WO2009105669A2 - Angiogenesis inhibition - Google Patents

Angiogenesis inhibition Download PDF

Info

Publication number
WO2009105669A2
WO2009105669A2 PCT/US2009/034709 US2009034709W WO2009105669A2 WO 2009105669 A2 WO2009105669 A2 WO 2009105669A2 US 2009034709 W US2009034709 W US 2009034709W WO 2009105669 A2 WO2009105669 A2 WO 2009105669A2
Authority
WO
WIPO (PCT)
Prior art keywords
vegf
heavy chain
vegf antagonist
antagonist
human
Prior art date
Application number
PCT/US2009/034709
Other languages
French (fr)
Other versions
WO2009105669A3 (en
Inventor
Abraham Scaria
Samuel Wadsworth
Original Assignee
Genzyme Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genzyme Corporation filed Critical Genzyme Corporation
Priority to BRPI0908496A priority Critical patent/BRPI0908496A2/en
Priority to JP2010547799A priority patent/JP2011512417A/en
Priority to CN2009801059368A priority patent/CN101951925A/en
Publication of WO2009105669A2 publication Critical patent/WO2009105669A2/en
Publication of WO2009105669A3 publication Critical patent/WO2009105669A3/en
Priority to IL207641A priority patent/IL207641A0/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/025Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a parvovirus

