WO2018140611A1 - Methods for treating disorders associated with angiogenesis and neovascularization - Google Patents

Methods for treating disorders associated with angiogenesis and neovascularization Download PDF

Info

Publication number
WO2018140611A1
WO2018140611A1 PCT/US2018/015257 US2018015257W WO2018140611A1 WO 2018140611 A1 WO2018140611 A1 WO 2018140611A1 US 2018015257 W US2018015257 W US 2018015257W WO 2018140611 A1 WO2018140611 A1 WO 2018140611A1
Authority
WO
WIPO (PCT)
Prior art keywords
months
patient
cnv
vegf inhibitor
immunoconjugate
Prior art date
Application number
PCT/US2018/015257
Other languages
French (fr)
Inventor
Gabriela BURIAN
William Greene
Kirk Dornbush
Original Assignee
Iconic Therapeutics, Inc.
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 Iconic Therapeutics, Inc. filed Critical Iconic Therapeutics, Inc.
Priority to US16/480,804 priority Critical patent/US20190388522A1/en
Priority to EP18745015.0A priority patent/EP3573641A4/en
Publication of WO2018140611A1 publication Critical patent/WO2018140611A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6437Coagulation factor VIIa (3.4.21.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • Age-related macular degeneration refers to the chronic, progressive degenerative pathology of the macula that results in loss of central vision.
  • AMD Age-related macular degeneration
  • Neovascular AMD also revered to as exudative or "wet” AMD
  • AMD is the leading cause of severe vision loss and blindness in elderly patients over the age of fifty in the industrialized world.
  • wet AMD In the United States alone, more than 1.5 million people suffer from wet AMD. It is expected that AMD incidence and prevalence will further increase with the ageing population, thus leading to a significant increase in the number of patients with wet AMD in the United States and worldwide.
  • Tissue factor is a cytokine receptor present on vascular endothelial cells. It is an integral membrane glycoprotein with an intracellular terminal domain, a transmembrane domain, and an extracellular binding domain for Factor VJJ (FVTI) and Factor VJJa (FWa). TF has been implicated in the process of angiogenesis and the inflammatory cascade of cytokine release, both processes in the pathogenesis of neovascular AMD and certain cancers.
  • FVTI Factor VJJ
  • FWa Factor VJJa
  • TF has been implicated in the process of angiogenesis and the inflammatory cascade of cytokine release, both processes in the pathogenesis of neovascular AMD and certain cancers.
  • Choroidal neovascularization is the process in which new blood vessels grow in the choroid layer of the eye, and is associated with wet AMD.
  • vascular endothelial growth factor therapies targeting vascular endothelial growth factor (VEGF) are currently the standard of clinical care for wet AMD.
  • VEGF vascular endothelial growth factor
  • the present invention provides a method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconj ugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTia) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best- corrected visual acuity
  • CNV mean choroidal neovascularization
  • the present invention provides a method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions, a composition comprising: (a) an effective amount of an immunoconj ugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTia) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • a composition comprising: (a) an effective amount of an immunoconj ugate dimer, wherein the monomer subunits of the dimer each comprises a
  • the present invention provides a method for reversing tumor neovascularization in an eye of a patient in need thereof, comprising administering to the patient in multiple dosing sessions a composition comprising: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • a composition comprising: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to
  • the VEGF inhibitor comprises an anti-VEGF antibody.
  • the immunoconjugate dimer is a homodimer. In some embodiments, the immunoconjugate dimer is a heterodimer.
  • the present invention provides a method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best- corrected visual acuity
  • CNV mean choroidal neovascularization
  • the present invention provides a method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor VHa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best- corrected visual acuity
  • CNV mean choroidal neovascularization
  • the present invention provides a method for preventing, inhibiting, or reversing tumor neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best- corrected visual acuity
  • CNV mean choroidal neovascularization
  • the mutated factor VII protein exhibits a decreased coagulation response in a mammalian host, as compared to a wild-type factor VII protein.
  • At least one of the monomer subunits of the immunoconjugate comprises a mutated human fVlla domain comprising a single point mutation at Lys341 or Ser 344.
  • the single point mutation is to an alanine.
  • the single point mutation is Lys341 to Ala341.
  • the single point mutation is Ser344 to Ala344.
  • the ocular neovascularization is secondary to proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma.
  • AMD wet age-related macular degeneration
  • ROI retinopathy of prematurity
  • neovascular glaucoma neovascular glaucoma.
  • the ocular neovascularization is choroidal neovascularization.
  • the choroidal neovascularization is secondary to wet AMD.
  • the patient has been previously diagnosed with wet AMD in the eye.
  • the eye of the patient has not been previously treated for choroidal neovascularization or wet AMD.
  • the patient has previously been treated for choroidal vascularization with anti-vascular endothelial growth factor (VEGF) therapy, laser therapy, or surgery.
  • VEGF anti-vascular endothelial growth factor
  • the administering comprises intravitreal injection at each dosing session. In some embodiments, the administering comprises suprachoroidal injection at each dosing session.
  • multiple dosing sessions comprise two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, fourteen or more, fifteen or more, sixteen or more, seventeen or more, eighteen or more, nineteen or more, or twenty or more dosing sessions.
  • the multiple dosing sessions comprise 12 to 24 dosing sessions.
  • the administering comprises intravitreal injection into the eye of the patient once every 28 days, once every 30 days, or once every 35 days.
  • the immunoconjugate comprises intravitreal the amino acid sequence of SEQ ID NO: 2 or 3. In a further embodiment, the immunoconjugate comprises the amino acid sequence of SEQ ID NO:2. In a further embodiment, the immunoconjugate comprises the amino acid sequence of SEQ ID NO: 3. In some embodiments, the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO:4. In some embodiments, the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO: 5. In some embodiments, the administering comprises intravenous or intratumoral administration.
  • the improved reduction in CNV lesion area or CNV exudation is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
  • the improved durability of treatment is increased by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
  • the retinal thickness of the eye of the patient is reduced in the eye of the patient, as compared to the retinal thickness of the eye of patients having been administered the VEGF inhibitor alone.
  • the retinal thickness is reduced by at least about 50 ⁇ , at least about 100 ⁇ , at least about 150 ⁇ , at least about 175 ⁇ , at least about 200 ⁇ , at least about 225 ⁇ , or at least about 250 ⁇ .
  • the retinal thickness is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
  • the decreased retinal thickness is decreased central retinal subfield thickness (CST), decreased center point thickness (CPT), or decreased central foveal thickness (CFT).
  • the intraocular pressure (IOP) in the eye of the patient is measured prior to each dosing session. In some embodiments, the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about SO minutes, or about 1 hour after each dosing session. In some embodiments, the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about SO minutes, or about 1 hour prior to each dosing session. In further embodiments, the IOP is measured via tonometry.
  • the VEGF inhibitor is present in the same composition as the immunoconjugate. In some embodiments, the VEGF inhibitor is present in a different composition than the immunoconjugate.
  • the anti-VEGF antibody is ranibizumab. In some embodiments, the dosage of ranibizumab is from about 0.2 mg to about 1 mg. In a further embodiment, the dosage of ranibizumab is 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, or 0.7 mg. In some embodiments, the ranibizumab is administered to the eye of the patient via an intravitreal injection.
  • the VEGF inhibitor is administered at a dosage from about 0.2 mg to about 0.7 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of about 0.6 mg. In some embodiments, wherein the VEGF inhibitor is administered at a dosage of 0.6 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of about 0.3 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of 0.3 mg.
  • the multiple dosing sessions comprise administration once per month. In a further embodiment, the multiple dosing sessions comprise administration once per month for the first three months, followed by monthly treatments in months 4-12 only when CNV activity is observed.
  • composition comprising the effective amount of the VEGF inhibitor is administered to the eye of the patient via an intravitreal injection.
  • the composition comprising the effective amount of the VEGF inhibitor is administered at each of the multiple dosing sessions.
  • each dosing session comprises the administration of between about 200 ug and about 600 ug of the immunoconj ugate dimer.
  • the administration is about 300 ug of the immunoconj ugate dimer.
  • the administration is about 600 pg of the immunoconjugate dimer.
  • the outcome is measured at least 6 months after beginning treatment
  • the patient is a human.
  • compositions for use in a method of treatment comprising an immunoconjugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and wherein the method of treatment further comprises administration of a VEGF inhibitor; and wherein the method of treatment comprises administration of multiple doses of the immunoconjugate and VEGF inhibitor.
  • the composition may be for use in treating neovascularization and/or angiogenesis.
  • the composition may be for use in treating ocular disorders.
  • the composition may be for use in treating ocular disorders associated with neovascularization and/or angiogenesis. In an embodiment, the composition may be for use in treating wet AMD, ocular neovascularization, or tumor neovascularization.
  • the monomer subunits of the immunoconjugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor protein is fused to only one of the Fc monomers.
  • compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best-corrected visual acuity
  • CNV mean choroidal neovascularization
  • compositions for use in treating, preventing, inhibiting, or reversing wet age-related macular degeneration comprisin the composition comprises (a) an immunoconj ugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVlla) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor.
  • a separate composition comprising a VEGF inhibitor may also be administered.
  • the monomer subunits of the immunoconj ugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers.
  • compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best-corrected visual acuity
  • CNV mean choroidal neovascularization
  • compositions for use in treating, preventing, inhibiting, or reversing ocular neovascularization comprisin the composition comprises (a) an immunoconjugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor.
  • an immunoconjugate dimer wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain
  • a VEGF inhibitor for compositions comprising only (a), a separate composition comprising a VEGF inhibitor may also be administered.
  • the monomer subunits of the immunoconjugate dimer each comprises a mutated human factor Wa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor W protein is fused to only one of the Fc monomers.
  • compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best-corrected visual acuity
  • CNV mean choroidal neovascularization
  • compositions for use in treating or reversing tumor neovascularization comprising (a) an immunoconj ugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (-Vila) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor.
  • a separate composition comprising a VEGF inhibitor may also be administered.
  • the monomer subunits of the immunoconj ugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the immunoconj ugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers.
  • compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
  • BCVA best-corrected visual acuity
  • CNV mean choroidal neovascularization
  • compositions for use in a method of treatment comprising an 0.3 mg, 0.6 mg, or 0.9 mg of an immunoconj ugate dimer, wherein monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated or fused to the human immunoglobulin Gl (IgGl) Fc domain; and wherein the method of treatment comprises administration of 0.5 mg of ranibizumab or 2.0 mg of aflibercept; and wherein multiple doses of the ranibizumab or aflibercept, an multiple does of the imm unoconj ugate dimer are administered monthly for at least three months; and wherein the composition is for use in treating ocular disorders associated with neovascularization and/or angiogenesis; and wherein the composition is for use in treating an eye of a patient in need thereof to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal
  • BCVA best-corrected visual acuity
  • FIG. 1 is a non-limiting diagram of two ICON-1 immunoconj ugate embodiments and one ICON- 1.5 immunoconj ugate embodiment of the present invention.
  • FIG. 2 is a graph of the rate of the intrinsic factor Xase complex (Fxase) hydrolysis (increase in absorbance at 405 nm - mOD/min) as a function of time.
  • FIG. 3 is a graph of thrombin generation by the known inhibitor of coagulation, active site inhibited FVTIa (FVIIai), as a function of time in normal pooled plasma.
  • FIG. 4 is a graph of thrombin generation by hl-conl, as a function of time in normal pooled plasma.
  • FIG. 5 is a graph of thrombin generation by human Factor Vila and hl-conl as a function of time in FVII-depleted plasma.
  • FIG. 6 is a graph of thrombin generation by hl-conl, as a function of time in rabbit plasma.
  • FIG. 7 is a graph of thrombin generation by hl-conl or FVIIai as a function of time in centrifuged rabbit plasma.
  • FIG. 8 is a graph showing the percent CNV in the pig as a function of intra vitreal dose of hl-conl.
  • Intravitreal injections 100 uL/eye
  • solutions of hl-conl (0.25, 0.5, 1.0 and 2.0 mg/mL) were injected into both eyes of mini-pigs on Day 10; control animals received 100 uL of formulation buffer.
  • the animals were sacrificed and the % CNV was determined.
  • FIG. 9 is a graph showing the percent CNV in the pig as a function of intravitreal dose of a 100 kDa fragment of hl-conl.
  • Intravitreal injections 100 uL/eye
  • solutions of hl-conl (0.25, 0.5, 1.0 and 2.0 mg/mL) were injected into both eyes of mini-pigs on Day 10; control animals received 100 uL of formulation buffer.
  • control animals received 100 uL of formulation buffer.
  • the animals were sacrificed and the % CNV was determined.
  • FIG. 10 is an image representing the distribution of patients within treatment arms with indications for dosing schedules that are further delineated by treatment induction and treatment extension.
  • FIG. 11 is a tabulation of the baseline CNV lesion area (mm 2 ) for the patients within each of the treatment arms compared with the mean CNV lesion area change from baseline.
  • FIG. 12 is a tabulation of the change in lesion size for the patients within each of the treatment arms, the total number of patients exhibiting the change (or no change), and the corresponding change in BCVA.
  • FIG. 13 is a graph depicting the proportional change/no change in lesion size for the patients within each of the treatment arms.
  • FIG. 14 is a graph displaying the mean change in CST over time in the study eyes in all treatment arms.
  • FIG. 15 is a graph displaying the mean change in BCVA score over time in the study eyes in all treatment arms.
  • FIG. 16 is a tabulation of the gain of BCVA from baseline at month 6 for patients in all treatment arms. The figure further identifies the proportion of patients with BCVA with >71 at month 6, proportion of patients with BCVA with ⁇ 33 letters at month 6, and the mean BCVA change in letter score at month 6.
  • FIG. 17 is a tabulation of the fluid type (I F, SRF, and Sub-RPE) at baseline for patients within each of the treatment arms. The figure further identifies the number of patients with no IRF, no SRF, no sub-RPE, and dry retina at 6 months for each of the treatment arms.
  • FIG. 18 is a graph identifying the percent of patients exhibiting dry retina within each of the treatment arms.
  • FIG. 19 is a tabulation of the patients within each of the treatment arms that received study drug retreatment, and a further indication of the number of retreatments.
  • FIG.20 is a graph identifying the time from treatment end to first retreatment for patients in each of the treatment arms.
  • FIG. 21 is a graph identifying the durability of treatment for patients within each of the treatment arms. Durability of treatment is represented as a proportion of the patients in each of the treatment arms that did not require retreatment
  • FIG. 22 is a graph depicting a dose-dependent reduction in lesion fluorescence in pigs that underwent bilateral laser induction of laser spots in the eyes; measured at day 14 post- induction.
  • the pigs were administered a vehicle control, ICON-1 at 300pg, ICON-1 at 6(X ⁇ g, ICON-1 at 900 ⁇ & or Eylea at 2mg.
  • FIG. 23 is a graph depicting a reduction in lesion fluorescence in pigs that underwent bilateral laser induction laser spots in the eyes; measured at day 14 post-induction.
  • the pigs were administered a vehicle control, ICON-1 at 600ug, Eylea at 2mg, or ICON-1 at 600pg and Eylea at 2mg.
  • a or “an” may refer to one or more of that entity, i.e. can refer to plural referents. As such, the terms “a” or “an”, “one or more” and “at least one” are used interchangeably herein.
  • reference to “an element” by the indefinite article “a” or “an” does not exclude the possibility that more than one of the elements is present, unless the context clearly requires that there is one and only one of the elements.
  • a "patient” refers to a warm-blooded animal, e.g., rat, mouse, sheep, cow, pig, guinea pig, non-human primate, human primate, wherein males and/or females may be a patient.
  • classic CNV means a well-defined CNV area that results in vision that is between 20/25 and 20/400, but may be worse than 20/800.
  • occupancy CNV means a poorly delineated CNV area that exhibits less leakage than classic CNV, and results in vision that is between 20/25 and 20/400, but may be worse than 20/800.
  • improved means at least a 5% improvement of the outcome for which "improved” modifies, as compared to a corresponding control, e.g., an improved reduction in mean CNV lesion area is at least a 5% improvement of the mean CNV lesion area as compared to the mean CNV lesion area under control conditions.
  • antibody includes any monoclonal antibody, polyclonal antibody, multispecific antibody, bispecific antibody, single-chain antibody, a single-chain variable fragment (scFv) of an antibody, FAB fragment of an antibody, and fragments thereof.
  • scFv single-chain variable fragment
  • CNV activity includes new or increased fluid and/or leakage, hemorrhage and/or lesion, persistent fluid, and decreased chorioretinal blood flow at the site of the perceived CNV lesion.
  • Pathologic angiogenesis the induction of the growth of existing blood vessels from the vessels in surrounding tissue, is observed in a variety of diseases, typically triggered by the release of specific growth factors for vascular endothelial cells.
  • Pathologic angiogenesis can result in neovascularization, i.e., the creation of new blood vessels, enabling solid tumor growth and metastasis, causing visual malfunction in ocular disorders, promoting leukocyte extravasation in inflammatory disorders, and/or influencing the outcome of cardiovascular diseases such as atherosclerosis.
  • a patient having a disease associated with neovascularization and/or angiogenesis such as cancer, rheumatoid arthritis, the exudative ("wet") form of macular degeneration, and/or atherosclerosis
  • administration may be local or systemic, depending upon the type of pathological condition involved in the therapy.
  • patient includes both humans and other species, including other mammal species.
  • the invention thus has both medical and veterinary applications.
  • immunoconj ugates are constructed using targeting and effector domains derived from the corresponding species.
  • the disease associated with neovascularization and/or angiogenesis is wet AMD.
  • the disease associated with neovascularization and/or angiogenesis is a cancer (ocular melanoma), atherosclerosis, rheumatoid arthritis, oculr melanoma, diabetic macular edema (DME), macular edema following retinal vein occlusion (RVO), proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), reinopathy of prematurity (ROP), or neovascular glaucoma.
  • cancer ocular melanoma
  • atherosclerosis rheumatoid arthritis
  • oculr melanoma oculr melanoma
  • DME diabetic macular edema
  • RVO retinal vein occlusion
  • proliferative diabetic retinopathy wet age-related macular degeneration
  • ROP reinopathy of prematur
  • immunoconjugate or “immunoconj ugates” refer to two chemically conjugated or fusion proteins: (1) ICON-1 (referred to interchangeably as hl-conl), a two-armed FVII-Fc fusion protein comprising two dimerized immunoglobulin (Ig) Fc monomers fused to two mutated FVII proteins; and (2) ICON- 1.5, a one-armed FW-Fc fusion protein comprising two dimerized immunoglobulin (Ig) Fc monomers, and a mutated FVII protein, wherein the mutated FVII protein is fused to only one of the Fc monomers. (See FIG. 1 for exemplary embodiments).
  • an immunoconj ugate has as an effector domain, an immunoglobulin Fc domain, and said effector domain is conjugated to a targeting domain comprising a mutant form of human factor VII.
  • an immunoconj ugate comprises an Fc domain of a human IgGl immunoglobulin conjugated to a targeting domain exhibiting decreased coagulation comprising a mutant form of factor W comprising one or two mutations selected from S344A and/or K341A, wherein the immunoconj ugate protein binds to tissue factor.
  • immunoconjugates of the present disclosure include immunoconjugates described in published international patent application WO/2017/18114S; and U.S. Patents 7,858,092; 8,388,974, 8,071,104; 7,887,809; and 6,924,359.
