the fusion proteins could be measured by use of the cAMP assay described in example 1 1.

The data fitting showed that the EC50 of EX-Fn8-p246-COL18NCl and EX-Fn8-p271 -COL18NC l were -0.1 I nM and 0.08nM, respectively.

SEQ ID NO: 30, EX-Fn8-p246-COL18NCl protein sequence, the Exenatide sequence is in Italic. The flexible loop between EX and Fn8 is in bold. The pCloud sequence p246 is underlined.

HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS SGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVTWAPPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLL PGT EYVVSVSSVYEQHESTPLRGRQKTGGGGSGGGSGSGSESGSGGGSTSSGTGSETESPGSG SESGSGPSSAGSGSGSESGSGSGSSGPGSTGGSGSESGSGPGSSGTGGTATGSGSESGSG PGSSGPGSTGSGSGSESGSGSGSSGTGSTGGSGSESGSGPGSPRPGSTGTGSGSESGSGP GSSERGSAGGSGSESGSGTSESSASGSTGGSGSESGSGSESGSGSGSESGSGPESPGSGG SGSESGSGTSGSTGSGSESGSGGGSGGGSGGGASSGVRLWATRQAMLGQVHEVPEGWLIF VAEQEELYVRVQNGFRKVQLEARTPLPRG

SEQ ID NO: 31 , EX-Fn8-p271-COL18NCl protein sequence, the Exenatide sequence is in Italic. The flexible unstructured linker between EX and Fn8 is in bold. The pCloud sequence p271 is underlined.

HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS SGGGSGGGSGGGSGGGSGSGGAVPPPTD

LRFTNIGPDTMRVT APPPSIDLTNFLVRYSPVKNEEDVAELSISPSDNAVVLTNLLPGT

EYVVSVSSVYEQHESTPLRGRQKTG

GGGSGGGSGGSGSESGSGGGSTSSGTGSETESPGGSG

SESGSGGPSSAGSGGSGSESGSGGSGSSGPGSTGGGSGSESGSGGPGSSGTGGTATG GSGSESGSGG PGSSGPGSTGSGGSGSESGSGGSGSSGTGSTGGGSGSESGSGGPGSPRPGSTGTGGSGSE SGSGGP GSSERGSAGGGSGSESGSGGTSESSASGSTGGGSGSESGSGGSESGSGGSGSESGSGGPE SPGSGGG SGSESGSGGTSGSTGGSGSESGSGGGGSGGGSGG

GASSGVRLWATRQAMLGQVHEVPEGWLI FVAEQEELYVRVQNGFRKVQLEARTPLPRG

Cited patents:

I . United States Patent 7557183 Polyethylene glycol linked GLP-1 compounds

2. United States Patent 8093356 Pegylated human interferon polypeptides

3. United States Patent 8058398 Modified G-CSF polypeptide

4. United States Patent 7052686 Pegylated interleukin-10

5. United States Patent 8030269 Calcitonin drug-oligomer conjugates, and uses thereof

6. United States Patent Application 20090325865 Liquid Formulations of

Pegylated Growth Hormone

7. United States Patent 8053561 Pegylated factor VIII

8. United States Patent 8053410 Pegylated factor VII glycoforms

9. United States Patent Application 20090312236 PEGYLATED INSULIN LISPRO COMPOUNDS

10. United States Patent 7271 149 GLP-1 fusion proteins

I I . United States Patent 6946134 Albumin fusion proteins

12. United States Patent 7785599 Albumin fusion proteins

13. WO201 1 140086 A2 Serum albumin binding molecules

14. EP2065402 Al Trimeric collagen scaffold antibodies

15. US7691815 B2 Methods for blocking TNF-alpha activity in mammals with trimeric soluble TNF receptors

Cited literatures:

1. Molineux, G., Pegylation: engineering improved biopharmaceuticals for oncology. Pharmacotherapy, 2003. 23(8 Pt 2): p.3S-8S.

2. Crawford, J., Clinical uses of pegylated pharmaceuticals in oncology. Cancer Treat Rev, 2002.28 Suppl A: p.7-11.

3. Pasut, G. and F.M. Veronese, PEG conjugates in clinical development or use as anticancer agents: an overview. Adv Drug Deliv Rev, 2009.61(13): p. 1177-88.

