Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSION PROTEIN COMPRISING A LIGAND BINDING DOMAIN OF VEGF AND PDGF
Document Type and Number:
WIPO Patent Application WO/2016/145189
Kind Code:
A1
Abstract:
Fusion proteins containing a PDGF binding portion, a VEGF binding portion, and an Fc antibody region are described. Also described are nucleic acids encoding the fusion proteins, compositions comprising the fusion proteins, and methods of using the fusion proteins for treating or preventing clinical conditions characterized by abnormal angiogenesis, such as vascular permeability, edema or inflammation.

Inventors:
ZEN KEVIN (US)
WU PAI-TZU (TW)
HER JENG-HORNG (TW)
CHEN HUANG-TSU (TW)
LEOU JIUN-SHYANG (TW)
HSU CHING-HSUAN (TW)
Application Number:
PCT/US2016/021762
Publication Date:
September 15, 2016
Filing Date:
March 10, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ALLGENESIS BIOTHERAPEUTICS INC
AP BIOSCIENCES INC
ZEN KEVIN (US)
International Classes:
C07K14/71; A61K38/16
Domestic Patent References:
WO2014160507A12014-10-02
WO2015109898A12015-07-30
Foreign References:
US20060234347A12006-10-19
CN102311502A2012-01-11
US20140315804A12014-10-23
Other References:
MATTHEW R KUDELKA; HANS E GROSSNIKLAUS; KENNETH JMANDELL: "Emergence of Dual VEGF and PDGF Antagonists in the Treatment of Exudative Age-Related Macular Degeneration", 2013, pages 15PP, XP002757499, Retrieved from the Internet [retrieved on 20010210]
CHAKRABARTI SANJUKTA ET AL: "Current protein-based anti-angiogenic therapeutics", MINI REVIEWS IN MEDICINAL CHEMISTRY,, vol. 14, no. 3, 1 January 2014 (2014-01-01), pages 291 - 312, XP009189956
ZHANG; MA, PROG RETIN EYE RES., vol. 26, no. 1, January 2007 (2007-01-01), pages L-37
SULLIVAN; BREKKEN, MABS., vol. 2, no. 2, March 2010 (2010-03-01), pages 165 - 75
BHISITKUK, BR J OPHTHALMOL., vol. 90, no. 12, December 2006 (2006-12-01), pages 1542 - 7
DANIEL ET AL., OPHTHALMOLOGY, vol. 121, no. 3, March 2014 (2014-03-01), pages 656 - 66
BENJAMIN ET AL., DEVELOPMENT, vol. 125, no. 9, May 1998 (1998-05-01), pages 1591 - 8
PATEL, RETINA., vol. 29, no. 6, June 2009 (2009-06-01), pages 545 - 8
DUAN ET AL., J BIOL CHEM., vol. 266, no. 1, 5 January 1991 (1991-01-05), pages 413 - 8
UENO ET AL., SCIENCE, vol. 252, no. 5007, 10 May 1991 (1991-05-10), pages 844 - 8
ILEIDARAN ET AL., FASEB J., vol. 9, no. 1, January 1995 (1995-01-01), pages 140 - 5
LOKKER ET AL., J BIOL CHEM., vol. 272, no. 52, 26 December 1997 (1997-12-26), pages 33037 - 44
LEPPANEN ET AL., BIOCHEMISTRY, vol. 39, no. 9, 7 March 2000 (2000-03-07), pages 2370 - 5
MARTIN ET AL., OPHTHALMOLOGY, vol. 119, 2012, pages 1388 - 1398
BLOCH ET AL., AM J OPHTHALMOL., vol. 156, no. 1, July 2013 (2013-07-01), pages 116 - 124
REINMUTH ET AL., FASEB J., vol. 15, no. 7, May 2001 (2001-05-01), pages 1239 - 41
ANDRAE ET AL., GENES DEV., vol. 22, no. 10, 15 May 2008 (2008-05-15), pages 1276 - 312
JO ET AL., AM J PATHOL., vol. 168, no. 6, June 2006 (2006-06-01), pages 2036 - 53
DUGEL, RETINA TODAY, March 2013 (2013-03-01), pages 65 - 71
HEIDARAN ET AL., FASEB J., vol. 9, no. 1, January 1995 (1995-01-01), pages 140 - 5
HOLASH, J. ET AL., PNAS, vol. 99, no. 17, 2002, pages 11393 - 98
HOLASH, J. ET AL., PROC NATL ACAD SCI U S A., vol. 99, no. 17, 20 August 2002 (2002-08-20), pages 11393 - 8
LU ET AL., AM J OBSTET GYNECOL., vol. 198, no. 4, 2008, pages 477.E1 - 477.E10
Attorney, Agent or Firm:
HSING, Weihong et al. (One Commerce Square2005 Market Street, Suite 220, Philadelphia PA, US)
Download PDF:
Claims:
CLAIMS

We claim:

1. A fusion protein comprising:

a. a first peptide comprising an extracellular ligand binding domain of a VEGF

receptor,

b. an Fc region of an antibody, and

c. a second peptide comprising an extracellular ligand binding domain of a PDGF receptor;

wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a); and

wherein the fusion protein is capable of binding to a VEGF and a PDGF and inhibiting the activity of the VEGF and the activity of the PDGF. 2. The fusion protein of claim 1, wherein:

a. the extracellular ligand binding domain of the VEGF receptor comprises an Ig- like domain D2 of a VEGFR1 and an Ig-like domain D3 of a VEGFR2;

b. the Fc region of the antibody comprises a CH2 and a CH3 region of IgGl ; and c. the extracellular ligand binding domain of the PDGF receptor comprises Ig-like domains D 1 to D3 of a PDGFR$.

3. The fusion protein of claim 2, wherein:

a. the extracellular ligand binding domain of the VEGF receptor comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 7;

b. the Fc region of the antibody comprises an amino acid sequence having at least

90% identity to SEQ ID NO: 12; and

c. the extracellular ligand binding domain of PDGF receptor comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 2.

4. The fusion protein of claim 3, wherein: a. the extracellular ligand binding domain of VEGF receptor comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 10;

b. the Fc region of the antibody comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 12 and 15; and

c. the extracellular ligand binding domain of PDGF receptor comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 2 and 5.

5. The fusion protein of any of claims 1- 4, further comprising a first linker peptide between the Fc region and the first or second peptide at the C -terminus o f the fusion protein, optionally a second linker peptide between the second or first peptide at the N-terminus of the fusion protein and the Fc region.

6. The fusion protein of claim 5, wherein the first linker peptide comprises one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 20, 22, 24, 26, 28, 30 and 32, and the second linker peptide comprises an amino acid sequence of SEQ ID NO: 18.

7. The fusion protein of any of claims 1- 6, wherein the fusion protein further comprises a signal peptide linked to N-terminus of the fusion protein.

8. A fusion protein comprising an amino acid sequence having at least 90% sequence

identity to SEQ ID NO: 38, SEQ ID NO: 40, amino acids 20-766 of SEQ ID NO: 42, amino acids 21-769 of SEQ ID NO: 44 or amino acids 20-768 of SEQ ID NO: 50.

9. The fusion protein of claim 8, comprising an amino acid sequence selected from the

group consisting of SEQ ID NO: 38, SEQ ID NO: 40, amino acids 20-766 of SEQ ID NO 42, amino acids 21-769 of SEQ ID NO: 44 and amino acids 20-768 of SEQ ID NO: 50.

10. An isolated nucleic acid molecule encoding the fusion protein of any one of claims 1 to 9.

1 1. A host cell comprising a nucleic acid molecule encoding the fusion protein of any one of claims 1 to 9.

12. A method of producing the fusion protein of any one of claims 1 to 9, comprising:

(1) culturing a host cell comprising a nucleic acid molecule encoding the fusion protein of any one of claims 1 to 9, under a condition that the fusion protein is produced; and

(2) recovering the fusion protein produced by the host cell.

13. A pharmaceutical composition comprising the fusion protein of any one of claims 1 to 9 and a pharmaceutically acceptable carrier.

14. A pharmaceutical composition comprising a nucleic acid molecule encoding the fusion protein of any one of claims 1 to 9 and a pharmaceutically acceptable carrier.

15. A method of treating or preventing a clinical condition selected from the group consisting of neovascularization, vascular permeability, edema and inflammation, the method comprising administering to a subject in need thereof an effective amount of the fusion protein of any one of claims 1 to 9.

16. A method of treating or preventing a clinical condition selected from the group consisting of choroidal neovascularization (CNV), wet age-related macular degeneration (AMD) and geographic atrophy, the method comprising administering to a subject in need thereof an effective amount of the fusion protein of any one of claims 1 to 9.

Description:
TITLE OF THE INVENTION

[0001] FUSION PROTEIN COMPRISING A LIGAND BINDING DOMAIN OF VEGF AND PDGF

CROSS REFERENCE TO RELATED APPLICATION

[0002] This application is entitled to priority pursuant to 35 U.S.C. § 1 19(e) to U.S.

Provisional Application No. 62/131,261 , filed March 11 , 2015, and the disclosure of which is incorporated by reference herein in its entirety.

REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY

[0003] This application contains a sequence listing, which is submitted electronically via

EFS-Web as an ASCII formatted sequence listing with a file name "688947-3WO_ST25", creation date of March 3, 2016, and having a size of about 88.9 k bytes. The sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.

FIELD OF THE INVENTION

[0004] The invention relates to fusion proteins comprising a PDGF binding portion, a

VEGF binding portion, and an Fc antibody region, nucleic acids and expression vectors encoding the fusion proteins, recombinant cells thereof, and compositions comprising the fusion proteins. Methods of using the fusion proteins to inhibit PDGF and VEGF functions are also provided.

BACKGROUND OF THE INVENTION

[0005] Angiogenesis, the formation of new blood vessels from pre-existing blood vessels, is a normal and vital process involved in fetal development and tissue repair. Angiogenesis is highly regulated by both angiogenic and anti-angiogenic factors, and it involves endothelial cell migration and proliferation, vessel maturation and remodeling, and degradation of the

extracellular matrix. Although it is an important process in normal growth and development, angiogenesis also plays a key role in tumor growth, ischemia and inflammation.

[0006] During rapid uncontrolled ocular angiogenesis, vascular permeability is increased, leading to vascular fragility and leakiness that results in hemorrhage and accumulation of fluids and protein exudates, and ultimately resulting in either vascular insufficiency or vascular overgrowth. Ocular angiogenesis can occur in a spectrum of ocular disorders such as age-related macular degeneration (AMD), proliferative diabetic neuropathy (PDR), and corneal

neovascularization. Both AMD and PDR can result in impairment of the structure and function of retinal neurons, ultimately causing vision loss. If left untreated, the abnormal blood vessels can lead to fibrous scarring, causing irreversible damage to retinal function that can eventually result in blindness (Zhang and Ma, Prog Retin Eye Res. 2007 Jan;26(l): l-37). Corneal neovascularization can similarly lead to a reduction in cornea transparency and vision loss.