  • ICON-1.5 and ICON-1 share similar degrees of binding and ADCC activity, and FXa conversion.
  • FIG. 1 provides the generalized structure of one embodiment of an immunoconj ugate that can be administered by the methods provided herein.
  • the mutated Factor Vila domain also referred to as the TF targeting domain
  • the IgGl Fc domain (also referred to as the effector domain) triggers a cytolytic response against cells which bind the immunoconj ugate, by the natural killer (NK) cell and complement pathways.
  • the IgGl Fc effector domain comprises both the CH2 and CH3 regions of the IgGl Fc region.
  • the reaction between FVHa and TF is species-specific (Janson et al., 1984; Schreiber et al., 2005; Peterson et al., 2005): murine FVII appears to be active in many heterologous species including rabbit, pig and human, whereas human FVIIa is only appreciably active in human, dog, rabbit and pig. Conversely, the human IgG Fc domain is active in both humans and mice. Accordingly, depending on the patient, the immunoconjugate is constructed using targeting and effector domains derived from the corresponding species, or from a species that is known to be active in the patient.
  • the mutated tissue factor targeting domain is derived from human Factor Vila conjugated to an effector domain comprising the Fc region of a human IgGl immunoglobulin.
  • the immunoconjugate is a protein of SEQ ID NO: 2.
  • the immunoconjugate is a protein of SEQ ID NO: 3.
  • the immunoconjugate is encoded by the mRNA sequence of SEQ ID NO: 1, 4, or 5.
  • the immunoconjugate described herein comprises two protein chains, each comprising a targeting domain joined to an effector domain via a linker or hinge region.
  • the linker or hinge region is naturally occurring, and in one embodiment, is of human origin.
  • the hinge region of an IgGl immunoglobulin for example the hinge region of the human IgGl immunoglobulin, in one embodiment, is used to link the targeting domain to the effector domain.
  • the hinge region of IgGl includes cysteine amino acids which form one or more disulfide bonds between the two monomer chains (e.g., as depicted in FIG. 1).
  • the immunoconjugate is a homodimer.
  • the immunoconjugate is a heterodimer, for example, an immunoconjugate comprising two monomers each having a targeting domain of a different amino acid sequence, but the same effector domains.
  • the amino acid sequences of the two targeting domains in one embodiment, differ by one amino acid, two or more amino acids, three or more amino acids or five or more amino acids.
  • each monomer subunit comprises an IgGl hinge region that links the targeting region and effector region of the immunoconjugate, and the monomer subunits of the immunoconj ugate heterodimer or the immunoconjugate homodimer are linked together via a disulfide bond between IgGl hinge regions.
  • the molecular weight of the ICON-1 immunoconjugate provided herein is from about ISO kDa to about 200 kDa. In another embodiment, the molecular weight of the immunoconjugate is about 1S7 kDa or 1S7 kDa.
  • the immunoconjugate in one embodiment is the immunoconjugate having the amino acid sequence set forth in SEQ ID NO: 2, also referred to herein as "hl-conl” or "ICON-1" (used interchangeably herein). In another embodiment, the immunoconjugate has the amino acid sequence set forth in SEQ ID NO: 3.
  • an immunoconjugate comprising a tissue factor targeting domain comprising a mutated Factor Vila domain.
  • the targeting domain comprises a mutated Factor Vila that has been mutated to inhibit initiation of the coagulation pathway without reducing binding affinity to tissue factor.
  • the mutation in Factor Vila is a single point mutation at residue 341.
  • the mutation is from Lys341 to Ala341.
  • other mutations that inhibit the coagulation pathway are encompassed by the immunoconj ugates provided herein.
  • the effector domain of the immunoconj ugates provided herein mediates both complement and natural killer (NK) cell cytotoxicity pathways.
  • compositions comprising the immunoconjugates of the invention.
  • methods of producing the immunoconjugate include expression in mammalian cells such as BHK cells.
  • cell lines may include HEK 293, CHO, and SP2/0.
  • Immunoconjugates may be generated by mammalian expression of the expression constructs.
  • the immunoconjugates are produced as fusion proteins (FVTi-Fc) or produced as chemical conjugates.
  • the immunoconjugate is post-translationally modified.
  • Post- translational modification includes: myristoylation, glypiation, palmitoylation, prenylation, lipoylation, acylation, alkylation, butrylation, gamma-carboxylation, glycosylation (N- glycosylation, O-glycosylation, fucosylation, and mannosylation), propionylation, succinylation, and sulfation.
  • the immunoconjugate dimer is administered in a co-therapeutic regimen with a VEGF inhibitor to treat a patient for one of the aforementioned diseases or disorders, for example, to treat wet AMD or another ocular disease associated with angiogenesis or neovascularization.
  • the VEGF inhibitor is administered in the same composition as the immunoconjugate dimer.
  • the immunoconjugate dimer and VEGF inhibitor are administered in separate compositions.
  • the VEGF inhibitor is administered prior to the immunoconjugate dimer.
  • the VEGF inhibitor is administered subsequent to the immunoconjugate dimer.
  • the VEGF inhibitor is administered simultaneously with the immunoconjugate dimer.
  • the VEGF inhibitor is an anti-VEGF antibody.
  • the VEGF inhibitor is ranibizumab or bevacizumab.
  • the VEGF in inhibitor is ranibizumab.
  • ranibizumab is administered at a dosage of 0.S mg or 0.3 mg per dosing session, and is administered as indicated in the prescribing information for LUCENTIS.
  • VEGF inhibitors may be selected from ranibizumab, bevacizumab, pazopanib, sunitinib, sorafenib, axitinib, regorafenib, ponatinib, cabozantinib, vandetanib, ramucirumab, lenvatinib, aflibercept, and ziv-aflibercept.
  • Administration methods encompassed by the methods provided herein include intravitreal injection, suprachoroidal injection, topical administration (e.g., eye drops), intravenous and intratumoral administration.
  • administration is via intravenous, intramuscular, intratumoral, subcutaneous, intrasynovial, intraocular, intraplaque, or intradermal injection of the immunoconjugate or of a replication-deficient adenoviral vector, or other viral vectors carrying a cDNA encoding a secreted form of the immunoconjugate.
  • the patient in need of treatment is administered one or more immunoconjugate dimers via intravitreal, intravenous or intratumoral injection, or injection at other sites, of one or more immunoconjugate proteins.
  • a patient in need of treatment is administered one or more immunoconjugate dimers via intravenous or intratumoral injection, or injection at other sites, of one or more expression vectors carrying a cDNA encoding a secreted form of one or more of the immunoconjugate dimers provided herein.
  • the patient is treated by intravenous or intratumoral injection of an effective amount of one or more replication-deficient adenoviral vectors, or one or more adeno-associated vectors carrying cDNA encoding a secreted form of one or more types of immunoconjugate proteins.
  • the patient in need of treatment is co-administered one or more immunoconjugate dimers and VEGF inhibitors via intravitreal, intravenous or intratumoral injection, or injection at other sites, of one or more immunoconjugate proteins and VEGF inhibitors.
  • a patient in need of treatment is co-administered one or more immunoconjugate dimers and VEGF inhibitors via intravenous or intratumoral injection, or injection at other sites, of one or more expression vectors carrying a cDNA encoding a secreted form of one or more of the immunoconjugate dimers provided herein.
  • a therapeutically effective amount of a therapeutic agent means a level or amount of a therapeutic agent needed to treat a condition or disease of the present disclosure, or the level or amount of a therapeutic agent that produces a therapeutic response or desired effect in the subject to which the therapeutic agent was administered; wherein a therapeutic agent is an immunoconjugate of the present disclosure.
  • the therapeutic agent may further include an immunoconjugate of the present disclosure and a VEGF inhibitor of the present disclosure.
  • a therapeutically effective amount of a therapeutic agent such as an immunoconjugate of the present disclosure and a VEGF inhibitor of the present disclosure, is an amount that is effective in reducing one or more symptoms of angiogenesis and/or neovascularization, as well as various forms of AMD.
  • pharmaceutical composition means a composition comprising a therapeutic agent
  • treatment means the following actions: (i) preventing a particular disease or disorder from occurring in a subject who may be predisposed to the disease or disorder but has not yet been diagnosed as having it; (ii) curing, treating, or inhibiting the disease, i.e., arresting its development; or (iii) ameliorating or reversing the disease by reducing or eliminating symptoms, conditions, and/or by causing regression of the disease.
  • a method of intravitreal injection is employed.
  • aseptic technique is employed when preparing the immunoconjugate dimer and/or VEGF inhibitor for injection, for example, via the use of sterile gloves, a sterile drape and a sterile eyelid speculum (or equivalent).
  • the patient is subjected to anesthesia and a broad-spectrum microbicide prior to the injection.
  • intravitreal injection of one or more of the VEGF inhibitors and/or immunoconjugate dimers, provided herein, for example the immunoconjugate dimer of SEQ ID NO: 2 is prepared by withdrawing the vial contents of the immunoconjugate dimer composition solution and/or the VEGF inhibitor composition solution through a 5-micron, 19-guage filter needle attached to a 1-cc tuberculin syringe.
  • the filter needle in a further embodiment, is then discarded and replaced with a sterile 30-gauge x 1 ⁇ 2-inch needle for the intravitreal injection.
  • the contents of the vial are expelled until the plunger tip is aligned with the line on the syringe that marks the appropriate dose for delivery.
  • the patient is monitored for elevation in intraocular pressure (IOP).
  • IOP intraocular pressure
  • the patient prior to and/or after the ocular injection, is monitored for elevation in IOP using tonometry.
  • the patient is monitored for increases in IOP via a check for perfusion of the optic nerve head immediately after the injection.
  • the patient is monitored for elevation in IOP.
  • the patient's IOP is substantially the same prior to intraocular injection of an immunoconjugate dimer and/or the VEGF inhibitor, as compared to after intraocular injection of the immunoconjugate dimer and/or VEGF inhibitor.
  • the patient's IOP varies by no more than 10%, no more than 20% or no more than 30% after intraocular injection, as compared to prior to intraocular injection (e.g., intravitreal injection).
  • the treatment methods provided herein in one embodiment, comprise a single administration of one of the immunoconjugate dimers and/or VEGF inhibitors provided herein (e.g., an immunoconjugate of SEQ ID NO: 2 or 3).
  • the treatment methods provided herein comprise multiple dosing sessions.
  • the multiple dosing sessions are multiple intraocular injections of one of the immunoconjugate dimers and/or VEGF inhibitors described herein.
  • the multiple dosing sessions in one embodiment comprise two or more, three or more, four or more or five or more dosing sessions.
  • each dosing session comprises intraocular injection of one of the immunoconj ugates and/or VEGF inhibitors described herein, or intratumoral injection of one of the immunoconj ugates and/or VEGF inhibitors described herein (i.e., either as the expressed protein or via a vector encoding the soluble immunoconjugate).
  • from about 2 to about 24 dosing sessions are employed, for example, from about 2 to about 24 intraocular dosing sessions (e.g., intravitreal or suprachoroidal injection).
  • from about 3 to about 30, or from about 5 to about 30, or from about 7 to about 30, or from about 9 to about 30, or from about 10 to about 30, or from about 12 to about 30 or from about 12 to about 24 dosing sessions are employed.
  • the dosing sessions are spaced apart by from about 10 days to about 60 days, or from about 10 days to about 50 days, or from about 10 days to about 40 days, or from about 10 days to about 30 days, or from about 10 days to about 20 days. In another embodiment, where multiple dosing sessions are employed, the dosing sessions are spaced apart by from about 20 days to about 60 days, or from about 20 days to about SO days, or from about 20 days to about 40 days, or from about 20 days to about 30 days.
  • the multiple dosing sessions are bi-weekly (e.g., about every 14 days), monthly (e.g., about every 30 days), or bi-monthly (e.g., about every 60 days). In yet another embodiment, the dosing sessions are spaced apart by about 28 days.
  • the multiple dosing sessions comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 dosing sessions, wherein the dosing sessions are spaced apart by 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 35 days, 40 days, 45 days, 50 days, 55 days, or 60 days.
  • an immunoconjugate dimer and/or VEGF inhibitor provided herein is administered to the eye of a patient in need of treatment of wet age-related macular degeneration (AMD).
  • the treatment comprises multiple dosing sessions of the immunoconjugate dimer and/or VEGF inhibitors.
  • the immunoconjugate dimer comprises monomer subunits that each include a mutated human factor VQa (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
  • the method of treating wet AMD comprises preventing, inhibiting or reversing choroidal neovascularization in the eye of the patient in need of treatment.
  • choroidal neovascularization is reversed by at least about 10%, at least about 20%, at least about 30% or at least about 40% after treatment, as compared to the choroidal neovascularization that was present in the afflicted eye of the patient prior to treatment
  • ocular disorders associated with ocular neovascularization are treatable with the immunoconj ugates and VEGF inhibitors and methods provided herein.
  • the ocular neovascularization in one embodiment, is choroidal neovascularization.
  • the ocular neovascularization is retinal neovascularization.
  • the ocular neovascularization is corneal neovascularization. Accordingly, an ocular disorder associated with choroidal, retinal or corneal neovascularization, in one embodiment, is treatable by one or more of the methods provided herein.
  • the method comprises administering to the eye of a patient in need thereof, the immunoconj ugate dimers and/or VEGF inhibitors described herein.
  • the treatment comprises multiple dosing sessions of the immunoconjugate dimer and/or VEGF inhibitors.
  • the immunoconj ugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
  • a patient in need of treatment of proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma is treated with the immunoconj ugates and/or VEGF inhibitors provided herein, for example, via intravitreal injection, suprachoroidal injection or topical administration (e.g., via eye drops) of the immunoconjugate and VEGF inhibitors into the affected eye.
  • Treatment in one embodiment occurs over multiple dosing sessions.
  • ocular neovascularization is said to be "associated with" or "secondary to" the respective disorder.
  • a patient in need of treatment of macular edema following retinal vein occlusion (RVO) is treated by one of the immunoconjugate dimers and VEGF inhibitors provided herein.
  • the method comprises administering to the patient a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVHa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • fVHa mutated factor Vila
  • the mutated fVIIa protein is a human mutated fVHa protein and is linked to the IgGl Fc domain via the hinge region of IgGl.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
  • the immunoconjugate dimer is administered to the patient over multiple dosing sessions, for example, via intravitreal administration at each dosing session.
  • a patient in need of treatment of diabetic macular edema is treated by one of the immunoconjugate dimers and VEGF inhibitors provided herein.
  • the method comprises administering to the patient a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer is administered to the patient over multiple dosing sessions. In even a further embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered intravitreally at each dosing session.
  • diabetic retinopathy is treated via one of the immunoconj ugates and VEGF inhibitors provided herein, in a patient in need thereof, for example, a patient with DME.
  • the method comprises administering to the patient, for example a DME patient, a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor VHa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • fVIIa mutated factor VHa
  • IgGl human immunoglobulin Gl
  • the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl .
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
  • the immunoconjugate dimer is administered to the patient over multiple dosing sessions.
  • the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions, for example, via intravitreal administration at each dosing session.
  • one or more of the immunoconjugates and VEGF inhibitors provided herein is used in a method to treat a disease or disorder associated with tumor neovascularization in a patient in need thereof, for example, a cancer patient
  • the method comprises administering to the patient, for example via intratumoral or intravenous injection, a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitor, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • fVTIa mutated factor Vila
  • the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
  • the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions.
  • the immunoconjugate dimer and VEGF inhibitors are used for treating a variety of cancers, particularly primary or metastatic solid tumors, including melanoma, renal, prostate, breast, ovarian, brain, neuroblastoma, head and neck, pancreatic, bladder, endometrial and lung cancer.
  • the cancer is a gynecological cancer.
  • the gynecological cancer is serous, clear cell, endometriod or undifferentiated ovarian cancer.
  • the immunoconjugate dimer and/or VEGF inhibitor in one embodiment is employed to target the tumor vasculature, particularly vascular endothelial cells, and/or tumor cells.
  • targeting the tumor vasculature offers several advantages for cancer immunotherapy with one or more of the immunoconjugate dimers and/or VEGF inhibitors described herein, as follows, (i) some of the vascular targets including tissue factor should be the same for all tumors; (ii) immunoconjugates targeted to the vasculature do not have to infiltrate a tumor mass in order to reach their targets; (iii) targeting the tumor vasculature should generate an amplified therapeutic response, because each blood vessel nourishes numerous tumor cells whose viability is dependent on the functional integrity of the vessel; and (iv) the vasculature is unlikely to develop resistance to an immunoconjugate, because that would require modification of the entire endothelium layer lining a vessel. Unlike previously described antiangiogenic methods that inhibit new vascular growth, immunoconjugate dimers provided herein elicit a cytolytic response to the neovasculature.
  • one or more of the immunoconjugates and VEGF inhibitors described herein is used in a method for treating atherosclerosis or rheumatoid arthritis.
  • the method comprises administering to the patient in need of treatment a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitor, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVna) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain.
  • the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl.
  • the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions.
  • a method for treating an ocular disorder with an immunoconjugate dimer and a VEGF inhibitor for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD
  • the patient subjected to the treatment method substantially maintains his or her vision subsequent to the treatment (e.g., the single dosing session or multiple dosing sessions), as measured by losing fewer than IS letters in a best-corrected visual acuity (BCVA) measurement, compared to the patient's BCVA measurement prior to undergoing treatment
  • the patient loses fewer than 10 letters, fewer than 8 letters, fewer than 6 letters or fewer than 5 letters in a BCVA measurement, compared to the patient's BCVA measurement prior to undergoing treatment
  • a patient having been administered an immunoconjugate and/or VEGF inhibitor of the present invention loses fewer than 10, 9, 8, 7, 6, or 5 letters in a BCVA measurement, compared to a patient's BCVA measurement prior to undergoing treatment In some embodiments, the patient loses fewer than about 10, about 9, about 8, about 7, about 6, or about 5 letters in a BCVA measurement, compared to a patient's BCVA measurement prior to undergoing treatment.
  • a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention loses fewer than between 15 and 5, 15 and 6, 15 and 7, 15 and 8, 15 and 9, 15 and 10, 10 and 5, 10 and 6, 10 and 7, 10 and 8, 10 and 9, 9 and 5, 9 and 6, 9 and 7, 9 and 8, 8 and 5, 8 and 6, 8 and 7, 7 and 5, 7 and 6, or 6 and 5 letters in a BCVA measurement.
  • a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention loses fewer than between about 15 and about 5, about 15 and about 6, about 15 and about 7, about 15 and about 8, about 15 and about 9, about 15 and about 10, about 10 and about 5, about 10 and about 6, about 10 and about 7, about 10 and about 8, about 10 and about 9, about 9 and about 5, about 9 and about 6, about 9 and about 7, about 9 and about 8, about 8 and about 5, about 8 and about 6, about 8 and about 7, about 7 and about 5, about 7 and about 6, or about 6 and about 5 letters in a BCVA measurement
  • a method for treating an ocular disorder with an immunoconj ugate dimer for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD
  • the patient subjected to the treatment method substantially maintains his or her vision subsequent to the treatment (e.g., the single dosing session or multiple dosing sessions), as measured by BCVA measurement
  • a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining 5, 6, 7, 8, 9, 10, 15, 20, or 25 or more letters in a best- corrected visual acuity (BCVA) measurement, compared to the patient's BCVA prior to the treatment.