Definitions

  • the invention relates to compositions and methods useful for inhibiting angiogenesis or treating pathological neovascularization e.g., asthma, arthritis, cancer, and macular degeneration.
  • pathological neovascularization e.g., asthma, arthritis, cancer, and macular degeneration.
  • Neovascularization is a key component of diseases like wet age-related macular degeneration (AMD), proliferative diabetic retinopathy, rheumatoid arthritis, osteoarthritis, and asthma. It also plays an important role in growth and spread of tumors. Neovascularization is regulated by an extraordinarily balance of pro- and anti- angiogenic factors.
  • VEGF Vascular endothelial growth factor
  • VEGF antagonists that inhibit or reduce VEGF activity include, but are not limited to, VEGF antibodies, soluble receptors, receptor fusion proteins, peptides and small molecules.
  • VEGF-R 1 Fit- 1
  • KDR VEGF-R2
  • a fusion protein has the formula X-Y-Z.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a 5 - 25 amino acid residue polypeptide.
  • Z is a CH3 region of an IgG heavy chain molecule.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues.
  • Z is a CH3 region of an IgG heavy chain molecule.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a 5 - 25 amino acid residue polypeptide.
  • Z is an Fc portion of an antibody molecule.
  • a fusion protein of the formula X-Y-Z is also provided.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues.
  • Z is an Fc portion of an antibody molecule.
  • Still another aspect of the invention is a method of multimerizing a polypeptide X.
  • a polypeptide X is linked to a polypeptide Z via a polypeptide Y to form polypeptide XYZ.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a 5 - 25 amino acid residue polypeptide.
  • Z is a CH3 region of an IgG heavy chain molecule.
  • Polypeptide XYZ which is formed multimerizes.
  • Yet another embodiment of the invention provides a method of m ⁇ ltimerizing a polypeptide X.
  • Polypeptide X is linked to a polypeptide Z via a moiety Y to form polymer XYZ.
  • X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor.
  • Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues.
  • Z is a CH3 region of an IgG heavy chain molecule.
  • Polypeptide XYZ which is so formed multimerizes.
  • a nucleic acid molecule encodes a fusion protein which comprises an lg-like domain 2 of VHGF-R 1 (FIt-I); a linker: and a multimerization domain.
  • the fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
  • a fusion protein comprises an Ig-like domain 2 of VEGF-Rl (FIt-I), a linker, and a multimerization domain.
  • the fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
  • an in vitro method is provided.
  • a nucleic acid molecule is delivered to an isolated mammalian cell.
  • the nucleic acid molecule encodes a fusion protein which comprises an Ig-like domain 2 of VEGF-Rl (FIt-I; a linker; and a multimerization domain.
  • the fusion protein comprises a sequence selected from the group consisting of SEQ D NO: 2, 8, 21, 23, and 25. Expression of the fusion protein is controlled by a promoter.
  • a cell is formed which expresses a fusion protein.
  • Still another embodiment of the invention is a method for delivering a fusion protein to a mammal.
  • a mammalian cell which expresses the fusion protein is delivered to a mammal.
  • the cell expresses and secretes the fusion protein thereby supplying the fusion protein to the mammal.
  • the fusion protein comprises an Ig-like domain 2 of VEGF-Rl (FIt-I). a linker, and a rnultimerization domain.
  • the fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
  • a fusion protein which comprises an Ig-like domain 2 of VEGF-RI (FIt-I), a linker, and a raultimerization domain is delivered to a mammal.
  • the fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
  • a nucleic acid construct which encodes said fusion protein can be delivered to the mammal, whereby the fusion protein is expressed by the mammal.
  • Another aspect of the invention is an improved method of treating an ocular disease in a mammal with a VEGF antagonist, including but not limited to, the fusion proteins of the present invention.
  • the improvement comprises providing a reduced amount of the VEGF antagonist that is therapeutically effective to treat the ocular disease.
  • the VEGF antagonist may be administered by protein or gene therapy, or the combination thereof.
  • the reduced amount may be maintained in the mammal for at least 6 months, e.g.. at least 9 months or at least i year.
  • the reduced amount may be from about 100 pg/ml to about 100 ⁇ g/ml, 1 ng/ml to about 95 ⁇ g/ml, 10 ng/ml to about 85 ⁇ g/m), 100 ng/ml to about 75 ⁇ g/rnl, from about 100 ng/ml to about 50 ⁇ g/ml, from about 1 ⁇ g/ml to about 25 ⁇ g/ml, from about 1 ⁇ g/ml to about 15 ⁇ g/ml. from about 1 ⁇ g/ml to about 10 ⁇ g/ml, or from about 1 ⁇ g/ml to about 4 ⁇ g/ml.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises administering to the mammal a nucleic acid encoding a VEGF antagonist.
  • the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 pg/ral to about 100 ⁇ g/ml, from about 1 ng/ml to about 95 ⁇ g/ml, from about 10 ng/ml to about 85 ⁇ g/ml, from about 100 ng/m!
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises administering to the mammal a viral vector encoding a VEGF antagonist at a therapeutically effective amount of from about 2x 10 6 to about 2x 10 12 drp per unit dose.
  • the therapeutically effective amount is from about 2x10 7 to about 2x 10 11 drp, from about 2x10* to about 2x10 10 drp per unit dose.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises administering to the mammal a soluble VEGF receptor or a variant at a therapeutically effective amount of from about 0.1 mg to about 50 mg per unit dose, preferably from about 0.1 mg to about 5 mg, e.g., from about 0.1 mg to about 1 mg, from about 0.1 mg to about 0.5 mg and from about 0.1 mg to about 0.25 mg.
  • the soluble VEGF receptor or its variant includes, but is not limited to, the fusion proteins of the present invention.
  • the soluble VEGF receptor is FLTl D29GLYFC (SEQ ID NO. 1 or 2) or D2-9GLY-CH3 (SEQ [D NO. 22 or 23).
  • the therapeutically effective amount of the soluble VEGF receptor is from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.5 mg to about 5 rag, or from about 0.5 mg to about 2.5 mg.
  • the soluble VEGF receptor or its variant may be administered repeatedly.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises administering to a diseased eye ⁇ f the mammal a nucleic acid encoding a VEGF antagonist of a fusion protein of the invention, where the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 ng/ml to about 75 ⁇ g/ml to treat the ocular disease.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises delivering Io a diseased eye of the mammal a nucleic acid encoding a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is expressed in transitional epithelial cells of the diseased eye at a therapeutically effective amount of from about 100 ng/ml to about 75 ⁇ g/ml.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises delivering to a diseased eye of the mammal by intravitreal injection (e.g., mid vitreal injection) a nucleic acid encoding a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 ng/ml to about 75 ⁇ g/ml.
  • the VEOF antagonist is expressed at a therapeutically effective amount of from about 100 pg/nil to about 100 ⁇ g/ml, 1 ng/ml to about 95 ⁇ g/ml, 10 ng/mi to about 85 ⁇ g/ml 100 ng/ml to about 50 ⁇ g/ml, from about 1 ⁇ g/ml to about 25 ⁇ g/ml, from about I ⁇ g/ml to about 15 ⁇ g/rnl, from about 1 ⁇ g/ml to about 10 ⁇ g/ml, or from about 1 ⁇ g/ml to about 4 ⁇ g/ml.
  • the nucleic acid encoding a VEGF antagonist is a viral vector.
  • the viral vector is AAV, e.g., AA V2.
  • the VEGF antagonist is expressed in the mammal for at least 6 months, e.g., at least 9 months or 1 year.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal.
  • the method comprises administering to a diseased eye of the mammal a VBGF antagonist of the formula X-Y-Z at a therapeutically effective amount of from about 2x 10 6 to about 2x 10 12 drp per unit dose.
  • the therapeutically effective amount is from about 2x10 7 to about 2x 1 ⁇ " drp, or from about 2x 10* to about 2x 10 10 dip per unit dose.
  • a second VIiGF antagonist is administered.
  • the second VEGF antagonist may be a fusion polypeptide comprising a VEGF receptor fused to an IgG heavy chain molecule, preferably a VEGF-Rl fused to the Fc or CH3 domain of IgG, more preferably, domain 2 of VEG-Rl fused to Fc or CH3.
  • a linker e.g., 9GIy, may be used to link the VEGF receptor and the IgG domain.
  • the second VEGF antagonist may be a VEGF antibody, e.g., bevacizumab or ranibizumab.
  • Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to a diseased eye of the mammal a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is at a therapeutically effective amount of from about 0.1 mg to about 50 mg per unit dose.
  • the therapeutically effective amount is from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.5 mg to about 5 mg, or from about 0.5 mg to about 2.5 mg.
  • Another aspect of the invention is a method of reducing haze in the eye in a mammal receiving gene therapy for the eye.
  • the method comprises administering TO the mammal (e.g., in the eye) gene therapy virions containing empty capsids not exceeding 90% of the total capsids.
  • the empty capsids do not exceed 80%, 70%, 60%, or 50% of the total capsids. In one embodiment, the gene therapy virions are substantially free of empty capsids.
  • the virions are AAV virions, e.g., AA V2 virions.
  • the ocular disease is selected from the group consisting of wet age-related macular degeneration (wet AMD), macular edema, retinal vein occlusions, retinopathy of prematurity, and diabetic retinopathy.
  • wet AMD wet age-related macular degeneration
  • macular edema macular edema
  • retinal vein occlusions retinal vein occlusions
  • retinopathy of prematurity retinopathy of prematurity
  • diabetic retinopathy diabetic retinopathy
  • agents for treating disease related to vascular proliferation and inflammation.
  • the agents may provide increased stability and bioavailability relative to natural forms of the proteins.
  • Biological activity of D2-9Gly-Fc vs. D2-Fc. 293 cells were grown in the starvation media (M 199 + 5% FCS) and transfected with plasmids containing D2- 9GIy-Fc and D2-Fc expression cassettes under control of CMV promoter.
  • Conditioned media (CM) was collected 72 h later.
  • HUVECs were seeded into 96 well plate (2E3 cells/well) in starvation media * VEGF (10 ng/mL) and 50 ul CM plus VEGF (10 ng/mL) was added 24 h later.
  • the controls (+/- VEGF) were incubated with CM from the control pEGFP (Clontech; pEGFP carries a red-shifted variant of wild-type green fluorescent protein (GFP) which has been optimized for brighter fluorescence and higher expression in mammalian cells) plasmid transfection.
  • the positive control was treated with 50 ng of Mt-I-IgG recombinant protein (R & D Systems).
  • the HUVECs were assayed for proliferation 3 days post treatment using CellTiter 96 ® AQ ueous reagent (Promega). The data represent the means of the average values of OD 490 of two experiments each assayed in triplicates.
  • Fig. 3 Western blot analysis of D2-9Gly-Fc and D2-Fc.
  • the size of both D2-9Gly-Fc and D2-Fcproteins appears to be twice as large while migrating in non-reducing gel as compared to migration in reducing gel.
  • the proteins were loaded from the conditioned media following 293 cell transfection of plasmids expressing D2-9Gly-Fc and D2-Fc were separated by SDS-electrophoresis and transferred to PVDF membrane. The blot was probed with goat anti-human anti-lgGl Fc and rabbit anti- goat IgG-HRP antibodies.
  • Fig. 4 sFlt-l hybrid proteins containing 9Gly linker and VEGF Ex3. Structure comparison of D2-9Gly-Ex3/CH3 to previously constructed proteins. All three proteins contain identical amino acid sequence of FIt-I domain 2, consisting of 24 aa of FIt-I signal peptide and 93 aa of FU-I domain 2. D2-9Gly-Ex3/CH3 contains 9 aa of 9GIy linker, 14 aa of VEGF Ex3 and 120 aa of the CH3 region of human IgGl heavy chain Fc.
  • Fig. 5 Biological activity of D2-9Gly-Ex3/CH3 vs. D2-9Gly-Fc. Protein D2-9Gly- Ex3/CH3, where domain 2 is connected to the CH3 region through 9GIy linker and VEGF Ex 3, is also efficiently inhibiting VEGF-dependent HUVECs proliferation as compared to control proteins D2-9Gly-Fc and D2-Fc. 50 ng of the recombinant FIt-I-IgG (R&D Systems) was used as a control.
  • Fig. 6 HUVECs proliferation assay comparing D2- ⁇ GIy 4 Ser) 3 -Fc protein activity with D2-9Gly-Fc and D2-9Gly-Ex3/CH3.
  • Fig. 7 Western blot. Proteins (non-reduced and reduced) from conditioned medium of transfected 293 cells (15 ul of CM) with plasmids expressing (1): D2-9Gly-Fc; (2): D2-(C ⁇ 4S)J-FC and (3) - EGFP proteins were separated by SDS-eiectrophoresis and transferred to PVDF membrane. The blot was probed with goat anti-human IgG1 Fc and rabbit anti-goat IgG-FIRP antibodies.
  • Fig. 8 Combinations of proteins with/without 9GIy linker or VEGF Ex3. Structure comparison of three novel proteins wilh or without 9GIy linker and/or VEGF Ex3, D2-9GIy-C ⁇ 3. D2-CH3 and D2-Ex3/CH3.
  • Fig. 9 HUVECs proliferation assay with the FIt-I (D2) constructs with 9GIy, Ex3 and CH3 combinations. Conditioned media from 293 cells (5 ul) containing proteins D2- Ex3/CH3, D2-9Gly-CI 13 and D2-CH3 were compared to D2-9Gly-Fc and D2-9Gly-
  • Fig. 10 Western blot. 293 cells were transfected with plasmids expressing: (1) D2- 9GIy-Fc; (2) D2-9Gly-CH3 (52/26 kDa); and (3) D2-CH3 (50/25 kDa). Proteins from 293 cells conditioned medium (15 ul of CM non-reduced and/or reduced) were separated by SDS-electrophoresis and transferred to PVDF membrane. The blot was probed with anti-human VEGF-Rl HRP conjugate (R&D Systems).
  • VEGF "in vitro" binding assay Conditioned media from 293 cells containing known concentrations of both D2-9Gly-Fc and FIt-I D(I -3) control soluble receptors (ranging in concentrations from 0.29 - 150 pM) were serially diluted and mixed with IO pM VCGF. The amount of unbound VEGF was then measured by ELISA. D2- 9GIy-Fc binds VEGF with higher affinity than all other constructs, "n” represents the number of independent experiments (transfection and binding assay).
  • Fig. 13A-13C show expression levels of FIt-I constructs having various linkers.
  • Fig. 13B shows dimerization or multimerization of FIt-I constructs having various linkers and a CH3 moiety of Fc of IgGl . The difference between the non- reduced and the reduced conditions indicates that the proteins had multimerized.
  • Fig. 13C shows the inhibitory bioactivity of indicated FIt-I constructs present in condition medium in a HUVEC proliferation assay in the presence of VEGF. Each of the constructs demonstrated inhibitory activity approaching proliferation levels of the HUVEC in the absence of VEGF.
  • FIt-I constructs Using a murine oxygen-induced retinopathy (OIR) model of retinal neovascularization (NV).
  • OIR oxygen-induced retinopathy
  • NV retinal neovascularization
  • one of the FIt-I constructs was administered to the mouse eyes and neovascularization was determined.
  • the mice were exposed to hyperoxic conditions.
  • the number of neovascular events was determined in the treated eyes compared to the events in the untreated eyes of the same animals. The animal was considered a "responder” if there was a greater than 50% reduction in neovascular events.
  • FIG. 15 An in situ hybridization assay shows the expression levels and location of AAV2-sFLTI D29GLYFC in a group of animals.
  • Il is a discovery of the present inventors that a FH-I Ig-like domain 2 without domains 1 and 3 is capable of efficiently binding VEGF and inhibiting VEGF-dependent endothelial cell proliferation.
  • Domain 2 can be covalently (inked to a muitimerization domain via a linker.
  • Linkers are typically polypeptide chains. The length of the chain may be 6, 7, 9, 11. 13, 15 or more amino acid residues, but typically is between 5 and 25 residues. Depending upon the length and side chain composition, a linker may have but need not have greater than average flexibility. Flexibility can be calculated using algorithms known in the art.
  • Multimerization domains are those portions of multimeric proteins which promote the association of subunits to form, for example, dimers, trimers, tctramers, etc.
  • Suitable recombinant proteins for efficiently binding VEGF and/or inhibiting VEGF-dependent endothelial cell proliferation are selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
  • the inventors have found lhat the multimerization domains and linkers can be used with a variety of other proteins or portions of proteins to induce multimerization.
  • Such proteins may be those which bind to Iigand or receptor only when multimcrized, or may be those whose binding affinity is enhanced when multiraerized.
  • Suitable proteins for muJtimerization include extracellular receptors (which include portions thereof), antibody variable regions, cytokines, chemokines, and growth factors. Suitable proteins include tyrosine kinase receptors and serine thereonine kinase receptors. Specific examples of extracellular receptors include EGF-receptor, G protein coupled receptors, FGF receptor, Fc receptors, T cell receptors, etc. Examples of antibody variable regions include Fab, F(ab') 2 , and ScFv. Examples of cytokines include GM-CSF, IL-l ⁇ , IL-1 ⁇ , IL-2, IL-3, IL-4, IL-5.
  • chemokines include BCA-I / BLC, BRAK, Chemokine CC-2, CTACK, CXCL-16, ELC. ENA, ENA-70, ENA-74 , ENA-78, Eotaxin, Exodus-2, Fractalkine, GCP-2, GRO, GRO alpha (MGSA), GRO-beta.
  • MCP-I MCP-2, MCP-3, MCP-4, MDC, MDC, MDC-2, MDC-4, MEC / CCL28, MIG, MIP, MIP-I alpha, MlP-I beta, MIP-I delta, MIP-3 / MPIF-I, MIP-3 alpha, MIP-3 bet , MIP-4 (PARC).
  • growth factors include Human Amphiregulin, Human Angiogenesis Proteins, Human ACE, Human Angiogenin, Human Angiopoietin, Human Angiostatin, Human Betacelluiin, Human BMP, Human BMP-13 / CDMP-2, Human BMP-14 /CDMP-I, Human BMP-2, Human BMP-3, Human BMP-4, Human BMP-5, Human BMP-6, Human BMP-7, Human BMP-8, Human BMP-9, Human Colony Stimulating Factors, Human fU3-Ligand, Human G- CSF, Human GM-CSF, Human M-CSF, Human Connective Tissue Growth Factor, Human Cripto-1, Human Cryptic, Human ECGF, Human EGF, Human EG-VEGF, Human Erythropoietin, Human Fet
  • Human PlGF Human PIGF-L Human PIGF-2, Human SCF, Human SMDF, Human Stem Cell Growth Factor, Human SCGF-alpha, Human SCGF-beta, Human Thrombopoietin, Human Transforming Growth Factor, Human TGF-alpha, Human TGF-beta, and Human VEGF.
  • FIt-I receptor protein has an extracellular portion which comprises seven Ig-like domains. These are located at residue numbers 32...123, 153...214, 230...327, 335...421, 428...553, S56...654, and 661...747 of Genbank accession no. P17948, see also SEQ ID NO: 15. Residue numbers 3-26 comprise a signal sequence.
  • FIt-I protein is encoded by the DNA sequence shown at Genbank accession no. NM_002019 (SEQ ID NO: 14).
  • Multimeriza ⁇ on domains can be used as are known in the art. Sequences of the Fc portion of IgGl or IgG2 lambda heavy chain can be used, for example, CH3 alone (aa 371-477) or both of CH2 and CH3 domains (aa 247-477). Fc portion of Ig molecules is that which is obtained by cleavage of whole antibody molecules with the enzyme papain. Other means can be used to obtain these portions. For the IgGl lambda heavy chain protein sequence, see Genbank accession no Y14737and SEQ ID NO: 10. Other Fc regions can be used for example from other IgG types and from IgA, IgM, IgD, or IgE antibodies.