4. Veronese, F.M. and A. Mero, The impact of PEGylation on biological therapies. BioDrugs, 2008.22(5): p.315-29.

5. Eliason, J.F., Pegylated cytokines: potential application in immunotherapy of cancer. BioDrugs, 2001.15(11): p.705-11. 6. Glaesner, W., et al., Engineering and characterization of the long-acting glucagon-like peptide-1 analogue LY2189265, an Fc fusion protein. Diabetes Metab Res Rev.26(4): p.287-96.

7. Picha, K.M., et al., Protein engineering strategies for sustained glucagon-like peptide-1 receptor-dependent control of glucose homeostasis. Diabetes, 2008.57(7): p. 1926-34.

8. Bush, M.A., et al., Safety, tolerability, pharmacodynamics and pharmacokinetics of albiglutide, a long-acting glucagon-like peptide-1 mimetic, in healthy subjects. Diabetes Obes Metab, 2009.11(5): p.498-505.

9. Matthews, J.E., et al., Pharmacodynamics, pharmacokinetics, safety, and tolerability of albiglutide, a long-acting glucagon-like peptide-1 mimetic, in patients with type 2 diabetes. J Clin Endocrinol Metab, 2008.93(12): p.4810-7. 10. Gamier, J., D.J. Osguthorpe, and B. Robson, Analysis of the accuracy and implications of simple methods for predicting the secondary structure of globular proteins. J Mol Biol, 1978. 120(1): p.97-120. 11. Todorovska, A., et al., Design and application of diabodies, triabodies and tetrabodies for cancer targeting. J Immunol Methods, 2001.248(1-2): p.47-66.

12. Sturniolo, T., et al., Generation of tissue-specific and promiscuous HLA ligand databases using DNA microarrays and virtual HLA class II matrices. Nat Biotechnol, 1999. 17(6): p.555-61.

13. Schellenberger, V., et al., A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat Biotechnol, 2009.27(12): p. 1186-90.

14. Sorensen, B., et al., Fibrinogen as a hemostatic agent. Semin Thromb Hemost, 2012.38(3): p.268-73.

15. Weisel, J.W. and R.I. Litvinov, Mechanisms of fibrin polymerization and clinical implications. Blood, 2013. 121(10): p. 1712-9.

16. Mosesson, M.W., Fibrinogen and fibrin structure and functions. J Thromb Haemost, 2005.3(8): p. 1894-904.

17. Gamier, J., J.F. Gibrat, and B. Robson, GOR method for predicting protein secondary structure from amino acid sequence. Methods Enzymol, 1996.266: p.540-53.

18. Pankov, R. and K.M. Yamada, Fibronectin at a glance. J Cell Sci, 2002.115(Pt 20): p.3861-3.

19. Mao, Y. and J.E. Schwarzbauer, Fibronectin fibrillogenesis, a cell-mediated matrix assembly process. Matrix Biol, 2005. 24(6): p.389-99.

20. Leahy, D.J., I. Aukhil, and H.P. Erickson, 2.0 A crystal structure of a four-domain segment of human fibronectin encompassing the RGD loop and synergy region. Cell, 1996. 84(1): p. 155-64.

21. de Pereda, J.M., G. Wiche, and R.C. Liddington, Crystal structure of a tandem pair of fibronectin type III domains from the cytoplasmic tail of integrin alpha6beta4. EMBO J, 1999.18(15): p.4087-95.

22. Ricard-Blum, S. and F. Ruggiero, The collagen superfamily: from the extracellular matrix to the cell membrane. Pathol Biol (Paris), 2005.53(7): p.430-42.

23. Khoshnoodi, J., et al., Molecular recognition in the assembly of collagens: terminal noncollagenous domains are key recognition modules in the formation of triple helical protomers. J Biol Chem, 2006.281(50): p.38117-21.

24. Gordon, M.K. and R.A. Hahn, Collagens. Cell Tissue Res.

339(1): p.247-57.

25. Shoulders, M.D. and R.T. Raines, Collagen structure and stability. Annu Rev Biochem, 2009.78: p.929-58.

26. Wirz, J. A., et al., Crystal structure of the human collagen XV trimerization domain: a potent trimerizing unit common to multiplexin collagens. Matrix Biol.30(1): p.9-15.

27. Boudko, S.P., et al., The NC2 domain of collagen IX provides chain selection and heterotrimerization. J Biol Chem.285(31): p.23721-31.