  • BCVA visual acuity
  • a patient having been administered an immunoconj ugate and/or VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining about 5, about 6, about 7, about 8, about 9, about 10, about 15, about 20, or about 25 or more letters in a best-corrected visual acuity (BCVA) measurement, compared to the patient's BCVA prior to the treatment.
  • BCVA best-corrected visual acuity
  • a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining greater than between S and 25, 5 and 20, 5 and 15, 5 and 10, 5 and 9, 5 and 8, 5 and 7, 5 and 6, 6 and 25, 6 and 20, 6 and 15, 6 and 10, 6 and 9, 6 and 8, 6 and 7, 7 and 25, 7 and 20, 7 and 15, 7 and 10, 7 and 9, 7 and 8, 8 and 25, 8 and 20, 8 and 15, 8 and 10, 8 and 9, 9 and 25, 9 and 20, 9 and 15, 9 and 10, 10 and 25, 10 and 20, 10 and 15, 15 and 25, 15 and 20, or 20 and 25 or more letters in a BCVA measurement, compared to the patient's BCVA prior to the treatment.
  • a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining greater than between about 5 and about 25, about 5 and about 20, about 5 and about 15, about 5 and about 10, about 5 and about 9, about 5 and about 8, about 5 and about 7, about 5 and about 6, about 6 and about 25, about 6 and about 20, about 6 and about 15, about 6 and about 10, about 6 and about 9, about 6 and about 8, about 6 and about 7, about 7 and about 25, about 7 and about 20, about 7 and about 15, about 7 and about 10, about 7 and about 9, about 7 and about 8, about 8 and about 25, about 8 and about 20, about 8 and about 15, about 8 and about 10, about 8 and about 9, about 9 and about 25, about 9 and about 20, about 9 and about 15, about 9 and about 10, about 10 and about 25, about 10 and about 20, about 10 and about 15, about 15 and about 25, about 15 and about 20, or about 20 and about 25 or more letters in a BCVA measurement, compared to the patient's BCVA prior to
  • a method for treating an ocular disorder with an immunoconj ugate dimer and a VEGF inhibitor for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD
  • the ocular neovascularization area e.g., the choroidal neovascularization area is reduced in the eye of the patient, as compared to the ocular neovascularization area (e.g., CNV area) prior to treatment.
  • treatment can include one dosing session or multiple dosing sessions, and reduction in ocular neovascularization ana (e.g., CNV area), in one embodiment, is assessed after individual dosing sessions, or multiple dosing sessions.
  • the ocular neovascularization area e.g., CNV area
  • the ocular neovascularization area is reduced by at least about 5%, or at least about 10%, or at least about 15%, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, as measured by fluorescein angiography.
  • a method for treating an ocular disorder with an immunoconjugate timer and a VEGF inhibitor
  • a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD the retinal thickness of the treated eye is reduced in the eye of the patient, as compared to the retinal thickness prior to treatment, as measured by optical coherence tomography (OCT).
  • OCT optical coherence tomography
  • treatment can include one dosing session or multiple dosing sessions, and reduction in retinal thickness, in one embodiment, is assessed after individual dosing sessions, or multiple dosing sessions.
  • the retinal thickness is reduced by at least about 5%, or at least about 10%, or at least about 15%, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, as measured by OCT.
  • the decreased retinal thickness is decreased central retinal subfield thickness (CSTX decreased center point thickness (CPT), or decreased central foveal thickness (CFT).
  • the immunoconjugate dimer and/or VEGF inhibitor is administered as a solution or a suspension.
  • the immunoconjugate composition and/or VEGF inhibitor composition in one embodiment, comprises argunne or protein A.
  • the immunoconjugate composition and/or VEGF inhibitor composition comprises argimne.
  • the argininc is present in the composition at from about 20 mM to about 40 mM, e.g, at 25 mM.
  • Other components of the composition include HEPES, sodium chloride, polysorbate-80, calcium chloride, or a combination thereof.
  • the immunoconjugate dimer and/or VEGF inhibitor is administered in a dose of between 10 pg and 600 pg, 10 pg and 500 pg, 10 pg and 400 pg, lOug and 300 ⁇ g, 10 ⁇ g and 200 ⁇ g, 10 ⁇ g and 100 ⁇ g, 10 ⁇ g and SO ⁇ , SO ⁇ g and 600 pg, SO ⁇ g and 500 ⁇ & 50 ⁇ g and 400 ⁇ & 50 ⁇ g and 300 ⁇ g, 50 ⁇ and 200 ⁇ & 50 ⁇ and ⁇ 100 ⁇ g and 600 ⁇ 3 ⁇ 4, 100 ⁇ g and 500 ⁇ 3 ⁇ 4 100 ⁇ g and 400 ⁇ 3 ⁇ 4 100 ⁇ 3 ⁇ 4 and 300 ⁇ 3 ⁇ 4, 100 ⁇ g and 200 ⁇ & 200 ⁇ g and 600 ⁇ 3 ⁇ 4 200 ⁇ 3 ⁇ 4 and 500 ⁇ & 200 ⁇ g and 400 ⁇ & 200 ⁇ g and 300 ⁇ 3 ⁇ 4, 100 ⁇ g and 200 ⁇ & 200 ⁇ g
  • the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of between about 10 ⁇ g and about 500 ⁇ g, about 10 ⁇ g and about 400 ⁇ g, about 10 ⁇ g and about 300 ⁇ g, about 10 ⁇ g and about 200 ⁇ g, about 10 ⁇ g and about 100 ⁇ g, about 10 ⁇ g and about 50 ⁇ g, about 50 ⁇ g and about 500 ug, about 50 ⁇ and about 400 ⁇ g, about 50 ug and about 300 ⁇ g, about 50 ⁇ g and about 200 ug, about 50 ug and about 100 ⁇ , about 100 ⁇ g and about 500 ⁇ g, about 100 ug and about 400 ⁇ g, about 100 ⁇ g and about 300 ⁇ g, about 100 ug and about 200 ⁇ , about 200 ⁇ g and about 500 ⁇ g, about 200 ug and about 400 ⁇ g, about 200 ⁇ g and about 300 ⁇ g, about 300 ug and about 200 ⁇ , about 200 ⁇ g and about 500
  • the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.40 mg, about 0.45 mg, about 0.50 mg, about 0.55 mg, about 0.60 mg, about 0.65 mg, about 0.7 mg, about 0.75 mg, about 0.8 mg, about 0.9 mg, about 0.95 mg, about 1 mg, about 1.05 mg, about 1.1 mg, about 1.15 mg, about 1.2 mg, about 1.25 mg, about 1.30 mg, about 1.35 mg, about 1.4 mg, about 1.45 mg, or about 1.50 mg.
  • the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.3 mg, 0.35 mg, 0.40 mg, 0.45 mg, 0.50 mg, 0.55 mg, 0.60 mg, 0.65 mg, 0.7 mg, 0.75 mg, 0.8 mg, 0.9 mg, 0.95 mg, 1 mg, 1.05 mg, 1.1 mg, 1.15 mg, 1.2 mg, 1.25 mg, 1.30 mg, 1.35 mg, 1.4 mg, 1.45 mg, or 1.50 mg.
  • the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose consisting of about 10ug, about 20 ⁇ g, about 30 ug, about 40 ⁇ g, about 50 ⁇ g, about 60 ug, about 70 ⁇ g, about 80 ⁇ g, about 90 ⁇ g, about 100 ⁇ g, about 125 ⁇ g, about 150 ⁇ g, about 175 ⁇ g, about 200 ug, about 225 ⁇ g, about 250 ⁇ g, about 275 ⁇ g, about 300 ⁇ g, about 325 ⁇ g, about 350 ⁇ g, about 375 ug, about 400 ⁇ g, about 425 ⁇ g, about 450 ug, about 475 ⁇ g, about S00 ug, about S2S ug, about SSO ug, about S7S ug, about 600 ug, about 625 ug, about 6S0 ⁇ g, about 675 ug, or about 700 ug,
  • the immunoconjugate dimer and/or VEGF inhibitor is administered in a solute volume of between 10 uL and 200 uL, 10 uL and 180 uL, 10 uL and 160 uL, 10 uL and 140 uL, 10 uL and 120 uL, 10 uL and 100 uL, 10 uL and 80 uL, 10 uL and 60 uL, 10uL and 40 uL, 10 uL and 20 uL, 10 uL and 15 uL, 20 uL and 200 uL, 20uL and 180 uL, 20 uL and 160 uL, 20uL and 140uL, 20 uL and 120 uL, 20 uL and 100 uL, 20 uL and 80 uL, 20 uL and 60 uL, 20 uL and 40 uL, 40 uL and 200uL, 40 uL and 180 uL, 40 uL and 180 uL, 40 uL and
  • the immunoconjugate dimer and/or VEGF inhibitor is administered in a solute volume consisting of about 10uL, about 15 uL, about 20 uL, about 25 uL, about 30 uL, about 35 uL, about 40 uL, about 45 uL, about 50 uL, about 55 uL, about 60 uL, about 65 uL, about 70 uL, about 75 uL, about 80 uL, about 85 uL, about 90 uL, about 95 uL, or about 100 uL.
  • the administration of an immunoconjugate dimer of the invention and VEGF inhibitor results in an improved outcome compared to administration of the VEGF inhibitor alone.
  • the improved outcome is greater than an additive effect of the dimer and VEGF inhibitor.
  • the improved outcome is synergistic as compared to treatment with the dimer or VEGF alone.
  • Outcome can be quantified by BVCA letter score; central subfield retinal thickness; thickness in the tissues/regions of the eye; CNV area, lesion area; CNV-associated exudation; CNV leakage area; volume of sub- retinal fluid; thickness of the central subfield sub-retinal hyper-reflective material; volume of total sub-retinal hyper-reflective material; presence or absence of intraretinal fluid, sub-retinal fluid, and/or sub-retinal pigment epithelium fluid; presence or absence of subfoveal and/or non- subfoveal cysts; atrophy and/or fibrosis; area of autofluorescence; area of discontinuous autofluorescence; volume of the central subfield pigment epithelium detachment; and integrity of the eye's outer nuclear layer, external limiting membrane, ellipsoid zone, and/or subfoveal retinal pigment epithelium.
  • a patient is administered, in multiple dosing sessions, an effective amount of (1) an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain and (2) a VEGF inhibitor.
  • the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF inhibitor prevents, inhibits, or reverses wet age-related macular degeneration (AMD) in an eye of a patient in need thereof.
  • ATD wet age-related macular degeneration
  • the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF inhibitor prevents, inhibits, or reverses ocular neovascularization in an eye of a patient in need thereof. In a further embodiment, the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF reverses tumor neovascularization in an eye of a patient in need thereof.
  • a BCVA letter score is determined in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. Ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the BCVA letter score baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months.
  • the CNV is classical CNV.
  • the CNV is occult.
  • the CNV is minimally classic.
  • the assessment BVCA letter score determinations occur as a last observation carried forward (LOCF) method.
  • a patient gains greater than 5, 10, 15, 20, 25, 30, 35, or 40 letters in the BCVA letter score at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments, a patient gains greater than about 5, about 10, about 15, about 20, about 25, about 30, about 35, or about 40 letters in the BCVA letter score at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater improvement in the BCVA letter score, compared to a patient who received the VEGF inhibitor monotherapy.
  • the central subfield retinal thickness in the eye is determined in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the central subfield retinal thickness baseline is determined, and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months.
  • the CNV is classical CNV.
  • the CNV is occult.
  • the central subfield retinal thickness determinations occur as a last observation carried forward (LOCF) method.
  • the central subfield retinal thickness determination is made utilizing sdOCT.
  • a patient exhibits an increase or decrease in the central subfield retinal thickness by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient exhibits an increase or decrease in the central subfield retinal thickness by at least 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • the patient exhibits an increase or decrease in the central subfield retinal thickness of the tissues and/or regions of the eye presented herein is an increase or decrease of at least about lOum, about 20um, about 30 um, about 40 um, about 50 um, about 60 um, about 70 um, about 80 um, about 90 um, about 100 um, about 125 um, about 150 um, about 175 um, about 200 um, about 225 um, about 250 um, about 275 um, about 300 um, about 325 um, about 350 um, about 375 um, about 400 um, about 425 um, about 450 um, about 475 um, about 500 um, about 525 um, about 550 um, about 575 um, about 600 um, about 625 um, about 650 um, about 675 um, or about 700 um.
  • the measure of thickness of the tissues and/or regions of the eye presented herein is an increase or decrease of at least lOum, 20um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, 100 um, 125 um, 150 um, 175 um, 200 um, 225 um, 250 um, 275 um, 300 um, 325 um, 350 um, 375 um, 400 um, 425 um, 450 um, 475 um, 500 um, 525 um, 550 um, 575 um, 600 um, 625 um, 650 um, 675 um, or 700 um.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the central subfield retinal thickness, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the CNV area is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the CNV area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determinations of the CNV areas occur as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the CNV area by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a decrease in the CNV area by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the CNV area, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the area of the lesion is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g.
  • the lesion area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determinations of the area of lesions occur as a last observation carried forward (LOCF) method. See FIG. 23 for synergistic decrease of lesion size, as measured by CTLF for combination administration of ICON-1 + VEGF inhibitor.
  • a patient exhibits a decrease in the area of lesions by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a decrease in the area of lesions by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater in the area of lesions, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the CNV-associated exudation is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the CNV-associated exudation baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months.
  • the CNV- associated exudation is classical CNV.
  • the CNV is occult CNV.
  • the determinations of the CNV areas occur as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the CNV-associated exudation by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient exhibits a decrease in the CNV-associated exudation by at least at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater improvement in the BCVA letter score, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the area of leaking CNV is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups.
  • the leakage area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the measure of the leakage area occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the area of leakage area by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV. In other embodiments, the CNV is occult.
  • a patient exhibits a decrease in the area of leakage area by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the area of leakage, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the volume of the sub-retinal fluid is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the measure of the volume of the sub- retinal fluid baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the measure of the measure of the volume of the sub-retinal fluid occurs as a last observation carried forward (LOCF) method. [00157] In some embodiments, a patient exhibits a decrease or increase in the volume of the sub-retinal fluid by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV.
  • a patient exhibits a decrease or increase in the volume of the sub-retinal fluid by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the volume of the sub-retinal fluid, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the thickness of the central subfield subretinal hyper-reflective material is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • ICON-1 immunoconj ugate dimer monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • immunoconjugate dimer and VEGF inhibitor therapy treatment groups e.g. anti-VEGF antibody, e.g. ranibizumab
  • the measure of the thickness of the central subfield subretinal hyper-reflective material baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the measure of the thickness of the central subfield subretinal hyper- reflective material occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease or increase in the thickness of the central subfield subretinal hyper-reflective material by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • the CNV is classical CNV. In other embodiments, the CNV is occult CNV.
  • a patient exhibits a decrease or increase in the thickness of the central subfield subretinal hyper-reflective material by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the thickness of the central subfield subretinal hyper-reflective material, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measure of the total subretinal hyper-reflective material volume is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • ICON-1 immunoconjugate dimer monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • immunoconjugate dimer and VEGF inhibitor therapy treatment groups e.g. anti-VEGF antibody, e.g. ranibizumab
  • the measure of the total volume of the subretinal hyper-reflective material baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • distinctions are made between subfoveal versus non-subfoveal.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the measure of the total volume of the subretinal hyper-reflective material occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease or increase in the total volume of the subretinal hyper-reflective material by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • distinctions are made between subfoveal versus non-subfoveal.
  • a patient exhibits a decrease or increase in the total volume of the subretinal hyper-reflective material by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • distinctions are made between subfoveal versus non-subfoveal.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the total volume of the subretinal hyper-reflective material, compared to a patient who received the VEGF inhibitor monotherapy.
  • the identification of the presence or absence is made for (1) intraretinal fluid, (2) subretinal fluid, (3) subretinal pigment epithelium fluid in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • ICON-1 immunoconj ugate dimer monotherapy e.g. anti- VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti- VEGF antibody, e.g. ranibizumab
  • immunoconjugate dimer and VEGF inhibitor therapy treatment groups e.g. anti- VEGF antibody, e.g. ranibizumab
  • the baseline determination of the presence or absence of fluid in said ocular locations is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determination of the presence or absence of fluid in said ocular locations occurs as a last observation carried forward (LOCF) method.
  • LOCF last observation carried forward
  • a patient exhibits a presence or absence of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a presence or absence of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid, compared to a patient who received the VEGF inhibitor monotherapy.
  • ICON-1 immunoconj ugate dimer monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • immunoconj ugate dimer and VEGF inhibitor therapy treatment groups e.g. anti-VEGF antibody, e.g. ranibizumab
  • the baseline determination of the presence or absence of subfoveal or non-subfoveal cysts is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determination of the presence or absence of said cysts occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a presence or absence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient exhibits a decrease in the presence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a presence or absence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient exhibits a decrease in the presence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits the presence or absence of or about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% decrease in subfoveal or non-subfoveal cysts, compared to a patient who received the VEGF inhibitor monotherapy.
  • the identification of atrophy and/or fibrosis is made for the eye in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups.
  • the baseline identification of atrophy and/or fibrosis is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the presence or atrophy and/or fibrosis occurs as a last observation carried forward (LOCF) method.
  • LOCF last observation carried forward
  • a patient exhibits a decrease in the atrophy and/or fibrosis of the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a decrease in the atrophy and/or fibrosis of the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the atrophy and/or fibrosis of the eye, compared to a patient who received the VEGF inhibitor monotherapy.
  • the total area of decreased autofluorescence is determined for the in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • the baseline determination of the area of decreased autofluorescence is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determination of the total area of decreased autofluorescence occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the total area of autofluorescence the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient exhibits a decrease in the total area of autofluorescence the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the total area of autofluorescence, compared to a patient who received the VEGF inhibitor monotherapy.
  • the total area of discontinuous autofluorescence in the eye is determined for a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups.
  • the baseline determination of the total area of discontinuous autofluorescence is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determination of the total area of discontinuous autofluorescence occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the total area of discontinuous autofluorescence the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a decrease in the total area of discontinuous autofluorescence the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the total area of discontinuous autofluorescence, compared to a patient who received the VEGF inhibitor monotherapy.
  • a measurement of the volume of the central subfield pigment epithelium detachment is determined for in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • ICON-1 immunoconjugate dimer monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • immunoconjugate dimer and VEGF inhibitor therapy treatment groups e.g. anti-VEGF antibody, e.g. ranibizumab
  • the baseline determination of the volume of the central subfield pigment epithelium detachment is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the volume of the central subfield pigment epithelium detachment occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits a decrease in the volume of the central subfield pigment epithelium detachment by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits a decrease in the volume of the central subfield pigment epithelium detachment by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the volume of the central subfield pigment epithelium detachment, compared to a patient who received the VEGF inhibitor monotherapy.
  • a determination of the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of an eye is made for a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups.
  • ICON-1 immunoconj ugate dimer monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • VEGF inhibitor monotherapy e.g. anti-VEGF antibody, e.g. ranibizumab
  • immunoconjugate dimer and VEGF inhibitor therapy treatment groups e.g. anti-VEGF antibody, e.g. ranibizumab
  • the baseline determination of the integrity of (1) - (4) is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • the CNV is classical CNV.