  • the multimcrization region of VEGF can also be used.
  • a DNA sequence encoding VHGF is shown at Genbank accession no. NM_003376and SEQ ID NO: 11.
  • An amino acid sequence of VEGF is shown at Genbank accession no. CAC19513and SEQ ID NO; 12.
  • the raultimerizalion region of VEGF (SEQ ID NO: 13), encoded by VEGF exon 3 (VEGF Ex3), is at about amino acid residues 75- 88 of VEGF protein (SEQ ID NO: 12).
  • Multiraerization domains will cause at least 5 %, 10 %, 20 %, 30 %, 40 %, 50 % 60 %, 75 %, 80 %, 85 %, 90 %, or 95 % of the monomelic fusion proteins to migrate on a non-denaturing polyacrylamidc gel at a rate appropriate for a multimer. Glycosylation can affect the migration of a protein in a gel. Although particular sequences are shown here, variants such as allelic variants can be used as well. Typically such variants will have at least 85 %, 90 %, 95 %, 97 %, 98 %, or 99 % identity with the disclosed sequence.
  • Multimerizaticn can be assayed, for example, using reducing and non-reducing geis, as demonstrated herein.
  • Multiraerization can also be assayed by detection of increased binding affinity of a protein for its ligand/receptor.
  • BiaCoreTM surface plasmon resonance assays can be used in this regard. These assays detect changes in mass by measuring changes in refractive index in an aqueous layer close to a sensor chip surface. Any method known in the art can be used to detect multiraerization.
  • Linker moieties according to the invention can be comprised of for example 5-100 amino acid residues, 5-75 amino acid residues, 5-50 amino acid residues, 5-25 amino acid residues, 5-20 amino acid residues, 5-15 amino acid residues, 5-10 amino acid residues, 5-9 amino acid residues.
  • linkers examples include: gly9 (SEQ ID NO: 27), gMSEQ ID NO: 28), ser 9 (SEQ ID NO: 29), gly 5 cyspro 2 cys (SEQ ID NO: 30), (gly 4 ser) 3 (SEQ ID NO: 31), SerCysValProLeuMetArgCysGlyGlyCysCysAsn (SEQ ID NO: 32), Pro Ser Cys VaI Pro Leu Met Aig Cys GIy GIy Cys Cys Asn (SEQ ID NO: 13), GIy Asp Leu lie TyT Arg Asn GIn Lys (SEQ ID NO: 26), and Gly 9 ProSerCysValProLeuMetArgCysGlyGlyCysCysAsn (SEQ ID NO: 34).
  • polypeptide linkers which can be used include a polyglycine of different lengths, including of 5, 7, or 30 residues. Additionally, other portions of Fit- 1 can be used as a linker, for example domain 3 of Fh-I . Sec SEQ ID NO: 15.
  • Linker moieties can also be made from other polymers, such as polyethylene glycol. Such linkers can have from 10 to 1000, 10-500, 10-250, 10-100, or 10-50 ethylene glycol monomer units. Suitable polymers should be of a size similar to the size occupied by the appropriate range of amino acid residues. A typical sized polymer would provide a spacing of from about 10-25 angstroms.
  • Fusion proteins according to the invention can be made by any means known in the art. While such proteins can be made synthetically, or by linking portions which are made, recombinant production can also be used.
  • a fused gene sequence can be produced using the standard tools of recombinant DNA.
  • the fused gene sequence can be inserted into a vector, for example a viral or plasmid vector, for replicating the fused gene sequence.
  • a promoter sequence which is functional in the ultimate recipient cell can be introduced upstream of the fused gene sequence. Promoters used can be constitutive, inducible or repressive. Examples of each type are well-known in the art.
  • the vector can be introduced into a host cell or mammal by any means known in the art.
  • Suitable vectors which can be used include adenovirus, adeno- associaled virus, retrovirus, lentivir ⁇ s, and plasmids. If the vector is in a viral vector and the vector has been packaged, then the virions can be used to infect cells. If naked DNA is used, then transfection or transformation procedures as are appropriate for the particular host cells can be used. Formulations of naked DNA utilizing polymers, liposomes, or nanospheres can be used for fusion gene delivery. Cells which can be transformed or transfected with recombinant constructs according to the invention may be any which are convenient to the artisan. Exemplary cell types which may be used include bacteria, yeast, insects, and mammalian cells.
  • cells of many tissue types may be chosen, as is convenient.
  • Exemplary cells which may be used arc fibroblasts, hepatocytes, endothelial cells, stem cells, hematopoietic cells, epithelial ceils, myocytes, neuronal cells, and keratinocytes. These cells can be used to produce protein in vitro, or can be delivered Io mammals including humans to produce the encoded proteins in vivo. This means of deliver)' is an alternative to delivering nucleic acid to a mammal, delivering viral vector to a mammal, and delivering fusion protein to a mammal
  • compositions of protein or nucleic acids can be in carriers, such as buffers, aqueous or lipophilic carriers, sterile or non-sterile, pyrogenic or non-pyrogenic vehicles.
  • Non-pyrogenic vehicles are useful for injectible formulations.
  • Formulations can be liquid or solid, for example, lyophilized. Formulations can also be administered as aerosols.
  • Compositions may contain one or more fusion proteins or one or more nucleic acids, or both fusion proteins and nucleic acids.
  • the fusion proteins and or nucleic acids in a composition may be homogeneous, in which case homomultimer proteins will form, or they may be heterogeneous in the composition, in which case heteromultimer proteins will form.
  • heteromulrimers typically the X moiety will vary between fusion proteins, but the Z moiety will be the same between fusion proteins.
  • Fusion proteins can be provided to a cell or mammalian host by any means known in the art. Protein can be delivered to the cell or host. Nucleic acid can be administered to the cell or host. Transformed or transfected cells can be administered to the cell or host. In the latter case, cells of the same genetic background are desired to reduce transplantation rejection.
  • Suitable cells for delivery' to mammalian host animals include any mammalian cell type from any organ, tumor, or cell line.
  • human, murine, goat ovine, bovine, dog, cat, and porcine cells can be used.
  • Suitable cell types for use include without limitation, fibroblasts, hepalccytes, endothelial cells, keratinocytes, hematopoietic cells, epithelial cells, myocytes, neuronal cells, and stem cells.
  • Means of delivery of fusion proteins or nucleic acids encoding fusion proteins include delivery of cells expressing the fusion proteins, deliver)' of the fusion proteins, and delivery of nucleic acids encoding the fusion proteins.
  • Fusion proteins, cells, or nucleic acids can be delivered directly to the desired organ or tumor, for example by injection, catheterization, or endoscopy. They can also be delivered intravenously, intrabronchially, intra-tumorally, intrathecally, intramuscularly, intraocularly, topically, subcutaneously, transdermals or per os.
  • Patients who can be effectively treated include those with wet age-related macular degeneration, proliferative diabetic retinopathy, rheumatoid arthritis, osteoarthritis, uveitis, asthma, and cancer.
  • the treatments will improve symptoms and/or markers of disease and/or disease severity.
  • Gene therapy viral vectors e.g., AAV2
  • AAV2 Gene therapy viral vectors
  • a secreted protein when delivered in the anterior or mid-vitreous space of the eye, transduce transitional epithelial cells in the eye (e.g., in non-human primate).
  • Secreted proteins produced from the transduced cells are available in the anterior chamber. Therefore, diseases in the front of the eye can be treated by intravitreal viral gene delivery. Examples of such front eye diseases include, but are not limited to, Dry Eye Syndrome, Sjogren's Syndrome, Uveitis, Corneal ⁇ ngiogenesis, Corneal Graft rejection.
  • secreted proteins include, but are not limited lo, sFLTI, IL-IO, ILlRa, ILl 8bp, soluble PDLl- Ig, CTLA4-Ig, soluble TNFR-Ig, soluble IL-!7R and soluble IL23R.
  • Nucleic acids can be delivered to mammals, and in particular to humans, in any desired vector. These include viral or non-viral vectors, including adenovirus vectors, adeno-associated virus vectors, retrovirus vectors, lentivirus vectors, and plasimd vectors. Exemplary types of viruses include HSV (herpes simplex virus), adenovirus, AAV (adeno associated virus), HIV (human immunodeficiency virus), BIV (bovine immunodeficiency virus), and MLV (murine leukemia virus). Nucleic acids can be administered in any desired format thai provides sufficiently efficient delivery levels, including in virus particles, in liposomes, in nanopartjcles, and complexed to polymers.
  • viral or non-viral vectors including adenovirus vectors, adeno-associated virus vectors, retrovirus vectors, lentivirus vectors, and plasimd vectors. Exemplary types of viruses include HSV (herpes simplex virus), adenovirus,
  • a fusion protein according to the invention can be administered to a patient. If a favorable response is observed, dien a nucleic acid molecule encoding the fusion protein can be administered for a long term effect. Alternatively, the protein and nucleic acid can be administered simultaneously or approximately simultaneously. In another alternative, an antibody or fusion protein for a ligand can be administered followed by or concomitantly with an antibody or fusion partner for a receptor. Another option employs a combination of nucleic acids in which one encodes an antibody and another encodes a fusion protein.
  • Some antibodies that can be employed in combination with the Fh-I constructs of the present invention are bevacizuraab and ranibizumab, both directed to VEGF. These are particularly useful for treating cancer and macular degeneration, respectively.
  • a "reduced amount" of a VEGF antagonist refers to a local concentration (inside a mammal) of the VEGF antagonist.
  • a local concentration is considered a reduced amount when it is lower than the local concentration of a commercially available ' reference VEGF antagonist" for the same disease indication.
  • a “reduced amount” may be 90%, 75%, 50%, 25%. 10% or 5% of the local concentration of a reference VEGF antagonist.
  • Ranibizumab (Luccntis) is a commercially available VEGF antagonist in the form of an anli-VEGF antibody for treating AMD.
  • a reduced amount for this VEGF antagonist is defined as a local concentration that is lower than the local concentration of Ranibizumab after the antibody is administered to the eye. It is estimated that the local concentration of Ranibizumab in the eye is about 100 ⁇ g/ml after a single dose.
  • the VEGF antagonist used to treat AMD according to the present invention is provided at a reduced amount, i.e., a local concentration lower than 100 ⁇ g/ml, e.g., at or lower than 95 ⁇ g/ml, 85 ⁇ g/ml, 75 ⁇ g/ml, 50 ⁇ g/ml, 25 ⁇ g/ml, 15 ⁇ g/ml, 10 ⁇ g/ml, or 5 ⁇ g/ml.
  • the VEGF antagonist of the present invention is a protein, it may be provided by either protein or gene therapy.
  • a "reference VEGF antagonist” refers to a commercially available VEGF antagonist.
  • the reference VEGF antagonist may be in any form as long as it antagonizes VEGF signaling.
  • the reference VEGF antagonist may be a fusion protein or other types of molecules such as VEGF antibodies.
  • a stock or preparation of viral virions e.g., gene therapy virions
  • viral vector particles packaged genomes
  • AAV vector particles packaged genomes
  • the vector particles comprise at least about 75% to 85%, more preferably about 90% of the virions present in the stock, even more preferably at least about 95%, or even 99% or more by weight of the virions present in the stock, or any integer between these ranges.
  • a stock is substantially free of empty capsids when from about 40% to about 1% or less, preferably about 25% to about 15% or less, more preferably about 10% or less, even more preferably about 5% to about 1% or less of the resulting stock comprises empty capsids.
  • Viral virions substantially free of empty capsids may be prepared, e.g., as described U.S. Patent No. 7.261,544, incorporated herein by reference in its entirety. Empty capsids may be measured by methods known in the art, e.g., Progen Capsid Elisa.
  • a gene delivery vehicle is any molecule that can carry inserted polynucleotides into a hosl cell.
  • gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, viruses, such as baculovinis, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosls, and may be used for gene therapy as well as for simple protein expression.
  • Gene deliver is terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene' " ) into a host cell, irrespective of the method used for the introduction.
  • exogenous polynucleotide sometimes referred to as a "transgene' "
  • Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfcction, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation. "gene gun” delivery and various other techniques used for the introduction of polynucleotides).
  • the introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a repHcon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • an extrachromosomal replicon e.g., a plasmid
  • a nuclear or mitochondrial chromosome e.g., a plasmid
  • a number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
  • the exogenous polynucleotide is inserted into a vector such as adenovirus, partially- deleted adenovirus, fully-deleted adenovirus, adeno-associated virus (AAV), retrovirus, lentivirus, naked plasmid, plasmid/liposome complex, etc. for delivery to the host via intravenous, intramuscular, intraportal or other route of administration.
  • Expression vectors which can be used in the methods and compositions of the present invention include, for example, viral vectors.
  • viral vectors One of the most frequently used methods of administration of gene therapy, both in vivo and ex vivo, is the use of viral vectors for delivery of the gene. Many species of virus are known, and many have been studied for gene therapy purposes.
  • the most commonly used viral vectors include those derived from adenoviruses, adeno-associated viruses (AAV) and retroviruses, including Antiviruses, such as human immunodeficiency virus (HIV).
  • Adenovirus is a non-enveloped, nuclear DNA virus with a genome of about 36 kb, which has been well-characterized through studies in classical genetics and molecular biology (Hurwiiz, M.S., Adenoviruses Virology. 3 rd edition, Fields et ai, eds., Raven Press, New York, 1996; Hitt, M.M. et ai. Adenovirus Vectors, The Development of Human Gene Therapy, Friedman, T. ed., Cold Spring Harbor Laboratory Press, New York 1999).
  • the viral genes are classified into early (designated E1-E4) and fate (designated L1-L5) transcriptional units, referring to the generation of two temporal classes of viral proteins.
  • the demarcation of these events is viral DNA replication.
  • the human adenoviruses are divided into numerous serotypes (approximately 47, numbered accordingly and classified into 6 groups: A, B, C, D, E and V), based upon properties including hemaghitination of red blood cells, oncogenicity, DNA and protein amino acid compositions and homologies, and antigenic relationships.
  • Recombinant adenoviral vectors have several advantages for use as gene delivery vehicles, including tropism for both dividing and non-dividing cells, minimal pathogenic potential, ability to replicate to high titer for preparation of vector stocks, and the potential to carry large inserts (Berkner, K.L., Curr. Top. Micro. Immunol. 158:39-66, 1992; Jolly, D., Cancer Genu Therapy 1 :51-64 1994).
  • Adenoviral vectors with deletions of various adenoviral gene sequences such as pseudoadenoviral vectors (PAVs) and partially-deleted adenoviral (termed “DeAd”), have been designed to take advantage of the desirable features of adenovirus which render it a suitable vehicle for delivery of nucleic acids to recipient cells.
  • PAVs pseudoadenoviral vectors
  • DeAd partially-deleted adenoviral
  • pseudoadenoviral vectors also known as 'gutless adenovirus' or mini-adenoviral vectors
  • PAVs are adenoviral vectors derived from the genome of an adenovirus that contain minimal cis-acting nucleotide sequences required for the replication and packaging of the vector genome and which can contain one or more transgenes
  • PAVs have been designed to take advantage of the desirable features of adenovirus which render it a suitable vehicle for gene delivery.
  • adenoviral vectors can generally carry inserts of up to 8kb in size by the deletion of regions which are dispensable for viral growth, maximal carrying capacity can be achieved with the use of adenoviral vectors containing deletions of most viral coding sequences, including PAVs. See U.S. Patent No. 5,882,877 of Gregory- et al; Kochanek et al, Proc. Natl. Acad. ScL USA 93:5731-5736, 1996; Parks et al, Proc. Natl. Acad ScL USA 93:13565-13570, 1996; Lieber et al. J. Virol.
  • PAVs which can accommodate up to about 36 kb of foreign nucleic acid, are advantageous because the carrying capacity of the vector is optimized, while the potential for host immune responses to the vector or the generation of replication- competent viruses is reduced.
  • PAV vectors contain the 5' inverted terminal repeat (ITR) and the 3' ITR nucleotide sequences that contain the origin of replication, and the cis-acting nucleotide sequence required for packaging of the PAV genome, and can accommodate one or more transgenes with appropriate regulatory elements, e.g. promoter, enhancers, etc.
  • adenoviral vectors provide a partially-deleted adenoviral (termed “DeAd ”) vector in which the majority of adenoviral early genes required for virus replication are deleted from the vector and placed within a producer cell chromosome under the control of a conditional promoter.
  • the deletable adenoviral genes that are placed in the producer cell may include £ A/El B, E2.
  • E4 only ORF6 and ORF6/7 need be placed into the cell
  • E3 may also be deleted from the vector, but since it is not required for vector production, it can be omitted from the producer cell.
  • adenoviral late genes normally under the control of the major late promoter (MLP), are present in the vector, but the MLP may be replaced by a conditional promoter.
  • Conditional promoters suitable for use in DeAd vectors and producer cell lines include those with the following characteristics: low basal expression in the uninduced state, such that cytotoxic or cytostatic adenovirus genes are not expressed at levels harmful to the cell; and high level expression in the induced state, such that sufficient amounts of viral proteins are produced to support vector replication and assembly.
  • Preferred conditional promoters suitable for use in DeAd vectors and producer cell lines include the dimerizer gene control system, based on the immunosuppressive agents FK506 and rapamycin, the ecdysone gene control system and the tetracycline gene control system. Also useful in the present invention may be ⁇ he GeoeSwitchTM technology (Valentis, Inc., Woodlands, TX) described in Abruzzese et al.. Hum. Gene Ther. 1999 10:1499-507, the disclosure of which is hereby incorporated herein by reference. The partially deleted adenoviral expression system is further described in WO99-'57296, the disclosure of which is hereby incorporated by reference herein.