[0007] Vascular endothelial growth factor (VEGF) plays an important role in

angiogenesis. The human VEGF family contains 6 members: VEGF-A VEGF-B, VEGF-C,

VEGF-D, VEGF-E and placental growth factor (PIGF). In addition, multiple isoforms of VEGF - A, VEGF-B and PIGF are generated through alternative RNA splicing (Sullivan and Brekken, MAbs. 2010 Mar-Apr;2(2): 165-75). VEGF-A is the prototypic member of the family and is the most well characterized. VEGF-A has been shown to serve as a mitogenic factor to endothelial cells, promote endothelial cell survival and proliferation, induce cell migration and increase microvascular permeability. The VEGF family of proteins activate the VEGF signaling pathway by binding to the extracellular region of cell surface VEGF receptors (VEGFRs) to activate the VEGF signaling pathway.

[0008] There are three types of VEGFR proteins: VEGFR1 , VEGFR2, and VEGFR3, and each contains an extracellular region comprising seven immunoglobulin (Ig)— like domains. The extracellular regions of VEGFRs bind to different VEGF proteins. For example, VEGFR- 1 (Flt- 1) binds to VEGF-A, VEGF-B, and PIGF, and can function as a decoy receptor for VEGFs or as a regulator of VEGFR-2. VEGFR-2 (KDR/Flk-1) binds all VEGF isoforms and is the predominant mediator of VEGF-induced angiogenesis signaling. VEGFR-3 (Flt-4) binds VEGF- C and VEGF-D, but not VEGF-A, and functions as a mediator of lymphangiogenesis.

[0009] The high molecular weight variants VEGF 20 6 and VEGF 1 8 9K are tightly bound to the extracellular membrane and do not interact with the VEGF receptors. While VEGF 165 is the predominant soluble variant, VEGF 121 and VEGF145 are also soluble valiants that bind to VEGFR1 and VEGFR2 receptors, as does the degradation product VEGF 110 (Bhisitkuk, Br J Ophthalmol. 2006 Dec;90(12): 1542-7). [0010] Blocking VEGF activity with antibodies, soluble VEGF receptors, or inhibitors of

VEGF tyrosine kinase activity are strategies that have been used to treat angiogenic-type disorders, such as AMD. Although anti-VEGF therapy generally stabilizes or improves visual function, it has been reported that sub-retinal scarring, or fibrosis, develops in approximately half of all treated eyes within two years of anti-VEGF treatment (Daniel et al., Ophthalmology. 2014 Mar;121 (3):656-66). In addition, targeting only VEGF prevents the formation of new blood vesicles, but it has no effect on newly-established blood vessels.

[0011] Recent data suggests that pericytes may play a role in anti-VEGF resistance, stabilization of new vessels, and scarring. Pericytes interact with endothelial cells and contribute to the establishment of the blood-retinal barrier. Importantly, pericytes provide survival signals to neovascular endothelial cells, making them resistant to VEGF depletion therapy (Benjamin et al, Development. 1998 May; 125(9): 1591-8; Patel, Retina. 2009 Jun;29(6 Suppl):S45-8).

Platelet-derived growth factor (PDGF) controls pericytes, driving their recruitment, proliferation and survival, and regulating the maturation of new vessels.

[0012] The human PDGF family contains four members: PDGF-A, PDGF-B, PDGF-C and PDGF-D. The four PDGF proteins form either homo- or heterodimers (for example, PDGF- AA, PDGF-AB, PDGF-BB, PDGF-CC and PDGF-DD), and they are inactive in their monomeric forms. The dimeric proteins bind to the extracellular region of cell surface PDGF receptors (PDGFRs) to activate the PDGF signaling pathway.

[0013] There are two types of PDGF receptors, PDGFR-a and PDGFR-β that form homo- or heterodimers (for example, PDGFR-αα, PDGFR-ββ and PDGFR-αβ) and contain extracellular regions comprising five Ig-like domains. The ligand-binding sites of the receptors are located to the first three Ig-like domains (Dl to D3).

[0014] The extracellular regions of the PDGFR dimers bind to different PDGF proteins. For example, PDGFR-αα specifically interacts with PDGF-AA, PDGF-AB, PDGF-BB and

PDGF-CC. PDGFR-αβ specifically interacts with PDGF-AB, PDGF-BB, PDGF-CC and PDGF- DD. PDGFR-ββ specifically interacts with PDGF-BB, and PDGF-DD. PDGF-BB, the only PDGF that can bind to all three receptor dimer forms with high affinity, has been shown to be able to induce pericytes proliferation and migration both in vitro and in vivo. An extracellular region consisting of all five Ig-like domains of PDGFR-β (Dl to D5) was previously shown to antagonize responses stimulated by PDGF-B (Duan et al., J Biol Chem. 1991 Jan 5;266(l ):413-8; Ueno et al., Science. 1991 May 10;252(5007):844-8). Studies using PDGFRJ3-Fc chimeric proteins demonstrated that Dl to D3 of human PDGFR-β are sufficient for high-affinity PDGF-B ligand binding (Heidaran et al., FASEB J. 1995 Jan;9(l): 140-5; Lokker et al., J Biol Chem. 1997 Dec 26;272(52):33037-44). Additionally, pre-dimerization of Dl to D3 of PDGFR-β fused to glutathione S-transferase (GST) improved binding affinity to PDGF-BB ligand compared to recombinant PDGFR-β D1-D3 protein (Leppanen et al., Biochemistry. 2000 Mar 7;39(9):2370- 5).

[0015] While the current anti-VEGF therapies are highly effective, intensive patient monitoring and frequent treatment are required to achieve optimal results. In addition, because these agents target symptoms of the disease and not the underlying cause, treatment must continue indefinitely. With suboptimal treatment, existing choroidal neovascular lesions (CNVs) will continue to grow and eventually mature into fibrotic scars leading to irreversible vision loss (Martin et al., Ophthalmology 2012; 1 19: 1388-1398; Bloch et al, Am J Ophthalmol. 2013 Jul; 156( l): l 16-124; Daniel et al., Ophthalmology. 2014 Mar;121(3):656-66). Agents that are able to block neovascularization and cause involution of the immature vasculature within the neovascular choroidal lesions have the potential to eliminate the source of the vascular leak and fibrosis, reducing or eliminating the need for intensive patient monitoring and continuous treatment.

[0016] Recently, a fusion protein comprising, from N-terminus to C-terminus, an extracellular portion of a PDGF receptor, an extracellular portion of a VEGF receptor, and a multimerization domain has been described (U.S. Patent Application Publication No.

2014/0315804). The fusion protein binds both PDGF and VEGF and inhibits their activities.

[0017] Despite the progress described in the art of dual inhibitors of PDGF and VEGF, there is a need in the art for improved formulations and treatments of angiogenic-type disorders.

BRIEF SUMMARY OF THE INVENTION

[0018] The invention satisfies this need by providing novel fusion proteins that simultaneously bind to both VEGF and PDGF, targeting both signaling pathways at the same time. The fusion proteins have been generated by fusing extracellular ligand binding domains derived from VEGF and PDGF receptors to a half-life prolonging Fc domain from IgGl . In specific embodiments of the invention, all of the components of the fusion proteins are of human origin and are therefore expected to be useful as non-immunogenic therapeutics in humans, i ' he fusion proteins are able to inhibit both VEGF- and PDGF-dependent cell growth in vitro, and they are able to reduce VEGF-induced retinal leakiness in an animal model.

[0019] There is increasing evidence that angiogenesis can occur in the absence of VEGF signaling, and that pericytes supply VEGF and other cell survival factors to the proliferating endothelial cells, conferring anti-VEGF resistance (Reinmuth et al., FASEB J. 2001

May; 15(7): 1239-41). A pericyte origin has also been suggested for myofibroblasts in scarring tissue and tumors. The PDGF signaling pathway is responsible for pericyte recruitment, survival and maturation (Andrae et al.. Genes Dev. 2008 May 15;22(10): 1276-312). Inhibition of PDGF receptor signaling by antibodies was shown to enhance the therapeutic effect of anti-VEGF treatment in multiple mouse models of ocular neovascularization (Jo et al., Am J Pathol. 2006 Jun;168(6):2036-53). A large phase 2 clinical trial of the anti-PDGF agent E10030 in combination with an anti-VEGF agent showed superior results over anti-VEGF monotherapy (Dugel, Retina Today, March 2013, 65-71). Thus, the fusion proteins of the invention are efficacious in treating angiogenic-type disorders, such as AMD and cancer. An additional benefit of the fusion proteins is that there is no need to inject two separate compositions, i.e. of an anti-PDGF agent and an anti-VEGF agent. Instead, a single composition comprising a fusion protein of both agents allows for a single injection, thereby decreasing the risk to patients for infections and injection trauma.

[0020] In one general aspect, the invention relates to a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C- terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a); and wherein the fusion protein is capable of binding to a VEGF -A molecule and a PDGF-BB molecule and inhibiting the activity of the VEGFR1, VEGFR2 and the activity of the PDGFR.

[0021] In an embodiment of the invention, the extracellular ligand binding domain of a

VEGF receptor is capable of binding to a VEGF ligand, and comprises one or more of Ig-like domains Dl -D7 of one or more VEGF receptors. Preferably, the extracellular ligand binding domain of the VEGF receptor comprises an Ig-like domain D2 of a first VEGF receptor and an Ig-like domain D3 of a second VEGF receptor, wherein the first and second VEGF receptors are the same or different VEGF receptors. In one embodiment, the extracellular ligand binding domain of the VEGF receptor comprises an Ig-like domain D2 of a VEGFR1 and an Ig-like domain D3 of a VEGFR2. In another embodiment, the extracellular ligand binding domain of the VEGF receptor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 7. More preferably, the extracellular ligand binding domain of the VEGF receptor comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 10.

[0022] In an embodiment of the invention, the extracellular ligand binding domain of a

PDGF receptor is capable of binding to a PDGF ligand and comprises one or more of Ig-like domains D1-D5 of one or more PDGF receptors. Preferably, the extracellular ligand binding domain of the PDGF receptor comprises Ig-like domains D1-D3 of one or more PDGF receptors. In one embodiment, the extracellular ligand binding domain of the PDGF receptor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 2. More preferably, the extracellular ligand binding domain of the PDGF receptor present comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 2 and 5.

[0023] In an embodiment of the invention, the Fc region of the antibody comprises a

CH2 and a CH3 region of IgGl . Preferably, the Fc region of the antibody comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 12. More preferably, the Fc region of the antibody comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 12 and 15.

[0024] In a preferred embodiment of the invention, the fusion protein comprises (a) an

Ig-like domain D2 of a VEGFR1 and an Ig-like domain D3 of a VEGFR2, (b) an Fc region of the antibody comprises a CI 12 and a CH3 region of IgGl, and (c) an Ig-like domains Dl to D3 of a PDGFR , wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a), more preferably in an order of

[0027] In another general aspect, the invention relates to an isolated nucleic acid molecule encoding a fusion protein of the invention.

[0028] In another general aspect, the invention relates to an expression vector comprising a nucleic acid molecule encoding a fusion protein of the invention.

[0029] In another general aspect, the invention relates to a recombinant host cell comprising a nucleic acid molecule encoding a fusion protein of the invention.

[0030] In another general aspect, the invention relates to a method of obtaining a fusion protein of the invention. The method comprises: (1) culturing a host cell comprising a nucleic acid molecule encoding the fusion protein under a condition that the fusion protein is produced; and (2) recovering the fusion protein produced by the host cell.