  • the CNV is occult CNV.
  • the determination of the integrity of (10 - (4) occurs as a last observation carried forward (LOCF) method.
  • a patient exhibits an increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
  • a patient exhibits an increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
  • a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye, compared to a patient who received the VEGF inhibitor monotherapy.
  • the immunoconjugate dimer utilized in treatments of the present disclosure is the ICON-1.5 immunoconjugate.
  • hl-conl SEQ ID NO:2
  • thrombogram CAT-like assay
  • CAT-like assay multidonor human citrate plasma from healthy individuals, human FVII- deficient plasma and normal rabbit citrate plasma were used.
  • Thrombin also referred to as Factor Ila, or activated blood coagulation factor ⁇
  • generation was initiated either with human relipidated TF (in human plasma) or with rabbit relipidated TF (in rabbit plasma).
  • hl-conl was maintained frozen at -70 °C until use. Each sample included 3.0 mg hl-conl/mL in formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCh, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
  • Human plasma FVIIa in 50% glycerol was purchased from Haematologic Technologies, Inc., 57 River Road, Essex Junction, VT 05452. It was stored at -20 °C until use. Before use, it was diluted to 10 nM in the formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaC12, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
  • Spectrozyme FXa #222
  • lipidated recombinant human TF reagent Catalog #4500L
  • lipidated recombinant rabbit TF were purchased from American Diagnostica, Inc. (Stamford, CT)
  • pooled normal human plasma Lit #IR 11-020711
  • rabbit plasma Lit #26731
  • congenital FVII-deficient plasma Catalog #0700 was purchased from George King Bio-Medical, Inc.
  • HBS buffer, pH 7.4 contained 150 mM NaCl, 2 mM CaCh and 20 mM HEPES.
  • FVJJai Active site inhibited FVTIa
  • PS 2-Dioleolyl-sn- Grycero-3-Phospho-L-Serine
  • PC 2-DioleoyJ-sn-Glycero-3-Phosphocholine
  • PCPS Phospholipid vesicles
  • Extrinsic FXase Lipidated recombinant human TF (0.1 nM) was incubated with either 5 nM plasma FVIIa or 5 nM hl-conl or mixture of both (each at 5 nM) and 100 ⁇ PCPS for 10 min at 37 °C. FX (4 ⁇ ) was added and at selected time points (0-5 min.) 10 ⁇ . aliquots of the reaction mixture were quenched into 170 ⁇ iL HBS-0.1% PEG-20 mM EDTA. Twenty ⁇ L of Spectrozyme FXa (0.2 mM) was added and the rate of substrate hydrolysis was measured as an increase in absorbance at 405 nm (mOD/min).
  • CTI Corn trypsin inhibitor
  • hl-conl, plasma FVIIa and FVIIai were added at selected concentrations.
  • Twenty ⁇ ⁇ of 5 pM TF and 20 ⁇ PCPS mixture (both concentrations final) were added to CTI-plasma and incubated for 3 min.
  • Thrombin generation was initiated by the addition of 20 ⁇ L of 2.5 mM ZGly-Gly- ArgAMC ⁇ HC1 in HBS containing 0.1 M CaCh.
  • Final concentration of substrate was 416 ⁇ and that of CaCh was 15 mM Thrombin generation curves were generated using Thrombinoscope BY software.
  • FXa-generating efficiency of two forms of FVIIa and of their mixture was determined in a chromogenic assay, hl-conl was less active than plasma FVIIa. Activity of hl- conl was 18% of that observed for plasma FVIIa. When both proteins were added at equimolar (5 nM) concentration, the rate of FXa generation in the middle between the rates observed for individual proteins, indicating that hl-conl competes with plasma FVIIa for the limited amount of TF (FIG. 2). These data also suggest that hl-conl has similar affinity for TF as plasma FVIIa.
  • FVIIai prolonged the lag phase of thrombin generation and significantly suppressed both the maximum rate of thrombin generation and the maximum levels of thrombin produced. No thrombin generation was observed in the absence of TF.
  • Thrombin generation in normal human plasma the effect of hl-conl
  • hl-conl was titrated into normal human plasma initiated with TF to generate thrombin. Varying concentrations of hl-conl was used, however even at extremely high hl-conl concentrations (1 ⁇ ), no inhibition of thrombin generation was observed (FIG. 4).
  • hl-conl does not compete with plasma FVIIa for TF in the citrate plasma environment, hl-conl has no pronounced (if any) effect on thrombin generation either in human plasma initiated with human TF or in rabbit plasma initiated with rabbit TF. It is not likely that hl-conl would cause bleeding or thrombotic complications.
  • hl-conl was provided by Laureate Pharma Inc., 201 E. College Ave, Princeton, NJ, 08540. hl-conl was maintained frozen at -70 °C until use: Lot PURIC1 080402 (SEC Fr 10- 14), two vials each containing 200 uL at 2.0 mg/mL, 1.0 mg/mL, 0.5 mg/mL and 0.25 mg/mL in formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCh, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
  • the formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCk, 25 mM Arginine, 0.01% Tween 80, pH 7.4) was used as the vehicle control.
  • Each pig was maintained in a separate cage within a communal environment that housed four pigs.
  • the lighting was computer controlled and set for a 6am to 6pm cycle.
  • the temperature average was 70-72 ° F, with a variation of +/- 1 degree.
  • Humidity was kept between 30 and 70%, with average humidity equal to 33%.
  • the animals were evaluated by the large animal husbandry supervisor and a licensed veterinary technician on arrival and a licensed veterinary technician once weekly until they were euthanized.
  • a veterinarian evaluated the animals to determine if there were any abnormalities or concerns.
  • the animals were quarantined for about 1 week prior to experiments. Feed and Water
  • hl-conl has limited cross-species activity and the pig is one of the few laboratory animal species in which it is active.
  • the vitreous cavity of the pig is approximately 3 mL, allowing intravitreal injection of reasonable volumes of test article.
  • the pig eye has retinal vascular similarities to humans in addition to several cone-dominant regions of the retina that are similar to the human macula.
  • the animals were anesthetized with a mixture of ketamine hydrochloride (40 mg/kg) and xylazine hydrochloride (10 mg/kg). Injections were administered using a strict sterile technique, which involved scrubbing the lids with a 5% povidone-iodine solution and covering the field with a sterile eye drape. A sterile lid speculum was used to maintain exposure of the injection site. All injections were performed 2 mm from the limbus through the pars plana, using a 30- gauge needle on a 1 mL tuberculin syringe. After injection, a drop of 2% cyclopentolate and antibiotic ointment was placed in the eye. The animals were examined daily for signs of conjunctival injection, increased intraocular pressure, anterior uveitis vitritis, or endophthalmitis, and were sacrificed on Day 14.
  • the pigs were anesthetized with an 8:1 mixture of ketamine and xylazine and perfused through the ear vein with 10 mL PBS containing 3 mg/mL fluorescein- labeled dextran with an average molecular weight 2 x 10 6 (Sigma, St Louis, MO, USA).
  • the eyes were enucleated and four stab incisions were made at the pars plana followed by fixation in 4% paraformaldehyde for 12 hours at 4 °C.
  • the cornea and the lens were removed, and the neurosensory retina was dissected from the eyecup and four radial cuts were made from the edge of the eyecup to the equator.
  • the choroid-retinal pigment epithelium (RPE) complex was separated from the sclera and flatmounted on a glass slide in Aquamount with the inner surface (RPE) facing upwards.
  • Flat mounts were stained with a monoclonal antibody against elastin (Sigma) and a Cy 3 -conjugated secondary antibody (Sigma) and examined with a confocal microscope (Zeiss LSM510, Thornwood, NY, USA).
  • the level of the Bruch's membrane was determined by confocal microscopy using the intense red signal within a series of z-stack images collected at and around the laser spot.
  • the presence of CNV was indicated by the branching linear green signals above the plane of Bruch's membrane. Absence of CNV was defined under very stringent criteria as the total absence of green fluorescence in the vessels in the spot (see Tezel et al., 2007. Ocular Immunol Inflamm 15, pp. 3-10.
  • Example 3 Tissye Cross-Reactivity Study of hl- ⁇ yfi ⁇ h Normal Human Tissues
  • IHC immunohistochemistry
  • TCR tissue cross-reactivity
  • hl-conl and hFVTia both bound with high and approximately equal affinity to lapidated hTF.
  • Lipidated rabbit tissue factor (rTF; Product # 4520L; Lot # 051017) purchased from American Diagnostica.
  • hl-conl 1 ml; 100 ⁇ g/ml; MW 157 kDa
  • Analyte binding to the lipidated TF was determined by subtracting the RU values noted in the reference flow cell 1 from flow cell 2 and 3. Binding of analytes to the TFs was monitored in real time to obtain on (ka) and off (kd) rates. The equilibrium dissociation constant (KD) was calculated from the observed ka and kd.
  • the chips were regenerated with 3 min pulses of 10 mM EDTA in HEPES buffer (20 mM HEPES, 150 mM NaCl, pH 7.4).
  • Capture of rTF to the chip - Flow cell 2 was coated with rabbit TF (>Resonance Units [RU] 10,000) by amine coupling.
  • Flow cell 3 was coated with human TF (>RU 8,000) by amine coupling.
  • the desired level of RM3 ⁇ 4X for the measurement of ligand- analyte interaction was based on the value determined by a previous experiment where rTF captured at 10,000 Resonance Units ("RU") gave binding of hl-conl with RMSX of 15 RU and hTF captured at 8,000 RU gave binding of hl-conl with RMHX of 10RU.
  • the amount of the analyte to be captured on the chip depended on the molecular weights of the interacting proteins. It is determined by the following formula:
  • RMax MWA / MWL ⁇ RL
  • MWA is the molecular weight of the analyte (157 kDa for hl-conl, 50 kDa for hFVIIa, and 150 kDa for IgGl).
  • MWL is the molecular weight of the ligand, in this assay it is expected to be very large (multiple of 35 kDa). Flow rate of the antibody solution
  • the flow rate used for capturing the ligand was 10 ⁇ 17 ⁇ .
  • the flow rate of 30 ⁇ . was used.
  • binding analysis was performed using saturating analyte concentrations of 0-500 nM for rabbit TF and 0-50 nM for human IF. Chi squared ( ⁇ 2 ) analysis was carried out between the actual sensorgram and the calculated on- and off-rates to determine the accuracy of the analysis.
  • ⁇ 2 value up to 2 is considered significant (accurate) and below 1 is highly significant (highly accurate).
  • hl-conl and hFVIIa both bound with high, and approximately equal, affinity to lipidated hTF.
  • Bom ligands also bound to lipidated rTF with approximately 10-fold lower affinities.
  • the study presented in this example is a randomized, double-masked, multicenter, active-controlled study. Patients enrolled in this study were naive to treatment for CNV. Patients were randomly assigned to one of the following three treatment arms in the selected study eye in a 1 : 1 : 1 ratio:
  • Randomization was stratified by best-corrected visual acuity (BCVA) letter score in the study eye at baseline ( ⁇ 54 letters versus >55 letters) and by study site.
  • BCVA best-corrected visual acuity
  • IVTT Intravitreal
  • any anatomical evidenced of increased CNV activity e.g., new or increased fluid and/or leakage, hemorrhage compared to the previous scheduled visit.
  • the masked physician made a determination if rescue treatment was needed according to the above criteria.
  • rescue treatment was administered to the study eye during a scheduled injection visit, to ensure that the study masking is maintained, the unmasked physician administers rescue treatment and the patient's scheduled study treatment/re-treatment was as follows.
  • ICON-1 monotherapy arm ICON-1 (0.3 mg) + rescue therapy (0.5 mg ranibizumab).
  • ranibizumab monotherapy arm ranibizumab (0.S mg) + sham injection.
  • Safety was evaluated by tracking of adverse events, clinical laboratory tests (serum chemistry, hematology and coagulation), vital signs measurements, abbreviated physical examinations, slit-lamp biomicroscopy, intraocular pressure (IOP) and dilated ophthalmoscopy.
  • Pharmacodynamic and biological activity were measured by means of BCVA by ETDRS visual acuity chart, spectral-domain optical coherence tomography (sdOCT), color fundus photography (CFP), fundus fluorescein angiography (FA), fundus autofluorescence (FAF), contrast sensitivity, and microperimetry.
  • Pharmacokinetic (PK) and immunogenicity was evaluated by means of measuring plasma concentrations of ICON-1 and anti-drug antibodies.
  • Example 6 Synergism of ICON- 1 and anti-VEGF Antibodies in Patients With Choroidal Neovascularization. Ocular Neovascularization, and Tumor Neovascularization
  • the study duration lasts for a period of twelve months, which includes a screening visit to determine in patient meet the study parameters, a baseline randomization visit at month 0, followed by monthly visits from months 1 through 12.
  • the baseline randomization visit identifies the CNV lesion area and exudation, and measures BCVA.
  • the study comprises three different groups: a control group that receives anti- VEGF alone, a group that receives doses of anti-VEGF and ICON-1 (0.3 mg), and a group that receives doses of anti-VEGF and ICON-1 (0.6 mg).
  • Patients in groups that receive both anti-VEGF and ICON-1 are expected to exhibit an improved best-corrected visual acuity (BCVA) outcome at month 3 and month 12 from the baseline determination, as compared to those receiving only the anti-VEGF treatment.
  • Patients in groups that receive both anti-VEGF and hl-conl are further expected to exhibit a reduction in mean CNV lesion area at month 3 and month 12 from the baseline determination, as compared to those receiving only the anti-VEGF treatment
  • Patients receiving both anti-VEGF and hl-conl are expected to exhibit at least a decrease in CNV activity such that there is no increase in the area of the lesion and there is no increase in the CNV-associated exudation.
  • Patients receiving both anti-VEGF and hl-conl are expected to exhibit a reduction of in the area of the lesion and a reduction in CNV-associated exudation.
  • Patients receiving both anti-VEGF and hl-conl are likely to exhibit regression of the lesion and CNV-associated exudation.
  • ICON-1 Dose-dependent responses of ICON-1 were determined in ten to twelve week old pigs (Swine/Hampshire Cross) which underwent bilateral laser induction to create ⁇ 6 single laser spots in each eye. Each group consisted of 4 animals, for a total of 48 laser spots per group.
  • ICON-1 at 30( ⁇ g, 600ug, and 900 ⁇ was administered intravitreally on day 7 post-laser treatment
  • Eylea (aflibercept) a VEGF inhibitor
  • FFA Fluorescein Angiography
  • ICON-1 administration was well tolerated with no dose related ocular toxicities and no systemic effects.
  • Example 8 Pharmacological study evaluating the efficacy of ICON- 1.5 in a model of laser-induced choroidal neovascularization (CN V).
  • This study evaluates the efficacy of intra vitreal injections of one-armed FVII-Fc immunoconj ugates, administered as monotherapy or in combination with anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA) compared to anti-VEGF monotherapy in a rabbit model of laser induced choroidal neovascularization (CNV).
  • anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA)
  • CNV laser induced choroidal neovascularization
  • Rabbits are lasered in both eyes (OU) on Day 0 (DO).
  • Test articles and vehicle are dosed bilaterally via intravitreal (IVT) injection on D7.
  • Ranibizumab (LUCENTIS) or aflibercept (EYLEA) are dosed on day 0 immediately after laser (DO).
  • anti-VEGF agents are injected on DO and one-armed FVII-Fc immunoconj ugates on D7.
  • Ocular Examination Mydriasis for ocular examination is done using topical 1% tropicamide HCL (one drop in each eye IS minutes prior to examination). Complete ocular examination (modified Hackett and McDonald) using a slit lamp biomicroscope and indirect ophthalmoscope to evaluate ocular surface morphology, anterior segment and posterior segment inflammation, cataract formation, and retinal changes are conducted by a veterinary ophthalmologist at baseline and D14.
  • FA Fluorescein angiography
  • Example 9 Pharmacological study evaluating the efficacy of ICON-1.5 in a swine model of laser-induced choroidal neovascularization (CN V)
  • This study evaluates the efficacy of intravitreal injections of one-armed FVll-Fc immunoconjugates, administered as monotherapy or in combination with anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA) compared to anti-VEGF monotherapy in a swine model of laser induced choroidal neovascularization (CNV).
  • anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA)
  • CNV laser induced choroidal neovascularization
  • Pigs are lasered in both eyes (OU) on Day 0 (DO).
  • Test articles and vehicle are dosed bilaterally via intravitreal (IVT) injection on D7.
  • Ranibizumab (LUCENTIS) or aflibercept (EYLEA) are dosed on day 0 immediately after laser (DO).
  • anti-VEGF agents are injected on DO and one-armed FVH-Fc immunoconjugates on D7.
  • Ocular Examination Mydriasis for ocular examination is done using topical 1% tropicamide HCL (one drop in each eye IS minutes prior to examination). Complete ocular examination (modified Hackett and McDonald) using a slit lamp biomicroscope and indirect ophthalmoscope to evaluate ocular surface morphology, anterior segment and posterior segment inflammation, cataract formation, and retinal changes are conducted by a veterinary ophthalmologist at baseline and D14.
  • FA Fluorescein angiography
  • Patents, patent applications, patent application publications, journal articles and protocols referenced herein are incorporated by reference in their entireties, for all purposes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Ophthalmology & Optometry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Endocrinology (AREA)
  • Biophysics (AREA)
  • Dermatology (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Dispersion Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided herein are methods and compositions for the treatment of diseases associated with angiogenesis and neovascularization. In one aspect, the invention relates to a method for treating a condition in an eye of a patient in need thereof comprising administering to the patient in multiple dosing sessions, an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor VIIa (fVIIa) protein conjugated to the human immunoglobulin G1 (IgG1) Fc domain and a VEGF inhibitor, wherein the administration results in an improved outcome compared to a patient having been administered the VEGF inhibitor alone.

Description

METHODS FOR TREATING DISORDERS ASSOCIATED WITH ANGIOGENESIS
AND NEOVASCULARIZATION
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application Serial No. 62/450,452, filed on January 25, 2017, which is herein incorporated by reference in its entirety.
STATEMENT REGARDING SEQUENCE LISTING
[0002] The Sequence Listing associated with this application is provided in text format in lieu of a paper copy, and is hereby incorporated by reference into the specification. The name of the text file containing the Sequence Listing is ICTH_006_00US_ST25. The text file is 24 KB, was created on January 25, 2017, and is being submitted electronically via EFS-Web.
BACKGROUND OF THE INVENTION
[0003] Age-related macular degeneration (AMD) refers to the chronic, progressive degenerative pathology of the macula that results in loss of central vision. According to the Macula Vision Research Foundation and the National Eye Institute, as many as fifteen million people in the United States suffer from some form of AMD, with similar numbers in Europe and other continents. Neovascular AMD (also revered to as exudative or "wet" AMD) is the leading cause of severe vision loss and blindness in elderly patients over the age of fifty in the industrialized world. In the United States alone, more than 1.5 million people suffer from wet AMD. It is expected that AMD incidence and prevalence will further increase with the ageing population, thus leading to a significant increase in the number of patients with wet AMD in the United States and worldwide.