  • Adeno-associated virus is a single-stranded human DNA parvovirus whose genome has a size of 4.6 kb.
  • the AAV genome contains two major genes: the rep gene, which codes for the rep proteins (Rep 76, Rep 68, Rep 52, and Rep 40) and the cap gene, which codes for AAV replication, rescue, transcription and integration, while the cap proteins form the AAV viral particle.
  • AAV derives its name from its dependence on an adenovirus or other helper virus (e.g., herpesvirus) to supply essential gene products that allow AAV to undergo a productive infection, i.e., reproduce itself in the host cell.
  • helper virus e.g., herpesvirus
  • AAV integrates as a provirus into the host cell's chromosome, until it is rescued by superinfection of the host cell with a helper virus, usually adenovirus (Muzyczka, Curr. Top. Micor. Immunol 158:97-127, 1992).
  • ITR inverted terminal repeat
  • the integration function of the ITR mediated by the rep protein in trans permits the AAV genome to integrate into a cellular chromosome after infection, in the absence of helper vims.
  • This unique property of the virus has relevance to the use of AAV in gene transfer, as it allows for a integration of a recombinant AAV containing a gene of interest into the cellular genome.
  • This predictability of integration site reduces the danger of random insertional events into the cellular genome that may activate or inactivate host genes or interrupt coding sequences, consequences that can limit the use of vectors whose integration of AAV, removal of this gene in the design of rAAV vectors may result in the altered integration patterns that have been observed with rAAV vectors (Ponnazhagan et al, Hum Gem Ther 8:275-284, 1997).
  • AAV AAV for gene transfer.
  • the host range of AAV is broad.
  • AAV can infect both quiescent and dividing cells.
  • AAV has not been associated with human disease, obviating many of the concerns that have been raised with retrovirus-derived gene transfer vectors.
  • Standard approaches to the generation of recombinant rAAV vectors have required the coordination of a series of intracellular events: transfection of the host cell with an rAAV vector genome containing a transgene of interest flanked by the AAV ITR sequences, transfection of the host cell by a plasmid encoding the genes for the AAV rep and cap proteins which are required in trans, and infection of the transfected cell with a helper virus to supply the non-AAV helper functions required in trans (M ⁇ zyczka, N.. Curr. Top. Micor. Immunol. 158:97-129, 1992).
  • the adenoviral (or other helper virus) proteins activate transcription of the AAV rep gene, and the rep proteins then activate transcription of the AAV cap genes.
  • the cap proteins then utilize the ITR sequences to package the rAAV genome into an rAAV viral particle. Therefore, the efficiency of packaging is determined, in part, by the availability of adequate amounts of the structural proteins, as well as the accessibility of any cis- acting packaging sequences required in the rAAV vector genome.
  • Retrovirus vectors are a common too) for gene delivery (Miller, Nature (1992) 357:455-460).
  • the ability of retrovirus vectors to deliver an unrearranged, single copy gene into a broad range of rodent, primate and human somatic cells makes retroviral vectors well suited for transferring genes to a cell.
  • Retroviruses are RNA viruses wherein the viral genome is RNA.
  • the genomic RNA is reverse transcribed into a DNA intermediate which is integrated very efficiently into the chromosomal DNA of infected cells.
  • This integrated DNA intermediate is referred to as a provirus.
  • Transcription of the provirus and assembly into infectious virus occurs in the presence of an appropriate helper virus or in a cell line containing appropriate sequences enabling encapsidation without coincident production of a contaminating helper virus.
  • a helper virus is not required for the production of the recombinant retrovirus if the sequences for encapsidation are provided by co-transfection with appropriate vectors.
  • the retroviral genome and the proviral DNA have three genes: the gag, the pol, and the env, which are flanked by two long terminal repeat (LTR) sequences.
  • the gag gene encodes the internal structural (matrix, capsid. and nucleocapsid) proteins; the pol gene encodes the RNA-directed DNA polymerase (reverse transcriptase) and the env gene encodes viral envelope glycoproteins.
  • the 5' and 3' LTRs serve to promote transcription and polyadenylation of the virion RNAs.
  • the LTR contains all other cis-acting sequences necessary for viral replication.
  • Lcntiviruses have additional genes including vit vpr, tat, rev, vpu, nef, and vpx (in HTV-L HIV-2 and'or S-V).
  • Adjacent to the 5' LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient encapsidation of viral RNA into particles (the Psi site). If the sequences necessary for encapsidation (or packaging of retroviral RN ⁇ into infectious virions) arc missing from the viral genome, the result is a cis defect which prevents encapsidation of genomic RNA. However, the resulting mutant is still capable of directing the synthesis of all varion proteins.
  • Lentiviruses are complex retroviruses which, in addition to the common retroviral genes gag, pol and env, contain other genes with regulatory or structural function. The higher complexity enables the lentivirus to modulate the life cycle thereof, as in the course of iatent infection.
  • a typical lentivirus is the human immunodeficiency virus (HIV), the etiologic agent of AIDS. In vivo, HIV can infect terminally differentiated cells that rarely divide, such as lymphocytes and macrophages In vitro.
  • HIV human immunodeficiency virus
  • HFV can infect primary cultures of monocyte-derived macrophages (MDM) as well as HeLa-Cd4 or T lymphoid cells arrested in the cell cycle by treatment with aphidicolin or gamma irradiation. Infection of cells is dependent on the active nuclear import of IiIV preintcgratton complexes through the nuclear pores of the target cells. That occurs by the interaction of multiple, partly redundant, molecular determinants in the complex with the nuclear import machinery of the target cell. Identified determinants include a functional nuclear localization signal (NLS) in the gag matrix (MA) protein, the karyophilic virion-associated protein, vpr, and a C-terminal phosphotyrosine residue in the gag MA protein.
  • NLS nuclear localization signal
  • MA gag matrix
  • vpr the karyophilic virion-associated protein
  • C-terminal phosphotyrosine residue in the gag MA protein.
  • retroviruses for gene therapy is described, for example, in United States Patent 6,013,516; and U.S. Patent 5,994,136. the disclosures of which are hereby incorporated herein by reference.
  • Other methods for delivery of DNA to cells do not use viruses for delivery.
  • cationic amphiphilic compounds can be used to deliver the nucleic acid of the present invention. Because compounds designed to facilitate intracellular delivery of biologically active molecules must interact with both non-polar and polar environments (in or on, for example, the plasma membrane, tissue fluids, compartments within the cell, and the biologically active molecular itself), such compounds are designed typically to contain both polar and non-polar domains.
  • araphiphiles Compounds having both such domains may be termed araphiphiles, and many lipids and synthetic lipids that have been disclosed for use in facilitating such intracellular deliver)' (whether for in vitro or in vivo application) meet this definition.
  • One particularly important class of such amphiphiles is the cationic amphiphiles.
  • cationic amphiphiles have polar groups that are capable of being positively charged at or around physiological pH, and this property is understood in the art to be important in defining how the amphiphiles interact with the many types of biologically active (therapeutic) molecules including, for example, negatively charged polynucleotides such as DNA.
  • compositions comprising cationic amphiphilic compounds for gene delivery is described, for example, in United States Patent 5,049,386; US 5,279,833; US 5,650,096; US 5,747,471; US 5,767,099; US 5,910,487; US 5,719,133 ; US 5,840,710; US 5,783,565; US 5,925,628; US 5,912,239; US 5,942,634; US 5,948,925: US 6,022,874; U.S. 5,994,317; U.S. 5,861,397; U.S. 5,952,916; U.S. 5,948,767; U.S. 5,939,401; and U.S. 5,935,936. the disclosures of which are hereby incorporated herein by reference.
  • nucleic acid of the present invention can be delivered using "naked DNA.”
  • Methods for delivering a non-infectious, non-integrating DNA sequence encoding a desired polypeptide or peptide operably linked to a promoter, free from association with transfection-facUitating proteins, viral particles, liposomal formulations, charged lipids and calcium phosphate precipitating agents arc described in U.S. Patent 5.580,859: U.S. 5,963,622; U.S. 5,910,488; the disclosures of which are hereby incorporated herein by reference.
  • adenovirus has been incorporated into the gene delivery systems to take advantage of its e ⁇ dosomolytic properties.
  • the reported combinations of viral and nonviral components generally involve either covalent attachment of the adenovirus to a gene delivery complex or co-internal ization of unbound adenovirus with cationic lipid: DNA complexes.
  • retinopathy of prematurity can be tested in an oxygen-induced retinopathy model in the mouse as described in Smith, Investigative Ophthalmology & Visual Science, 35: 101-111, 1994.
  • Laser-induced choroidal neovascularization in a mouse can be used as a model for human choroidal neovascularization (CNV) occurs in diseases such as age-related macular degeneration. Tobe, American Journal of Pathology 153: 1641-1646. 1998.
  • Other models of CNV have been developed in primates, rals, minipigs, and rabbits.
  • Mouse models of age-related macular degeneration have been developed in genetically-deficient mice. Mice deficient in either monocyte chemoattractant protein- 1 or C-C chemokine receptor-2 develop features of age-related macular degeneration. Ambati, Nature Med. 9: 1390-1397, 2003.
  • a VEGF antagonist of the present invention for the prevention or treatment of disease, the appropriate dosage of a VEGF antagonist of the present invention (protein or gene therapy) will depend on the type of disease to be treated, the severity and course of the disease, whether the VEGF antagonist is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the VEGF antagonist, and the discretion of the attending physician.
  • the VEGF antagonist is suitably administered to the patient at one time or over a series of treatments.
  • the compositions of the present invention are administered in a therapeutically effective or synergistic amount.
  • a therapeutically effective amount is such that co-administration of a VEGF antagonist and one or more other therapeutic agents, or administration of a composition of the present invention, results in reduction or inhibition of the targeting disease or condition.
  • a therapeutically synergistic amount is that amount of a VEGF antagonist and one or more other therapeutic agents necessary to synergistically or significantly reduce or eliminate conditions or symptoms associated with a particular disease.
  • the VEGF antagonist may be administered so that the local concentration provided is about 100 pg/ml to about 100 ⁇ g/ml, 1 ng/ml to about 95 ⁇ g/ml, 10 ng/mt to about 85 ⁇ g/ml, 100 ng/ml to about 75 ⁇ g/ml, from about 100 ng/ml to about 50 ⁇ g/ml, from about 1 ⁇ g/ml to about 25 ⁇ g/ml, from about 1 ⁇ g/ml to about 15 ⁇ g/ml, from about 1 ⁇ g/ml to about 10 ⁇ g/ml, or from about 1 ⁇ g/ml to about 4 ⁇ g/ml.
  • the dose may be from about 2x10 6 to about 2x10 12 , from about 2x 10 7 to about 2x 10 11 drp. or from about 2x10 8 to about 2x10 10 drp per unit dose.
  • the dose may be from about 0.1 mg to about 50 mg, from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.1 mg to about 5 mg, from about 0.1 mg to about 4 mg, from about 0.1 mg to about 3 rag, from about 0.1 mg to about 2.5 mg, from about 0.1 mg to about 2 mg, from about 0.1 mg to about 3.5 mg, from about 0.1 mg to about 1 mg, from about 0.1 mg to about 0.75 mg, from about 0.1 mg to about 0.5 mg, from about 0.1 mg to about 0.25 mg. from about 0.25 mg to about 5 mg, from about 0.5 mg to about 5 mg. from about 1 mg to about 5 mg, from about 1.5 mg to about 5 mg, from about 2 mg to about 5 mg, from about 2.5 mg to about 5 mg, from about 0.5 mg to about 5 mg, or from about 0.5 mg to about 2.5 mg per unit dose.
  • the VEGF antagonist of the present invention may be administered as a single dose or repeatedly. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. The progress of the therapy of the invention is monitored by conventional techniques and assays.
  • an animal or a patient develops an immune response to the VEGF antagonist- one may try to reduce the immune response (inflammation and/or haze or antibodies), e.g., by using available immune suppessors, when necessary.
  • the immune response inflammation and/or haze or antibodies
  • reducing the number of empty viral capsid helps reducing the immune response.
  • a reduction of the immune response against the viral vector helps to increase the expression of the VEGF antagonist (e.g., D29GLYFC), and therefore enhances the therapeutic efficacy of the VEGF antagonist.
  • Example 2 We tested an isolated FIt-I FIt-I Ig-like domain 2 connected to the IgGl Fc region by a flexible poiyglycme 9-tner linker (D2-9Gly-Fc).
  • the D2-9Gly-Fc fusion protein is capable of efficiently binding VEGF and of inhibiting VEGF-dependent human umbilical vein endothelial cell (HUVEC) proliferation. See Fig. 2.
  • FIt-I Ig-like domain 2 is linked directly to the IgGl heavy chain (Fc) to form D2-Fc, only minimal VEGF binding was observed. See Fig. 2.
  • ⁇ n intravitr ⁇ al injection of AAV vector (1 x 10 8 to 1 x 10 9 particles in a volume of 0.0005 ml.) is administered to newborn (PO) or 1 day old (Pl) C57BL/6 mice.
  • Retinal neovascularization (NV) is induced in C57BL/6 mice by exposing P7 pups and their nursing dam to hyperoxia for 5 days. The pups are returned to room air on Pl 2 and are euthanized at Pl 7 (time of peak NV).
  • NV Retinal neovascularization
  • Cohorts of animals treated with the AAV vectors coding for the anti-angiogenic agents are compared to cohorts treated with vectors coding for irrelevant transgenes or with vectors that do not code for a transgene.
  • the average number of endothelial cell nuclei in each treated eye is compared to each animal's untreated fellow eye.
  • VEGF A when present as a dimer, binds to FIt-I through acidic residues (amino acids 63-67 of the mature protein) that allows a possible mechanism for ligand-induced dimerization of receptor (Keyt et al., 1996).
  • a dimerization of domain 2 of FIt-I was used as a strategy to restore the binding of domain 2 of FlM to VEGF A. Fusion with a fragment of IgG heavy chain can be used for dimerization of proteins (Davis-Smyth et al., 1996).
  • amino acids 75-88 i.e., PSCVPLMRCGGCCN; SEQ ID NO: 13
  • V ⁇ GF A SEQ ID NO: 12
  • the third protein, D2-9Gly-Ex3/CH3 contains the polyglycine 9-mer (9GIy) linker and the multimerization domain of VEGF (aa PSCVPLMRCGGCCN; SEQ ID NO: 13; VEGF Ex3 ), but it also contains the CH3 region of human IgGl heavy chain Fc (aa 371-477 of the SEQ ID NO: 10).
  • the protein D2-Fc did not show efficient inhibitory activity in the HUVEC proliferation assay (Fig. 5) and by implication did not bind to VEGF 165 efficiently.
  • the third hybrid protein, D2-9GIy-Ex3/CH3, which comprises domain 2 of Fh-I fused to the CH3 region via both the 9GIy linker and the dimerization region of VEGF 165 (Ex 3) did demonstrate inhibitory activity in a VEGF-dependent HUVECs proliferation assay (Fig. 5). This implies that this hybrid protein binds to VEGF165 efficiently.
  • D2-(GlY 4 Ser) 3 -Fc was further characterized in HUVECs proliferation assay. Biological activity of D2-(Gly 4 Ser)rFc as measured by inhibition of HUVEC proliferation was similar to that of D2-9Gly-Fc and D2-9Gly-Ex3/CH3 (Fig. 6).
  • the D2-(Gly 4 Scr)rFc construct was further characterized by Western blot and compared to D2-9Gly-Fc (Fig.9). Both constructs are present mostly in a dimer form and the monomer forms were detected after separation of reduced samples.
  • the FIt-I ELISA data of conditioned media from transfected 293 cells has shown similar FIt-I levels for D2-9Gly-Ex3/CH3, D2-9Gly-CH3 and D2-Ex3/CH3 and D2- CH3 (70-90 ng/ml) and a little higher (-150 ng/ml) for the least active form of Ul- CH3.
  • Western blot of D2-9Gly-CH3 and D2-CH3 constructs (Fig.10 ) is showing a prevalence of dlmer forms in non-reduced conditions.
  • VEGF binding assay allows us to compare the relative VEGF binding affinities of our soluble VEGF receptors in a cell free system.
  • conditioned media containing known concentrations of soluble receptor (ranging in concentrations from 0.29 - 150 pM) were serially diluted and mixed with 10 pM VEGF. The amount of unbound VEGF was then measured by ELlSA.
  • D2- 9GIy-Fc binds VEGF with higher affinity to bind VEGF at receptor concentrations from 0.001 to - 0.2 pM than all other constructs.
  • D2-CH3 has the lowest affinity to bind VEGF (Fig. 11).
  • the AAV2-SFLT1 D29GLYFC genomic components consist of two AAV2 ITRs and the expression cassette of sFLTlD29GLYFC.
  • a hybrid chicken ⁇ -actin promoter is used for driving the expression of sFLTlD29GLYFC and consists of a CMV IE enhancer, the chicken ⁇ -actin promoter, and a 94 bp fragment of exon 1 that partially belongs to the ⁇ -actin transcript.
  • the hybrid chicken ⁇ -actin/rabbit ⁇ -globin intron is also a part of the CBA promoter, and it contains a number of cryptic transcription factors binding sites.
  • the poly-adenylation region is obtained from the SV40 viral genome.
  • test article AAV2-sFLTH)29GLYFC, and the AAV2 vehicle control, phosphate- buffered saline containing 0.014% Tween ® 20, Lot No. NBR 15045-108, were prepared as a stock frozen liquid in polypropylene vials and a prefo ⁇ n ⁇ lated, aqueous solution, respectively. Solutions of the test and vehicle articles were prepared to yield dosing solutions at appropriate concentrations for achieving the intended doses at the desired dose volumes.
  • AAV2-SFLT1D29GLYFC (2 x 10 8 , 2 x 10 9 , and 2 x 10 10 in 50 ⁇ i dose volume) gave dose-dependent expression of SFLT1D29GLYFC as measured in the aqueous and vitreous fluids and these levels were persistent for at least one year following administration of the AAV vector.
  • Inflammatory changes consisting of lymphocytes or plasma cells in the trabeclar raeshwork and the iridocorneal angle, inflammatory cells in the vitreous, or perivascular lymphocytes ion the retina were observed in some animals (either with AAV2-sFLTlD29GLYFC or with the control virions) with vitreal haze. No tissue destruction or reorganization was observed. The animal with most persistent inflammation had lowest expression of the transgene.
  • Lucentis was administered 11 weeks after AAV2- sFLT1D29GLYFC (doses of 2 x 10 8 , 2 x 10 9 , and 2 x 10 10 ), with no adverse effects observed.