[0031] In another general aspect, the invention relates to a pharmaceutical composition comprising a fusion protein of the invention and a pharmaceutically acceptable carrier.

[0032] In another general aspect, the invention relates to a pharmaceutical composition comprising a nucleic acid molecule encoding a fusion protein of the invention and a

pharmaceutically acceptable carrier.

[0033] In another general aspect, the invention relates to a method of reducing the activity of the VEGFR and the activity of the PDGFR, the method comprising administering to a subject in need thereof an effective amount of a fusion protein of the invention.

[0034] In another general aspect, the invention relates to a method of treating or preventing a clinical condition selected from the group consisting of tissue vascularization, vascular permeability, edema and inflammation, the method comprising administering to a subject in need thereof an effective amount of a fusion protein according to an embodiment of the invention.

[0035] In another general aspect, the invention relates to a method of treating or preventing a clinical condition selected from the group consisting of choroidal

neovascularization (CNV), wet age-related macular degeneration (AMD) and geographic atrophy, the method comprising administering to a subject in need thereof an effective amount of the fusion protein of according to embodiments of the invention.

[0036] In an embodiment of the invention, the fusion protein is administered as an isolated protein or as an expression vector. [0037] In an embodiment of the invention, the clinical condition is selected from the group consisting of brain edema, stroke, cancer, psoriasis, arthritis, asthma, generalized edema associated with burns, ascites and pleural effusion associated with tumors, inflammation or trauma, chronic airway inflammation, capillary leak syndrome, sepsis, kidney disease associated with increased leakage of protein, rheumatoid arthritis, inflammatory arthritis, osteoarthritis, atherosclerosis, psoriasis, ocular inflammation and/or ocular angiogenesis, including aged- related macular degeneration, proliferative and nonproliferative diabetic retinopathy, corneal neovascularization, rubeosis iridis and neovascular glaucoma.

[0038] In another general aspect, the invention relates to a dimeric antagonist for PDGF and VEGF comprising a fusion protein of the invention.

[0039] In another general aspect, the invention relates to a protein conjugate comprising a fusion protein of the invention bound to at least one ligand selected from the group consisting of PDGF-BB and VEGF- A.

[0040] Other aspects, features and advantages of the invention will be apparent from the following disclosure, including the detailed description of the invention and its preferred embodiments and the appended claims.

BRIEF DESCRIPTION OF THE DRAWINGS

[0041] The foregoing summary, as well as the following detailed description of the invention, will be better understood when read in conjunction with the appended drawings. It should be understood that the invention is not limited to the precise embodiments shown in the drawings.

[0042] In the drawings:

[0043] Figure 1 shows the structural design of exemplary bi-functional fusion proteins according to embodiments of the invention, Fusion Protein 2 (top) and Fusion Protein 1 (bottom), designed to inhibit both PDGF and VEGF pathways simultaneously: PDGFR extracellular Ig- like domain (PID) represents the extracellular Ig-like domains Dl to D3 of PDGFRP; Fc represents IgGl CH2 and CH3 domains; VEGFR extracellular Ig-like domain (VID) represents the extracellular Ig-like domain D2 of VEGFR1 and the extracellular Ig-like domain D3 of VEGFR2; [0044] Figure 2A and Figure 2B show SDS-PAGE gel analysis of the purified bi- functional Fc Fusion Proteins 1 and 2, respectively; Figure 2C shows SDS-PAGE gel analysis of the non-reduced and reduced purified bi-functional Fc fusion proteins, respectively: Lane M = marker, Lanes 1 (non-reduced) and 2 (reduced) = Positive Control 2; Lanes 3 (non-reduced) and 4 (reduced) - Fusion Protein 5; Lanes 5 (non-reduced) and 6 (reduced) = Fusion Protein 3;

[0045] Figure 3 shows the results of a direct ligand binding assay of three test samples: purified Positive Control 1 (·), Fusion Protein 3 (A), and Fusion Protein 5 (■) against VEGF 165. Wells were pre-coated with VEGF 165 and incubated with various concentrations of test samples; the amount of bound test sample was detected using FIRP conjugated goat anti-human IgGl Fc specific antibody, and the OD4S0 readings were plotted against test sample concentrations;

[0046] Figure 4 shows the results of a direct ligand binding assay of three test samples: purified Positive Control 2 (·), Fusion Protein 3 ( A), and Fusion Protein 5 (■) against PDGF- BB. Wells were pre-coated with PDGF-BB and incubated with various concentrations of test sample; the amount of test sample was detected using FIRP conjugated goat an ti -human IgGl Fc specific antibody, and the OD450 readings were plotted against test sample concentrations;

[0047] Figure 5 A and Figure 5B show the affinity assessment of Fusion Protein 1 against

VEGF 1 6 5 and PDGF-BB in solution, respectively, in a competitive binding assay. Various concentrations of Fusion Protein 1 were incubated overnight in solution with a fixed

concentration of either VEGF 1 65 or PDGF-BB, the concentrations of free VEGFi 65 or PDGF-BB were determined using a quantitative sandwich enzyme-linked immunoassay (ELISA) assay, and plotted against Fusion Protein 1 concentrations;

[0048] Figure 6A and Figure 6B show the affinity assessment of Fusion Protein 2 against

VEGF 1 6 5 and PDGF-BB in solution, respectively, in a competitive binding assay. Various concentrations of Fusion Protein 2 were incubated overnight in solution with a fixed

concentration of either VEGF 165 or PDGF-BB; the concentrations of free VEGF 165 or PDGF-BB were determined using a quantitative sandwich ELISA assay and plotted against Fusion Protein 2 concentrations;

[0049] Figure 7 shows the inhibitory effect of Positive Control 1 (·), Fusion Protein 3

(A), and Fusion Protein 5 (■) on the VEGF-dependent growth of HUVEC cells. OD490 readings were plotted against test sample concentrations; and [0050] Figure 8 shows the inhibitory effect of Positive Control 2 (·), Fusion Protein 3

(A), and Fusion Protein 5 (■) on the PDGF-dependent growth of B ALB/3 T3 cells; OD490 readings were plotted against test sample concentrations.

DETAILED DESCRIPTION OF THE INVENTION

[0051] Various publications, articles and patents are cited or described in the background and throughout the specification; each of these references is herein incorporated by reference in its entirety. Discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is for the purpose of providing context for the invention. Such discussion is not an admission that any or all of these matters form part of the prior art with respect to any inventions disclosed or claimed.

[0052] Unless defined otherwise, all technical and scientific terms used herein have the same meaning commonly understood to one of ordinary skill in the art to which this invention pertains. Otherwise, certain terms used herein have the meanings as set in the specification. All patents, published patent applications and publications cited herein are incorporated by reference as if set forth fully herein. It must be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise.

[0053] The invention relates to a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a); and wherein the fusion protein is capable of binding to a VEGF-A and a PDGF-BB and inhibiting the activity of the VEGFR1, VEGFR2 and the activity of the PDGFR.

[0054] It is surprisingly discovered during the present invention that the orientation of the extracellular ligand binding domains of the VEGF and PDGF receptors with respect to the other components in the fusion protein, such as the antibody Fc region, has an impact on the binding affinity of the fusion protein for the VEGF and PDGF ligands. A fusion protein according to an embodiment of the invention, has optimized affinity of the fusion protein for both of the ligands and may have an increased efficacy of the fusion proteins. [0055] As used herein, the phrase "fusion protein" refers to a protein having two or more portions covalently linked together, where each of the portions is derived from different proteins.

[0056] As used herein, the term "VEGF" refers to any vascular endothelial growth factor protein that regulates the VEGF signaling pathway. Thus, the term VEGF can refer to VEGF- A VEGF-B, VEGF-C, VEGF-D, VEGF-E, P1GF, or isoforms thereof.

[0057] As used herein, the terms "VEGF receptor" and "VEGFR" refer to any receptor that binds to a VEGF ligand. Thus, the term VEGF receptor can refer to a VEGFR 1 , VEGFR2 or VEGFR3.

[0058] As used herein, the phrase "extracellular ligand binding domain" refers to any region of a receptor protein that is located on the outside of the cell and is able to bind to its ligand.

[0059] Accordingly, the extracellular ligand binding domain of the VEGF receptor that is present in the fusion protein of the invention can be from any VEGFR, including, but not limited to, VEGFR1 , VEGFR2, and VEGFR3. The seven extracellular IgG-like domains of the VEGFR proteins are numbered 1 , 2, 3, 4, 5, 6 and 7, from the N- to C-terminus of the extracellular region, and are alternatively referred to as Dl, D2, D3, D4, D5, D6 and D7. The extracellular ligand binding domain of a VEGFR that is present in the fusion protein of the invention can comprise one or more of any of the seven IgG-like domains from the extracellular region of one or more of any VEGFR protein. For example, the extracellular ligand binding domain of a VEGF receptor that is present in the fusion protein of the invention can be one or more of Dl, D2, D3, D4, D5, D6, or D7 from one or more of VEGFR 1 , VEGFR2, or VEGFR3.

[0060] In a preferred embodiment, the extracellular ligand binding domain of the VEGF receptor that is present in the fusion protein comprises an Ig-like domain D2 of a VEGFR1 and an Ig-like domain D3 of a VEGFR2. Preferably, the extracellular ligand binding domains of the VEGFR comprises one or more mutations that increases its binding to a VEGF. In a more preferred embodiment, the extracellular ligand binding domain of the VEGF receptor that is present in the fusion protein comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 7. In an even more preferred embodiment, the extracellular ligand binding domain of the VEGF receptor that is present in the fusion protein comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 10. |0061] The extracellular ligand binding domain of the VEGF receptor that is present in the fusion protein of the invention can be from any animal, such as a human or another suitable mammal, such as a mouse, rabbit, rat, pig, dog, or a primate. In a preferred embodiment, the VEGFR is from a human.

[0062] As used herein, the term "PDGF" refers to any plasma-derived growth factor protein that regulates the PDGF signaling pathway. Thus, the term PDGF can refer to PDGF- A, PDGF-B, PDGF-C or PDGF-D.

[0063] As used herein, the terms "PDGF receptor" and "PDGFR" refer to any receptor that binds to a PDGF ligand.

[0064] The extracellular ligand binding domain of the PDGF receptor that is present in the fusion protein of the invention can be from any PDGFR, including, but not limited to, PDGFR-a and PDGFR-β. The five extracellular IgG-like domains of the PDGFR proteins are numbered 1, 2, 3, 4 and 5, from the N- to C-terminus of the extracellular region and are alternatively referred to as Dl, D2, D3, D4 and D5. The extracellular ligand binding domain of a PDGFR that is present in the fusion protein of the invention can be one or more of any of the five IgG-like domains from the extracellular region of one or more of any PDGFR protein. For example, the extracellular ligand binding domain of a PDGF receptor that is present in the fusion protein of the invention can be one or more of Dl, D2, D3, D4, or D5 from one or more of PDGFR-a or PDGFR-β.