[0004] Tissue factor (TF) is a cytokine receptor present on vascular endothelial cells. It is an integral membrane glycoprotein with an intracellular terminal domain, a transmembrane domain, and an extracellular binding domain for Factor VJJ (FVTI) and Factor VJJa (FWa). TF has been implicated in the process of angiogenesis and the inflammatory cascade of cytokine release, both processes in the pathogenesis of neovascular AMD and certain cancers. [0005] Choroidal neovascularization (CNV) is the process in which new blood vessels grow in the choroid layer of the eye, and is associated with wet AMD. Therapies targeting vascular endothelial growth factor (VEGF) are currently the standard of clinical care for wet AMD. However, due to the multifaceted aspects of choroidal neovascularization and AMD pathogenesis, targeting VEGF alone is most likely insufficient to halt the progression of the disease towards the advanced CNV-associated degenerative processes.
[0006] There is an unmet medical need for new therapeutic strategies for choroidal neovascularization and age-related macular degeneration. The present invention addresses this and other needs.
SUMMARY OF THE TNVE TION
[0007] In one aspect, the present invention provides a method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconj ugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTia) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0008] In one aspect, the present invention provides a method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions, a composition comprising: (a) an effective amount of an immunoconj ugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTia) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone. [0009] In one aspect, the present invention provides a method for reversing tumor neovascularization in an eye of a patient in need thereof, comprising administering to the patient in multiple dosing sessions a composition comprising: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0010] In some embodiments, the VEGF inhibitor comprises an anti-VEGF antibody. In some embodiments, the immunoconjugate dimer is a homodimer. In some embodiments, the immunoconjugate dimer is a heterodimer.
[0011] In one aspect, the present invention provides a method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0012] In one aspect, the present invention provides a method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor VHa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0013] In one aspect, the present invention provides a method for preventing, inhibiting, or reversing tumor neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor; wherein the administration results in an improved best- corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0014] In some embodiments, the mutated factor VII protein exhibits a decreased coagulation response in a mammalian host, as compared to a wild-type factor VII protein.
[0015] In some embodiments, at least one of the monomer subunits of the immunoconjugate comprises a mutated human fVlla domain comprising a single point mutation at Lys341 or Ser 344. In a further embodiment, the single point mutation is to an alanine. In a further embodiment, the single point mutation is Lys341 to Ala341. In a further embodiment, the single point mutation is Ser344 to Ala344.
[0016] In some embodiments, the ocular neovascularization is secondary to proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma. In some embodiments, the ocular neovascularization is choroidal neovascularization. In some embodiments, the choroidal neovascularization is secondary to wet AMD.
[0017] In some embodiments, the patient has been previously diagnosed with wet AMD in the eye. In a further embodiment, the eye of the patient has not been previously treated for choroidal neovascularization or wet AMD. In some embodiments, the patient has previously been treated for choroidal vascularization with anti-vascular endothelial growth factor (VEGF) therapy, laser therapy, or surgery. [0018] In some embodiments, the administering comprises intravitreal injection at each dosing session. In some embodiments, the administering comprises suprachoroidal injection at each dosing session.
[0019] In some embodiments, multiple dosing sessions comprise two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, fourteen or more, fifteen or more, sixteen or more, seventeen or more, eighteen or more, nineteen or more, or twenty or more dosing sessions. In some embodiments, wherein the multiple dosing sessions comprise 12 to 24 dosing sessions. In some embodiments, the administering comprises intravitreal injection into the eye of the patient once every 28 days, once every 30 days, or once every 35 days.
[0020] In some embodiments, the immunoconjugate comprises intravitreal the amino acid sequence of SEQ ID NO: 2 or 3. In a further embodiment, the immunoconjugate comprises the amino acid sequence of SEQ ID NO:2. In a further embodiment, the immunoconjugate comprises the amino acid sequence of SEQ ID NO: 3. In some embodiments, the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO:4. In some embodiments, the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO: 5. In some embodiments, the administering comprises intravenous or intratumoral administration.
[0021] In some embodiments, the improved reduction in CNV lesion area or CNV exudation is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%. In some embodiments, the improved durability of treatment is increased by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
[0022] In some embodiments, subsequent to the multiple dosing sessions, the retinal thickness of the eye of the patient is reduced in the eye of the patient, as compared to the retinal thickness of the eye of patients having been administered the VEGF inhibitor alone. In some embodiments, the retinal thickness is reduced by at least about 50 μπι, at least about 100 μπι, at least about 150 μπι, at least about 175 μπι, at least about 200 μπι, at least about 225 μηι, or at least about 250 μιη. In some embodiments, the retinal thickness is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%. In a further embodiment, the decreased retinal thickness is decreased central retinal subfield thickness (CST), decreased center point thickness (CPT), or decreased central foveal thickness (CFT).
[0023] In some embodiments, the intraocular pressure (IOP) in the eye of the patient is measured prior to each dosing session. In some embodiments, the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about SO minutes, or about 1 hour after each dosing session. In some embodiments, the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about SO minutes, or about 1 hour prior to each dosing session. In further embodiments, the IOP is measured via tonometry.
[0024] In some embodiments, the VEGF inhibitor is present in the same composition as the immunoconjugate. In some embodiments, the VEGF inhibitor is present in a different composition than the immunoconjugate. In some embodiments, the anti-VEGF antibody is ranibizumab. In some embodiments, the dosage of ranibizumab is from about 0.2 mg to about 1 mg. In a further embodiment, the dosage of ranibizumab is 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, or 0.7 mg. In some embodiments, the ranibizumab is administered to the eye of the patient via an intravitreal injection.
[0025] In some embodiments, the VEGF inhibitor is administered at a dosage from about 0.2 mg to about 0.7 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of about 0.6 mg. In some embodiments, wherein the VEGF inhibitor is administered at a dosage of 0.6 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of about 0.3 mg. In some embodiments, the VEGF inhibitor is administered at a dosage of 0.3 mg.
[0026] In some embodiments, the multiple dosing sessions comprise administration once per month. In a further embodiment, the multiple dosing sessions comprise administration once per month for the first three months, followed by monthly treatments in months 4-12 only when CNV activity is observed.
[0027] In some embodiments, wherein the composition comprising the effective amount of the VEGF inhibitor is administered to the eye of the patient via an intravitreal injection. In some embodiments, the composition comprising the effective amount of the VEGF inhibitor is administered at each of the multiple dosing sessions. In some embodiments, each dosing session comprises the administration of between about 200 ug and about 600 ug of the immunoconj ugate dimer. In some embodiments, the administration is about 300 ug of the immunoconj ugate dimer. In some embodiments, the administration is about 600 pg of the immunoconjugate dimer.
[0028] In some embodiments, the outcome is measured at least 6 months after beginning treatment In some embodiments, the patient is a human.
[0029] In another aspect, there is provided a composition for use in a method of treatment, wherein the composition comprises an immunoconjugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and wherein the method of treatment further comprises administration of a VEGF inhibitor; and wherein the method of treatment comprises administration of multiple doses of the immunoconjugate and VEGF inhibitor. In an embodiment, the composition may be for use in treating neovascularization and/or angiogenesis. In an embodiment, the composition may be for use in treating ocular disorders. In an embodiment, the composition may be for use in treating ocular disorders associated with neovascularization and/or angiogenesis. In an embodiment, the composition may be for use in treating wet AMD, ocular neovascularization, or tumor neovascularization. In an embodiment, the monomer subunits of the immunoconjugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In another embodiment, the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor protein is fused to only one of the Fc monomers. The compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0030] In another aspect, there is provided a composition for use in treating, preventing, inhibiting, or reversing wet age-related macular degeneration (AMD), wherein the composition comprises (a) an immunoconj ugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVlla) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor. For compositions comprising only (a), a separate composition comprising a VEGF inhibitor may also be administered. In an embodiment, the monomer subunits of the immunoconj ugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In another embodiment, the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers. The compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0031] In another aspect, there is provided a composition for use in treating, preventing, inhibiting, or reversing ocular neovascularization, wherein the composition comprises (a) an immunoconjugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor. For compositions comprising only (a), a separate composition comprising a VEGF inhibitor may also be administered. In an embodiment, the monomer subunits of the immunoconjugate dimer each comprises a mutated human factor Wa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In another embodiment, the immunoconjugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor W protein is fused to only one of the Fc monomers. The compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone. [0032] In another aspect, there is provided a composition for use in treating or reversing tumor neovascularization, wherein the composition comprises (a) an immunoconj ugate dimer, wherein at least one of the monomer subunits of the dimer comprises a mutated human factor Vila (-Vila) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and optionally (b) a VEGF inhibitor. For compositions comprising only (a), a separate composition comprising a VEGF inhibitor may also be administered. In an embodiment, the monomer subunits of the immunoconj ugate dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In another embodiment, the immunoconj ugate comprises two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers. The compositions may be for use in treating an eye of a patient in need thereof, and an effective amount of the composition may be administered to the patient in multiple dosing sessions to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
[0033] In another aspect, there is provided a composition for use in a method of treatment, wherein the composition comprises an 0.3 mg, 0.6 mg, or 0.9 mg of an immunoconj ugate dimer, wherein monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated or fused to the human immunoglobulin Gl (IgGl) Fc domain; and wherein the method of treatment comprises administration of 0.5 mg of ranibizumab or 2.0 mg of aflibercept; and wherein multiple doses of the ranibizumab or aflibercept, an multiple does of the imm unoconj ugate dimer are administered monthly for at least three months; and wherein the composition is for use in treating ocular disorders associated with neovascularization and/or angiogenesis; and wherein the composition is for use in treating an eye of a patient in need thereof to result in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, improved durability of treatment, or no exudative presence of subretinal fluid, intraretinal fluid, and/or rubretinal pigment epithelium fluid compared to patients having been administered the VEGF inhibitor alone. BRIEF DESCRIPTION OF THE FIGURES
[0034] FIG. 1 is a non-limiting diagram of two ICON-1 immunoconj ugate embodiments and one ICON- 1.5 immunoconj ugate embodiment of the present invention.
[0035] FIG. 2 is a graph of the rate of the intrinsic factor Xase complex (Fxase) hydrolysis (increase in absorbance at 405 nm - mOD/min) as a function of time.
[0036] FIG. 3 is a graph of thrombin generation by the known inhibitor of coagulation, active site inhibited FVTIa (FVIIai), as a function of time in normal pooled plasma.
[0037] FIG. 4 is a graph of thrombin generation by hl-conl, as a function of time in normal pooled plasma.
[0038] FIG. 5 is a graph of thrombin generation by human Factor Vila and hl-conl as a function of time in FVII-depleted plasma.
[0039] FIG. 6 is a graph of thrombin generation by hl-conl, as a function of time in rabbit plasma.
[0040] FIG. 7 is a graph of thrombin generation by hl-conl or FVIIai as a function of time in centrifuged rabbit plasma.
[0041] FIG. 8 is a graph showing the percent CNV in the pig as a function of intra vitreal dose of hl-conl. Intravitreal injections (100 uL/eye) of solutions of hl-conl (0.25, 0.5, 1.0 and 2.0 mg/mL) were injected into both eyes of mini-pigs on Day 10; control animals received 100 uL of formulation buffer. On Day 14 the animals were sacrificed and the % CNV was determined.
[0042] FIG. 9 is a graph showing the percent CNV in the pig as a function of intravitreal dose of a 100 kDa fragment of hl-conl. Intravitreal injections (100 uL/eye) of solutions of hl-conl (0.25, 0.5, 1.0 and 2.0 mg/mL) were injected into both eyes of mini-pigs on Day 10; control animals received 100 uL of formulation buffer. On Day 14 the animals were sacrificed and the % CNV was determined. [0043] FIG. 10 is an image representing the distribution of patients within treatment arms with indications for dosing schedules that are further delineated by treatment induction and treatment extension.
[0044] FIG. 11 is a tabulation of the baseline CNV lesion area (mm2) for the patients within each of the treatment arms compared with the mean CNV lesion area change from baseline.
[0045] FIG. 12 is a tabulation of the change in lesion size for the patients within each of the treatment arms, the total number of patients exhibiting the change (or no change), and the corresponding change in BCVA.
[0046] FIG. 13 is a graph depicting the proportional change/no change in lesion size for the patients within each of the treatment arms.
[0047] FIG. 14 is a graph displaying the mean change in CST over time in the study eyes in all treatment arms.
[0048] FIG. 15 is a graph displaying the mean change in BCVA score over time in the study eyes in all treatment arms.
[0049] FIG. 16 is a tabulation of the gain of BCVA from baseline at month 6 for patients in all treatment arms. The figure further identifies the proportion of patients with BCVA with >71 at month 6, proportion of patients with BCVA with < 33 letters at month 6, and the mean BCVA change in letter score at month 6.
[0050] FIG. 17 is a tabulation of the fluid type (I F, SRF, and Sub-RPE) at baseline for patients within each of the treatment arms. The figure further identifies the number of patients with no IRF, no SRF, no sub-RPE, and dry retina at 6 months for each of the treatment arms.
[0051] FIG. 18 is a graph identifying the percent of patients exhibiting dry retina within each of the treatment arms.
[0052] FIG. 19 is a tabulation of the patients within each of the treatment arms that received study drug retreatment, and a further indication of the number of retreatments. [0053] FIG.20 is a graph identifying the time from treatment end to first retreatment for patients in each of the treatment arms.
[0054] FIG. 21 is a graph identifying the durability of treatment for patients within each of the treatment arms. Durability of treatment is represented as a proportion of the patients in each of the treatment arms that did not require retreatment
[0055] FIG. 22 is a graph depicting a dose-dependent reduction in lesion fluorescence in pigs that underwent bilateral laser induction of laser spots in the eyes; measured at day 14 post- induction. The pigs were administered a vehicle control, ICON-1 at 300pg, ICON-1 at 6(X^g, ICON-1 at 900μ& or Eylea at 2mg.
[0056] FIG. 23 is a graph depicting a reduction in lesion fluorescence in pigs that underwent bilateral laser induction laser spots in the eyes; measured at day 14 post-induction. The pigs were administered a vehicle control, ICON-1 at 600ug, Eylea at 2mg, or ICON-1 at 600pg and Eylea at 2mg.
DETAILED DESCRIPTION OF THE INVENTION
[0057] The term "a" or "an" may refer to one or more of that entity, i.e. can refer to plural referents. As such, the terms "a" or "an", "one or more" and "at least one" are used interchangeably herein. In addition, reference to "an element" by the indefinite article "a" or "an" does not exclude the possibility that more than one of the elements is present, unless the context clearly requires that there is one and only one of the elements.
[0058] Reference throughout this specification to "one embodiment", "an embodiment", "one aspect", or "an aspect" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics can be combined in any suitable manner in one or more embodiments. [0059] As used herein, in particular embodiments, the terms "about" or "approximately" when preceding a numerical value indicates the value plus or minus a range of 10%.
[0060] As used herein, a "patient" refers to a warm-blooded animal, e.g., rat, mouse, sheep, cow, pig, guinea pig, non-human primate, human primate, wherein males and/or females may be a patient.
[0061] As used herein, "classic CNV" means a well-defined CNV area that results in vision that is between 20/25 and 20/400, but may be worse than 20/800.
[0062] As used herein, "occult CNV" means a poorly delineated CNV area that exhibits less leakage than classic CNV, and results in vision that is between 20/25 and 20/400, but may be worse than 20/800.
[0063] As used herein, "improved" means at least a 5% improvement of the outcome for which "improved" modifies, as compared to a corresponding control, e.g., an improved reduction in mean CNV lesion area is at least a 5% improvement of the mean CNV lesion area as compared to the mean CNV lesion area under control conditions.
[0064] As used herein, "antibody" includes any monoclonal antibody, polyclonal antibody, multispecific antibody, bispecific antibody, single-chain antibody, a single-chain variable fragment (scFv) of an antibody, FAB fragment of an antibody, and fragments thereof.
[0065] As used herein, "CNV activity" includes new or increased fluid and/or leakage, hemorrhage and/or lesion, persistent fluid, and decreased chorioretinal blood flow at the site of the perceived CNV lesion.
Angiogenesis
[0066] Pathologic angiogenesis, the induction of the growth of existing blood vessels from the vessels in surrounding tissue, is observed in a variety of diseases, typically triggered by the release of specific growth factors for vascular endothelial cells. Pathologic angiogenesis can result in neovascularization, i.e., the creation of new blood vessels, enabling solid tumor growth and metastasis, causing visual malfunction in ocular disorders, promoting leukocyte extravasation in inflammatory disorders, and/or influencing the outcome of cardiovascular diseases such as atherosclerosis.
[0067] In one aspect of the present invention, methods for treating a patient having a disease associated with neovascularization and/or angiogenesis, such as cancer, rheumatoid arthritis, the exudative ("wet") form of macular degeneration, and/or atherosclerosis are provided. As described herein, administration may be local or systemic, depending upon the type of pathological condition involved in the therapy. As used herein, the term "patient" includes both humans and other species, including other mammal species. The invention thus has both medical and veterinary applications. In veterinary compositions and treatments, immunoconj ugates are constructed using targeting and effector domains derived from the corresponding species.
[0068] In the aspects provided herein, methods for treating a patient for a disease associated with angiogenesis and/or neovascularization are provided. In one embodiment, the disease associated with neovascularization and/or angiogenesis is wet AMD. In other embodiments, the disease associated with neovascularization and/or angiogenesis is a cancer (ocular melanoma), atherosclerosis, rheumatoid arthritis, oculr melanoma, diabetic macular edema (DME), macular edema following retinal vein occlusion (RVO), proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), reinopathy of prematurity (ROP), or neovascular glaucoma.
Immunoconiugates
[0069] As used herein, "immunoconjugate" or "immunoconj ugates" refer to two chemically conjugated or fusion proteins: (1) ICON-1 (referred to interchangeably as hl-conl), a two-armed FVII-Fc fusion protein comprising two dimerized immunoglobulin (Ig) Fc monomers fused to two mutated FVII proteins; and (2) ICON- 1.5, a one-armed FW-Fc fusion protein comprising two dimerized immunoglobulin (Ig) Fc monomers, and a mutated FVII protein, wherein the mutated FVII protein is fused to only one of the Fc monomers. (See FIG. 1 for exemplary embodiments).
[0070] As used herein, "conjugated proteins" and "fusion proteins" are used interchangeably, and one of ordinary skill in the art would be aware of the metes and bounds of the distinctions between conjugated proteins and fusion proteins. [0071] In some embodiments, an immunoconj ugate has as an effector domain, an immunoglobulin Fc domain, and said effector domain is conjugated to a targeting domain comprising a mutant form of human factor VII. In some embodiments, an immunoconj ugate comprises an Fc domain of a human IgGl immunoglobulin conjugated to a targeting domain exhibiting decreased coagulation comprising a mutant form of factor W comprising one or two mutations selected from S344A and/or K341A, wherein the immunoconj ugate protein binds to tissue factor. In some embodiments, immunoconjugates of the present disclosure include immunoconjugates described in published international patent application WO/2017/18114S; and U.S. Patents 7,858,092; 8,388,974, 8,071,104; 7,887,809; and 6,924,359.
[0072] ICON-1.5 and ICON-1 share similar degrees of binding and ADCC activity, and FXa conversion.