Abstract

Fusion proteins comprising a functional polypeptide, a linker and an IgG heavy chain molecule, and vectors encoding such fusion proteins, are provided for inhibiting angiogenesis in individuals with pathological conditions related to neovascularization. In particular, vectors encoding a fusion VEGF antagonist comprising a VEGF receptor fused to an IgG heavy chain molecule is provided to treat ocular diseases in a mammal.

Description

ANGIOGENESIS INHIBITION
FIELD OF THE INVENTION
The invention relates to compositions and methods useful for inhibiting angiogenesis or treating pathological neovascularization e.g., asthma, arthritis, cancer, and macular degeneration.
BACKGROUND OF THE INVENTION
Pathological neovascularization is a key component of diseases like wet age-related macular degeneration (AMD), proliferative diabetic retinopathy, rheumatoid arthritis, osteoarthritis, and asthma. It also plays an important role in growth and spread of tumors. Neovascularization is regulated by an exquisite balance of pro- and anti- angiogenic factors.
Vascular endothelial growth factor (VEGF) is known to be necessary for neovascularization. Inhibition or reduction of VEGF activity by VEGF antagonists has been shown to inhibit neovascularization in animal models of AMD, arthritis and in various tumor models. VEGF antagonists that inhibit or reduce VEGF activity include, but are not limited to, VEGF antibodies, soluble receptors, receptor fusion proteins, peptides and small molecules.
VEGF-R 1 (Fit- 1 ) and VEGF-R2 (KDR) proteins have been shown to bind VEGF with high affinity. Both FIt-I and KDR have seven Ig-like domains in their extracellular region. Domain 2 has been shown to be essential for VEGF binding. Fusions of each of the full-length, soluble receptor (domains 1-7) and domains 1-3 to IgG Fc bind VEGF efficiently. IgG Fc fusions to Ig-like domain 2 alone was, however, incapable of binding VEGF, as was a combination of Ig-like domain 1 and 2. Davis-Smyth, 1996. Therefore, Ig-like domains 1 and 3 seem to be required along with domain 2 for efficient VEGF binding.
BRIEF SUMMARY OF THE INVENTION
According to one embodiment of the invention a fusion protein is provided. The fusion protein has the formula X-Y-Z. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a 5 - 25 amino acid residue polypeptide. Z is a CH3 region of an IgG heavy chain molecule.
Another embodiment of the invention is a polypeptide of the formula X-Y-Z. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues. Z is a CH3 region of an IgG heavy chain molecule.
Yet another aspect of the invention is a fusion protein of the formula X-Y-Z. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a 5 - 25 amino acid residue polypeptide. Z is an Fc portion of an antibody molecule.
A fusion protein of the formula X-Y-Z is also provided. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues. Z is an Fc portion of an antibody molecule.
Still another aspect of the invention is a method of multimerizing a polypeptide X. A polypeptide X is linked to a polypeptide Z via a polypeptide Y to form polypeptide XYZ. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a 5 - 25 amino acid residue polypeptide. Z is a CH3 region of an IgG heavy chain molecule. Polypeptide XYZ which is formed multimerizes.
Yet another embodiment of the invention provides a method of mυltimerizing a polypeptide X. Polypeptide X is linked to a polypeptide Z via a moiety Y to form polymer XYZ. X comprises a polypeptide selected from the group consisting of an extracellular receptor, an antibody variable region, a cytokine, a chemokine, and a growth factor. Y consists essentially of a linker moiety which provides the spatial separation of 5-25 amino acid residues. Z is a CH3 region of an IgG heavy chain molecule. Polypeptide XYZ which is so formed multimerizes.
In one embodiment of the invention a nucleic acid molecule is provided. The nucleic acid molecule encodes a fusion protein which comprises an lg-like domain 2 of VHGF-R 1 (FIt-I); a linker: and a multimerization domain. The fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
In another embodiment of the invention a fusion protein is provided. The fusion protein comprises an Ig-like domain 2 of VEGF-Rl (FIt-I), a linker, and a multimerization domain. The fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25. In another embodiment of the invention an in vitro method is provided. A nucleic acid molecule is delivered to an isolated mammalian cell. The nucleic acid molecule encodes a fusion protein which comprises an Ig-like domain 2 of VEGF-Rl (FIt-I; a linker; and a multimerization domain. The fusion protein comprises a sequence selected from the group consisting of SEQ D NO: 2, 8, 21, 23, and 25. Expression of the fusion protein is controlled by a promoter. A cell is formed which expresses a fusion protein.
Still another embodiment of the invention is a method for delivering a fusion protein to a mammal. A mammalian cell which expresses the fusion protein is delivered to a mammal. The cell expresses and secretes the fusion protein thereby supplying the fusion protein to the mammal. The fusion protein comprises an Ig-like domain 2 of VEGF-Rl (FIt-I). a linker, and a rnultimerization domain. The fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25.
Another aspect of the invention is a method for supplying a fusion protein to a mammal. A fusion protein which comprises an Ig-like domain 2 of VEGF-RI (FIt-I), a linker, and a raultimerization domain is delivered to a mammal The fusion protein comprises a sequence selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25. Alternatively, a nucleic acid construct which encodes said fusion protein can be delivered to the mammal, whereby the fusion protein is expressed by the mammal.
Another aspect of the invention is an improved method of treating an ocular disease in a mammal with a VEGF antagonist, including but not limited to, the fusion proteins of the present invention. The improvement comprises providing a reduced amount of the VEGF antagonist that is therapeutically effective to treat the ocular disease. The VEGF antagonist may be administered by protein or gene therapy, or the combination thereof. The reduced amount may be maintained in the mammal for at least 6 months, e.g.. at least 9 months or at least i year.
In some embodiments of the present invention, the reduced amount may be from about 100 pg/ml to about 100 μg/ml, 1 ng/ml to about 95 μg/ml, 10 ng/ml to about 85 μg/m), 100 ng/ml to about 75 μg/rnl, from about 100 ng/ml to about 50 μg/ml, from about 1 μg/ml to about 25 μg/ml, from about 1 μg/ml to about 15 μg/ml. from about 1 μg/ml to about 10 μg/ml, or from about 1 μg/ml to about 4 μg/ml.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to the mammal a nucleic acid encoding a VEGF antagonist. In some embodiments, the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 pg/ral to about 100 μg/ml, from about 1 ng/ml to about 95 μg/ml, from about 10 ng/ml to about 85 μg/ml, from about 100 ng/m! to about 75 μg/ml, from about 100 ng/mi to about 50 μg/ml, from about 1 μg/ml to about 25 μg/ml, from about 1 μg/ml to about 15 μg/rnl, from about 1 μg/ml to about 10 μg/ml, or from about 1 μg/ml to about 4 μg/ml to treat the ocular disease.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to the mammal a viral vector encoding a VEGF antagonist at a therapeutically effective amount of from about 2x 106 to about 2x 1012 drp per unit dose.
In some embodiments, the therapeutically effective amount is from about 2x107 to about 2x 1011 drp, from about 2x10* to about 2x1010 drp per unit dose.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to the mammal a soluble VEGF receptor or a variant at a therapeutically effective amount of from about 0.1 mg to about 50 mg per unit dose, preferably from about 0.1 mg to about 5 mg, e.g., from about 0.1 mg to about 1 mg, from about 0.1 mg to about 0.5 mg and from about 0.1 mg to about 0.25 mg. The soluble VEGF receptor or its variant includes, but is not limited to, the fusion proteins of the present invention.
In some embodiments, the soluble VEGF receptor is FLTl D29GLYFC (SEQ ID NO. 1 or 2) or D2-9GLY-CH3 (SEQ [D NO. 22 or 23).
In some embodiments, the therapeutically effective amount of the soluble VEGF receptor is from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.5 mg to about 5 rag, or from about 0.5 mg to about 2.5 mg. The soluble VEGF receptor or its variant may be administered repeatedly.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to a diseased eye υf the mammal a nucleic acid encoding a VEGF antagonist of a fusion protein of the invention, where the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 ng/ml to about 75 μg/ml to treat the ocular disease.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises delivering Io a diseased eye of the mammal a nucleic acid encoding a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is expressed in transitional epithelial cells of the diseased eye at a therapeutically effective amount of from about 100 ng/ml to about 75 μg/ml.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises delivering to a diseased eye of the mammal by intravitreal injection (e.g., mid vitreal injection) a nucleic acid encoding a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is expressed at a therapeutically effective amount of from about 100 ng/ml to about 75 μg/ml.
In some embodiments, the VEOF antagonist is expressed at a therapeutically effective amount of from about 100 pg/nil to about 100 μg/ml, 1 ng/ml to about 95 μg/ml, 10 ng/mi to about 85 μg/ml 100 ng/ml to about 50 μg/ml, from about 1 μg/ml to about 25 μg/ml, from about I μg/ml to about 15 μg/rnl, from about 1 μg/ml to about 10 μg/ml, or from about 1 μg/ml to about 4 μg/ml.
In one embodiment, the nucleic acid encoding a VEGF antagonist is a viral vector. In another embodiment, the viral vector is AAV, e.g., AA V2.
In one embodiment, the VEGF antagonist is expressed in the mammal for at least 6 months, e.g., at least 9 months or 1 year.
Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to a diseased eye of the mammal a VBGF antagonist of the formula X-Y-Z at a therapeutically effective amount of from about 2x 106 to about 2x 1012 drp per unit dose.
In some embodiments, the therapeutically effective amount is from about 2x107 to about 2x 1 θ" drp, or from about 2x 10* to about 2x 1010 dip per unit dose.
In some embodiment, a second VIiGF antagonist is administered. The second VEGF antagonist may be a fusion polypeptide comprising a VEGF receptor fused to an IgG heavy chain molecule, preferably a VEGF-Rl fused to the Fc or CH3 domain of IgG, more preferably, domain 2 of VEG-Rl fused to Fc or CH3. A linker, e.g., 9GIy, may be used to link the VEGF receptor and the IgG domain.
The second VEGF antagonist may be a VEGF antibody, e.g., bevacizumab or ranibizumab. Another aspect of the invention is a method of treating an ocular disease in a mammal. The method comprises administering to a diseased eye of the mammal a VEGF antagonist of the formula X-Y-Z, wherein the VEGF antagonist is at a therapeutically effective amount of from about 0.1 mg to about 50 mg per unit dose.
Tn some embodiment the therapeutically effective amount is from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.5 mg to about 5 mg, or from about 0.5 mg to about 2.5 mg.
Another aspect of the invention is a method of reducing haze in the eye in a mammal receiving gene therapy for the eye. The method comprises administering TO the mammal (e.g., in the eye) gene therapy virions containing empty capsids not exceeding 90% of the total capsids.
In some embodiments, the empty capsids do not exceed 80%, 70%, 60%, or 50% of the total capsids. In one embodiment, the gene therapy virions are substantially free of empty capsids.
In one embodiment, the virions are AAV virions, e.g., AA V2 virions.
In some embodiments, the ocular disease is selected from the group consisting of wet age-related macular degeneration (wet AMD), macular edema, retinal vein occlusions, retinopathy of prematurity, and diabetic retinopathy.
These and other embodiments of the invention which will be described in more detail below provide the art with methods and agents for treating disease related to vascular proliferation and inflammation. The agents may provide increased stability and bioavailability relative to natural forms of the proteins.
BRIEF DESCRIPTION OF THE DRA WINGS Fig. 1. Flexible region of 9-Gly linker in D2-9Gly-Fc construct. The predicted relative flexibility by Karpus and Schultz (19S5) metbod shows the polyglycine 9-meτ (9-Gly) linker (aa 94 to 103) in D2-9Gly-Fc protein as a region with greater flexibility than the average (>1) as compared to D2-Fc construct that does not contain 9-Gly linker. Both fusion proteins contain identical amino acid sequences enclosed in boxes: sp - signal peptide (aa -24 to -1), FIt-I domain 2 (aa 1 to 93) and IgGl-Fc residues (244 aa). The arrow represents the signal peptidase cleavage site as predicted using the SignaiP V2.0 program (Nielsen et a!.. 1997).
Fig. 2. Biological activity of D2-9Gly-Fc vs. D2-Fc. 293 cells were grown in the starvation media (M 199 + 5% FCS) and transfected with plasmids containing D2- 9GIy-Fc and D2-Fc expression cassettes under control of CMV promoter. Conditioned media (CM) was collected 72 h later. HUVECs were seeded into 96 well plate (2E3 cells/well) in starvation media * VEGF (10 ng/mL) and 50 ul CM plus VEGF (10 ng/mL) was added 24 h later. The controls (+/- VEGF) were incubated with CM from the control pEGFP (Clontech; pEGFP carries a red-shifted variant of wild-type green fluorescent protein (GFP) which has been optimized for brighter fluorescence and higher expression in mammalian cells) plasmid transfection. The positive control was treated with 50 ng of Mt-I-IgG recombinant protein (R & D Systems). The HUVECs were assayed for proliferation 3 days post treatment using CellTiter 96® AQueous reagent (Promega). The data represent the means of the average values of OD490 of two experiments each assayed in triplicates.
Fig. 3. Western blot analysis of D2-9Gly-Fc and D2-Fc. The size of both D2-9Gly-Fc and D2-Fcproteins appears to be twice as large while migrating in non-reducing gel as compared to migration in reducing gel. The proteins were loaded from the conditioned media following 293 cell transfection of plasmids expressing D2-9Gly-Fc and D2-Fc were separated by SDS-electrophoresis and transferred to PVDF membrane. The blot was probed with goat anti-human anti-lgGl Fc and rabbit anti- goat IgG-HRP antibodies.
Fig. 4. sFlt-l hybrid proteins containing 9Gly linker and VEGF Ex3. Structure comparison of D2-9Gly-Ex3/CH3 to previously constructed proteins. All three proteins contain identical amino acid sequence of FIt-I domain 2, consisting of 24 aa of FIt-I signal peptide and 93 aa of FU-I domain 2. D2-9Gly-Ex3/CH3 contains 9 aa of 9GIy linker, 14 aa of VEGF Ex3 and 120 aa of the CH3 region of human IgGl heavy chain Fc.
Fig. 5. Biological activity of D2-9Gly-Ex3/CH3 vs. D2-9Gly-Fc. Protein D2-9Gly- Ex3/CH3, where domain 2 is connected to the CH3 region through 9GIy linker and VEGF Ex 3, is also efficiently inhibiting VEGF-dependent HUVECs proliferation as compared to control proteins D2-9Gly-Fc and D2-Fc. 50 ng of the recombinant FIt-I-IgG (R&D Systems) was used as a control.
Fig. 6. HUVECs proliferation assay comparing D2-{GIy4Ser)3-Fc protein activity with D2-9Gly-Fc and D2-9Gly-Ex3/CH3.
Fig. 7. Western blot. Proteins (non-reduced and reduced) from conditioned medium of transfected 293 cells (15 ul of CM) with plasmids expressing (1): D2-9Gly-Fc; (2): D2-(CΪ4S)J-FC and (3) - EGFP proteins were separated by SDS-eiectrophoresis and transferred to PVDF membrane. The blot was probed with goat anti-human IgG1 Fc and rabbit anti-goat IgG-FIRP antibodies.
Fig. 8. Combinations of proteins with/without 9GIy linker or VEGF Ex3. Structure comparison of three novel proteins wilh or without 9GIy linker and/or VEGF Ex3, D2-9GIy-CΗ3. D2-CH3 and D2-Ex3/CH3.
Fig. 9. HUVECs proliferation assay with the FIt-I (D2) constructs with 9GIy, Ex3 and CH3 combinations. Conditioned media from 293 cells (5 ul) containing proteins D2- Ex3/CH3, D2-9Gly-CI 13 and D2-CH3 were compared to D2-9Gly-Fc and D2-9Gly-
Ex3/CH3.
Fig. 10 Western blot. 293 cells were transfected with plasmids expressing: (1) D2- 9GIy-Fc; (2) D2-9Gly-CH3 (52/26 kDa); and (3) D2-CH3 (50/25 kDa). Proteins from 293 cells conditioned medium (15 ul of CM non-reduced and/or reduced) were separated by SDS-electrophoresis and transferred to PVDF membrane. The blot was probed with anti-human VEGF-Rl HRP conjugate (R&D Systems).
Fig. 11. VEGF "in vitro" binding assay. Conditioned media from 293 cells containing known concentrations of both D2-9Gly-Fc and FIt-I D(I -3) control soluble receptors (ranging in concentrations from 0.29 - 150 pM) were serially diluted and mixed with IO pM VCGF. The amount of unbound VEGF was then measured by ELISA. D2- 9GIy-Fc binds VEGF with higher affinity than all other constructs, "n" represents the number of independent experiments (transfection and binding assay).
Fig. 12. The binding kinetics of the soluble FIt-I constructs were measured by surface plasmon resonance with a BIΛcore instrument. sFlt-1 constructs were immobilized onto a sensor chip, and VEGF165 was injected at concentrations ranging from 0.2 to 15 nM. The sensorgrams were evaluated using the BlA Evaluation program, the rate constants Ka and Kd were determined and the dissociation constant (KD) calculated from the ratio of Kd/Ka = KD. The lower KD value means better affinity.
Fig. 13A-13C. Fig 13A shows expression levels of FIt-I constructs having various linkers. Fig. 13B shows dimerization or multimerization of FIt-I constructs having various linkers and a CH3 moiety of Fc of IgGl . The difference between the non- reduced and the reduced conditions indicates that the proteins had multimerized. Fig. 13C shows the inhibitory bioactivity of indicated FIt-I constructs present in condition medium in a HUVEC proliferation assay in the presence of VEGF. Each of the constructs demonstrated inhibitory activity approaching proliferation levels of the HUVEC in the absence of VEGF.
Fig. 14. Using a murine oxygen-induced retinopathy (OIR) model of retinal neovascularization (NV). one of the FIt-I constructs was administered to the mouse eyes and neovascularization was determined. The mice were exposed to hyperoxic conditions. The number of neovascular events was determined in the treated eyes compared to the events in the untreated eyes of the same animals. The animal was considered a "responder" if there was a greater than 50% reduction in neovascular events.
Fig. 15. An in situ hybridization assay shows the expression levels and location of AAV2-sFLTI D29GLYFC in a group of animals.
DETAILED DESCRIPTION OF THE INVENTION
Il is a discovery of the present inventors that a FH-I Ig-like domain 2 without domains 1 and 3 is capable of efficiently binding VEGF and inhibiting VEGF-dependent endothelial cell proliferation. Domain 2 can be covalently (inked to a muitimerization domain via a linker. Linkers are typically polypeptide chains. The length of the chain may be 6, 7, 9, 11. 13, 15 or more amino acid residues, but typically is between 5 and 25 residues. Depending upon the length and side chain composition, a linker may have but need not have greater than average flexibility. Flexibility can be calculated using algorithms known in the art. Multimerization domains are those portions of multimeric proteins which promote the association of subunits to form, for example, dimers, trimers, tctramers, etc. Suitable recombinant proteins for efficiently binding VEGF and/or inhibiting VEGF-dependent endothelial cell proliferation are selected from the group consisting of SEQ ID NO: 2, 8, 21, 23, and 25. Moreover, the inventors have found lhat the multimerization domains and linkers can be used with a variety of other proteins or portions of proteins to induce multimerization. Such proteins may be those which bind to Iigand or receptor only when multimcrized, or may be those whose binding affinity is enhanced when multiraerized. Suitable proteins for muJtimerization include extracellular receptors (which include portions thereof), antibody variable regions, cytokines, chemokines, and growth factors. Suitable proteins include tyrosine kinase receptors and serine thereonine kinase receptors. Specific examples of extracellular receptors include EGF-receptor, G protein coupled receptors, FGF receptor, Fc receptors, T cell receptors, etc. Examples of antibody variable regions include Fab, F(ab')2, and ScFv. Examples of cytokines include GM-CSF, IL-lα, IL-1 β, IL-2, IL-3, IL-4, IL-5. IL-6, IL-7, IL-8, JL-10, IL-12, IL-18, 1L-21, IL-23, IFN-α, IFN-β, IFN-γ, MIP- lα, MlP-lβ. TGF-β, TNFo, and TNF-β. Examples of chemokines include BCA-I / BLC, BRAK, Chemokine CC-2, CTACK, CXCL-16, ELC. ENA, ENA-70, ENA-74 , ENA-78, Eotaxin, Exodus-2, Fractalkine, GCP-2, GRO, GRO alpha (MGSA), GRO-beta. GRO-gamma, HCC-I, HCC-4, 1-309, IP-IO, I-TAC, LAG-I , LD78-beta, LEC / NCC-4, LL-37, Lymphotactin, MCP. MCAF (MCP-I), MCP-2, MCP-3, MCP-4, MDC, MDC, MDC-2, MDC-4, MEC / CCL28, MIG, MIP, MIP-I alpha, MlP-I beta, MIP-I delta, MIP-3 / MPIF-I, MIP-3 alpha, MIP-3 bet , MIP-4 (PARC). MIP-5, NAP-2 , PARC , PF-4, RANTES, RANTES-2, SDF-I alpha, SDF-I beta, TARC, and TECK. Examples of growth factors include Human Amphiregulin, Human Angiogenesis Proteins, Human ACE, Human Angiogenin, Human Angiopoietin, Human Angiostatin, Human Betacelluiin, Human BMP, Human BMP-13 / CDMP-2, Human BMP-14 /CDMP-I, Human BMP-2, Human BMP-3, Human BMP-4, Human BMP-5, Human BMP-6, Human BMP-7, Human BMP-8, Human BMP-9, Human Colony Stimulating Factors, Human fU3-Ligand, Human G- CSF, Human GM-CSF, Human M-CSF, Human Connective Tissue Growth Factor, Human Cripto-1, Human Cryptic, Human ECGF, Human EGF, Human EG-VEGF, Human Erythropoietin, Human Fetuin, Human FOF, Human FGF-I, Human FGF- 10, Human FGF-16, Human FGF-17, Human FGF-IS, Human FCF- 19, Human FGF- 2, Human FGP-20, Human FGF-3, Human FGF-4, Human FGF-5, Human FGF-6, Human FGF-7 / KGF, Human FGF-8, Human FGF-9, Human FGF-acidic, Human FGF-basic, Human GDF-I K Human GDF- 15, Human Growth Hormone Releasing Factor, Human HB-EGF, Human Heregulin, Human HGF, Human 3GF, Human IGF-I, Human IGF-II, Human Inhibin, Human KGF, Human LCGF, Human LIF, Human Miscellaneous Growth Factors, Human MSP, Human Myostatin, Human Myostatin Propeptide, Human Nerve Growth Factor, Human Oncostatin M, Human PD-ECGF, Human PDGF, Human PDGF (AA Homodimer), Human PDGF (AB Heterodimer), Human PDGF (BB Homodimer), Human PDGF (CC Homodimer), Human PIGF. Human PlGF. Human PIGF-L Human PIGF-2, Human SCF, Human SMDF, Human Stem Cell Growth Factor, Human SCGF-alpha, Human SCGF-beta, Human Thrombopoietin, Human Transforming Growth Factor, Human TGF-alpha, Human TGF-beta, and Human VEGF.
FIt-I receptor protein has an extracellular portion which comprises seven Ig-like domains. These are located at residue numbers 32...123, 153...214, 230...327, 335...421, 428...553, S56...654, and 661...747 of Genbank accession no. P17948, see also SEQ ID NO: 15. Residue numbers 3-26 comprise a signal sequence. FIt-I protein is encoded by the DNA sequence shown at Genbank accession no. NM_002019 (SEQ ID NO: 14).
Multimerizaύon domains can be used as are known in the art. Sequences of the Fc portion of IgGl or IgG2 lambda heavy chain can be used, for example, CH3 alone (aa 371-477) or both of CH2 and CH3 domains (aa 247-477). Fc portion of Ig molecules is that which is obtained by cleavage of whole antibody molecules with the enzyme papain. Other means can be used to obtain these portions. For the IgGl lambda heavy chain protein sequence, see Genbank accession no Y14737and SEQ ID NO: 10. Other Fc regions can be used for example from other IgG types and from IgA, IgM, IgD, or IgE antibodies. The multimcrization region of VEGF can also be used. A DNA sequence encoding VHGF is shown at Genbank accession no. NM_003376and SEQ ID NO: 11. An amino acid sequence of VEGF is shown at Genbank accession no. CAC19513and SEQ ID NO; 12. The raultimerizalion region of VEGF (SEQ ID NO: 13), encoded by VEGF exon 3 (VEGF Ex3), is at about amino acid residues 75- 88 of VEGF protein (SEQ ID NO: 12). Multiraerization domains will cause at least 5 %, 10 %, 20 %, 30 %, 40 %, 50 % 60 %, 75 %, 80 %, 85 %, 90 %, or 95 % of the monomelic fusion proteins to migrate on a non-denaturing polyacrylamidc gel at a rate appropriate for a multimer. Glycosylation can affect the migration of a protein in a gel. Although particular sequences are shown here, variants such as allelic variants can be used as well. Typically such variants will have at least 85 %, 90 %, 95 %, 97 %, 98 %, or 99 % identity with the disclosed sequence.