[0065] In a preferred embodiment, the extracellular ligand binding domain of the PDGF receptor that is present in the fusion protein comprises Ig-like domains Dl to D3 of a PDGFRp. Preferably, the extracellular ligand binding domains of the PDGFR comprises one or more mutations that increases its binding to a PDGF. In a more preferred embodiment, the extracellular ligand binding domain of the PDGF receptor that is present in the fusion protein comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 2. In an even more preferred embodiment, the extracellular ligand binding domain of the PDGF receptor that is present in the fusion protein comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 2 and 5.

[0066] The extracellular ligand binding domain of the PDGF receptor that is present in the fusion protein of the invention can be from any animal, such as a human or another suitable mammal, such as a mouse, rabbit, rat, pig, dog, or a primate. In a preferred embodiment, the PDGFR is from a human.

[0067] As used herein, the term "Fc region" of an antibody refers to the "fragment, crystallizable" region of an antibody that is composed of two heavy chains that comprise two or three constant domains, depending on the class of the antibody. The term is used to define a C- terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes both native and variant Fc regions. The Fc region of an antibody can serve as a multimerization domain, which is a domain that promotes the association of subunits into multimers, such as dimers, trimers, tetramers, etc. Preferably, the Fc region comprises one or more mutations that promote the association of subunits into multimers.

[0068] The Fc region of an antibody that is present in the fusion protein of the invention can be from any isotype (e.g., IgG, IgM, IgA, IgD or IgE), any subclass (e.g., IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, etc.), any allotype, or any engineered mutant, such as knob and hole Fc fragments. In a preferred embodiment, the Fc region of an antibody that is present in the fusion protein of the invention comprises a CH2 and a CH3 region of IgGl . In a more preferred embodiment, the Fc region of an antibody that is present in the fusion protein of the invention comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 12. In an even more preferred embodiment, the Fc region of an antibody that is present in the fusion protein of the invention comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 12 and l5.

[0069] The Fc region of an antibody that is present in the fusion protein of the invention can be from any animal, such as a human or another suitable mammal, such as a mouse, rabbit, rat, pig, dog, or a primate. In a preferred embodiment, the Fc region is from a human antibody.

[0070] According to embodiments of the invention, components of the fusion protein can be linked by a linking moiety such as a peptide linker. Preferably, the linker increases flexibility of the fusion protein components, helps insure correct folding, minimizes steric hindrance and does not interfere significantly with the structure of each functional component within the fusion protein. In some embodiments, the peptide linker comprises 2 to 20 amino acids. In some embodiments, the peptide linker comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15. 16, 17, 18, 19, or 20 amino acids. Preferably, the fusion protein comprises a first linker peptide between the Fc region and the first or second peptide at the C-terminus of the fusion protein, and optionally a second linker peptide between the second or first peptide at the N-terminus of the fusion protein and the Fc region. In preferred embodiments of the invention, the first linker peptide comprises one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 20, 22, 24, 26, 28, 30 and 32. and the second linker peptide comprises an amino acid sequence of SEQ ID NO: 18.

[0071] According to embodiments of the invention, the fusion protein further comprises a signal peptide linked to N-terminus of the fusion protein to ensure the secretion of the fusion protein from the cell. Any signal peptide that is recognized and processed by host cells can be used. In preferred embodiments, the signal peptide includes but is not limited to an amino acid sequence of SEQ ID NOs: 34 or 36.

[0072] In a preferred embodiment, the fusion protein of the invention comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 38 or at least 90% identity to SEQ ID NO: 40, preferably at least 90% identity to amino acids number 20 to 766 of SEQ ID NO: 42, amino acids number 21 to 769 of SEQ ID NO: 44 or amino acids number 20 to 768 of SEQ ID NO: 50. The fusion protein can include one or more mutations or single nucleotide

polymorphisms (SNPs) in the VEGF receptor domains and/or PDGF receptor domains. There can also include one or more mutations or SNPs that occur at the Fc region of an antibody in accordance with embodiments of the invention.

[0073] In a more preferred embodiment, the fusion protein of the invention comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 38, SEQ ID NO: 40, amino acids 20-766 of SEQ ID NO: 42, and amino acids 21-769 of SEQ ID NO:44 and amino acids 20-768 of SEQ ID NO: 50.

[00741 In a general aspect, the fusion protein of the invention binds to a VEGF and a

PDGF and inhibits the activity of the VEGF and the PDGF receptors. As used herein, the term "binds to" refers to the binding of an extracellular domain of a receptor protein to a ligand that results in one or more of inhibiting, blocking, neutralizing, reducing, abrogating or interfering with the ligand' s activities. In certain embodiments, the extracellular domain of a VEGF or a PDGF receptor protein inhibits ligand activities by binding to the ligand, i.e., VEGF or PDGF, respectively, and sequestering the ligand from binding to other molecules, such as other VEGF receptors and PDGF receptors. In certain other embodiments, the extracellular domain of a

VEGF or a PDGF receptor protein inhibits ligand activities by binding to the ligand, i.e., VEGF or PDGF, respectively, and preventing the ligand from triggering downstream signaling events in the cells.

[0075] As used herein, the term "inhibition" or "inhibit" in the context of ligand activity as used herein refers to a property of an extracellular domain of a receptor protein that reduces the activity of the ligand as analyzed by various functional assays, including, but not limited to, binding assays, cell growth assays, competitive binding assays, and in vivo assays.

[0076] The fusion proteins or components of the fusion protein disclosed herein can be characterized or assessed for biological activities including, but not limited to, affinity to a target binding partner (e.g., a PDGF and/or VEGF family protein), competitive binding (e.g., blocking a PDGF or VEGF from binding to a PDGFR or VEGFR), inhibitory activity (e.g., inhibiting the activation of PDGF or VEGF signaling pathways), inhibition of cell proliferation, inhibition of tumor growth, and inhibition of angiogenesis (e.g., inhibition of choroidal neovascularization). In some embodiments, the fusion proteins or fusion protein components disclosed herein can be assessed for biological activity in vivo or in vitro.

[0077] The invention also provides an isolated nucleic acid molecule encoding a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a). According to embodiments of the invention, the nucleic acid molecule encoding the fusion protein can be codon-optimized for expression in a particular type of host cell, such as Chinese hamster ovary cells. According to preferred embodiments of the invention, the nucleic acid molecule encoding the fusion protein comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 37, 39, 41, 43 and 49.

[0078] According to other embodiments of the invention, the nucleic acid molecule encoding the fusion protein can be in an expression vector. Expression vectors include, but are not limited to, vectors for recombinant protein expression and vectors for delivery of nucleic acids into a subject for expression in a tissue of the subject, such as viral vectors. Examples of viral vectors suitable for use with the invention include, but are not limited to adenoviral vectors, adeno-associated virus vectors, lentiviral vectors, etc. The vector can also be a non-viral vector. Examples of non-viral vectors include, but are not limited to plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, bacteriophages, etc. The vector may include any element to establish a conventional function of an expression vector, for example, a promoter, ribosome binding element, terminator, enhancer, selection marker, or an origin of replication.

[0079] According to other embodiments of the invention, the nucleic acid molecule encoding the fusion protein can be codon optimized for improved recombinant expression from a desired host cell using methods known in the art in view of the present disclosure.

[0080] The invention also provides a host cell comprising a nucleic acid molecule encoding a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a). Host cells include, but are not limited to, host cells for recombinant protein expression and host cells for delivery of the nucleic acid into a subject for expression in a tissue of the subject. Examples of host cells suitable for use with the invention include, but are not limited to Chinese hamster ovary (CHO) cells, Human Embryonic Kidney 293 (HEK-293), etc.

[0081] The invention also provides a method of producing a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C- terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a). In a general aspect, the method comprises (1) culturing a host cell comprising a nucleic acid molecule encoding the fusion protein under a condition that the fusion protein is produced; and (2) recovering the fusion protein produced by the host cell. The fusion protein can be purified further using methods known in the art.

[0082] In some embodiments, the fusion protein is expressed in host cells and purified therefrom using a combination of one or more standard purification techniques, including, but not limited to, Protein A affinity chromatography, Protein G affinity chromatography, buffer exchange, size exclusion chromatography, ultrafiltration, and dialysis.

[0083] The invention also provides a pharmaceutical composition comprising a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a). Compositions of the invention comprise a therapeutically effective amount of the fusion protein.

[0084] The term "therapeutically effective amount" means an amount of a therapeutically active compound needed to elicit the desired biological or clinical effect. According to embodiments of the invention, "a therapeutically effective amount" i s an amount sufficient to effect beneficial or desired results, including clinical results. A therapeutically effective amount can be administered in one or more administrations. In terms of a disease state, an effective amount is an amount sufficient to ameliorate, stabilize, or delay development of a disease.

According to specific embodiments of the invention, a therapeutically effective amount is an amount of a fusion protein needed to treat or prevent a disorder characterized by abnormal angiogenesis, such as a disease characterized by vascular permeability, edema, inflammation, retinopathies, fibrosis or cancer.

[0085] In some embodiments, the pharmaceutical composition comprising a fusion protein comprises a fusion protein formulated in a buffer at a protein concentration from about 0.5 to about 100 mg/mL, preferably about 40 to about 80 mg/mL, such as about 40, 50, 60, 70 or 80 mg/mL, most preferably about 40 ± about 5 mg/mL. In other preferred embodiments, the fusion protein is formulated in a buffer at a protein concentration of more than about 40 mg/mL, preferably about 80 ± about 10 mg/mL.

[0086] In particular embodiments, the buffer is a phosphate buffer with a pH of about 6.5 to 8, more preferably about 7 to 7.5, even more preferably about 7.2. The phosphate buffer comprises about 5 to 20 mM sodium phosphate, such as 5, 10, 15 or 20 mM sodium phosphate, more preferably about 10 mM sodium phosphate; about 20 to 60 mM sodium chloride, more preferably about 40 mM sodium chloride; about 1 to 10% weight-per- volume (w/v) sucrose, more preferably about 5% w/v sucrose; and about 0.01 to 0.05% w/v of a surfactant, more preferably about 0.03% w/v polysorbate 20.

[0087] In other particular embodiments, the buffer is a histidine buffer with a pH of about 5 to 8, more preferably about 6 to 7, most preferably about 6.8. The histidine buffer comprises about 10 to 50 mM histidine, such as 10, 20, 30, 40 or 50 mM histidine, more preferably about 25 mM histidine; about 10 to 30 mM sodium chloride, such as 10, 20 or 30 mM sodium chloride, more preferably about 20 mM sodium chloride; about 1 to 10% w/v sucrose, such as 1, 2, 4, 6, 8 or 10% w/v sucrose, more preferably about 6% w/v sucrose; and about 0.01 to 0.05% w/v of a surfactant, more preferably about 0.03% w/v polysorbate 20.

[0088] The invention also provides a composition comprising a nucleic acid molecule encoding a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor; wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a), preferably in an order of (c)-(b)-(a).

[0089] Compositions comprising a nucleic acid molecule encoding a fusion protein of the invention can comprise a delivery vehicle for introduction of the nucleic acid molecule into a cell for expression of the fusion protein. Examples of nucleic acid delivery vehicles include liposomes, biocompatible polymers, including natural polymers and synthetic polymers, lipoproteins, polypeptides, polysaccharides, lipopolysaccharides, artificial viral envelopes, metal particles, and bacteria, viruses, such as baculoviruses, adenoviruses and retroviruses,

bacteriophages, cosmids, plasmids, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic hosts.