[0073] In one aspect provided herein, a protein comprising a mutated FVII protein (targeting domain) conjugated to a human IgGl Fc region (effector domain) is provided. FIG. 1 provides the generalized structure of one embodiment of an immunoconj ugate that can be administered by the methods provided herein. The mutated Factor Vila domain (also referred to as the TF targeting domain), in the aspects provided herein, binds tissue factor with high affinity and specificity, but does not initiate coagulation, or minimizes coagulation normally associated with tissue factor binding. The IgGl Fc domain (also referred to as the effector domain) triggers a cytolytic response against cells which bind the immunoconj ugate, by the natural killer (NK) cell and complement pathways. In one embodiment, the IgGl Fc effector domain comprises both the CH2 and CH3 regions of the IgGl Fc region.
[0074] Table 1: Description of Sequences
Figure imgf000017_0001
Figure imgf000018_0001
[0075] The reaction between FVHa and TF is species-specific (Janson et al., 1984; Schreiber et al., 2005; Peterson et al., 2005): murine FVII appears to be active in many heterologous species including rabbit, pig and human, whereas human FVIIa is only appreciably active in human, dog, rabbit and pig. Conversely, the human IgG Fc domain is active in both humans and mice. Accordingly, depending on the patient, the immunoconjugate is constructed using targeting and effector domains derived from the corresponding species, or from a species that is known to be active in the patient. For example, in the human treatment methods provided herein, the mutated tissue factor targeting domain is derived from human Factor Vila conjugated to an effector domain comprising the Fc region of a human IgGl immunoglobulin. For example, in one embodiment, the immunoconjugate is a protein of SEQ ID NO: 2. In a further embodiment, the immunoconjugate is a protein of SEQ ID NO: 3. In one embodiment, the immunoconjugate is encoded by the mRNA sequence of SEQ ID NO: 1, 4, or 5.
[0076] In one embodiment, the immunoconjugate described herein comprises two protein chains, each comprising a targeting domain joined to an effector domain via a linker or hinge region. In a further embodiment, the linker or hinge region is naturally occurring, and in one embodiment, is of human origin. The hinge region of an IgGl immunoglobulin, for example the hinge region of the human IgGl immunoglobulin, in one embodiment, is used to link the targeting domain to the effector domain. In one embodiment, the hinge region of IgGl includes cysteine amino acids which form one or more disulfide bonds between the two monomer chains (e.g., as depicted in FIG. 1).
[0077] In one embodiment, the immunoconjugate is a homodimer. However, in another embodiment, the immunoconjugate is a heterodimer, for example, an immunoconjugate comprising two monomers each having a targeting domain of a different amino acid sequence, but the same effector domains. The amino acid sequences of the two targeting domains, in one embodiment, differ by one amino acid, two or more amino acids, three or more amino acids or five or more amino acids. In one embodiment, each monomer subunit comprises an IgGl hinge region that links the targeting region and effector region of the immunoconjugate, and the monomer subunits of the immunoconj ugate heterodimer or the immunoconjugate homodimer are linked together via a disulfide bond between IgGl hinge regions.
[0078] In one embodiment, the molecular weight of the ICON-1 immunoconjugate provided herein is from about ISO kDa to about 200 kDa. In another embodiment, the molecular weight of the immunoconjugate is about 1S7 kDa or 1S7 kDa. For example, the immunoconjugate in one embodiment is the immunoconjugate having the amino acid sequence set forth in SEQ ID NO: 2, also referred to herein as "hl-conl" or "ICON-1" (used interchangeably herein). In another embodiment, the immunoconjugate has the amino acid sequence set forth in SEQ ID NO: 3.
[0079] As provided throughout, in embodiments described herein, an immunoconjugate comprising a tissue factor targeting domain comprising a mutated Factor Vila domain is provided. The targeting domain comprises a mutated Factor Vila that has been mutated to inhibit initiation of the coagulation pathway without reducing binding affinity to tissue factor. In one embodiment, the mutation in Factor Vila is a single point mutation at residue 341. In a further embodiment, the mutation is from Lys341 to Ala341. However, other mutations that inhibit the coagulation pathway are encompassed by the immunoconj ugates provided herein. The effector domain of the immunoconj ugates provided herein, in one embodiment, mediates both complement and natural killer (NK) cell cytotoxicity pathways.
[0080] Also provided herein are pharmaceutical compositions comprising the immunoconjugates of the invention.
Immunoconiugate Production
[0081] In some embodiments, methods of producing the immunoconjugate include expression in mammalian cells such as BHK cells. In further embodiments, cell lines may include HEK 293, CHO, and SP2/0. Immunoconjugates may be generated by mammalian expression of the expression constructs. In some embodiments, the immunoconjugates are produced as fusion proteins (FVTi-Fc) or produced as chemical conjugates.
[0082] In some embodiments, the immunoconjugate is post-translationally modified. Post- translational modification includes: myristoylation, glypiation, palmitoylation, prenylation, lipoylation, acylation, alkylation, butrylation, gamma-carboxylation, glycosylation (N- glycosylation, O-glycosylation, fucosylation, and mannosylation), propionylation, succinylation, and sulfation.
VEGF Inhibitors
[0083] As provided herein, the immunoconjugate dimer is administered in a co-therapeutic regimen with a VEGF inhibitor to treat a patient for one of the aforementioned diseases or disorders, for example, to treat wet AMD or another ocular disease associated with angiogenesis or neovascularization.
[0084] In one embodiment, the VEGF inhibitor is administered in the same composition as the immunoconjugate dimer.
[0085] However, in another embodiment, the immunoconjugate dimer and VEGF inhibitor are administered in separate compositions. In some embodiments, the VEGF inhibitor is administered prior to the immunoconjugate dimer. In some embodiments, the VEGF inhibitor is administered subsequent to the immunoconjugate dimer. In some embodiments, the VEGF inhibitor is administered simultaneously with the immunoconjugate dimer.
[0086] In one embodiment, the VEGF inhibitor is an anti-VEGF antibody. In one embodiment, the VEGF inhibitor is ranibizumab or bevacizumab. In a another embodiment, the VEGF in inhibitor is ranibizumab. In another embodiment, ranibizumab is administered at a dosage of 0.S mg or 0.3 mg per dosing session, and is administered as indicated in the prescribing information for LUCENTIS.
[0087] In some embodiments, VEGF inhibitors may be selected from ranibizumab, bevacizumab, pazopanib, sunitinib, sorafenib, axitinib, regorafenib, ponatinib, cabozantinib, vandetanib, ramucirumab, lenvatinib, aflibercept, and ziv-aflibercept.
Administering the Immunoconjugate and VEGF Inhibitor
[0088] Administration methods encompassed by the methods provided herein include intravitreal injection, suprachoroidal injection, topical administration (e.g., eye drops), intravenous and intratumoral administration. In another embodiment, administration is via intravenous, intramuscular, intratumoral, subcutaneous, intrasynovial, intraocular, intraplaque, or intradermal injection of the immunoconjugate or of a replication-deficient adenoviral vector, or other viral vectors carrying a cDNA encoding a secreted form of the immunoconjugate. In one embodiment, the patient in need of treatment is administered one or more immunoconjugate dimers via intravitreal, intravenous or intratumoral injection, or injection at other sites, of one or more immunoconjugate proteins. Alternatively, in one embodiment, a patient in need of treatment is administered one or more immunoconjugate dimers via intravenous or intratumoral injection, or injection at other sites, of one or more expression vectors carrying a cDNA encoding a secreted form of one or more of the immunoconjugate dimers provided herein. In some embodiments, the patient is treated by intravenous or intratumoral injection of an effective amount of one or more replication-deficient adenoviral vectors, or one or more adeno-associated vectors carrying cDNA encoding a secreted form of one or more types of immunoconjugate proteins. In one embodiment, the patient in need of treatment is co-administered one or more immunoconjugate dimers and VEGF inhibitors via intravitreal, intravenous or intratumoral injection, or injection at other sites, of one or more immunoconjugate proteins and VEGF inhibitors. Alternatively, in one embodiment, a patient in need of treatment is co-administered one or more immunoconjugate dimers and VEGF inhibitors via intravenous or intratumoral injection, or injection at other sites, of one or more expression vectors carrying a cDNA encoding a secreted form of one or more of the immunoconjugate dimers provided herein.
[0089] As used herein, "effective amount" or "therapeutically effective amount" means a level or amount of a therapeutic agent needed to treat a condition or disease of the present disclosure, or the level or amount of a therapeutic agent that produces a therapeutic response or desired effect in the subject to which the therapeutic agent was administered; wherein a therapeutic agent is an immunoconjugate of the present disclosure. The therapeutic agent may further include an immunoconjugate of the present disclosure and a VEGF inhibitor of the present disclosure. Thus, a therapeutically effective amount of a therapeutic agent, such as an immunoconjugate of the present disclosure and a VEGF inhibitor of the present disclosure, is an amount that is effective in reducing one or more symptoms of angiogenesis and/or neovascularization, as well as various forms of AMD. [0090] As used herein, "pharmaceutical composition" means a composition comprising a therapeutic agent
[0091] As used herein, "treatment", "treating", and the like, mean the following actions: (i) preventing a particular disease or disorder from occurring in a subject who may be predisposed to the disease or disorder but has not yet been diagnosed as having it; (ii) curing, treating, or inhibiting the disease, i.e., arresting its development; or (iii) ameliorating or reversing the disease by reducing or eliminating symptoms, conditions, and/or by causing regression of the disease.
[0092] In one embodiment, a method of intravitreal injection is employed. In a further embodiment, aseptic technique is employed when preparing the immunoconjugate dimer and/or VEGF inhibitor for injection, for example, via the use of sterile gloves, a sterile drape and a sterile eyelid speculum (or equivalent). In one embodiment, the patient is subjected to anesthesia and a broad-spectrum microbicide prior to the injection.
[0093] In one embodiment, intravitreal injection of one or more of the VEGF inhibitors and/or immunoconjugate dimers, provided herein, for example the immunoconjugate dimer of SEQ ID NO: 2 is prepared by withdrawing the vial contents of the immunoconjugate dimer composition solution and/or the VEGF inhibitor composition solution through a 5-micron, 19-guage filter needle attached to a 1-cc tuberculin syringe. The filter needle in a further embodiment, is then discarded and replaced with a sterile 30-gauge x ½-inch needle for the intravitreal injection. The contents of the vial are expelled until the plunger tip is aligned with the line on the syringe that marks the appropriate dose for delivery.
[0094] In one method of ocular injection, e.g., intravitreal or suprachoroidal injection, prior to and/or after the injection, the patient is monitored for elevation in intraocular pressure (IOP). For example, in one embodiment, prior to and/or after the ocular injection, the patient is monitored for elevation in IOP using tonometry. In another embodiment, the patient is monitored for increases in IOP via a check for perfusion of the optic nerve head immediately after the injection. In one embodiment, prior to ocular injection of one of the immunoconjugate dimers and/or VEGF inhibitors provided herein, for example about 20 minutes, about 30 minutes, about 40 minutes, about 50 minutes or about 1 hour prior to the ocular injection, the patient is monitored for elevation in IOP. In another embodiment, after ocular injection of one of the immunoconjugate dimers and/or VEGF inhibitors provided herein, for example, about 10 minutes, about 20 minutes, about 30 minutes, about 40 minutes, about SO minutes or about 1 hour after the intraocular injection, the patient is monitored for elevation in IOP. In one embodiment, the patient's IOP is substantially the same prior to intraocular injection of an immunoconjugate dimer and/or the VEGF inhibitor, as compared to after intraocular injection of the immunoconjugate dimer and/or VEGF inhibitor. In one embodiment, the patient's IOP varies by no more than 10%, no more than 20% or no more than 30% after intraocular injection, as compared to prior to intraocular injection (e.g., intravitreal injection).
[0095] The treatment methods provided herein in one embodiment, comprise a single administration of one of the immunoconjugate dimers and/or VEGF inhibitors provided herein (e.g., an immunoconjugate of SEQ ID NO: 2 or 3). However, in another embodiment, the treatment methods provided herein comprise multiple dosing sessions. In a further embodiment, the multiple dosing sessions are multiple intraocular injections of one of the immunoconjugate dimers and/or VEGF inhibitors described herein. The multiple dosing sessions, in one embodiment comprise two or more, three or more, four or more or five or more dosing sessions. In a further embodiment, each dosing session comprises intraocular injection of one of the immunoconj ugates and/or VEGF inhibitors described herein, or intratumoral injection of one of the immunoconj ugates and/or VEGF inhibitors described herein (i.e., either as the expressed protein or via a vector encoding the soluble immunoconjugate).
[0096] In one embodiment, from about 2 to about 24 dosing sessions are employed, for example, from about 2 to about 24 intraocular dosing sessions (e.g., intravitreal or suprachoroidal injection). In a further embodiment, from about 3 to about 30, or from about 5 to about 30, or from about 7 to about 30, or from about 9 to about 30, or from about 10 to about 30, or from about 12 to about 30 or from about 12 to about 24 dosing sessions are employed.
[0097] In one embodiment, where multiple dosing sessions are employed, the dosing sessions are spaced apart by from about 10 days to about 60 days, or from about 10 days to about 50 days, or from about 10 days to about 40 days, or from about 10 days to about 30 days, or from about 10 days to about 20 days. In another embodiment, where multiple dosing sessions are employed, the dosing sessions are spaced apart by from about 20 days to about 60 days, or from about 20 days to about SO days, or from about 20 days to about 40 days, or from about 20 days to about 30 days. In even another embodiment, the multiple dosing sessions are bi-weekly (e.g., about every 14 days), monthly (e.g., about every 30 days), or bi-monthly (e.g., about every 60 days). In yet another embodiment, the dosing sessions are spaced apart by about 28 days.
[0098] In one embodiment, the multiple dosing sessions comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 dosing sessions, wherein the dosing sessions are spaced apart by 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 35 days, 40 days, 45 days, 50 days, 55 days, or 60 days.
[0099] The immunoconj ugates and/or VEGF inhibitors provided herein are amenable for use in any disease or disorder in which angiogenesis and/or neovascularization is implicated. For example, in one aspect, an immunoconjugate dimer and/or VEGF inhibitor provided herein is administered to the eye of a patient in need of treatment of wet age-related macular degeneration (AMD). In one embodiment, the treatment comprises multiple dosing sessions of the immunoconjugate dimer and/or VEGF inhibitors. As provided throughout, the immunoconjugate dimer comprises monomer subunits that each include a mutated human factor VQa (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
[00100] In one embodiment, the method of treating wet AMD comprises preventing, inhibiting or reversing choroidal neovascularization in the eye of the patient in need of treatment. In a further embodiment, choroidal neovascularization is reversed by at least about 10%, at least about 20%, at least about 30% or at least about 40% after treatment, as compared to the choroidal neovascularization that was present in the afflicted eye of the patient prior to treatment
[00101] Other ocular disorders associated with ocular neovascularization are treatable with the immunoconj ugates and VEGF inhibitors and methods provided herein. The ocular neovascularization, in one embodiment, is choroidal neovascularization. In another embodiment the ocular neovascularization is retinal neovascularization. In yet another embodiment, the ocular neovascularization is corneal neovascularization. Accordingly, an ocular disorder associated with choroidal, retinal or corneal neovascularization, in one embodiment, is treatable by one or more of the methods provided herein. In a further embodiment, the method comprises administering to the eye of a patient in need thereof, the immunoconj ugate dimers and/or VEGF inhibitors described herein. In a further embodiment, the treatment comprises multiple dosing sessions of the immunoconjugate dimer and/or VEGF inhibitors. In a further embodiment, the immunoconj ugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3.
[00102] For example, in one embodiment, a patient in need of treatment of proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma is treated with the immunoconj ugates and/or VEGF inhibitors provided herein, for example, via intravitreal injection, suprachoroidal injection or topical administration (e.g., via eye drops) of the immunoconjugate and VEGF inhibitors into the affected eye. Treatment in one embodiment occurs over multiple dosing sessions. With respect to the aforementioned disorders, ocular neovascularization is said to be "associated with" or "secondary to" the respective disorder.
[00103] In one embodiment, a patient in need of treatment of macular edema following retinal vein occlusion (RVO) is treated by one of the immunoconjugate dimers and VEGF inhibitors provided herein. In one embodiment, the method comprises administering to the patient a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVHa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the mutated fVIIa protein is a human mutated fVHa protein and is linked to the IgGl Fc domain via the hinge region of IgGl. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer is administered to the patient over multiple dosing sessions, for example, via intravitreal administration at each dosing session.
[00104] In another embodiment, a patient in need of treatment of diabetic macular edema (DME) is treated by one of the immunoconjugate dimers and VEGF inhibitors provided herein. In one embodiment, the method comprises administering to the patient a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer is administered to the patient over multiple dosing sessions. In even a further embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered intravitreally at each dosing session.
[00105] In yet another embodiment, diabetic retinopathy is treated via one of the immunoconj ugates and VEGF inhibitors provided herein, in a patient in need thereof, for example, a patient with DME. In one embodiment, the method comprises administering to the patient, for example a DME patient, a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitors, wherein the monomer subunits of the dimer each comprise a mutated factor VHa (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl . In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer is administered to the patient over multiple dosing sessions. In even a further embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions, for example, via intravitreal administration at each dosing session.
[00106] In one embodiment of the invention, one or more of the immunoconjugates and VEGF inhibitors provided herein is used in a method to treat a disease or disorder associated with tumor neovascularization in a patient in need thereof, for example, a cancer patient In one embodiment, the method comprises administering to the patient, for example via intratumoral or intravenous injection, a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitor, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions.
[00107] In cancer treatments, the immunoconjugate dimer and VEGF inhibitors are used for treating a variety of cancers, particularly primary or metastatic solid tumors, including melanoma, renal, prostate, breast, ovarian, brain, neuroblastoma, head and neck, pancreatic, bladder, endometrial and lung cancer. In one embodiment, the cancer is a gynecological cancer. In a further embodiment, the gynecological cancer is serous, clear cell, endometriod or undifferentiated ovarian cancer. The immunoconjugate dimer and/or VEGF inhibitor in one embodiment is employed to target the tumor vasculature, particularly vascular endothelial cells, and/or tumor cells. Without wishing to be bound by theory, targeting the tumor vasculature offers several advantages for cancer immunotherapy with one or more of the immunoconjugate dimers and/or VEGF inhibitors described herein, as follows, (i) some of the vascular targets including tissue factor should be the same for all tumors; (ii) immunoconjugates targeted to the vasculature do not have to infiltrate a tumor mass in order to reach their targets; (iii) targeting the tumor vasculature should generate an amplified therapeutic response, because each blood vessel nourishes numerous tumor cells whose viability is dependent on the functional integrity of the vessel; and (iv) the vasculature is unlikely to develop resistance to an immunoconjugate, because that would require modification of the entire endothelium layer lining a vessel. Unlike previously described antiangiogenic methods that inhibit new vascular growth, immunoconjugate dimers provided herein elicit a cytolytic response to the neovasculature.
[00108] In another embodiment, one or more of the immunoconjugates and VEGF inhibitors described herein is used in a method for treating atherosclerosis or rheumatoid arthritis. In one embodiment, the method comprises administering to the patient in need of treatment a composition comprising an effective amount of an immunoconjugate dimer and/or VEGF inhibitor, wherein the monomer subunits of the dimer each comprise a mutated factor Vila (fVna) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain. In a further embodiment, the mutated fVIIa protein is a human mutated fVIIa protein and is linked to the IgGl Fc domain via the hinge region of IgGl. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2 or 3. In yet a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 2. In a further embodiment, the immunoconjugate dimer has the amino acid sequence of SEQ ID NO: 3. In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered to the patient over multiple dosing sessions.