Multimerizaticn can be assayed, for example, using reducing and non-reducing geis, as demonstrated herein. Multiraerization can also be assayed by detection of increased binding affinity of a protein for its ligand/receptor. BiaCore™ surface plasmon resonance assays can be used in this regard. These assays detect changes in mass by measuring changes in refractive index in an aqueous layer close to a sensor chip surface. Any method known in the art can be used to detect multiraerization.
Linker moieties according to the invention can be comprised of for example 5-100 amino acid residues, 5-75 amino acid residues, 5-50 amino acid residues, 5-25 amino acid residues, 5-20 amino acid residues, 5-15 amino acid residues, 5-10 amino acid residues, 5-9 amino acid residues. Examples of useful linkers include: gly9 (SEQ ID NO: 27), gMSEQ ID NO: 28), ser9(SEQ ID NO: 29), gly5cyspro2cys (SEQ ID NO: 30), (gly4ser)3 (SEQ ID NO: 31), SerCysValProLeuMetArgCysGlyGlyCysCysAsn (SEQ ID NO: 32), Pro Ser Cys VaI Pro Leu Met Aig Cys GIy GIy Cys Cys Asn (SEQ ID NO: 13), GIy Asp Leu lie TyT Arg Asn GIn Lys (SEQ ID NO: 26), and Gly9ProSerCysValProLeuMetArgCysGlyGlyCysCysAsn (SEQ ID NO: 34). Other polypeptide linkers which can be used include a polyglycine of different lengths, including of 5, 7, or 30 residues. Additionally, other portions of Fit- 1 can be used as a linker, for example domain 3 of Fh-I . Sec SEQ ID NO: 15. Linker moieties can also be made from other polymers, such as polyethylene glycol. Such linkers can have from 10 to 1000, 10-500, 10-250, 10-100, or 10-50 ethylene glycol monomer units. Suitable polymers should be of a size similar to the size occupied by the appropriate range of amino acid residues. A typical sized polymer would provide a spacing of from about 10-25 angstroms.
Fusion proteins according to the invention can be made by any means known in the art. While such proteins can be made synthetically, or by linking portions which are made, recombinant production can also be used. A fused gene sequence can be produced using the standard tools of recombinant DNA. The fused gene sequence can be inserted into a vector, for example a viral or plasmid vector, for replicating the fused gene sequence. A promoter sequence which is functional in the ultimate recipient cell can be introduced upstream of the fused gene sequence. Promoters used can be constitutive, inducible or repressive. Examples of each type are well-known in the art. The vector can be introduced into a host cell or mammal by any means known in the art. Suitable vectors which can be used include adenovirus, adeno- associaled virus, retrovirus, lentivirυs, and plasmids. If the vector is in a viral vector and the vector has been packaged, then the virions can be used to infect cells. If naked DNA is used, then transfection or transformation procedures as are appropriate for the particular host cells can be used. Formulations of naked DNA utilizing polymers, liposomes, or nanospheres can be used for fusion gene delivery. Cells which can be transformed or transfected with recombinant constructs according to the invention may be any which are convenient to the artisan. Exemplary cell types which may be used include bacteria, yeast, insects, and mammalian cells. Among mammalian cells, cells of many tissue types may be chosen, as is convenient. Exemplary cells which may be used arc fibroblasts, hepatocytes, endothelial cells, stem cells, hematopoietic cells, epithelial ceils, myocytes, neuronal cells, and keratinocytes. These cells can be used to produce protein in vitro, or can be delivered Io mammals including humans to produce the encoded proteins in vivo. This means of deliver)' is an alternative to delivering nucleic acid to a mammal, delivering viral vector to a mammal, and delivering fusion protein to a mammal
Compositions of protein or nucleic acids can be in carriers, such as buffers, aqueous or lipophilic carriers, sterile or non-sterile, pyrogenic or non-pyrogenic vehicles. Non-pyrogenic vehicles are useful for injectible formulations. Formulations can be liquid or solid, for example, lyophilized. Formulations can also be administered as aerosols. Compositions may contain one or more fusion proteins or one or more nucleic acids, or both fusion proteins and nucleic acids. The fusion proteins and or nucleic acids in a composition may be homogeneous, in which case homomultimer proteins will form, or they may be heterogeneous in the composition, in which case heteromultimer proteins will form. In the case of heteromulrimers, typically the X moiety will vary between fusion proteins, but the Z moiety will be the same between fusion proteins.
Fusion proteins can be provided to a cell or mammalian host by any means known in the art. Protein can be delivered to the cell or host. Nucleic acid can be administered to the cell or host. Transformed or transfected cells can be administered to the cell or host. In the latter case, cells of the same genetic background are desired to reduce transplantation rejection.
Suitable cells for delivery' to mammalian host animals include any mammalian cell type from any organ, tumor, or cell line. For example, human, murine, goat ovine, bovine, dog, cat, and porcine cells can be used. Suitable cell types for use include without limitation, fibroblasts, hepalccytes, endothelial cells, keratinocytes, hematopoietic cells, epithelial cells, myocytes, neuronal cells, and stem cells.
Means of delivery of fusion proteins or nucleic acids encoding fusion proteins include delivery of cells expressing the fusion proteins, deliver)' of the fusion proteins, and delivery of nucleic acids encoding the fusion proteins. Fusion proteins, cells, or nucleic acids can be delivered directly to the desired organ or tumor, for example by injection, catheterization, or endoscopy. They can also be delivered intravenously, intrabronchially, intra-tumorally, intrathecally, intramuscularly, intraocularly, topically, subcutaneously, transdermals or per os. Patients who can be effectively treated include those with wet age-related macular degeneration, proliferative diabetic retinopathy, rheumatoid arthritis, osteoarthritis, uveitis, asthma, and cancer. The treatments will improve symptoms and/or markers of disease and/or disease severity.
Gene therapy viral vectors (e.g., AAV2) encoding a secreted protein, when delivered in the anterior or mid-vitreous space of the eye, transduce transitional epithelial cells in the eye (e.g., in non-human primate). Secreted proteins produced from the transduced cells are available in the anterior chamber. Therefore, diseases in the front of the eye can be treated by intravitreal viral gene delivery. Examples of such front eye diseases include, but are not limited to, Dry Eye Syndrome, Sjogren's Syndrome, Uveitis, Corneal Λngiogenesis, Corneal Graft rejection. Examples of secreted proteins include, but are not limited lo, sFLTI, IL-IO, ILlRa, ILl 8bp, soluble PDLl- Ig, CTLA4-Ig, soluble TNFR-Ig, soluble IL-!7R and soluble IL23R.
Nucleic acids can be delivered to mammals, and in particular to humans, in any desired vector. These include viral or non-viral vectors, including adenovirus vectors, adeno-associated virus vectors, retrovirus vectors, lentivirus vectors, and plasimd vectors. Exemplary types of viruses include HSV (herpes simplex virus), adenovirus, AAV (adeno associated virus), HIV (human immunodeficiency virus), BIV (bovine immunodeficiency virus), and MLV (murine leukemia virus). Nucleic acids can be administered in any desired format thai provides sufficiently efficient delivery levels, including in virus particles, in liposomes, in nanopartjcles, and complexed to polymers.
Combinations of protein and nucleic acid treatments can be used. For example, a fusion protein according to the invention can be administered to a patient. If a favorable response is observed, dien a nucleic acid molecule encoding the fusion protein can be administered for a long term effect. Alternatively, the protein and nucleic acid can be administered simultaneously or approximately simultaneously. In another alternative, an antibody or fusion protein for a ligand can be administered followed by or concomitantly with an antibody or fusion partner for a receptor. Another option employs a combination of nucleic acids in which one encodes an antibody and another encodes a fusion protein. Some antibodies that can be employed in combination with the Fh-I constructs of the present invention (whether in the protein or nucleic acid form) are bevacizuraab and ranibizumab, both directed to VEGF. These are particularly useful for treating cancer and macular degeneration, respectively.
The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, Second Edition (Sambrook et al., 1989); Current Protocols In Molecular Biology (F.M. Ausubel et al., cds., 1987); Oligonucleotide Synthesis (MJ. Gait ed, 1984); Animal Cell Culture (RJ. Freshney, ed., 1987). Methods In Enzymolυgy (Academic Press, Inc.); Handbook Of Experimental Immunology (DM. Wei & CC. Blackwell, eds.); Gene Transfer Vectors For Mammalian Cells (J. M. Miller & M.P Calos. eds., 1987); PCR: The Polymerase Cftain Reaction, (Mullis et al., eds., 1994); Current Protocols In Immunology (J .E. Coligan ct al., eds., 1991 ); Antibodies: A Laboratory Manual (F.. Harlow and D. Lane eds. (1988)); and PCR 2: A Practical Approach (MJ. MacPherson, B.D. Hames and G.R. Taylor eds. (1995)).
As used herein, a "reduced amount" of a VEGF antagonist, according to the present invention, refers to a local concentration (inside a mammal) of the VEGF antagonist. A local concentration is considered a reduced amount when it is lower than the local concentration of a commercially available ' reference VEGF antagonist" for the same disease indication. For example, a "reduced amount" may be 90%, 75%, 50%, 25%. 10% or 5% of the local concentration of a reference VEGF antagonist. As an example, Ranibizumab (Luccntis) is a commercially available VEGF antagonist in the form of an anli-VEGF antibody for treating AMD. When a VEGF antagonist is used to treat AMD according to the present invention, a reduced amount for this VEGF antagonist is defined as a local concentration that is lower than the local concentration of Ranibizumab after the antibody is administered to the eye. It is estimated that the local concentration of Ranibizumab in the eye is about 100 μg/ml after a single dose. Accordingly, the VEGF antagonist used to treat AMD according to the present invention is provided at a reduced amount, i.e., a local concentration lower than 100 μg/ml, e.g., at or lower than 95 μg/ml, 85 μg/ml, 75 μg/ml, 50 μg/ml, 25 μg/ml, 15 μg/ml, 10 μg/ml, or 5 μg/ml. Where the VEGF antagonist of the present invention is a protein, it may be provided by either protein or gene therapy.
As used herein, a "reference VEGF antagonist" refers to a commercially available VEGF antagonist. The reference VEGF antagonist may be in any form as long as it antagonizes VEGF signaling. For example, the reference VEGF antagonist may be a fusion protein or other types of molecules such as VEGF antibodies. A stock or preparation of viral virions (e.g., gene therapy virions) comprising viral vector particles (packaged genomes) is "substantially free of empty capsids when at least about 50%-99% or more of the virions present in the stock are virions with packaged genomes (i.e., AAV vector particles). Preferably, the vector particles comprise at least about 75% to 85%, more preferably about 90% of the virions present in the stock, even more preferably at least about 95%, or even 99% or more by weight of the virions present in the stock, or any integer between these ranges. Thus, a stock is substantially free of empty capsids when from about 40% to about 1% or less, preferably about 25% to about 15% or less, more preferably about 10% or less, even more preferably about 5% to about 1% or less of the resulting stock comprises empty capsids. Viral virions substantially free of empty capsids may be prepared, e.g., as described U.S. Patent No. 7.261,544, incorporated herein by reference in its entirety. Empty capsids may be measured by methods known in the art, e.g., Progen Capsid Elisa.
A gene delivery vehicle is any molecule that can carry inserted polynucleotides into a hosl cell. Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, viruses, such as baculovinis, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosls, and may be used for gene therapy as well as for simple protein expression.
Gene deliver)', gene transfer and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene'") into a host cell, irrespective of the method used for the introduction. Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfcction, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation. "gene gun" delivery and various other techniques used for the introduction of polynucleotides). The introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a repHcon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome. A number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
The exogenous polynucleotide is inserted into a vector such as adenovirus, partially- deleted adenovirus, fully-deleted adenovirus, adeno-associated virus (AAV), retrovirus, lentivirus, naked plasmid, plasmid/liposome complex, etc. for delivery to the host via intravenous, intramuscular, intraportal or other route of administration. Expression vectors which can be used in the methods and compositions of the present invention include, for example, viral vectors. One of the most frequently used methods of administration of gene therapy, both in vivo and ex vivo, is the use of viral vectors for delivery of the gene. Many species of virus are known, and many have been studied for gene therapy purposes. The most commonly used viral vectors include those derived from adenoviruses, adeno-associated viruses (AAV) and retroviruses, including Antiviruses, such as human immunodeficiency virus (HIV).
Adenovirus is a non-enveloped, nuclear DNA virus with a genome of about 36 kb, which has been well-characterized through studies in classical genetics and molecular biology (Hurwiiz, M.S., Adenoviruses Virology. 3rd edition, Fields et ai, eds., Raven Press, New York, 1996; Hitt, M.M. et ai. Adenovirus Vectors, The Development of Human Gene Therapy, Friedman, T. ed., Cold Spring Harbor Laboratory Press, New York 1999). The viral genes are classified into early (designated E1-E4) and fate (designated L1-L5) transcriptional units, referring to the generation of two temporal classes of viral proteins. The demarcation of these events is viral DNA replication. The human adenoviruses are divided into numerous serotypes (approximately 47, numbered accordingly and classified into 6 groups: A, B, C, D, E and V), based upon properties including hemaghitination of red blood cells, oncogenicity, DNA and protein amino acid compositions and homologies, and antigenic relationships.
Recombinant adenoviral vectors have several advantages for use as gene delivery vehicles, including tropism for both dividing and non-dividing cells, minimal pathogenic potential, ability to replicate to high titer for preparation of vector stocks, and the potential to carry large inserts (Berkner, K.L., Curr. Top. Micro. Immunol. 158:39-66, 1992; Jolly, D., Cancer Genu Therapy 1 :51-64 1994). Adenoviral vectors with deletions of various adenoviral gene sequences, such as pseudoadenoviral vectors (PAVs) and partially-deleted adenoviral (termed "DeAd"), have been designed to take advantage of the desirable features of adenovirus which render it a suitable vehicle for delivery of nucleic acids to recipient cells.
In particular, pseudoadenoviral vectors (PAVs), also known as 'gutless adenovirus' or mini-adenoviral vectors, are adenoviral vectors derived from the genome of an adenovirus that contain minimal cis-acting nucleotide sequences required for the replication and packaging of the vector genome and which can contain one or more transgenes (See, U.S. Patent No. 5,882,877 which covers pseudoadenoviral vectors (PAV) and methods for producing PAV, incorporated herein by reference). PAVs have been designed to take advantage of the desirable features of adenovirus which render it a suitable vehicle for gene delivery. While adenoviral vectors can generally carry inserts of up to 8kb in size by the deletion of regions which are dispensable for viral growth, maximal carrying capacity can be achieved with the use of adenoviral vectors containing deletions of most viral coding sequences, including PAVs. See U.S. Patent No. 5,882,877 of Gregory- et al; Kochanek et al, Proc. Natl. Acad. ScL USA 93:5731-5736, 1996; Parks et al, Proc. Natl. Acad ScL USA 93:13565-13570, 1996; Lieber et al. J. Virol. 70:8944-8960, 1996; Fisher et al, Virology 217:11-22, 1996; U.S. Patent No. 5,670,488; PCT Publication No. WO96/33280, published October 24, 1996; PCT Publication No. WO96/40955, published December 19, 19%; PCT Publication No. WO97/25446, published July 19, 1997; PCT Publication No. WO95/29993, published November 9, 1995; PCT Publication No. WO97/00326, published January 3, 1997: Morral et a/., Hum. Gene Ther. 10:2709-2716, 1998. Such PAVs, which can accommodate up to about 36 kb of foreign nucleic acid, are advantageous because the carrying capacity of the vector is optimized, while the potential for host immune responses to the vector or the generation of replication- competent viruses is reduced. PAV vectors contain the 5' inverted terminal repeat (ITR) and the 3' ITR nucleotide sequences that contain the origin of replication, and the cis-acting nucleotide sequence required for packaging of the PAV genome, and can accommodate one or more transgenes with appropriate regulatory elements, e.g. promoter, enhancers, etc.
Other, partially deleted adenoviral vectors provide a partially-deleted adenoviral (termed "DeAd ") vector in which the majority of adenoviral early genes required for virus replication are deleted from the vector and placed within a producer cell chromosome under the control of a conditional promoter. The deletable adenoviral genes that are placed in the producer cell may include £1 A/El B, E2. E4 (only ORF6 and ORF6/7 need be placed into the cell), plX and plVa2. E3 may also be deleted from the vector, but since it is not required for vector production, it can be omitted from the producer cell. The adenoviral late genes, normally under the control of the major late promoter (MLP), are present in the vector, but the MLP may be replaced by a conditional promoter. Conditional promoters suitable for use in DeAd vectors and producer cell lines include those with the following characteristics: low basal expression in the uninduced state, such that cytotoxic or cytostatic adenovirus genes are not expressed at levels harmful to the cell; and high level expression in the induced state, such that sufficient amounts of viral proteins are produced to support vector replication and assembly. Preferred conditional promoters suitable for use in DeAd vectors and producer cell lines include the dimerizer gene control system, based on the immunosuppressive agents FK506 and rapamycin, the ecdysone gene control system and the tetracycline gene control system. Also useful in the present invention may be {he GeoeSwitch™ technology (Valentis, Inc., Woodlands, TX) described in Abruzzese et al.. Hum. Gene Ther. 1999 10:1499-507, the disclosure of which is hereby incorporated herein by reference. The partially deleted adenoviral expression system is further described in WO99-'57296, the disclosure of which is hereby incorporated by reference herein.
Adeno-associated virus (AAV) is a single-stranded human DNA parvovirus whose genome has a size of 4.6 kb. The AAV genome contains two major genes: the rep gene, which codes for the rep proteins (Rep 76, Rep 68, Rep 52, and Rep 40) and the cap gene, which codes for AAV replication, rescue, transcription and integration, while the cap proteins form the AAV viral particle. AAV derives its name from its dependence on an adenovirus or other helper virus (e.g., herpesvirus) to supply essential gene products that allow AAV to undergo a productive infection, i.e., reproduce itself in the host cell. In the absence of helper virus, AAV integrates as a provirus into the host cell's chromosome, until it is rescued by superinfection of the host cell with a helper virus, usually adenovirus (Muzyczka, Curr. Top. Micor. Immunol 158:97-127, 1992).
Interest in AAV as a gene transfer vector results from several unique features of its biology. At both ends of lhe AAV genome is a nucleotide sequence known as an inverted terminal repeat (ITR), which contains the cis-acting nucleotide sequences required for virus replication, rescue, packaging and integration. The integration function of the ITR mediated by the rep protein in trans permits the AAV genome to integrate into a cellular chromosome after infection, in the absence of helper vims. This unique property of the virus has relevance to the use of AAV in gene transfer, as it allows for a integration of a recombinant AAV containing a gene of interest into the cellular genome. Therefore, stable genetic transformation, ideal for many of the goals of gene transfer, may be achieved by use of rAAV vectors. Furthermore, the site of integration for AAV is well-established and has been localized to chromosome 19 of humans (Kotin et al, Proc. Natl. Acad. ScL 87:2211-2215, 1990). This predictability of integration site reduces the danger of random insertional events into the cellular genome that may activate or inactivate host genes or interrupt coding sequences, consequences that can limit the use of vectors whose integration of AAV, removal of this gene in the design of rAAV vectors may result in the altered integration patterns that have been observed with rAAV vectors (Ponnazhagan et al, Hum Gem Ther 8:275-284, 1997).
There are other advantages to the use of AAV for gene transfer. The host range of AAV is broad. Moreover, unlike retroviruses, AAV can infect both quiescent and dividing cells. In addition, AAV has not been associated with human disease, obviating many of the concerns that have been raised with retrovirus-derived gene transfer vectors.
Standard approaches to the generation of recombinant rAAV vectors have required the coordination of a series of intracellular events: transfection of the host cell with an rAAV vector genome containing a transgene of interest flanked by the AAV ITR sequences, transfection of the host cell by a plasmid encoding the genes for the AAV rep and cap proteins which are required in trans, and infection of the transfected cell with a helper virus to supply the non-AAV helper functions required in trans (Mυzyczka, N.. Curr. Top. Micor. Immunol. 158:97-129, 1992). The adenoviral (or other helper virus) proteins activate transcription of the AAV rep gene, and the rep proteins then activate transcription of the AAV cap genes. The cap proteins then utilize the ITR sequences to package the rAAV genome into an rAAV viral particle. Therefore, the efficiency of packaging is determined, in part, by the availability of adequate amounts of the structural proteins, as well as the accessibility of any cis- acting packaging sequences required in the rAAV vector genome.
Retrovirus vectors are a common too) for gene delivery (Miller, Nature (1992) 357:455-460). The ability of retrovirus vectors to deliver an unrearranged, single copy gene into a broad range of rodent, primate and human somatic cells makes retroviral vectors well suited for transferring genes to a cell.