[0090] The invention also relates to use of the pharmaceutical compositions described herein to treat or prevent a condition, disease or disorder characterized by abnormal angiogenesis, such as a disease characterized by neovascularization, vascular permeability, edema, or inflammation, retinopathies, fibrosis or cancer. According to embodiments of the invention, a method of treating a condition, disease or disorder characterized by abnormal angiogenesis in a subject comprises administering to the subject in need of the treatment a pharmaceutical compositions of the invention. Any of the pharmaceutical compositions described herein can be used in a method of the invention, including pharmaceutical compositions comprising the fusion protein or pharmaceutical compositions comprising a nucleic acid encoding the fusion protein. Preferably, a pharmaceutical composition of the invention is administered to a subject via the vitreous, conjunctiva, tenon, retrobulbar, or sclera for ophthalmology-related diseases, and into blood or tissues for systemic diseases. [0091] As used herein, "subject" means any animal, preferably a mammal, most preferably a human, who will be or has been treated by a method according to an embodiment of the invention. The term "mammal'' as used herein, encompasses any mammal. Examples of mammals include, but are not limited to, cows, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, non-human primates (NHPs) such as monkeys or apes, humans, etc., more preferably a human.

[0092] A "condition, disease or disorder characterized by abnormal angiogenesis" or an

"angiogenic-type disorder" as used herein, shall have the same meaning, and refers to any disorder related to abnormal blood vessel production, including excessive, insufficient, or abnormal angiogenesis. Examples of angiogenic-type disorders that can be treated according to a method of the invention include, but are not limited to, diseases characterized by

neovascularization, vascular permeability, edema, or inflammation. These include, but are not limited to, ocular inflammation and/or ocular angiogenesis, including aged-related macular degeneration (such as wet AMD, dry AMD or geographic atrophy), proliferative and

nonproliferative diabetic retinopathy, ocular disease characterized by neovascularization (such as corneal neovascularization or choroidal neovascularization), uveitis (such as anterior uveitis or posterior uveitis), retinitis pigmentosa, diabetic retinopathy, rubeosis iridis, neovascular glaucoma, inflammatory disease, rheumatoid arthritis, inflammatory arthritis, osteoarthritis, autoimmune disease, and cancer. In preferred embodiments of the invention, the angiogenic- type disorder to be treated is retinopathies, fibrosis or cancer.

[0093] In other embodiments, the angiogenic-type disorder that can be treated according to a method of the invention include, but are not limited to, brain edema, stroke, psoriasis, asthma, generalized edema associated with burns, ascites and pleural effusion associated with tumors, inflammation or trauma, chronic airway inflammation, capillary leak syndrome, sepsis, kidney disease associated with increased leakage of protein.

[0094] The terms "treat," "treating," and "treatment" as used herein refer to

administering a composition to a subject to achieve a desired therapeutic or clinical outcome in the subject. In one embodiment, the terms "treat," "treating," and "treatment" refer to

administering a pharmaceutical composition of the invention to reduce, alleviate or slow the progression or development of an angiogenic-type disorder, such as vascular permeability, edema or inflammation. In yet another embodiment, the terms "treat," "treating," and "treatment" refer to administering a pharmaceutical composition of the invention to inhibit or reduce corneal neovascularization and/or leaky vasculature in the eye. In yet another embodiment, the terms "treat," "treating," and "treatment" refer to administering a pharmaceutical composition of the invention to slow the progression or development of new blood vessels in the cornea (i.e., corneal neovascularization) or site of interest. In particular embodiments of the invention, when used with reference to AMD, the terms "treat," "treating," and "treatment" refer to preventing or reducing VEGF-induced retinal leakiness, and to preventing or reducing ocular scarring and fibrosis related to angiogenesis. In particular embodiments of the invention, when used with reference to cancer, the terms "treat," "treating," and "treatment" refer to reducing the proliferation of, de-differentiation of, or spread of cancerous cells. Treating a tumor according to the present invention includes a reduction in tumor size, a reduction in tumor growth, and a reduction in tumor metastasis. As used herein, the term "tumor" refers to abnormal tissue masses, and includes both benign and malignant masses.

[0095] According to embodiments of the invention, a pharmaceutical composition can be administered by any method known to those skilled in the art in view of the present disclosure, such as by topical administration, intravitreous injection, suprachoroidal or subconjunctival injection. In a preferred embodiment, the ophthalmic formulation is intravitreally administered. The pharmaceutical composition can be administered to any part of the eye, and is preferably administered to the vitreous of the eye for the treatment of angiogenic-type disorders. The pharmaceutical composition can be administered to any part of the body, and is preferably administered to the blood or tissue/organ for the treatment of angiogenic-type disorders.

[0096] Parameters such as the dosage amount, frequency of administration, and duration of administration of a pharmaceutical composition to a subject according to an embodiment of the invention are not limited in any particular way. The optimum values of such parameters can depend on a variety of factors, such as the subject to be treated, the particular angiogenic-type disease to be treated, the severity of the disease, etc., and one of ordinary skill in the art will be able to determine the optimum values for such parameters in order to achieve the desired therapeutic or clinical outcome. For example, a pharmaceutical composition can be administered once per day, or more than once per day, such as twice, three times, four times, etc. An exemplary and non-limiting dosing regimen comprises administering a pharmaceutical composition intravitreally once for a duration of one month. [0097] In other embodiments, the invention may be administered together with other anti-angiogenic agents, such as anti-hepatocyte growth factor (IIGF), anti-HGF receptor (HGFR), anti-fibroblast growth factor (FGF), anti-FGF receptor (FGFR), anti-inflammatory

(corticosteroid, non-steroidal anti-inflammatory drugs), immunomodulatory, antibiotic, and anti- cancer agents.

[0098] In a general aspect, the invention provides a dimeric antagonist for PDGF and

VEGF comprising the fusion protein of the invention. Each fusion protein in the dimer comprises any fusion protein disclosed herein. In one embodiment, the dimeric fusion protein comprises two identical fusion proteins of the invention. In another embodiment, the dimeric fusion protein comprises two different fusion proteins of the invention. In another embodiment, the dimeric fusion protein comprises at least one fusion protein comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 38, SEQ ID NO: 40, amino acids 20-766 of SEQ ID NO: 42, amino acids 21-769 of SEQ ID NO:44 and amino acids 20-768 of SEQ ID NO: 50, or an amino acid sequence having at least 90% identity to the amino acid sequence selected from the group consisting of SEQ ID NOs: 38, SEQ ID NO:40, amino acids 20-766 of SEQ ID NO: 42, amino acids 21-769 of SEQ ID NO:44 and amino acids 20-768 of SEQ ID NO: 50.

[0099] In another general aspect, the invention provides a protein conjugate comprising the fusion protein of the invention bound to at least one ligand selected from the group consisting of PDGF and VEGF.

EMBODIMENTS

[00100] Embodiment 1 is a fusion protein comprising (a) a first peptide comprising an extracellular ligand binding domain of a VEGF receptor, (b) an Fc region of an antibody, and (c) a second peptide comprising an extracellular ligand binding domain of a PDGF receptor;

wherein the fusion protein is arranged from N-terminus to C-terminus in an order selected from the group consisting of (a)-(b)-(c) and (c)-(b)-(a); and wherein the fusion protein is capable of binding to a VEGF and a PDGF and inhibiting the activity of the VEGF and the activity of the PDGF.

[00101] Embodiment 2 is a fusion protein according to embodiment 1 , wherein the extracellular ligand binding domain of a VEGF receptor is capable of binding to a VEGF ligand and comprises one or more selected from the group consisting of Ig-like domains D1 -D7 of a VEGF receptor.

|00102] Embodiment 3 is a fusion protein according to embodiment 1, wherein the extracellular ligand binding domain of a PDGF receptor is capable of binding to a PDGF ligand and comprises one or more selected from the group consisting of Ig-like domains D1-D5 of a PDGF receptor.

[00103] Embodiment 4 is a fusion protein according to embodiment 1, wherein (a) the extracellular ligand binding domain of the VEGF receptor comprises an Ig-like domain D2 of a first VEGFR, preferably VEGFR1. and an Ig-like domain D3 of a second VEGFR, preferably VEGFR2; (b) the Fc region of the antibody comprises a CH2 and a CH3 region of IgGl; and (c) the extracellular ligand binding domain of the PDGF receptor comprises Ig-like domains Dl to D3 of a PDGFR, preferably PDGFR .

[00104] Embodiment 5 is a fusion protein according to embodiment 4, wherein: (a) the extracellular ligand binding domain of the VEGF receptor comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 7; (b) the Fc region of the antibody comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 12; and (c) the

extracellular ligand binding domain of PDGF receptor comprises an amino acid sequence having at least 90% identity to SEQ ID NO: 2.

[00105] Embodiment 6 is a fusion protein according to embodiment 5, wherein: (a) the extracellular ligand binding domain of VEGF receptor comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 7 and 10; (b) the Fc region of the antibody comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 12 and 15; and (c) the extracellular ligand binding domain of PDGF receptor comprises the amino acid sequence selected from the group consisting of SEQ ID NOs: 2 and 5.

[00106] Embodiment 7 is a fusion protein according to any of embodiments 1 to 6, further comprising a first linker peptide between the Fc region and the first or second peptide at the C- terminus of the fusion protein, and optionally a second linker peptide between the second or first peptide at the N-terminus of the fusion protein and the Fc region.

[00107] Embodiment 8 is a fusion protein according to embodiment 7, wherein the first linker peptide comprises one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 20, 22, 24, 26, 28, 30 and 32, and the second linker peptide comprises an amino acid sequence of SEQ ID NO: 18.

[00108] Embodiment 9 is a fusion protein according to embodiments 1 to 8, wherein the fusion protein further comprises a signal peptide linked to the N-terminus of the fusion protein.

[00109] Embodiment 10 is a fusion protein according to embodiment 9, wherein the signal peptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 34 and 36.

[00110] Embodiment 1 1 is a fusion protein according to any of embodiments 1 -10, wherein the fusion protein is arranged from N-terminus to C -terminus in an order of (c)-(b)-(a);

[00111] Embodiment 12 is a fusion protein comprising an amino acid sequence having at least 90% identity to SEQ ID NO: 38 or 40 or having at least 90% identity to amino acids 20-766 of SEQ ID NO: 42, amino acids 21 -769 of SEQ ID NO: 44 or amino acids 20-768 of SEQ ID NO: 50.

[00112] Embodiment 13 is a fusion protein according to embodiment 12, comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 38, SEQ ID NO: 40, amino acids 20-766 of SEQ ID NO: 42, amino acids 21-769 of SEQ ID NO: 44 and amino acids 20-768 of SEQ ID NO: 50.

[00113] Embodiment 14 is an isolated nucleic acid molecule encoding the fusion protein of any one of embodiments 1 to 13.

[00114] Embodiment 1 5 is an isolated nucleic acid molecule according to embodiment 14, wherein the fusion protein further comprises a signal peptide linked to the N-terminus of the fusion protein.

[00115] Embodiment 16 is an isolated nucleic acid molecule according to embodiment 15, wherein the signal peptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 34 and 36.

[001 16] Embodiment 17 is an isolated nucleic acid molecule according to any of embodiments 14-16, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 37, 39, 41 , 43 and 49.