[00109] In one embodiment of a method for treating an ocular disorder with an immunoconjugate dimer and a VEGF inhibitor, for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD, the patient subjected to the treatment method substantially maintains his or her vision subsequent to the treatment (e.g., the single dosing session or multiple dosing sessions), as measured by losing fewer than IS letters in a best-corrected visual acuity (BCVA) measurement, compared to the patient's BCVA measurement prior to undergoing treatment In a further embodiment, the patient loses fewer than 10 letters, fewer than 8 letters, fewer than 6 letters or fewer than 5 letters in a BCVA measurement, compared to the patient's BCVA measurement prior to undergoing treatment
[00110] In some embodiments, a patient having been administered an immunoconjugate and/or VEGF inhibitor of the present invention loses fewer than 10, 9, 8, 7, 6, or 5 letters in a BCVA measurement, compared to a patient's BCVA measurement prior to undergoing treatment In some embodiments, the patient loses fewer than about 10, about 9, about 8, about 7, about 6, or about 5 letters in a BCVA measurement, compared to a patient's BCVA measurement prior to undergoing treatment.
[00111] In some embodiments, a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention loses fewer than between 15 and 5, 15 and 6, 15 and 7, 15 and 8, 15 and 9, 15 and 10, 10 and 5, 10 and 6, 10 and 7, 10 and 8, 10 and 9, 9 and 5, 9 and 6, 9 and 7, 9 and 8, 8 and 5, 8 and 6, 8 and 7, 7 and 5, 7 and 6, or 6 and 5 letters in a BCVA measurement.
[00112] In some embodiments, a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention loses fewer than between about 15 and about 5, about 15 and about 6, about 15 and about 7, about 15 and about 8, about 15 and about 9, about 15 and about 10, about 10 and about 5, about 10 and about 6, about 10 and about 7, about 10 and about 8, about 10 and about 9, about 9 and about 5, about 9 and about 6, about 9 and about 7, about 9 and about 8, about 8 and about 5, about 8 and about 6, about 8 and about 7, about 7 and about 5, about 7 and about 6, or about 6 and about 5 letters in a BCVA measurement
[00113] In another embodiment of a method for treating an ocular disorder with an immunoconj ugate dimer, for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD, the patient subjected to the treatment method substantially maintains his or her vision subsequent to the treatment (e.g., the single dosing session or multiple dosing sessions), as measured by BCVA measurement
[00114] In some embodiments, a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining 5, 6, 7, 8, 9, 10, 15, 20, or 25 or more letters in a best- corrected visual acuity (BCVA) measurement, compared to the patient's BCVA prior to the treatment. In some embodiments, a patient having been administered an immunoconj ugate and/or VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining about 5, about 6, about 7, about 8, about 9, about 10, about 15, about 20, or about 25 or more letters in a best-corrected visual acuity (BCVA) measurement, compared to the patient's BCVA prior to the treatment.
[00115] In some embodiments, a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining greater than between S and 25, 5 and 20, 5 and 15, 5 and 10, 5 and 9, 5 and 8, 5 and 7, 5 and 6, 6 and 25, 6 and 20, 6 and 15, 6 and 10, 6 and 9, 6 and 8, 6 and 7, 7 and 25, 7 and 20, 7 and 15, 7 and 10, 7 and 9, 7 and 8, 8 and 25, 8 and 20, 8 and 15, 8 and 10, 8 and 9, 9 and 25, 9 and 20, 9 and 15, 9 and 10, 10 and 25, 10 and 20, 10 and 15, 15 and 25, 15 and 20, or 20 and 25 or more letters in a BCVA measurement, compared to the patient's BCVA prior to the treatment.
[00116] In some embodiments, a patient having been administered an immunoconj ugate and a VEGF inhibitor of the present invention regains his or her vision subsequent to the treatment, as measured by gaining greater than between about 5 and about 25, about 5 and about 20, about 5 and about 15, about 5 and about 10, about 5 and about 9, about 5 and about 8, about 5 and about 7, about 5 and about 6, about 6 and about 25, about 6 and about 20, about 6 and about 15, about 6 and about 10, about 6 and about 9, about 6 and about 8, about 6 and about 7, about 7 and about 25, about 7 and about 20, about 7 and about 15, about 7 and about 10, about 7 and about 9, about 7 and about 8, about 8 and about 25, about 8 and about 20, about 8 and about 15, about 8 and about 10, about 8 and about 9, about 9 and about 25, about 9 and about 20, about 9 and about 15, about 9 and about 10, about 10 and about 25, about 10 and about 20, about 10 and about 15, about 15 and about 25, about 15 and about 20, or about 20 and about 25 or more letters in a BCVA measurement, compared to the patient's BCVA prior to the treatment.
[00117] In one embodiment of a method for treating an ocular disorder with an immunoconj ugate dimer and a VEGF inhibitor, for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD, the ocular neovascularization area, e.g., the choroidal neovascularization area is reduced in the eye of the patient, as compared to the ocular neovascularization area (e.g., CNV area) prior to treatment. As provided herein, treatment can include one dosing session or multiple dosing sessions, and reduction in ocular neovascularization ana (e.g., CNV area), in one embodiment, is assessed after individual dosing sessions, or multiple dosing sessions. In a further embodiment, the ocular neovascularization area (e.g., CNV area) is reduced by at least about 5%, or at least about 10%, or at least about 15%, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, as measured by fluorescein angiography.
[00118] In one embodiment of a method for treating an ocular disorder with an immunoconjugate (timer and a VEGF inhibitor, for example, a method for treating wet AMD, diabetic retinopathy, diabetic macular edema, or choroidal neovascularization secondary to an ocular disorder such as wet AMD, the retinal thickness of the treated eye is reduced in the eye of the patient, as compared to the retinal thickness prior to treatment, as measured by optical coherence tomography (OCT). As provided herein, treatment can include one dosing session or multiple dosing sessions, and reduction in retinal thickness, in one embodiment, is assessed after individual dosing sessions, or multiple dosing sessions. In a further embodiment, the retinal thickness is reduced by at least about 5%, or at least about 10%, or at least about 15%, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, as measured by OCT. In a further embodiment, the decreased retinal thickness is decreased central retinal subfield thickness (CSTX decreased center point thickness (CPT), or decreased central foveal thickness (CFT).
[00119] In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered as a solution or a suspension. The immunoconjugate composition and/or VEGF inhibitor composition, in one embodiment, comprises argunne or protein A. In a further embodiment, the immunoconjugate composition and/or VEGF inhibitor composition comprises argimne. In even a further embodiment, the argininc is present in the composition at from about 20 mM to about 40 mM, e.g, at 25 mM. Other components of the composition, in one embodiment, include HEPES, sodium chloride, polysorbate-80, calcium chloride, or a combination thereof.
[00120] In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered in a dose of between 10 pg and 600 pg, 10 pg and 500 pg, 10 pg and 400 pg, lOug and 300 μg, 10 μg and 200 μg, 10 μg and 100 μg, 10 μg and SO μ^, SO μg and 600 pg, SO μg and 500 μ& 50 μg and 400 μ& 50 μg and 300 μg, 50 μ§ and 200 μ& 50 μ§ and ΙΟΟμ^ 100 μg and 600 μ¾, 100 μg and 500 μ¾ 100 μg and 400 μ¾ 100 μ¾ and 300 μ¾, 100 μg and 200 μ& 200 μg and 600 μ¾ 200 μ¾ and 500 μ& 200 μg and 400 μ& 200 μg and 300 μ¾ 300 μg and 600 μ¾, 300 μg and 500 μ§, 300 μg and 400 μ§, 400 μ% and 600 μ£, 400 μg and 500 μ§.
[00121] In one embodiment, the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of between about 10 μg and about 500 μg, about 10 μg and about 400 μg, about 10μg and about 300 μg, about 10 μg and about 200 μg, about 10 μg and about 100 μg, about 10 μg and about 50 μg, about 50 μg and about 500 ug, about 50 μ§ and about 400 μg, about 50 ug and about 300 μg, about 50 μg and about 200 ug, about 50 ug and about 100μ§, about 100 μg and about 500 μg, about 100 ug and about 400 μg, about 100 μg and about 300 μg, about 100 ug and about 200 μ§, about 200 μg and about 500 μg, about 200 ug and about 400 μg, about 200 μg and about 300 μg, about 300 ug and about 500 μg, about 300 μg and about 400 μg, or about 400 μg and about 500 μg
[00122] In one embodiment, the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.40 mg, about 0.45 mg, about 0.50 mg, about 0.55 mg, about 0.60 mg, about 0.65 mg, about 0.7 mg, about 0.75 mg, about 0.8 mg, about 0.9 mg, about 0.95 mg, about 1 mg, about 1.05 mg, about 1.1 mg, about 1.15 mg, about 1.2 mg, about 1.25 mg, about 1.30 mg, about 1.35 mg, about 1.4 mg, about 1.45 mg, or about 1.50 mg.
[00123] In one embodiment, the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose of 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.3 mg, 0.35 mg, 0.40 mg, 0.45 mg, 0.50 mg, 0.55 mg, 0.60 mg, 0.65 mg, 0.7 mg, 0.75 mg, 0.8 mg, 0.9 mg, 0.95 mg, 1 mg, 1.05 mg, 1.1 mg, 1.15 mg, 1.2 mg, 1.25 mg, 1.30 mg, 1.35 mg, 1.4 mg, 1.45 mg, or 1.50 mg.
[00124] In one embodiment, the immunoconj ugate dimer and/or VEGF inhibitor is administered in a dose consisting of about 10ug, about 20μg, about 30 ug, about 40 μg, about 50 μg, about 60 ug, about 70 μg, about 80 μg, about 90 μg, about 100 μg, about 125 μg, about 150 μg, about 175 μg, about 200 ug, about 225 μg, about 250 μg, about 275 μg, about 300 μg, about 325 μg, about 350 μg, about 375 ug, about 400 μg, about 425 μg, about 450 ug, about 475 μg, about S00 ug, about S2S ug, about SSO ug, about S7S ug, about 600 ug, about 625 ug, about 6S0 μg, about 675 ug, or about 700 ug,
[00125] In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered in a solute volume of between 10 uL and 200 uL, 10 uL and 180 uL, 10 uL and 160 uL, 10 uL and 140 uL, 10 uL and 120 uL, 10 uL and 100 uL, 10 uL and 80 uL, 10 uL and 60 uL, 10uL and 40 uL, 10 uL and 20 uL, 10 uL and 15 uL, 20 uL and 200 uL, 20uL and 180 uL, 20 uL and 160 uL, 20uL and 140uL, 20 uL and 120 uL, 20 uL and 100 uL, 20 uL and 80 uL, 20 uL and 60 uL, 20 uL and 40 uL, 40 uL and 200uL, 40 uL and 180 uL, 40 uL and 160 uL, 40uL and 140 uL, 40 uL and 120 uL, 40 uL and 100 uL, 40 uL and 80 uL, 40 uL and 60 uL, 60 uL and 200 uL, 60 uL and 180 uL, 60 uL and 160 uL, 60 uL and 140 uL, 60 uL and 120 uL, 60 uL and 100 uL, 60 uL and 80 uL, 80 uL and 200 uL, 80 uL and 180 uL, 80 uL and 160 uL, 80 uL and 140 uL, 80 uL and 120 uL, 80 uL and 100 uL, 100 uL and 200 uL, 100 uL and 180 uL, 100 uL and 160 uL, 100 uL and 140 uL, 100 uL and 120 uL, 120 uL and 200 uL, 120 uL and 180 uL, 120 uL and 160uL, 120 uL and 140 uL, 140 uL and 200 uL, 140 uL and 180 uL, 140 uL and 160 uL, 160 uL and 200 uL, 160 uL and 180 uL, or 180 uL and 200 uL.
[00126] In one embodiment, the immunoconjugate dimer and/or VEGF inhibitor is administered in a solute volume consisting of about 10uL, about 15 uL, about 20 uL, about 25 uL, about 30 uL, about 35 uL, about 40 uL, about 45 uL, about 50 uL, about 55 uL, about 60 uL, about 65 uL, about 70 uL, about 75 uL, about 80 uL, about 85 uL, about 90 uL, about 95 uL, or about 100 uL.
[00127] One exemplary composition of the present invention is provided in Table 2 below.
Figure imgf000033_0001
Treatment Outcomes
[00128] As provided herein, the administration of an immunoconjugate dimer of the invention and VEGF inhibitor results in an improved outcome compared to administration of the VEGF inhibitor alone. In some embodiments, the improved outcome is greater than an additive effect of the dimer and VEGF inhibitor. In some embodiments, the improved outcome is synergistic as compared to treatment with the dimer or VEGF alone. Outcome can be quantified by BVCA letter score; central subfield retinal thickness; thickness in the tissues/regions of the eye; CNV area, lesion area; CNV-associated exudation; CNV leakage area; volume of sub- retinal fluid; thickness of the central subfield sub-retinal hyper-reflective material; volume of total sub-retinal hyper-reflective material; presence or absence of intraretinal fluid, sub-retinal fluid, and/or sub-retinal pigment epithelium fluid; presence or absence of subfoveal and/or non- subfoveal cysts; atrophy and/or fibrosis; area of autofluorescence; area of discontinuous autofluorescence; volume of the central subfield pigment epithelium detachment; and integrity of the eye's outer nuclear layer, external limiting membrane, ellipsoid zone, and/or subfoveal retinal pigment epithelium.
[00129] In some embodiments, a patient is administered, in multiple dosing sessions, an effective amount of (1) an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVTIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain and (2) a VEGF inhibitor. In a further embodiment, the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF inhibitor prevents, inhibits, or reverses wet age-related macular degeneration (AMD) in an eye of a patient in need thereof. In a further embodiment, the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF inhibitor prevents, inhibits, or reverses ocular neovascularization in an eye of a patient in need thereof. In a further embodiment, the administration, in multiple dosing sessions, of the immunoconjugate dimer and the VEGF reverses tumor neovascularization in an eye of a patient in need thereof.
[00130] The administration of both an immunoconjugate dimer and a VEGF inhibitor results in superior clinical outcomes as compared to VEGF inhibitor monotherapy, as detailed herein. [00131] In one embodiment, a BCVA letter score is determined in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. Ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the BCVA letter score baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult. In other embodiments, the CNV is minimally classic. In one embodiment, the assessment BVCA letter score determinations occur as a last observation carried forward (LOCF) method.
[00132] In some embodiments, a patient gains greater than 5, 10, 15, 20, 25, 30, 35, or 40 letters in the BCVA letter score at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments, a patient gains greater than about 5, about 10, about 15, about 20, about 25, about 30, about 35, or about 40 letters in the BCVA letter score at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00133] In some embodiments, a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater improvement in the BCVA letter score, compared to a patient who received the VEGF inhibitor monotherapy.
[00134] In one embodiment, the central subfield retinal thickness in the eye is determined in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the central subfield retinal thickness baseline is determined, and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult. In one embodiment, the central subfield retinal thickness determinations occur as a last observation carried forward (LOCF) method. In one embodiment, the central subfield retinal thickness determination is made utilizing sdOCT.
[00135] In some embodiments, a patient exhibits an increase or decrease in the central subfield retinal thickness by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00136] In some embodiments, a patient exhibits an increase or decrease in the central subfield retinal thickness by at least 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00137] In one embodiment, the patient exhibits an increase or decrease in the central subfield retinal thickness of the tissues and/or regions of the eye presented herein is an increase or decrease of at least about lOum, about 20um, about 30 um, about 40 um, about 50 um, about 60 um, about 70 um, about 80 um, about 90 um, about 100 um, about 125 um, about 150 um, about 175 um, about 200 um, about 225 um, about 250 um, about 275 um, about 300 um, about 325 um, about 350 um, about 375 um, about 400 um, about 425 um, about 450 um, about 475 um, about 500 um, about 525 um, about 550 um, about 575 um, about 600 um, about 625 um, about 650 um, about 675 um, or about 700 um.
[00138] In one embodiment, the measure of thickness of the tissues and/or regions of the eye presented herein is an increase or decrease of at least lOum, 20um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, 100 um, 125 um, 150 um, 175 um, 200 um, 225 um, 250 um, 275 um, 300 um, 325 um, 350 um, 375 um, 400 um, 425 um, 450 um, 475 um, 500 um, 525 um, 550 um, 575 um, 600 um, 625 um, 650 um, 675 um, or 700 um. [00139] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the central subfield retinal thickness, compared to a patient who received the VEGF inhibitor monotherapy.
[00140] In one embodiment, a measure of the CNV area is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the CNV area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determinations of the CNV areas occur as a last observation carried forward (LOCF) method.
[00141] In some embodiments, a patient exhibits a decrease in the CNV area by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00142] In some embodiments, a patient exhibits a decrease in the CNV area by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00143] In some embodiments, a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the CNV area, compared to a patient who received the VEGF inhibitor monotherapy. [00144] In one embodiment, a measure of the area of the lesion is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the lesion area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determinations of the area of lesions occur as a last observation carried forward (LOCF) method. See FIG. 23 for synergistic decrease of lesion size, as measured by CTLF for combination administration of ICON-1 + VEGF inhibitor.
[00145] In some embodiments, a patient exhibits a decrease in the area of lesions by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00146] In some embodiments, a patient exhibits a decrease in the area of lesions by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00147] In some embodiments, a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater in the area of lesions, compared to a patient who received the VEGF inhibitor monotherapy.
[00148] In one embodiment, a measure of the CNV-associated exudation is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the CNV-associated exudation baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months. In some embodiments the CNV- associated exudation is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determinations of the CNV areas occur as a last observation carried forward (LOCF) method.
[00149] In some embodiments, a patient exhibits a decrease in the CNV-associated exudation by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00150] In some embodiments, a patient exhibits a decrease in the CNV-associated exudation by at least at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00151] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater improvement in the BCVA letter score, compared to a patient who received the VEGF inhibitor monotherapy.
[00152] In one embodiment, a measure of the area of leaking CNV (leakage area) is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the leakage area baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the measure of the leakage area occurs as a last observation carried forward (LOCF) method.
[00153] In some embodiments, a patient exhibits a decrease in the area of leakage area by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult.
[00154] In some embodiments, a patient exhibits a decrease in the area of leakage area by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00155] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the area of leakage, compared to a patient who received the VEGF inhibitor monotherapy.
[00156] In one embodiment, a measure of the volume of the sub-retinal fluid is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the measure of the volume of the sub- retinal fluid baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the measure of the measure of the volume of the sub-retinal fluid occurs as a last observation carried forward (LOCF) method. [00157] In some embodiments, a patient exhibits a decrease or increase in the volume of the sub-retinal fluid by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV.
[00158] In some embodiments, a patient exhibits a decrease or increase in the volume of the sub-retinal fluid by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00159] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the volume of the sub-retinal fluid, compared to a patient who received the VEGF inhibitor monotherapy.
[00160] In one embodiment, a measure of the thickness of the central subfield subretinal hyper-reflective material is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the measure of the thickness of the central subfield subretinal hyper-reflective material baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the measure of the thickness of the central subfield subretinal hyper- reflective material occurs as a last observation carried forward (LOCF) method. [00161] In some embodiments, a patient exhibits a decrease or increase in the thickness of the central subfield subretinal hyper-reflective material by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV.