28. Boudko, S.P., J. Engel, and H.P. Bachinger, Trimerization and triple helix stabilization of the collagen XIX NC2 domain. J

Biol Chem, 2008.283(49): p.34345-51.

29. Kvansakul, M., et al., Crystal structure of the collagen alphal(VIII) NCI trimer. Matrix Biol, 2003.22(2): p.145-52.

30. Bogin, O., et al., Insight into Schmid metaphyseal chondrodysplasia from the crystal structure of the collagen X

NCI domain trimer. Structure, 2002.10(2): p. 165-73.

31. Sundaramoorthy, M., et al., Crystal structure of NCI domains.

Structural basis for type IV collagen assembly in basement membranes. J Biol Chem, 2002.277(34): p.31142-53.

32. Boudko, S.P., et al., Crystal structure of human collagen XVIII trimerization domain: A novel collagen trimerization Fold. J Mol Biol, 2009.392(3): p.787-802. 33. Ghai, R., et al., Clq and its growing family. Immunobiology, 2007.212(4-5): p.253-66.

34. Schaffler, A. and C. Buechler, CTRP family: linking immunity to metabolism. Trends Endocrinol Metab.23(4): p. 194-204. 35. Kishore, U. and K.B. Reid, Clq: structure, function, and receptors. Immunopharmacology, 2000.49(1-2): p. 159-70.

36. Farrah, T. and C.A. Smith, Emerging cytokine family. Nature, 1992.358(6381): p.26.

37. Cabal-Hierro, L. and P.S. Lazo, Signal transduction by tumor necrosis factor receptors. Cell Signal.24(6): p. 1297-305.

38. Zelensky, A.N. and J.E. Gready, The C-type lectin-like domain superfamily. FEBS J, 2005.272(24): p.6179-217.

39. Feinberg, H., et al., Trimeric structure of langerin. J Biol Chem.285(17): p. 13285-93.

40. Hakansson, K., et al., Crystal structure of the trimeric alpha-helical coiled-coil and the three lectin domains of human lung surfactant protein D. Structure, 1999. 7(3): p. 255-64.

41. Ng, K.K., et al., Orientation of bound ligands in mannose-binding proteins. Implications for multivalent ligand recognition. J Biol Chem, 2002.277(18): p. 16088-95.

42. Nielsen, B.B., et al., Crystal structure of tetranectin, a trimeric plasminogen-binding protein with an alpha-helical coiled coil. FEBS Lett, 1997.412(2): p.388-96.

43. Hoist, J. J., T. Vilsboll, and C.F. Deacon, The incretin system and its role in type 2 diabetes mellitus. Mol Cell Endocrinol, 2009.297(1-2): p. 127-36.

44. Baggio, L.L. and D.J. Drucker, Biology of incretins: GLP-1 and GIP. Gastroenterology, 2007.132(6): p.2131-57.

45. Connaris, H., P.R. Crocker, and G.L. Taylor, Enhancing the receptor affinity of the sialic acid-binding domain of Vibrio cholerae sialidase through multivalency. J Biol Chem, 2009. 284(11): p.7339-51.

46. Kim, D., et al., Heptameric Targeting Ligands against EGFR and HER2 with High Stability and Avidity. PLoS One.7(8): p. e43077.

47. O'Reilly, M.K. and J.C. Paulson, Multivalent ligands for siglecs. Methods Enzymol.478: p.343-63.

48. Burcelin, R., et al., Encapsulated, genetically engineered cells, secreting glucagon-like peptide-1 for the treatment of non-insulin-dependent diabetes mellitus. Ann N Y Acad Sci, 1999.875: p.277-85.

49. Diez, J.J. and P. Iglesias, The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur J Endocrinol, 2003.148(3): p.293-300.

50. Shapiro, L. and P.E. Scherer, The crystal structure of a complement-1 q family protein suggests an evolutionary link to tumor necrosis factor. Curr Biol, 1998.8(6): p.335-8.

51. Aaltonen, K.J., et al., Systematic review and meta-analysis of the efficacy and safety of existing TNF blocking agents in treatment of rheumatoid arthritis. PLoS One.7(1): p. e30275. 52. Stewart, M.W., Aflibercept (VEGF-TRAP): the next anti-VEGF drug. Inflamm Allergy Drug Targets. 10(6): p. 497-508.