[00117] Embodiment 18 is an expression vector comprising a nucleic acid molecule encoding the fusion protein of any one of embodiments 1 to 13. [00118] Embodiment 19 is an expression vector according to embodiment 18, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 37, 39, 41, 43 and 49.

|00119] Embodiment 20 is a host cell comprising a nucleic acid molecule encoding the fusion protein of any one of embodiments 1 to 13.

[00120] Embodiment 21 is a method of producing the fusion protein of any one of embodiments 1 to 13, comprising: (1) culturing a host cell comprising a nucleic acid molecule encoding the fusion protein of any one of embodiments 1 to 13 under a condition that the fusion protein is produced; and (2) recovering the fusion protein produced by the host cell.

[00121] Embodiment 22 is a pharmaceutical composition comprising the fusion protein of any one of embodiments 1 to 13 and a pharmaceutically acceptable carrier.

[00122] Embodiment 23 is a pharmaceutical composition according to embodiment 22, wherein the composition comprises 40 to 80 mg/mL of the fusion protein formulated in a buffer comprising about 5-100 mM histidne, about 1-10% w/v sucrose, and about 0.005-0.1% w/v polysorbate 20, at a pH of about 6.3 to 7.3.

[00123] Embodiment 24 is a pharmaceutical composition comprising a nucleic acid molecule encoding the fusion protein of any one of embodiments 1 to 13 and a pharmaceutically acceptable carrier such as a lipid carrier (e.g., Lipofectamine), chemicals (e.g., polyethleneimine), or an electroporation buffer.

[00124] Embodiment 25 is a pharmaceutical composition according to embodiment 24, wherein the composition comprises plasmids with expression cassette and a pharmaceutically acceptable earner such as a lipid carrier (e.g., Lipofectamine), chemicals (e.g., polyethleneimine), or an electroporation buffer.

[00125] Embodiment 26 is a method of treating or preventing a clinical condition selected from the group consisting of neovascularization, vascular permeability, edema and inflammation, the method comprising administering to a subject in need thereof an effective amount of the fusion protein of any one of embodiments 1 to 13, or the pharmaceutical composition of any of Embodiments 22-25.

[00126] Embodiment 27 is the method of embodiment 26, wherein the fusion protein is administered as an isolated protein in the pharmaceutical composition of any of Embodiments 22-23. [00127] Embodiment 28 is the method of embodiment 26, wherein the fusion protein is administered in the pharmaceutical composition of any of Embodiments 24 and 25.

[00128] Embodiment 29 is a method of any one of embodiments 26 to 28, wherein the clinical condition is selected from the group consisting of brain edema, stroke, cancer, psoriasis, arthritis, asthma, generalized edema associated with burns, ascites and pleural effusion associated with tumors, inflammation or trauma, chronic airway inflammation, capillary leak syndrome, sepsis, kidney disease associated with increased leakage of protein, ocular

inflammation and/or ocular angiogenesis, including aged-related macular degeneration, diabetic retinopathy, uveitis and corneal neovascularization.

[00129] Embodiment 30 is a method of treating or preventing a clinical condition selected from the group consisting of choroidal neovascularization (CNV), wet age-related macular degeneration (AMD) and geographic atrophy, the method comprising administering to a subject in need thereof an effective amount of the fusion protein of any one of embodiments 1 to 13, or the pharmaceutical composition of any of Embodiments 22-25.

[00130] Embodiment 31 is the method of embodiment 30, wherein the fusion protein is administered as an isolated protein in the pharmaceutical composition of any of Embodiments 22-23.

[00131] Embodiment 32 is the method of embodiment 30, wherein the fusion protein is administered in the pharmaceutical composition of any of Embodiments 24 and 25.

[00132] Embodiment 33 is a dimeric antagonist for PDGF and VEGF comprising the fusion protein of any one of embodiments 1 to 13.

[00133] Embodiment 34 is a protein conjugate comprising the fusion protein of any one of embodiments 1 to 13 bound to at least one ligand selected from the group consisting of PDGF and VEGF.

EXAMPLES

[00134] The following examples of the invention are to further illustrate the nature of the invention. It should be understood that the following examples do not limit the invention and that the scope of the invention is to be determined by the appended claims.

[00135] Example 1 - Generation, Expression and Purification and Analysis of Fusion

Proteins [00136] A PDGFR extracellular Ig-like domain (PID) (SEQ ID NO: 2) (Heidaran et al,

FASEB J. 1995 Jan;9(l):140-5; Lokker et al, J Biol Chem. 1997 Dec 26;272(52):33037-44) having the Ig-like domains D1-D3 of PDGFRp, and a VEGFR extracellular Ig-like domain (VID) (SEQ ID NO: 7) having the Ig-like domain D2 of VEGFR- 1 (VEGFR- 1_D2) and the Ig- like domain D3 of VEGFR-2 (VEGFR-2_D3) (Holash, J., et al, PNAS, 2002, 99 (17): 1 1393 - 98) were incorporated into fusion proteins. A short flexible peptide linker, GGGGGS (SEQ ID NO: 20) was placed between the C-terminus of the Fc region (SEQ ID NO: 12) and the N- terminal module (either PID or VID) to ensure correct folding and minimize steric hindrance. A signal peptide (e.g., SEQ ID NO: 34 or SEQ ID NO: 36) was included to ensure that the produced Fusion Protein 2 or Fusion Protein 1 would be secreted. The Fc region of human IgGl was incorporated to cause dimerization of the fusion protein, mimicking in vivo receptor dimerization, and to allow for easy purification of the expressed fusion proteins.

|00137] Coding sequences of Fusion Proteins 1 and 2 having the amino acid sequences of

SEQ ID NOs: 38 and 40, respectively, were transfected into and expressed in human embryonic kidney cell line (HEK293F). The secreted fusion proteins were purified from the cell culture supernatant using one- step Protein G chromatography. The proteins were captured by Protein G affinity column (Thermo-Fisher Scientific), eluted with low pH (3.5) buffer, and neutralized with Tris-HCl. As shown in Figures 2A-2C greater than 90% purity was achieved using the single step purification method. Figures 2A-2C also show that purified Fusion Proteins 1, 2, 3 and 5 have the predicted weight (MW - 180 kDa) and properly dimerized when expressed in mammalian cells.

[00138] During development, the coding sequence of Fusion Protein 1 , SEQ ID NO: 37, was incorporated into an expression vector. The results of competition binding assays of purified Fusion Protein 1 against PDGF and of a PDGF-dependent BATB/3T3 cell growth inhibition assay of Fusion Protein 1 indicate that there is a positional effect on the binding ability of Fusion Protein 1, e.g., it binds PDGF-BB with much lower affinity (see, e.g., Tables 4 and 6).

[00139] Therefore, Fusion Protein 2 was constructed by rearranging the orientation of VID and PID. The coding sequence of Fusion Protein 2, SEQ ID NO 39, was incorporated into an expression vector. HEK293F host cells were transfected with the expression vector using a suitable amount of expression vector DNA. A stable clone was selected based on protein expression levels evaluated by an anti-Fc EI JSA, and a research cell bank (RCB) was generated from the clone with the optimal levels of fusion protein expression. The Fusion Protein 2 RCB was used for shake flask protein production, and the protein was purified from the culture supernatant using Protein A affinity resin. The protein was buffer-exchanged to phosphate buffer with excipients. The final concentration of the purified Fusion Protein 2 was

approximately 20 mg/mL.

[00140] Mutagenesis of Fusion Protein 2 was earned out to generate a variant of Fusion Protein 2. i.e., Fusion Protein 3, which comprises a deleted lysine residue ( 528) at the C- terminus of IgGl Fc and a phenylalanine-to-serine (F150S) mutation in the PID of Fusion Protein 2. A 2-step PCR mutagenesis method was used to first delete the lysine and then change the phenylalanine to a serine. The final construct comprises a coding sequence of SEQ ID NO 43. CHO-S host cells were transfected with the expression vector using a suitable amount of expression vector DNA. A first stable clone was selected based on protein expression levels evaluated by an anti-Fc ELISA. This was used to produce Fusion Protein 3 having the amino acid sequence of amino acids 21-769 of SEQ ID NO: 44 (amino acids 1-20 of SEQ ID NO: 44 is a signal peptide sequence that was cleaved off during protein synthesis). Monoclonality issues were detected by FACS, so a round of re-cloning was performed. A second stable clone was selected based on expression levels, and the clone was used to generate a Fusion Protein 4 RCB. The Fusion Protein 4 RCB was used for both shake flask studies and bioreactor production. A vial of Fusion Protein 4 RCB was thawed and expanded in a 1L shake flask to produce Fusion Protein 4, which has the same amino acid sequence of Fusion Protein 3. Six batches of purified protein were prepared using Protein A affinity resin (one step purification). The protein was buffer-exchanged to histidine buffer with excipients. The final concentration of Fusion Protein 4 was approximately 20 mg/mL.

[00141] To obtain a sufficient amount of Fusion Protein 4, a 7L bioreactor (BR) was run with a final working volume of about 5 L. Platform operation procedures, parameter settings, and control strategies were used, and the medium type and feed schedule were followed. The final product titer was 0.21 g/L.

[00142] Purified fusion proteins samples were further processed by clarification, protein A affinity chromatography, a first ultrafiltration step, size exclusion chromatography, a second ultrafiltration step, dialysis, and a third ultrafiltration step to obtain the final product. [00143] To generate Fusion Protein 5, which has the amino acid sequence of amino acids

20-768 of SEQ ID NO: 50 (amino acids 1-19 of SEQ ID NO: 50 is a signal peptide sequence that was cleaved off during protein synthesis), the DNA sequence encoding Fusion Protein 5 was codon-optimizing for expression in CHO cells. The synthesized codon-optimized DNA, with a nucleic acid sequence of SEQ ID NO: 49, was cloned into an expression vector. CHO Kl host cells were seeded at 2 x 105 cells/mL in CD CHO (Gibco 12490-003) containing 4mM

Glutamine (J.T Baker 2078-06) and 1 % HT Supplement (Gibco 11067-030) 72 hours before transfection. The host cells were incubated in an Infors shaker (36.5°C, 75 % humidity, 6 % C02, 1 10 RPM) and counted for cell density before use. A suitable amount of expression plasmid DNA was added into the host cells (1L or 5L working volume), and polymer-based transfection reagent was added. The transfected cultures were incubated in an Infors shaker (36.5°C, 75 % humidity, 6 % C02, 110 RPM) for 4 hours and a proprietary feed solution was added. The transfected cultures were then incubated in an Infors shaker (32°C, 75 % humidity, 6 % C02, 1 10 RPM). The transfected cultures were harvested on day 10 after transfection. The supernatants were purified for generation of research materials. The purification process included clarification, Protein A affinity chromatography, concentration by Amicon Ultracel, size exclusion chromatography, dialysis by Slide- A-Lyzer, and final concentration by Amicon Ultracel in the formulation buffer.