Retroviruses are RNA viruses wherein the viral genome is RNA. When a host cell is infected with a retrovirus, the genomic RNA is reverse transcribed into a DNA intermediate which is integrated very efficiently into the chromosomal DNA of infected cells. This integrated DNA intermediate is referred to as a provirus. Transcription of the provirus and assembly into infectious virus occurs in the presence of an appropriate helper virus or in a cell line containing appropriate sequences enabling encapsidation without coincident production of a contaminating helper virus. A helper virus is not required for the production of the recombinant retrovirus if the sequences for encapsidation are provided by co-transfection with appropriate vectors.
The retroviral genome and the proviral DNA have three genes: the gag, the pol, and the env, which are flanked by two long terminal repeat (LTR) sequences. The gag gene encodes the internal structural (matrix, capsid. and nucleocapsid) proteins; the pol gene encodes the RNA-directed DNA polymerase (reverse transcriptase) and the env gene encodes viral envelope glycoproteins. The 5' and 3' LTRs serve to promote transcription and polyadenylation of the virion RNAs. The LTR contains all other cis-acting sequences necessary for viral replication. Lcntiviruses have additional genes including vit vpr, tat, rev, vpu, nef, and vpx (in HTV-L HIV-2 and'or S-V). Adjacent to the 5' LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient encapsidation of viral RNA into particles (the Psi site). If the sequences necessary for encapsidation (or packaging of retroviral RNΛ into infectious virions) arc missing from the viral genome, the result is a cis defect which prevents encapsidation of genomic RNA. However, the resulting mutant is still capable of directing the synthesis of all varion proteins.
Lentiviruses are complex retroviruses which, in addition to the common retroviral genes gag, pol and env, contain other genes with regulatory or structural function. The higher complexity enables the lentivirus to modulate the life cycle thereof, as in the course of iatent infection. A typical lentivirus is the human immunodeficiency virus (HIV), the etiologic agent of AIDS. In vivo, HIV can infect terminally differentiated cells that rarely divide, such as lymphocytes and macrophages In vitro. HFV can infect primary cultures of monocyte-derived macrophages (MDM) as well as HeLa-Cd4 or T lymphoid cells arrested in the cell cycle by treatment with aphidicolin or gamma irradiation. Infection of cells is dependent on the active nuclear import of IiIV preintcgratton complexes through the nuclear pores of the target cells. That occurs by the interaction of multiple, partly redundant, molecular determinants in the complex with the nuclear import machinery of the target cell. Identified determinants include a functional nuclear localization signal (NLS) in the gag matrix (MA) protein, the karyophilic virion-associated protein, vpr, and a C-terminal phosphotyrosine residue in the gag MA protein. The use of retroviruses for gene therapy is described, for example, in United States Patent 6,013,516; and U.S. Patent 5,994,136. the disclosures of which are hereby incorporated herein by reference. Other methods for delivery of DNA to cells do not use viruses for delivery. For example, cationic amphiphilic compounds can be used to deliver the nucleic acid of the present invention. Because compounds designed to facilitate intracellular delivery of biologically active molecules must interact with both non-polar and polar environments (in or on, for example, the plasma membrane, tissue fluids, compartments within the cell, and the biologically active molecular itself), such compounds are designed typically to contain both polar and non-polar domains. Compounds having both such domains may be termed araphiphiles, and many lipids and synthetic lipids that have been disclosed for use in facilitating such intracellular deliver)' (whether for in vitro or in vivo application) meet this definition. One particularly important class of such amphiphiles is the cationic amphiphiles. In general, cationic amphiphiles have polar groups that are capable of being positively charged at or around physiological pH, and this property is understood in the art to be important in defining how the amphiphiles interact with the many types of biologically active (therapeutic) molecules including, for example, negatively charged polynucleotides such as DNA.
The use of compositions comprising cationic amphiphilic compounds for gene delivery is described, for example, in United States Patent 5,049,386; US 5,279,833; US 5,650,096; US 5,747,471; US 5,767,099; US 5,910,487; US 5,719,133 ; US 5,840,710; US 5,783,565; US 5,925,628; US 5,912,239; US 5,942,634; US 5,948,925: US 6,022,874; U.S. 5,994,317; U.S. 5,861,397; U.S. 5,952,916; U.S. 5,948,767; U.S. 5,939,401; and U.S. 5,935,936. the disclosures of which are hereby incorporated herein by reference.
In addition, nucleic acid of the present invention can be delivered using "naked DNA." Methods for delivering a non-infectious, non-integrating DNA sequence encoding a desired polypeptide or peptide operably linked to a promoter, free from association with transfection-facUitating proteins, viral particles, liposomal formulations, charged lipids and calcium phosphate precipitating agents arc described in U.S. Patent 5.580,859: U.S. 5,963,622; U.S. 5,910,488; the disclosures of which are hereby incorporated herein by reference.
Gene transfer systems that combine viral and nonviral components have also been reported. Cristiano et al., (1993) Proc. Natl. Acad. Sci. USA 90:11548; Wu et al (1994) J. Biol. Chem. 269:11542; Wagner et al. (1992) Proc. Natl. Acad. Sci. USA 89:6099; Yoshimura et al. (1993) J. Biol. Chem. 268:2300; Curie! et al. (1991) Proc. Natl. Acad. Sd. USA 88:8850; Kupfer et al. (1994) Human Gene Ther. 5:1437; and Gottschalk et al. (1994) Gene Ther. 1:185. In most cases, adenovirus has been incorporated into the gene delivery systems to take advantage of its eπdosomolytic properties. The reported combinations of viral and nonviral components generally involve either covalent attachment of the adenovirus to a gene delivery complex or co-internal ization of unbound adenovirus with cationic lipid: DNA complexes.
For delivery of DNA and protein to the eye, administration will typically be local. This has the advantage of limiting the amount of DNA that needs to be administered and limiting systemic side-effects. Many possible modes of delivery can be used, including, but not limited to: topical administration on the cornea by a gene gun; subconjunctival injection, intracanieral injection, via eye drops to the cornea, injection into the anterior chamber via the termporai limbus, intrastromal injection, corneal application combined with electrical pulses, intracorneal injection, subretinal injection, intravitreal injection (e.g., front, mid or back viireal injection), and intraocular injection. Alternatively cells can be transfected or transduced ex vivo and delivered by intraocular implantation. See, Auricchio. MoI. Ther. 6: 490-494, 2002; Bennett, Nature Med. 2: 649-654, 1996; Borras, Experimental Eye Research 16: 643- 652, 2003; Chaum, Survey of Ophthalmology 47: 449-469, 2002; Carapochiaro, Expert Opinions in Biological Thenφy 2: 537-544 (2002); Lai, Gene Therapy 9: 804 813. 2002; Pleyer, Progress in Retinal and Eye Research, 22: 277-293,.2003. The effects of various proposed therapeutic agents and administrations can be tested in suitable animal models for particular diseases. For example, retinopathy of prematurity can be tested in an oxygen-induced retinopathy model in the mouse as described in Smith, Investigative Ophthalmology & Visual Science, 35: 101-111, 1994. Laser-induced choroidal neovascularization in a mouse can be used as a model for human choroidal neovascularization (CNV) occurs in diseases such as age-related macular degeneration. Tobe, American Journal of Pathology 153: 1641-1646. 1998. Other models of CNV have been developed in primates, rals, minipigs, and rabbits. Mouse models of age-related macular degeneration have been developed in genetically-deficient mice. Mice deficient in either monocyte chemoattractant protein- 1 or C-C chemokine receptor-2 develop features of age-related macular degeneration. Ambati, Nature Med. 9: 1390-1397, 2003.
For the prevention or treatment of disease, the appropriate dosage of a VEGF antagonist of the present invention (protein or gene therapy) will depend on the type of disease to be treated, the severity and course of the disease, whether the VEGF antagonist is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the VEGF antagonist, and the discretion of the attending physician. The VEGF antagonist is suitably administered to the patient at one time or over a series of treatments. In a combination therapy regimen, the compositions of the present invention are administered in a therapeutically effective or synergistic amount. As used herein, a therapeutically effective amount is such that co-administration of a VEGF antagonist and one or more other therapeutic agents, or administration of a composition of the present invention, results in reduction or inhibition of the targeting disease or condition. A therapeutically synergistic amount is that amount of a VEGF antagonist and one or more other therapeutic agents necessary to synergistically or significantly reduce or eliminate conditions or symptoms associated with a particular disease. Depending on the type and severity of the disease, the VEGF antagonist may be administered so that the local concentration provided is about 100 pg/ml to about 100 μg/ml, 1 ng/ml to about 95 μg/ml, 10 ng/mt to about 85 μg/ml, 100 ng/ml to about 75 μg/ml, from about 100 ng/ml to about 50 μg/ml, from about 1 μg/ml to about 25 μg/ml, from about 1 μg/ml to about 15 μg/ml, from about 1 μg/ml to about 10 μg/ml, or from about 1 μg/ml to about 4 μg/ml. When the VEGF antagonist is delivered by gene therapy through viral virions, the dose may be from about 2x106 to about 2x1012, from about 2x 107 to about 2x 1011 drp. or from about 2x108 to about 2x1010 drp per unit dose. When the VEGF antagonist is administered by protein therapy, the dose may be from about 0.1 mg to about 50 mg, from about 0.1 mg to about 25 mg, from about 0.1 mg to about 10 mg, from about 0.1 mg to about 5 mg, from about 0.1 mg to about 4 mg, from about 0.1 mg to about 3 rag, from about 0.1 mg to about 2.5 mg, from about 0.1 mg to about 2 mg, from about 0.1 mg to about 3.5 mg, from about 0.1 mg to about 1 mg, from about 0.1 mg to about 0.75 mg, from about 0.1 mg to about 0.5 mg, from about 0.1 mg to about 0.25 mg. from about 0.25 mg to about 5 mg, from about 0.5 mg to about 5 mg. from about 1 mg to about 5 mg, from about 1.5 mg to about 5 mg, from about 2 mg to about 5 mg, from about 2.5 mg to about 5 mg, from about 0.5 mg to about 5 mg, or from about 0.5 mg to about 2.5 mg per unit dose.
The VEGF antagonist of the present invention may be administered as a single dose or repeatedly. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. The progress of the therapy of the invention is monitored by conventional techniques and assays.
Where an animal or a patient develops an immune response to the VEGF antagonist- one may try to reduce the immune response (inflammation and/or haze or antibodies), e.g., by using available immune suppessors, when necessary. When gene therapy is used according to some embodiments of the invention, reducing the number of empty viral capsid helps reducing the immune response. According to some imbodiraents, a reduction of the immune response against the viral vector helps to increase the expression of the VEGF antagonist (e.g., D29GLYFC), and therefore enhances the therapeutic efficacy of the VEGF antagonist.
While the invention has been described with respect to specific examples including presently preferred modes of carrying out the invention, those skilled in the art will appreciate that there are numerous variations and permutations of the above described systems and techniques that fall within the spirit and scope of the invention as set forth in the appended claims. All references disclosed herein are expressly incorporated by reference,
EXAMPLES
Example 1
Two constructs were generated: the first, D2-9Gly-Fc, containing a polyglycine 9-mer (9GIy) linker and the second. D2-Fc, with the same sequence except the 9GIy linker (Fig- 1).
We analyzed the amino acid sequences of D2-9Gly-Fc and D2-Fc proteins using the Protein Analysis Toolbox of the sequence analysis program MacVector 6.5.1. (IBI, New Haven, CT). The polyglycine 9-mer linker in the D2-9Gly-Fc sequence was identified as a region with higher than average flexibility by the flexibility prediction method of Karpus and Schultz (1985) Naturwiss, 72: 212-213. No such region was detected in the D2-Fc sequence (Fig. 1).
Example 2 We tested an isolated FIt-I FIt-I Ig-like domain 2 connected to the IgGl Fc region by a flexible poiyglycme 9-tner linker (D2-9Gly-Fc). The D2-9Gly-Fc fusion protein is capable of efficiently binding VEGF and of inhibiting VEGF-dependent human umbilical vein endothelial cell (HUVEC) proliferation. See Fig. 2. In contrast, when FIt-I Ig-like domain 2 is linked directly to the IgGl heavy chain (Fc) to form D2-Fc, only minimal VEGF binding was observed. See Fig. 2. Both the dimerization via IgGl Fc and the insertion of a flexible linker appear to facilitate VEGF binding to FIt- I domain 2. The presence of diraeric forms in both D2-9Gly-Fc and D2-Fc were confirmed by the Western blot analysis. See Fig. 3.
Example 3
Λn intravitrεal injection of AAV vector (1 x 108 to 1 x 109 particles in a volume of 0.0005 ml.) is administered to newborn (PO) or 1 day old (Pl) C57BL/6 mice. Retinal neovascularization (NV) is induced in C57BL/6 mice by exposing P7 pups and their nursing dam to hyperoxia for 5 days. The pups are returned to room air on Pl 2 and are euthanized at Pl 7 (time of peak NV). (Smith LEH, Weslowski E. McLellan A, Kostyk SK, D'Amato R, Sullivan and D'Amore PA. Oxygen-Induced Retinopathy in the Mouse. Invest Opth Vis ScL 1994;35:l0l~l 11.) Entire paraffin embedded eyes are serially cross sectioned at 5 micron intervals. The degree of NV is determined by counting the number of endothelial cell nuclei internal to the inner limiting membrane in sections taken every 100 microns.
Cohorts of animals treated with the AAV vectors coding for the anti-angiogenic agents are compared to cohorts treated with vectors coding for irrelevant transgenes or with vectors that do not code for a transgene. The average number of endothelial cell nuclei in each treated eye is compared to each animal's untreated fellow eye.
Example 4 Generation of D2-9Gly-Ex3/CH3
Domain 2 of FU-I has been shown to be essential for VEGF165 binding. However, it was demonstrated thai FIt-I domain 2 aione was incapable of binding VEGF A. (Davis-Smyth et a!.. 1996.) VEGF A, when present as a dimer, binds to FIt-I through acidic residues (amino acids 63-67 of the mature protein) that allows a possible mechanism for ligand-induced dimerization of receptor (Keyt et al., 1996).
Therefore, a dimerization of domain 2 of FIt-I was used as a strategy to restore the binding of domain 2 of FlM to VEGF A. Fusion with a fragment of IgG heavy chain can be used for dimerization of proteins (Davis-Smyth et al., 1996). Here we demonstrate that amino acids 75-88 (i.e., PSCVPLMRCGGCCN; SEQ ID NO: 13) of VΕGF A ( SEQ ID NO: 12) increase the biological activity of sFlt-1 hybrid proteins.
Initially, three hybrid proteins were engineered: D2-9Gly-Fc, D2-Fc and D2-9Gly- Ex3/CH3 (Fig. 4). AH three hybrid proteins contain the same FIt-I domain D2 as D2- 9GIy-Fc. No VEGF binding was observed with D2-Fc, which does not contain the polyglycine 9-mer (9GIy) linker. The third protein, D2-9Gly-Ex3/CH3, contains the polyglycine 9-mer (9GIy) linker and the multimerization domain of VEGF (aa PSCVPLMRCGGCCN; SEQ ID NO: 13; VEGF Ex3 ), but it also contains the CH3 region of human IgGl heavy chain Fc (aa 371-477 of the SEQ ID NO: 10).
The protein D2-Fc did not show efficient inhibitory activity in the HUVEC proliferation assay (Fig. 5) and by implication did not bind to VEGF165 efficiently. However, the third hybrid protein, D2-9GIy-Ex3/CH3, which comprises domain 2 of Fh-I fused to the CH3 region via both the 9GIy linker and the dimerization region of VEGF 165 (Ex 3), did demonstrate inhibitory activity in a VEGF-dependent HUVECs proliferation assay (Fig. 5). This implies that this hybrid protein binds to VEGF165 efficiently. Example 5
Using linker (GIy4SeO3 in FlM D2 construct
The use of several polyglycine linkers has been previously described for improvement of protein features (Mouz et al., 1996; Qiu et al., 1998). For the next construct we have used another type of linker, the 15-mer (Gly-Gly-Gly-Gly-Ser)3 (Huston et al., 1988). D2-(Gly4Ser)3-Fc protein was generated and it contains F(M domain 2, (GIy4Ser)3 linker and the Fc region of human IgG I heavy chain..
D2-(GlY4Ser)3-Fc was further characterized in HUVECs proliferation assay. Biological activity of D2-(Gly4Ser)rFc as measured by inhibition of HUVEC proliferation was similar to that of D2-9Gly-Fc and D2-9Gly-Ex3/CH3 (Fig. 6).
The D2-(Gly4Scr)rFc construct was further characterized by Western blot and compared to D2-9Gly-Fc (Fig.9). Both constructs are present mostly in a dimer form and the monomer forms were detected after separation of reduced samples.
Example 6
Role of 9GIy or VEGF Ex3 in FlM (D2) constructs
In order to investigate the role of 9GIy linker or VEGF dimerizing sequence Ex3 on soluble receptor VEGF binding, three other constructs were generated: D2-9Gly-CH3, D2-CH3 and D2-Ex3/CH3 (Fig. 8). All three constructs were generated and like all the previous constructs were also put under control of C-MV promoter. Their VEGF blocking activity was assessed in HUVECs proliferation assay (Fig. 9). The HUVEC proliferation assay of proteins containing lhe CH3 region of IgGl has shown that D2-9Gly-CH3 (without Ex3) and protein D2-Ex3/CH3 (without 9GIy linker) had similar VEGF blocking potency as compared to the parental D2-9Gly- Ex3/CH3 protein. However, protein D2-CH3 appeared to be the weakest VEGF inhibitor from all of them (Fig. 9).
The FIt-I ELISA data of conditioned media from transfected 293 cells has shown similar FIt-I levels for D2-9Gly-Ex3/CH3, D2-9Gly-CH3 and D2-Ex3/CH3 and D2- CH3 (70-90 ng/ml) and a little higher (-150 ng/ml) for the least active form of Ul- CH3. Western blot of D2-9Gly-CH3 and D2-CH3 constructs (Fig.10 ) is showing a prevalence of dlmer forms in non-reduced conditions.
Example 7
D2-9G Iy-Fc binds VEGF better than all constructs
VEGF binding assay allows us to compare the relative VEGF binding affinities of our soluble VEGF receptors in a cell free system.
Briefly, conditioned media containing known concentrations of soluble receptor (ranging in concentrations from 0.29 - 150 pM) were serially diluted and mixed with 10 pM VEGF. The amount of unbound VEGF was then measured by ELlSA. D2- 9GIy-Fc binds VEGF with higher affinity to bind VEGF at receptor concentrations from 0.001 to - 0.2 pM than all other constructs. D2-CH3 has the lowest affinity to bind VEGF (Fig. 11).
Example 8
The AAV2-SFLT1 D29GLYFC genomic components consist of two AAV2 ITRs and the expression cassette of sFLTlD29GLYFC. A hybrid chicken β-actin promoter is used for driving the expression of sFLTlD29GLYFC and consists of a CMV IE enhancer, the chicken β-actin promoter, and a 94 bp fragment of exon 1 that partially belongs to the β-actin transcript. The hybrid chicken β-actin/rabbit β-globin intron is also a part of the CBA promoter, and it contains a number of cryptic transcription factors binding sites. The poly-adenylation region is obtained from the SV40 viral genome.
The test article. AAV2-sFLTH)29GLYFC, and the AAV2 vehicle control, phosphate- buffered saline containing 0.014% Tween® 20, Lot No. NBR 15045-108, were prepared as a stock frozen liquid in polypropylene vials and a prefoπnυlated, aqueous solution, respectively. Solutions of the test and vehicle articles were prepared to yield dosing solutions at appropriate concentrations for achieving the intended doses at the desired dose volumes.
When injected intravitreally into the eyes of cynomolgus monkeys, we found that AAV2-SFLT1D29GLYFC (2 x 108, 2 x 109, and 2 x 1010 in 50 μi dose volume) gave dose-dependent expression of SFLT1D29GLYFC as measured in the aqueous and vitreous fluids and these levels were persistent for at least one year following administration of the AAV vector.
We performed laser-CNV experiments in non-human primates (NHP) and showed that SFLT1D29GLYFC was very effective at inhibiting choroidal neovascularization in this NHP model. In one set of the experiments, 2 x 108 and 2 x 109 vector particles injected into the anterior/mid vitreous space resulted in dose-dependent expression, but no efficacy was observed in this set of experiments. When 2 x 1010 SFI.T1D29GLYFC was injected into the mid vitreous space (mid vitreal injection), it was expressed in the injected eyes (e.g., transitional epithelial cells and retinal ganglion cells showed mRNA expression) and therapeutically effective concentrations were measured in the aqueous humor : 111 ng/ml, 277 ng/ml, 885 ng/ml, 967ng/ml, 1,057 ng/ml, and 1,584 ng/ml. Levels in the vitreous humor were approximately three-fold higher than in the aqueous humor. These concentrations were effective in inhibiting laser induced choroidal neovascularizaton (CNV). Figure 15 shows the location of expression in another group of animals that received 2 x 1010 SFLT1D29GLYFC.
Mild to moderate vitrcai haze was observed in high dose group, which was resolved over time in most animals. No anti-transgene T-ςell response was detected although there was anti-AAV T-celi response was observed in some animals. Animals receiving control AAV2 virions also developed haze at high dose, suggesting that the haze was not due to the transgene sFLT, but the viral capsids instead. Vitreal haze was scored and compared to the systems developed by Nussenblatt et al. (1985, Opthalmology 92: 467). Inflammatory changes consisting of lymphocytes or plasma cells in the trabeclar raeshwork and the iridocorneal angle, inflammatory cells in the vitreous, or perivascular lymphocytes ion the retina were observed in some animals (either with AAV2-sFLTlD29GLYFC or with the control virions) with vitreal haze. No tissue destruction or reorganization was observed. The animal with most persistent inflammation had lowest expression of the transgene.
In a different group of animals, no animals had pre-existing AAV titers in aqueous or serum. Serum and aqueous and vitreous humor were evaluated for the presence of sFLT01 protein, as well as antibodies against sFLTOl and the vector, AA V2. Intermittent, inconsistent positivity was observed in several animals throughout the course of this study using this assay, including some animals in the vehicle control group. There were no findings of antibodies against sFLTOl in the serum. There was a dose and time dependent increase in animals that possessed a titer against AA V2 in the serum. The animal with highest aqueous titer had lowest expression, and the animal with lowest aqueous titer had highest expression. Some animals were evaluated for changes in additional clinical signs, qualitative food consumption, physical examination), body weight, ophthalmic exams, fluorescein angiography, electrorelinography, tonometry. After termination, a full necropsy was conducted on the animals, and tissues were collected and pererved. Tissues from control and high-dose monkeys were processed and examined microscopically. No effect on retina electrical activity of the retina was observed.
For some animals, Lucentis was administered 11 weeks after AAV2- sFLT1D29GLYFC (doses of 2 x 108, 2 x 109, and 2 x 1010), with no adverse effects observed.
References Davis-Smyth, et al., EMBO 1. 15, 1996, 4919 Huston, J. S., et al. (1991) Methods Enzymol 203, 46-88 Huston, J. S., et al (1988) Proc. Natl. Acad Sd. USA, 85, 5879-5883. Johnson, S., et al. (1991) Methods Enzymol 203, 88-98 karpus, P. A, et al. (1985) Naturwiss., 72. 212-213. Keyt, B. A., etal ( 1996) J. Biol. Chem. 271 : 5638 - 5646. Kortt, A. A., et al. ( 1997) Protein Engng, 10, 423-433. Lee, Y-L., et al (1998) Human Gene Therapy, 9, 457-465 Mouz N., et al. (1996) Proc. Natl Acad. Sci. USA, 93, 9414-9419. Nielsen, et al. (1997) Protein Eng., 10, 1 Qiu, H., et al (1998) J. Biol. Chem. 273: 11173-11176.