[00144] Fusion Protein 6, having the amino acids 20-766 of SEQ ID NO: 42, was derived from Fusion Protein 5 by deleting the first two amino acids (QG) of Fusion Protein 5, wherein amino acids 1-19 of SEQ ID NO: 42 is a signal peptide sequence that was cleaved off during protein synthesis. The DNA insert was generated by PCR using Fusion Protein 5 expression vector as a template, and the DNA sequence encoding Fusion Protein 6, with a nucleic acid sequence of SEQ ID NO: 41 , was cloned into an expression vector. The culturing and purification procedures were the same as for Fusion Protein 5. The final concentration of Fusion Protein 6 was approximately 80 mg/mL in the designated formulation buffer (based on the absorption at 280 nm).

[00145] Collectively, these data indicate that fusion proteins having simultaneous anti-

VEGF and anti-PDGF activities have been constructed, expressed, purified and characterized.

[00146] Example 2: Binding Affinity of the Fusion Proteins to VEGFi 65 [00147] Λ direct binding enzyme-linked immunosorbent assay (EL1SA) was used to measure the binding affinity of fusion proteins of the invention to VEGF 16 5, a splice variant of VEGF-A. A synthesized VEGF Trap was used as Positive Control 1.

[00148] VEGF Trap is a soluble VEGF receptor that was engineered for therapeutic use and is currently approved by FDA to treat AMD. VEGF Trap contains the second Ig-like domain (D2) of VEGFR1 fused to the third Ig-like domain (D3) of VEGFR2 fused to the Fc region of human IgGl (Holash, J., et al, Proc Natl Acad Sci U S A. 2002 Aug 20;99(17): 11393- 8). VEGF Trap targets VEGF-A, VEGF-B, and P1GF.

[00149] 100 μί, of a coating solution (1 μg/mL VEGF165 in lx phosphate buffered saline (PBS), pH 7.2) were added to each well of a 96-well ELISA plate, and the plate was incubated overnight at 4°C. The wells were washed twice with 400 of lx PBS buffer, and excess liquid was carefully removed with a paper towel.

[00150] 400 of a blocking solution (5 g non-fat skim milk in 100 mL lx PBS) were added to each well, and the plate was incubated at room temperature for 1 hour. The wells were washed twice with l PBS buffer.

[00151] Fusion protein and control samples were serially diluted three-fold in blocking solution, with a highest protein concentration of 10 mM. 100 μΤ of the serially diluted samples were added to each well. The plate was covered and incubated on a plate shaker (-100 rpm) for 1 hour at room temperature. The wells were washed three times with wash buffer (l PBS, 0.05% Tween-20).

[00152] 100 μΐ^ of 1 :2500 diluted horseradish peroxidase-conjugated goat anti-human

IgG Fc specific antibodies in blocking solution were added to each well. The plates were sealed and incubated on a plate shaker for 1 hour at room temperature. The plates were washed three times with wash buffer.

[00153] 100 μΐ of 3,5,3',5'-Tetramethylbenzidine (TMB) were added to each well, and the plates were incubated for 3 to 5 minutes to allow for the reaction to take place. To stop the reaction, 100 μΕ of stop solution (IN HC1) were added to each well.

[00154] The optical density (OD) of each well was determined using an ELISA plate reader at an absorbance wavelength of 450 nm. The absorbance was plotted against the protein concentration of the fusion protein or the control, and the concentration at which the signal was half the maximal effective concentration (EC 5 0) was determined. [00155] The binding affinity, expressed as the EC50 value, was between 0.22 and 0.93 nM for the tested fusion proteins of the invention. The EL1S A results are shown in Table 1.

Table 1

[00156] Results from this Example showed that fusion proteins according to embodiments of the invention, such as Fusion Proteins 1 to 5, bind VEGF 16 5 with a high affinity. See also Figure 3.

[00157] Example 3 - Binding Affinity of the Fusion Proteins to PDGF

[00158] A direct binding ELISA was used to measure the binding affinity of fusion proteins of the invention to PDGF. A synthesized PDGF Trap was used as Positive Control 2.

[00159] PDGF Trap is a soluble PDGF receptor that was engineered for use as a positive control. PDGF Trap contains the second Ig-like domain (Dl to D3) of PDGFRp fused to the Fc region of human IgGl . (Lu et al. Am J Obstet Gynecol, 2008, 198(4): 477.el-el 0). PDGF Trap targets PDGF-BB, PDGF-DD, and PDGF-AB.

[00160] 100 μΕ of a coating solution (1 μg/mE PDGF-BB in lx phosphate buffered saline

(PBS), pH 7.2) were added to each well of a 96-well ELISA plate, and the plate was incubated overnight at 4°C. The wells were washed twice with 400 μΕ of lx PBS buffer, and excess liquid was carefully removed with a paper towel.

[00161] 400 μΐ, of a blocking solution (1 g bovine serum albumin in 100 mL of lx PBS) were added to each well, and the plate was incubated at room temperature for 1 hour. The wells were washed twice with lx PBS buffer.

[00162] Fusion protein and control samples were serially diluted three-fold in blocking solution, with a highest protein concentration of 10 mM. 100 μΕ of the serially diluted samples were added to each well. The plate was covered and incubated on a plate shaker (-100 rpm) for 1 hour at room temperature. The wells were washed three times with wash buffer (lx PBS, 0.05% Tween-20). [00163] 100 μΐ of 1 :2500 diluted horseradish peroxidase-conju gated goat anti-human

IgG Fc specific antibodies in blocking solution were added to each well. The plates were sealed and incubated on a plate shaker for 1 hour at room temperature. The plates were washed tliree times with wash buffer.

[00164] 100 μΐ, of 3,5,3',5'-Tetramethylbenzidine (TMB) were added to each well, and the plates were incubated for 3 to 5 minutes to allow for the reaction to take place. To stop the reaction, 100 μΐ. of stop solution (IN HC1) were added to each well.

[00165] The optical density (OD) of each well was determined using an ELISA plate reader at an absorbance wavelength of 450 nm. The absorbance was plotted against the protein concentration of the fusion protein or the control, and the concentration of the fusion protein at which the signal was half the maximal effective concentration (EC 5 0) was determined.

[00166] The binding affinity, expressed as the EC 5 0 value, was approximately 0.16 to 2.5 nM for the tested fusion proteins of the invention. The ELISA results are shown in Table 2.

Table 2

[00167] Results from this Example showed that fusion proteins of the invention, e.g.,

Fusion Proteins 1 to 5, bind PDGF with a high affinity. See also Figure 4.

[00168] Example 4 - Competitive Binding of the Fusion Proteins to VEGF K , 5

[00169] A competitive binding assay was used to assess the binding affinity of fusion proteins of the invention to VEGF165. A synthesized VEGF Trap was used as Positive Control 1.

[00170] Fusion protein and control samples were serially diluted three-fold in blocking solution, with a highest protein concentration of 10 mM. Equal volumes of the diluted samples were incubated with 10 pM of VEGF 1 5 for a final concentration of 5 pM VEGF (R&D System) overnight at room temperature.

[00171] 50 μΕ of assay diluent from the Quantikine ELISA Human VEGF kit (R&D

Systems, Inc.) were added to each well of a 96-well plate. 200 μΐ, of the standards, controls, or fusion proteins were added to the appropriate wells in duplicate. The plates were sealed and incubated for 2 hours at room temperature and then washed three times with wash buffer.

[00172] 200 of VEGFi 6 5 conjugate provided in the kit was added to each well, and the plates were sealed and incubated for 2 hours at room temperature. The plates were washed three times.

[00173] 200 iL of substrate solution provided in the kit were added to each well, and the plates were sealed and incubated for 20 minutes at room temperature. To stop the reaction, 50 μΤ of stop solution provided in the kit were added to each well.

[00174] The OD of each well was determined using an ELISA plate reader at an absorbance wavelength of 450 nm and 540 nm (or 570 nm). The concentration of unbound VEGFi6 5 (free VEGF 1 65) was plotted against the protein concentration of the fusion protein or the control, and the concentration of the fusion protein at which the signal from the free VEGF165 was reduced by 50% (IC50) was determined.

[00175] The IC50 value of the fusion protein of the invention was determined to be approximately 3.78 to 4.67 pM. The results of the competitive binding assay are shown in Table 3.

Table 3

[00176] Results from this Example confirmed that fusion proteins of the invention, e.g., Fusion Proteins 1 , 2 and 3, bind VEGF 1 6 5 with a high affinity. See also Figures 5 A and 6A.

[00177] Example 5 - Competitive Binding of the Fusion Proteins to PDGF-BB

[00178] A competitive binding assay was used to assess the binding affinity of fusion proteins of the invention to PDGF-BB. A synthesized PDGF Trap was used as Positive Control 2.

[00179] Fusion protein and control samples were serially diluted three-fold in blocking solution, with a highest protein concentration of 1 0 mM. Equal volumes of the diluted samples were incubated with 20 pM of PDGF-BB for a final concentration of 10 pM overnight at room temperature.

[00180] 100 μΙ of assay diluent from the Quantikine ELISA Human PDGF-BB kit were added to each well of a 96-well plate. 100 μΐ. of the standards, controls, or fusion proteins were added to the appropriate wells in duplicate. The plates were sealed and incubated for 2 hours at room temperature and then washed four times with wash buffer.

[00181] 200 μΙ of PDGF-BB conjugate provided in the kit w r ere added to each well, and the plates were sealed and incubated for 1.5 hours at room temperature. The plates were washed four times.

[00182] 200 of substrate solution provided in the kit were added to each well, and the plates were sealed and incubated for 20 minutes at room temperature. To stop the reaction, 50 μΐ, of stop solution provided in the kit were added to each well.

[00183] The OD of each well was determined using an ELISA plate reader at an

absorbance wavelength of 450 nm and 540 nm (or 570 nm). Free PDGF-BB was plotted against the protein concentration of the fusion protein or the control, and the concentration of the fusion protein at which the signal from the free PDGF-BB was reduced by 50% (IC 5 o) was determined.

[00184] The IC 5 0 value of the fusion protein of the invention was determined to be approximately 0.125-200 nM. The results of the competitive binding assay are shown in Table 4.

Tabic 4

[00185] Results from this Example confirmed that fusion proteins of the invention, e.g.,

Fusion Proteins 2 and 3, bind PDGF with a high affinity. See also Figures 5B and 6B.

[00186] Example 6 - Inhibition of HUVEC Proliferation by the Fusion Proteins

[00187] A human umbilical vein endothelial cell (HUVEC) proliferation assay was carried out to test the functionality of the fusion proteins of the invention. A synthesized VEGF Trap was used as Positive Control 1. [00188] 100 of a coating solution (1% gelatin in double distilled water) were added to each well of a 96-well ELISA plate, and the plate was incubated for 2 hours or overnight at 37°C. The wells were washed twice with lx PBS buffer.

[00189] 3500 counts of human umbilical vein endothelial cells in endothelial cell growth medium were added to each well, and the plate was incubated overnight at 37°C.

[00190] Fusion protein samples were diluted in assay buffer (Medium-199 lx Earle's Salts, 10% fetal bovine serum, 10 mM HEPES, lx antibiotic/antimycotic). with a highest protein concentration of 300 nM. The fusion protein samples were mixed with VEGF 16 s (8 ng/mL), and the mixtures were incubated overnight at room temperature. The wells were then washed with ΙΟΟ μΙ οΐ lx PBS.

[00191] 100 μΐ, of the VEGFi 6 s/sample mixture were added to each well, and the plates were incubated for 72 hours at 37°C with 5% C0 2. Following incubation, 10 μΕ MTS detection reagent (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-( 4-sulfophenyl)-2H- tetrazolium) + phenazine methosulfaie in distilled PBS) were added to each well, and the plates were incubated at 37°C for 2.5 hours.

[00192] The OD of each well was determined using an ELISA plate reader at an absorbance wavelength of 490 nm. The absorbance was plotted against the protein concentration of the fusion protein or the control, and the concentration at which the cell proliferation was inhibited by 50% (IC 50 ) was determined.

[00193] The inhibition of cell proliferation (IC 5 o) was determined to be between 0.058 and

0.285 nM for the tested fusion proteins of the invention. The results of the proliferation assay are shown in Table 5.

Table 5

[00194] Results from this Example showed that fusion proteins of the invention, e.g.,

Fusion Proteins 1 to 5, inhibited VEGF -dependent growth of ITUVEC cells. See also Figure 7. [00195] Example 7 - Inhibition of BALB/3T3 Proliferation by the Fusion Proteins

[00196] A cell proliferation assay using 3T3 fibroblasts derived from BALB mice was carried out to test the functionality of the fusion proteins of the invention. A synthesized PDGF Trap was used as Positive Control 2.

[00197] 4000 counts of mouse 3T3 fibroblast cells in DMEM (1 mM sodium pyruvate, 4 mM L-glutamine, 10% bovine serum albumin, lx antibiotic/antimycotic) were added to each well, and the plate was incubated overnight at 37°C.

[00198] Fusion protein samples were diluted in assay buffer (1 mM sodium pyruvate, 4 mM L-glutamine, 0.5% bovine serum albumin, lx antibiotic/antimycotic), with a highest protein concentration of 300 nM. The fusion protein samples were mixed with PDGF (8 ng/mL), and the mixtures were incubated overnight at room temperature. The wells were then washed with 200 μΐ of l PBS.

[00199] Cells were starved with assay buffer at 37°C with 5% C0 2 for 4 hours. 100 of the PDGF/sample mixture were added to each well, and the plates were incubated for 72 hours at 37°C with 5% C0 2. Following incubation, 10 MTS detection reagent were added to each well, and the plates were incubated at 37°C for 2.5 hours.

[00200] The OD of each well was determined using an ELISA plate reader at an absorbance wavelength of 490 nm. The absorbance was plotted against the protein concentration of the fusion protein or the control, and the concentration at which the cell proliferation was inhibited by 50% (IC 5 o) was determined.

[00201] The IC 5 o was calculated determined to be between 0.45 and 1000 nM for the tested fusion proteins of the invention. The results of the proliferation assay are shown in Table 6.

Table 6

[00202] Results from this Example showed that fusions of the invention, e.g., Fusion

Proteins 1 to 5, inhibited PDGF-dependent growth of BALB/3T3 cells. See also Figure 8.

[00203] Example 8 - Inhibition of VEGF-Induced Leakage in Dutch Belted Rabbits by the Fusion Proteins

[00204] Fusion proteins of the invention were tested in an in vivo model of retinal neovascularization to determine their efficacy in preventing vascular leakage. In this model, VEGF is intravitreally injected to the vitreous of rabbit eyes to induce uncontrolled

neovascularization of the retina and subsequent leakage. Avastin®, a recombinant humanized monoclonal antibody that blocks angiogenesis by inhibiting VEGF-A, and Eylea, a recombinant fusion protein consisting of portions of human VEGFRl and VEGFR2 fused to the Fc portion of human IgGl , were used as Positive Controls 3 and 4.

[00205] Dutch Belted rabbits were anesthetized using isoflurane (3-5%), and their eyes were treated with ophthalmic Betadine solution. The rabbits' eyes were then washed with sterile saline, and lidocaine hydrochloride (2% injectable) or proparacaine (0.5%) was applied to the ocular surface.

[00206] On Day 1, Dutch Belted rabbits were intravitreally injected with fusion proteins of the invention, vehicle (negative) control, or reference (positive) controls at predetermined doses using a BD 300 μΕ insulin syringe (31 ga x 5/16 inch). The needle was inserted through the dorsotemporal quadrant of the eye, approximately 3-4 mm poseterior to the limb and 3-4 mm lateral to the dorsal rectus muscles, and 50 μΕ of solution was delivered. On Day 3, VEGF 165 was injected into the same eyes.

[00207] Fluorescein angiograms (FAs) were conducted on all dosage groups 3 days after

VEGF-induction (Day 6) to assess leakiness and tortuosity using a scale from 0 (normal) to 4 (severe).

[00208] Signs of ocular irritation were scored using the Draize scoring system prior to fusion protein dosing, prior to VEGF induction, and prior to FA assessments. According to the Draize analysis, all of the rabbit eyes were normal prior to the initiation of dosing, and no drug- related findings were evident during the course of the study. The findings that were scored using the Draize system were transient and observed in all of the dose groups, and were thus likely due to the procedure associated with the intravitreal dose administration. [00209] FAs associated with the vehicle control group had the highest mean score (2.58) associated with retinal vasculature leakiness and tortuosity. The two reference positive control groups had mean scores of 0.25 and 0, indicating a significant reduction in retinal vasculature leakiness and tortuosity. The tested fusion proteins of the invention had a mean score of 0.167, showing effectiveness in reducing VEGF-induced retinal leakiness and tortuosity comparable to the positive controls. The results of the in vivo assay are shown in Table 7.

Table 7

[00210] Example 9 - Dose-Response Inhibition of VEGF-induced Leakage in Dutch Belted Rabbits by the Fusion Proteins

[00211] Fusion proteins of the invention were tested in an in vivo model of retinal neovascularization at varying doses to determine their dose-response effectiveness in preventing vascular leakage. In this model, VEGF 165 is intravitreally injected to the vitreous of rabbit eyes to induce uncontrolled neovascularization of the retina and subsequent leakage.

[00212] On Day 1 , Dutch Belted rabbits were intravitreally injected with Fusion Protein 5 according to an embodiment of the invention at various doses, vehicle (negative) control, or reference (positive) controls. VEGF-induction was carried out on day 3.

[00213] FAs were conducted on all dosage groups 3 days after the VEGF-induction (Day 6) to assess leakiness and tortuosity using a scale from 0 (normal) to 4 (severe).

|00214] Signs of ocular irritation were noted using the Draize scoring system prior to fusion protein dosing, prior to VEGF-induction, and prior to FA assessments. According to the Draize analysis, all of the rabbit eyes were normal prior to the initiation of dosing, and no drug- related findings were evident during the course of the study. The findings that were scored using the Draize system were transient and observed in all of the dose groups, and were thus likely due to the procedure associated with the intravitreal dose administration. [00215] For the first VEGF-induction, FAs associated with vehicle control group had the highest mean score (3.4) associated with retinal vasculature leakiness and tortuosity. The two reference positive control groups had mean scores of 0, indicating a significant reduction in retinal vasculature leakiness and tortuosity. The tested fusion protein of the invention (Fusion Protein 5) had scores of 0.08, 0.42, and 0.17 at doses of 100, 500 and 1000 μg, respectively, showing effectiveness in reducing VEGF-induced retinal leakiness and tortuosity comparable to the positive controls.

[00216] The results of the dose-response in vitro assay are shown in Table 8.

Table 8

[00217] Example 10 - Reduction of Lesion Size in Laser-induced Choroidal

Neovascularization (CNV) in Rats by the Fusion Proteins

[00218] The eyes of Brown Norway will be dilated with a 1% Cyclogyl solution and protected from light. Following dilation, the rats will be anesthetized using a ketamine and xylazine mixture. Three lesion burns will be introduced to the retina of each eye using a laser at 532 nm on Day 1.

[00219] On Day 3, the animals will be anesthetized with a ketamine and xylazine mixture, their eyes will be dilated, and 5 of fusion proteins according to embodiments of the invention, vehicle (negative) control, or reference (positive) controls at predetermined doses will be intravitreally injected into both eyes of an animal using a Hamilton syringe with 33 gauge needle.

[00220] On Day 22, the animals will receive an IP injection of 10% fluorescein sodium at

1 μΕ/g of body weight. Fundus images will be taken prior to lesion introduction, after lesion burns to confirm successful lesions, and on Day 22 using a Micron III small animal funduscope (Phoenix Research). Lesion size will be determined and compared across dosage groups. [00221] Example 11 - Reduction of Lesion Size in Laser-induced CNV in Monkeys by the Fusion Proteins

[00222] A laser-induced CNV model will be established in monkeys. Six to nine burns will be introduced around the macula of each eye using 532 nm diode laser photocoagulation, and 0.5 mg of fusion proteins of the invention will be intravitreally injected on the same day.

[00223] The animals will be sedated with intravenous 2.5% soluble pentobarbitone (1 mL/kg) 20 days later. The eyelids will be fixed to keep the eyes open, and color photographs will be taken using a fundus camera.

[00224] Fluorescein dye (20% fluorescein sodium; 0.05mL/kg) will then be injected into a vein of a lower extremity. Photographs will be taken at several time points after injection of the dye, including the arterial phase, early arteriovenous phase, and several late arteriovenous phases, to monitor leakage of fluorescein associated with CNV lesions.

[00225] Example 12 - Inhibition of Human Tumor Growth in Xenograft Mice by the Fusion Proteins

[00226] Various human cancer cells, such as human hepatocellular carcinoma Hep3B cells (ATCC# HB-8064) and human colorectal cancer LoVo cells (ATCC# CCL-229), can be used to establish xenograft models in nude mice.

[00227] In order to assess the inhibitory effects of the fusion proteins of the invention on the tumor growth, tumor cells will be implanted into nude mice, and various concentrations of fusion proteins according to embodiments of the invention, ranging from 0.1 to 10 mg/kg, will be administered to the mice intravenously twice weekly. The tumor growth will be measured weekly for up to 7 weeks.

[00228] Example 13 - Pharmacokinetic Assessment of the Fusion Proteins in Rats and Monkeys

[00229] The pharmacokinetics of the fusion proteins of the invention will be assessed in animals. A range of 10 to 300 mg/kg fusion proteins according to embodiments of the invention will be administered to rats or monkeys via subcutaneous injection or intravenous injection.

Blood samples will be obtained at different time points after the injection for up to 15 days. The concentrations of the fusion proteins in the blood samples will be determined using an ELISA method, and pharmacokinetic parameters will be calculated. [00230] Example 14 - Ocular Pharmacokinetic Assessment of the Fusion Proteins in

Rabbits and Monkeys

[00231] The pharmacokinetics of the fusion proteins of the invention will be assessed in animals. A range of 0.1 to 4 mg per eye of fusion proteins according to embodiments of the invention will be administered to rabbits or monkeys via intravitreal injection. Ocular tissues and blood samples will be obtained at different time points after the injection for up to 28 days. The concentrations of the fusion proteins in the ocular tissues and blood samples will be determined using an ELISA method, and pharmacokinetic parameters will be calculated. [00232] While the invention has been described in detail, and with reference to specific embodiments thereof, it will be apparent to one of ordinary skill in the art mat various changes and modifications can be made therein without departing from the spirit and scope of the invention.