Claims

We claim: 1. A method of treating an ocular disease in a mammal, said method comprising administering to a diseased eye of said mammal a viral vector encoding a VIr)GF antagonist at a therapeutically effective amount of from about 2x 108 to about 2x1010 dip.
2. The method of claim 1 , wherein said viral vector is an AAV vector.
3. The method of claim 2, wherein said AAV vector is AAV2.
4. The method of claim I , wherein said VEGF antagonist is a fusion polypeptide comprising a VEGF receptor rused to an JgG heavy chain molecule.
5. The method of claim 4, wherein said VEGF receptor comprises VECrF-Rl (FLT-I ).
6. The method of claim 4, wherein said VEGF receptor comprises an IgG-like domain 2 of VEGF-Rl (FLT-I D2).
7. The method of claim 4, wherein said IgG heavy chain molecule comprises an Fc portion of the heavy chain.
8. The method of claim 4, wherein said IgG heavy chain molecule comprises a CH3 portion of the heavy chain.
9. The method of claim 4, wherein said VEGF receptor is fused to said IgG heavy chain molecule through an amino acid linker.
10. The method of claim 9, wherein said amino acid linker is a 9GIy linker.
11. The method of claim 4, wherein said fusion polypeptide is FLT- 1 D2-9GLY-FC or FLT-1D2-9GLY-CH3.
12. The method of claim 1 , wherein said VEGF antagonist is administered to the diseased eye by intravitreal injection.
13. The method of claim 12, wherein said VF.GF antagonist is administered to the diseased eye by mid vitreal injection.
14. The method of claim 1 , wherein said VEGF antagonist is expressed in transitional epithelial cells of the diseased eye.
15. The method of claim 1 , wherein said VEGF antagonist is expressed in the diseased eye for at least 6 months.
16. The method of claim 15, wherein said VEGF antagonist is expressed in the diseased eye for at least 1 year.
17. The method of claim 1 , wherein said ocular disease is selected from the group consisting of wet age-related macular degeneration (wet AMD), macular edema, retinal vein occlusions, retinopathy of prematurity, and diabetic retinopathy.
18. The method of claim 1 , further comprising administering a second VEGF antagonist.
19. The method of ciaim 18, wherein said second VEGF antagonist is a fusion polypeptide comprising a VEGF receptor fused to an IgG heavy chain molecule.
20. The method of claim 18. wherein said protein is a VEGF antibody.
21. The method of claim 20, wherein said VEGF antibody is bevacizumab or ranibizumab.
22. A method of treating an ocular disease in a mammal, said method comprising administering to a diseased eye of said mammal a nucleic acid encoding a VEGF antagonist, wherein said VEGF antagonist is expressed at a therapeutically effective amount of from about 100 ng/ml to about 1,500 ng/ml to treat said ocular disease.
23. The method of claim 22, wherein said viral vector is an AAV vector.
24. The method of claim 23, wherein said AAV vector is AA V2.
25. The method of claim 22, wherein said VEGF antagonist is a fusion polypeptide comprising a VEGF receptor fused to an IgG heavy chain molecule.
26. The method of claim 25, wherein said VEGF receptor comprises VEGF-Rl (FLT-I).
27. The method of claim 25, wherein said VEGF receptor comprises an igG-like domain 2 of VEGF-Rl (FLT-1D2).
28. The method of claim 25, wherein said IgG heavy chain molecule comprises an Fc portion of the heavy chain.
29. The method of claim 25, wherein said IgG heavy chain molecule comprises a CH3 portion of the heavy chain.
30. The method of claim 25, wherein said VEGF receptor is fused to said IgG heavy chain molecule through an amino acid linker.
31. The method of claim 30. wherein said amino acid linker is a 9GIy linker.
32. The method of claim 25, wherein said fusion polypeptide is FLT- 1 D2-9GLY-FC or FLT-1D2-9GLY-CH3.
33. The method of claim 22, wherein said VEGF antagonist is administered to the diseased eye by intravitreal injection.
34. The method of claim 33, wherein said VEGF antagonist is administered to the diseased eye by mid vitreal injection.
35. The method of claim 22, wherein said VEGF antagonist is expressed in transitional epithelial cells of the diseased eye.
36. The method of claim 22, wherein said VEGF antagonist is expressed in the diseased eye for at least 6 months.
37. The method of claim 36, wherein said VEGF antagonist is expressed in the diseased eye for at least I year.
38. The method of claim 22, wherein said ocular disease is selected from the group consisting of wet age-reiated macular degeneration (wet AMD), macular edema, retinal vein occlusions, retinopathy of prematurity, and diabetic retinopathy.
39. The method of claim 22, further comprising administering a second VEGF antagonist.
40. The method of claim 39, wherein said second VEGF antagonist is a fusion polypeptide comprising a VEGF receptor fused to an IgG heavy chain molecule.
41. The method of claim 39, wherein said protein is a VEGF antibody.
42. The method of claim 41 , wherein said VBGF antibody is bevacizumab or ranibizumab.
43. A method of reducing haze in the eye in a mammal receiving AAV gene therapy for the eye, said method comprising administering to said mammal gene therapy AAV virions containing empty capsids not exceeding 90% of the total capsids.
44. The method of claim 43, wherein said gene therapy AAV virions contain empty capsids not exceeding 80%, 70%, 60%, or 50% of the total capsids.
45. The method of claim 44, wherein said gene therapy AAV virions are substantially free of empty capsids.
46. The method of claim 43, wherein said AAV is AA V2.
PCT/US2009/034709 2008-02-20 2009-02-20 Angiogenesis inhibition WO2009105669A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
BRPI0908496A BRPI0908496A2 (en) 2008-02-20 2009-02-20 angiogenesis inhibition
JP2010547799A JP2011512417A (en) 2008-02-20 2009-02-20 Inhibition of angiogenesis
CN2009801059368A CN101951925A (en) 2008-02-20 2009-02-20 Angiogenesis inhibition
IL207641A IL207641A0 (en) 2008-02-20 2010-08-16 Compositions comprising nucleic acids encoding vegf antagonists for use in treating ocular disease

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US3007208P 2008-02-20 2008-02-20
US61/030,072 2008-02-20
US3454608P 2008-03-07 2008-03-07
US61/034,546 2008-03-07
US5663208P 2008-05-28 2008-05-28
US61/056,632 2008-05-28

Publications (2)

Publication Number Publication Date
WO2009105669A2 true WO2009105669A2 (en) 2009-08-27
WO2009105669A3 WO2009105669A3 (en) 2009-12-17

Family

ID=40986215

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/034709 WO2009105669A2 (en) 2008-02-20 2009-02-20 Angiogenesis inhibition

Country Status (5)

Country Link
JP (1) JP2011512417A (en)
CN (1) CN101951925A (en)
BR (1) BRPI0908496A2 (en)
IL (1) IL207641A0 (en)
WO (1) WO2009105669A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160507A1 (en) * 2013-03-13 2014-10-02 Genzyme Corporation Fusion proteins comprising pdgf and vegf binding portions and methods of using thereof
WO2016005381A1 (en) * 2014-07-10 2016-01-14 Bayer Pharma Aktiengesellschaft Pdgfrbeta-fc fusion proteins and uses thereof
US9834604B2 (en) 2011-06-30 2017-12-05 Genzyme Corporation Inhibitors of T-cell activation
US9943573B2 (en) 2012-05-15 2018-04-17 Avalanche Australia Pty Ltd. Treatment of ocular neovascularization using anti-VEGF proteins
US10000741B2 (en) 2014-03-17 2018-06-19 Adverum Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
RU2712623C2 (en) * 2015-03-31 2020-01-30 Илдонг Фарм. Ко., Лтд. Pharmaceutical composition for preventing and treating eye diseases, containing, as active ingredient, fusion protein in which tissue-penetrating peptide and anti-vascular endothelial growth factor preparation are fused
US10584328B2 (en) 2015-12-23 2020-03-10 Adverum Biotechnologies, Inc. Mutant viral capsid libraries and related systems and methods
TWI687225B (en) * 2014-02-06 2020-03-11 美商健臻公司 Compositions and methods for treating and preventing macular degeneration
US11021519B2 (en) 2015-03-02 2021-06-01 Adverum Biotechnologies, Inc. Compositions and methods for intravitreal delivery of polynucleotides to retinal cones
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108697792A (en) * 2015-11-19 2018-10-23 珠海泰瑞尚生物医药科技有限公司 Method and composition for combining VEGF
KR101685532B1 (en) * 2016-04-26 2016-12-13 한국프라임제약주식회사 A VEGFR fusion protein
GB201806333D0 (en) * 2018-04-18 2018-05-30 Glaxosmithkline Ip Dev Ltd Parvovirus vector production
CN108671229B (en) * 2018-05-08 2022-03-25 华博生物医药技术(上海)有限公司 Pharmaceutical composition preparation of recombinant human vascular endothelial growth factor receptor-antibody fusion protein
CN114181972A (en) * 2021-11-23 2022-03-15 上海本导基因技术有限公司 Lentiviral vectors suitable for gene therapy of refractory angiogenic eye diseases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060088936A1 (en) * 2002-05-01 2006-04-27 Warrington Kenneth H Raav expression systems for genetic modification of specific capsid proteins
US20070224178A1 (en) * 2004-09-13 2007-09-27 Abraham Scaria Multimeric constructs

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060088936A1 (en) * 2002-05-01 2006-04-27 Warrington Kenneth H Raav expression systems for genetic modification of specific capsid proteins
US20070224178A1 (en) * 2004-09-13 2007-09-27 Abraham Scaria Multimeric constructs

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BAINBRIDGE ET AL.: 'Gene therapy for ocular angiogenesis'' CLINICAL SCIENCE vol. 104, 2003, pages 561 - 575 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9834604B2 (en) 2011-06-30 2017-12-05 Genzyme Corporation Inhibitors of T-cell activation
US11028170B2 (en) 2011-06-30 2021-06-08 Genzyme Corporation Inhibitors of T-cell activation
US10004788B2 (en) 2012-05-15 2018-06-26 Avalanche Australia Pty Ltd. Treatment of ocular neovascularization using anti-VEGF proteins
US9943573B2 (en) 2012-05-15 2018-04-17 Avalanche Australia Pty Ltd. Treatment of ocular neovascularization using anti-VEGF proteins
US11084864B2 (en) 2013-03-13 2021-08-10 Genzyme Corporation Fusion proteins comprising PDGF and VEGF binding portions and methods of using thereof
CN105188733B (en) * 2013-03-13 2020-05-08 建新公司 Fusion proteins comprising PDGF and VEGF binding moieties and methods of use thereof
EP2968461A1 (en) * 2013-03-13 2016-01-20 Genzyme Corporation Fusion proteins comprising pdgf and vegf binding portions and methods of using thereof
EP2968461B1 (en) * 2013-03-13 2022-09-21 Genzyme Corporation Fusion proteins comprising pdgf and vegf binding portions and methods of using thereof
US9637534B2 (en) 2013-03-13 2017-05-02 Genzyme Corporation Fusion proteins comprising PDGF and VEGF binding portions and methods of using thereof
US10183983B2 (en) 2013-03-13 2019-01-22 Genzyme Corporation Nucleic acids encoding fusion proteins comprising PDGF and VEGF binding portions and methods of using thereof
WO2014160507A1 (en) * 2013-03-13 2014-10-02 Genzyme Corporation Fusion proteins comprising pdgf and vegf binding portions and methods of using thereof
CN105188733A (en) * 2013-03-13 2015-12-23 建新公司 Fusion proteins comprising PDGF and VEGF binding portions and methods of using thereof
TWI687225B (en) * 2014-02-06 2020-03-11 美商健臻公司 Compositions and methods for treating and preventing macular degeneration
US11248214B2 (en) 2014-03-17 2022-02-15 Adverum Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
US10000741B2 (en) 2014-03-17 2018-06-19 Adverum Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
WO2016005381A1 (en) * 2014-07-10 2016-01-14 Bayer Pharma Aktiengesellschaft Pdgfrbeta-fc fusion proteins and uses thereof
US11021519B2 (en) 2015-03-02 2021-06-01 Adverum Biotechnologies, Inc. Compositions and methods for intravitreal delivery of polynucleotides to retinal cones
US11052130B2 (en) 2015-03-31 2021-07-06 Ildong Pharm Co., Ltd. Method for treating eye diseases with a fusion protein of a tissue-penetrating peptide and anti-vascular endothelial growth factor
RU2712623C2 (en) * 2015-03-31 2020-01-30 Илдонг Фарм. Ко., Лтд. Pharmaceutical composition for preventing and treating eye diseases, containing, as active ingredient, fusion protein in which tissue-penetrating peptide and anti-vascular endothelial growth factor preparation are fused
US11738064B2 (en) 2015-03-31 2023-08-29 Ildong Pharm Co., Ltd. Pharmaceutical composition for preventing and treating eye diseases, containing as active ingredient, fusion protein in which tissue-penetrating peptide and anti-vascular endothelial growth factor preparation are fused
US10584328B2 (en) 2015-12-23 2020-03-10 Adverum Biotechnologies, Inc. Mutant viral capsid libraries and related systems and methods
US11427931B2 (en) 2015-12-23 2022-08-30 Adverum Biotechnologies, Inc. Mutant viral capsid libraries and related systems and methods
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Also Published As

Publication number Publication date
WO2009105669A3 (en) 2009-12-17
JP2011512417A (en) 2011-04-21
CN101951925A (en) 2011-01-19
IL207641A0 (en) 2010-12-30
BRPI0908496A2 (en) 2019-01-15

Similar Documents

Publication Publication Date Title
US9815892B2 (en) Multimeric constructs
WO2009105669A2 (en) Angiogenesis inhibition
PT1871667E (en) Mobile on-wing engine washing and water reclamation system
US20230322873A1 (en) Methods to produce rod-derived cone viability factor (rdcvf)
MXPA04008937A (en) Methods of modulating cd200 receptors.
WO2010019263A2 (en) Soluble flt constructs for treating cancers
JP7065085B2 (en) Use of itrizumab to reduce phosphorylation of CD6
WO2021143733A1 (en) Fusion protein, preparation method therefor and use thereof
JP2017526625A (en) Denatured latent related protein constructs
TW202019476A (en) Agent for treating ocular disease involving retinal fibrosis
US20230365640A1 (en) Clec2 fusion protein and uses thereof
CA3228666A1 (en) Compositions and methods for transgene expression
CA3222464A1 (en) Retinal disorders
KR20230095097A (en) GDF15 modulator for use in inhibition of ocular tissue fibrosis
CN114805587A (en) CAR-NK cells and their use in the treatment of ageing and fibrosis of organs and tissues

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980105936.8

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09713015

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2834/KOLNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 207641

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2010547799

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/009178

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009713015

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0908496

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100819