[00162] In some embodiments, a patient exhibits a decrease or increase in the thickness of the central subfield subretinal hyper-reflective material by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00163] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the thickness of the central subfield subretinal hyper-reflective material, compared to a patient who received the VEGF inhibitor monotherapy.
[00164] In one embodiment, a measure of the total subretinal hyper-reflective material volume is taken in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the measure of the total volume of the subretinal hyper-reflective material baseline is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments, distinctions are made between subfoveal versus non-subfoveal. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the measure of the total volume of the subretinal hyper-reflective material occurs as a last observation carried forward (LOCF) method.
[00165] In some embodiments, a patient exhibits a decrease or increase in the total volume of the subretinal hyper-reflective material by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In some embodiments, distinctions are made between subfoveal versus non-subfoveal.
[00166] In some embodiments, a patient exhibits a decrease or increase in the total volume of the subretinal hyper-reflective material by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments, distinctions are made between subfoveal versus non-subfoveal.
[00167] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease in the total volume of the subretinal hyper-reflective material, compared to a patient who received the VEGF inhibitor monotherapy.
[00168] In one embodiment, the identification of the presence or absence is made for (1) intraretinal fluid, (2) subretinal fluid, (3) subretinal pigment epithelium fluid in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti- VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the baseline determination of the presence or absence of fluid in said ocular locations is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the presence or absence of fluid in said ocular locations occurs as a last observation carried forward (LOCF) method.
[00169] In some embodiments, a patient exhibits a presence or absence of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00170] In some embodiments, a patient exhibits a presence or absence of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00171] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase or decrease of (1) intraretinal fluid, (2) subretinal fluid, and/or (3) subretinal pigment epithelium fluid, compared to a patient who received the VEGF inhibitor monotherapy.
[00172] In one embodiment, the identification of the presence or absence of subfoveal or non-subfoveal cysts in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the baseline determination of the presence or absence of subfoveal or non-subfoveal cysts is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the presence or absence of said cysts occurs as a last observation carried forward (LOCF) method. [00173] In some embodiments, a patient exhibits a presence or absence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment In some embodiments, a patient exhibits a decrease in the presence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00174] In some embodiments, a patient exhibits a presence or absence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment In some embodiments, a patient exhibits a decrease in the presence of subfoveal or non-subfoveal cysts at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00175] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits the presence or absence of or about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% decrease in subfoveal or non-subfoveal cysts, compared to a patient who received the VEGF inhibitor monotherapy.
[00176] In one embodiment, the identification of atrophy and/or fibrosis is made for the eye in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments the baseline identification of atrophy and/or fibrosis is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the presence or atrophy and/or fibrosis occurs as a last observation carried forward (LOCF) method. [00177] In some embodiments, a patient exhibits a decrease in the atrophy and/or fibrosis of the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00178] In some embodiments, a patient exhibits a decrease in the atrophy and/or fibrosis of the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00179] In some embodiments, a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the atrophy and/or fibrosis of the eye, compared to a patient who received the VEGF inhibitor monotherapy.
[00180] In one embodiment, the total area of decreased autofluorescence is determined for the in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments, the baseline determination of the area of decreased autofluorescence is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the total area of decreased autofluorescence occurs as a last observation carried forward (LOCF) method.
[00181] In some embodiments, a patient exhibits a decrease in the total area of autofluorescence the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, S months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00182] In some embodiments, a patient exhibits a decrease in the total area of autofluorescence the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00183] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the total area of autofluorescence, compared to a patient who received the VEGF inhibitor monotherapy.
[00184] In one embodiment, the total area of discontinuous autofluorescence in the eye is determined for a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconj ugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments, the baseline determination of the total area of discontinuous autofluorescence is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the total area of discontinuous autofluorescence occurs as a last observation carried forward (LOCF) method.
[00185] In some embodiments, a patient exhibits a decrease in the total area of discontinuous autofluorescence the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. [00186] In some embodiments, a patient exhibits a decrease in the total area of discontinuous autofluorescence the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00187] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the total area of discontinuous autofluorescence, compared to a patient who received the VEGF inhibitor monotherapy.
[00188] In one embodiment, a measurement of the volume of the central subfield pigment epithelium detachment is determined for in a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconjugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments, the baseline determination of the volume of the central subfield pigment epithelium detachment is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the volume of the central subfield pigment epithelium detachment occurs as a last observation carried forward (LOCF) method.
[00189] In some embodiments, a patient exhibits a decrease in the volume of the central subfield pigment epithelium detachment by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. [00190] In some embodiments, a patient exhibits a decrease in the volume of the central subfield pigment epithelium detachment by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment.
[00191] In some embodiments, a patient treated with an immunoconj ugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater decrease in the volume of the central subfield pigment epithelium detachment, compared to a patient who received the VEGF inhibitor monotherapy.
[00192] In one embodiment, a determination of the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of an eye is made for a patient or a population of patients wherein patients are grouped into one of the following three groups: (1) ICON-1 immunoconj ugate dimer monotherapy, (2) VEGF inhibitor monotherapy (e.g. anti-VEGF antibody, e.g. ranibizumab), and (3) immunoconjugate dimer and VEGF inhibitor therapy treatment groups. In some embodiments, the baseline determination of the integrity of (1) - (4) is determined and is then repeated following the commencement of treatment at 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. In some embodiments the CNV is classical CNV. In other embodiments, the CNV is occult CNV. In one embodiment, the determination of the integrity of (10 - (4) occurs as a last observation carried forward (LOCF) method.
[00193] In some embodiments, a patient exhibits an increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment. [00194] In some embodiments, a patient exhibits an increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye by at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% at 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 12 months after beginning treatment
[00195] In some embodiments, a patient treated with an immunoconjugate dimer and a VEGF inhibitor exhibits about a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even a 100% greater increase in the integrity of the (1) outer nuclear layer, (2) external limiting membrane, (3) ellipsoid zone, and (4) subfoveal retinal pigment epithelium of the eye, compared to a patient who received the VEGF inhibitor monotherapy.
[00196] In some embodiments, the immunoconjugate dimer utilized in treatments of the present disclosure is the ICON-1.5 immunoconjugate.
EXAMPLES
[00197] The present invention is further illustrated by reference to the following Examples. However, it should be noted that these Examples, like the embodiments described above, are illustrative and are not to be construed as restricting the scope of the invention in any way.
Example 1 - Evaluation Of hl-conl In in vitro Thrombin Generation Assays
[00198] The effect of hl-conl (SEQ ID NO:2) in a thrombin generation assay in plasma was tested. Specifically, the effect of hl-conl on thrombin generation in plasma in a tissue factor initiated reaction using a thrombogram (CAT-like) assay (Hemker et al. 2002. Pathophysiol. Haemost Thromb. 32, pp. 249-253; Mann et al. 2007. J. Thrornb Haemost. 5, pp. 2055-2061, each incorporated by reference herein in its entirety for all purposes) was evaluated. For the CAT-like assays, multidonor human citrate plasma from healthy individuals, human FVII- deficient plasma and normal rabbit citrate plasma were used. Thrombin (also referred to as Factor Ila, or activated blood coagulation factor Π) generation was initiated either with human relipidated TF (in human plasma) or with rabbit relipidated TF (in rabbit plasma).
[00199] hl-conl was maintained frozen at -70 °C until use. Each sample included 3.0 mg hl-conl/mL in formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCh, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
[00200] Human plasma FVIIa in 50% glycerol was purchased from Haematologic Technologies, Inc., 57 River Road, Essex Junction, VT 05452. It was stored at -20 °C until use. Before use, it was diluted to 10 nM in the formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaC12, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
[00201] Spectrozyme FXa (#222), lipidated recombinant human TF reagent (Catalog #4500L) and lipidated recombinant rabbit TF were purchased from American Diagnostica, Inc. (Stamford, CT), pooled normal human plasma (Lot #IR 11-020711) and rabbit plasma (Lot #26731) were purchased from Innovative Research Novi, ML 48377), congenital FVII-deficient plasma (Catalog #0700) was purchased from George King Bio-Medical, Inc. (Overland Park, KS) and human factor X (hFX) (#HCX-0050) and Phe-Pro-Arg-chloromethylketone (FPRck; Catalog #FPRCK-01), corn trypsin inhibitor (CTl; Catalog #CTI-01) were purchased from Haematologic Technologies, Inc (Essex Junction, VT, USA). Fluorogenic substrate Z-CHy-Gly- Arg-AMC HCl was purchased from Bachem (Torrance, CA) and ethylenediaminetetraacetic acid disodium salt dihydrate (EDTA; #E5134), NaCl (#S7653) and HEPES (#H3375) were purchased from Sigma (St. Louis, MO). HBS buffer, pH 7.4 contained 150 mM NaCl, 2 mM CaCh and 20 mM HEPES.
[00202] Active site inhibited FVTIa (FVJJai) was produced in house. 1, 2-Dioleolyl-sn- Grycero-3-Phospho-L-Serine (PS; #840035) and 1, 2-DioleoyJ-sn-Glycero-3-Phosphocholine (PC; #850375) were purchased from Avanti Polar Lipids, Inc. (Alabaster, AL, USA). Phospholipid vesicles (PCPS) composed of 25% PS and 75% PC were prepared as described in Higgins and Mann 1983, incorporated by reference herein in its entirety for all purposes.
Extrinsic FXase [00203] Lipidated recombinant human TF (0.1 nM) was incubated with either 5 nM plasma FVIIa or 5 nM hl-conl or mixture of both (each at 5 nM) and 100 μΜ PCPS for 10 min at 37 °C. FX (4 μΜ) was added and at selected time points (0-5 min.) 10 μΐ. aliquots of the reaction mixture were quenched into 170 \iL HBS-0.1% PEG-20 mM EDTA. Twenty μL of Spectrozyme FXa (0.2 mM) was added and the rate of substrate hydrolysis was measured as an increase in absorbance at 405 nm (mOD/min).
Thrombin generation (CAT -like) assay
[00204] Corn trypsin inhibitor (CTI) at a final 0.1 mg/mL concentration was added to citrate plasma and 80 μί^ of this plasma was transferred into Immulon® 96- well plate (Thermo Electron Co., Waltham MA). When desired, hl-conl, plasma FVIIa and FVIIai were added at selected concentrations. Twenty μΐ^ of 5 pM TF and 20 μΜ PCPS mixture (both concentrations final) were added to CTI-plasma and incubated for 3 min. Thrombin generation was initiated by the addition of 20 μL of 2.5 mM ZGly-Gly- ArgAMC · HC1 in HBS containing 0.1 M CaCh. Final concentration of substrate was 416 μΜ and that of CaCh was 15 mM Thrombin generation curves were generated using Thrombinoscope BY software.
Results
Comparison of hl-conl with plasma FVIIa in the extrinsic FXase
[00205] FXa-generating efficiency of two forms of FVIIa and of their mixture was determined in a chromogenic assay, hl-conl was less active than plasma FVIIa. Activity of hl- conl was 18% of that observed for plasma FVIIa. When both proteins were added at equimolar (5 nM) concentration, the rate of FXa generation in the middle between the rates observed for individual proteins, indicating that hl-conl competes with plasma FVIIa for the limited amount of TF (FIG. 2). These data also suggest that hl-conl has similar affinity for TF as plasma FVIIa.
Thrombin generation in normal human plasma: the effect of FVIIai
[00206] It was hypothesized that due to the low activity of the hl-conl tissue factor (TF) complex in the extrinsic FXase, hl-conl could act as an inhibitor by binding TF into an inefficient complex and preventing formation of an efficient complex between plasma FVIIa and TF. To test this hypothesis, the effect of a known inhibitor of coagulation, i.e., active site inhibited FVlIa (Kjalke et al. 1997), on thrombin generation in normal human plasma was evaluated. FVIIai at 1 nM concentration had no effect on thrombin generation initiated with lipidated human TF (FIG. 3). However, at 10 nM, FVIIai prolonged the lag phase of thrombin generation and significantly suppressed both the maximum rate of thrombin generation and the maximum levels of thrombin produced. No thrombin generation was observed in the absence of TF.
Thrombin generation in normal human plasma: the effect of hl-conl
[00207] hl-conl was titrated into normal human plasma initiated with TF to generate thrombin. Varying concentrations of hl-conl was used, however even at extremely high hl-conl concentrations (1 μΜ), no inhibition of thrombin generation was observed (FIG. 4).
Thrombin generation in congenital FVII-deficient human plasma
[00208] No thrombin generation was observed upon the addition of lipidated human TF to congenital FVII-deficient plasma, indicating that there no detectable functional FVHa in that plasma (FIG. 5). An addition of 0.1 nM plasma FVIIa together with TF produced thrombin generation profile slightly lower than that observed in normal human plasma. An addition of 0.1 nM hl-conl alone in the presence of TF led to the initiation of thrombin generation, however the process was significantly delayed and suppressed (FIG. 5). This result was consistent with the observation of low hl-conl activity in the extrinsic FXase. The addition of both plasma FVIIa and hl-conl at equimolar concentrations (0.1 nM) did not impair thrombin generation initiated with plasma FVHa alone.
Thrombin generation in normal rabbit plasma
[00209] Thrombin generation in rabbit plasma was initiated with lipidated rabbit TF. The addition of 1 nM hl-conl to this plasma had no pronounced effect on thrombin generation (FIG. 6). Similarly, no pronounced effect was observed when 10 nM FVIIai was added. At higher hl- conl concentrations (10-1000 nM) some suppression in thrombin generation was observed. However, the control experiment with no TF added led to thrombin generation, suggesting an endogenous presence of TF. Thrombin generation in centrifuged rabbit plasma
[00210] After centrifugation of rabbit plasma, an endogenous thrombin generating activity did not disappear completely, but was significantly decreased (FIG. 7). No suppression in TF- triggered thrombin generation was observed when 10 nM FVIIai was added. Similarly, no suppression was observed when 1-100 nM hl-conl was added and only a limited decrease in thrombin generation was observed when high concentration (1 μΜ) hl-conl was added (FIG. 7). These data indicate that at physiologically-relevant concentrations hl-conl does not compete for rabbit TF with rabbit FVIIa.
Conclusions
[00211] hl-conl does not compete with plasma FVIIa for TF in the citrate plasma environment, hl-conl has no pronounced (if any) effect on thrombin generation either in human plasma initiated with human TF or in rabbit plasma initiated with rabbit TF. It is not likely that hl-conl would cause bleeding or thrombotic complications.
Example 2 - Effects of Treatment With hl-conl on Choroidal Neovascularization in the Pig
[00212] In this study, hl-conl activity in a porcine wet AMD model (Kiilgaard et al., 2005. Acta. Ophthalmol. Scand. 83, pp. 697-704, incorporated by reference herein in its entirety for all purposes) and the optimal dose for the activity was examined. Additionally, the safety of hl-conl when administered by intravitreal injection was determined.
[00213] In this study intravitreal injection of hl-conl was demonstrated to result in the destruction of established laser-induced CNV in this porcine model. The injections of hl-conl were well tolerated and the effects were dose-related, with and EDso of 13.5 pg/dose. A major breakdown product of hl-conl (lOOkDa) was tested and was also well-tolerated and effective with an EDso of 16.2 ug/dose.
Test Articles hl-conl [00214] hl-conl was provided by Laureate Pharma Inc., 201 E. College Ave, Princeton, NJ, 08540. hl-conl was maintained frozen at -70 °C until use: Lot PURIC1 080402 (SEC Fr 10- 14), two vials each containing 200 uL at 2.0 mg/mL, 1.0 mg/mL, 0.5 mg/mL and 0.25 mg/mL in formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCh, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
100 kD Fragment of hl-conl
[00215] The following samples of the 100 kD fragment of hl-conl were provided by Laureate Pharma Inc. 201 E. College Ave, Princeton, NJ, 08540. The fragment was maintained frozen at -70 °C until use: Lot PURIC1 080402 (SEC Fr 15), two vials each containing 200 uL at 2.0 mg/mL, 1.0 mg/mL, 0.5 mg/mL and 0.25 mg/mL in formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCh, 25 mM Arginine, 0.01% Tween 80, pH 7.4).
Control Article
[00216] The formulation buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaCk, 25 mM Arginine, 0.01% Tween 80, pH 7.4) was used as the vehicle control.
Test Animals
[00217] Two studies were conducted, each with groups of five (one group per test article) Yucatan miniature pigs (Sus scrofa), 10-12 weeks old, each weighting approximately 20 kilograms were bought from Professional Veterinary Research (Brownstown, IN, USA).
Husbandry
[00218] Each pig was maintained in a separate cage within a communal environment that housed four pigs. The lighting was computer controlled and set for a 6am to 6pm cycle. The temperature average was 70-72 ° F, with a variation of +/- 1 degree. Humidity was kept between 30 and 70%, with average humidity equal to 33%. The animals were evaluated by the large animal husbandry supervisor and a licensed veterinary technician on arrival and a licensed veterinary technician once weekly until they were euthanized. A veterinarian evaluated the animals to determine if there were any abnormalities or concerns. The animals were quarantined for about 1 week prior to experiments. Feed and Water
[00219] Daily feed and water were provided to the miniature pigs. They were bedded on hay that served as a feed supplement. The feed was Purina #5084, Laboratory Porcine Grower Diet, Manufactured by Purina Mills, LLC, 555 Maryville University Drive, Suite 500, St. Louis, Missouri 63141, and fed at 2% body weight per day. The water was 0.5 micron filtered tap water. It was not routinely analyzed for contaminants except by the water company and reports are reviewed annually.
Justification of Species
[00220] hl-conl has limited cross-species activity and the pig is one of the few laboratory animal species in which it is active. The vitreous cavity of the pig is approximately 3 mL, allowing intravitreal injection of reasonable volumes of test article. The pig eye has retinal vascular similarities to humans in addition to several cone-dominant regions of the retina that are similar to the human macula.
Methods
Laser-Induced Choroidal Neovascularization
[00221] Under general anesthesia, the pupils of the animals were dilated with 1% tropicamide and 2.5% phenylephrine. An indirect ophthalmoscope with a double-frequency YAG laser (532 nm) was used to deliver 74 spots per eye using a 2.2 D lens and the following laser parameters: laser power 1000-1500 mW, duration 0.1 seconds, and repetition rate 500 msec. The laser treatment was designed to yield a microrupture of the Bruch's membrane, generating CNV at 60-70% of the laser spots within two weeks (Bora et al, 2003, incorporated by reference herein in its entirely for all purposes).
Study Design
[00222] The study design is summarized in Table 3 below.
Figure imgf000056_0001
Figure imgf000057_0001
[00223] Choroidal neovascularization was induced on Day 0 in both eyes of two groups of 5 pigs. On Day 10, 100 uL of solutions of hl-conl (Study 1) or its 100 kD fragment (Study 2) at 0.25, 0.5, 1.0 or 2.0 mg/mL were administered by intravitreal injection into both eyes of the pigs as shown in Table 3. On Day 10, 100 uL of formulation buffer was administered by intravitreal injection into both eyes of the control pigs.
Test and Control Articles Administration
[00224] The animals were anesthetized with a mixture of ketamine hydrochloride (40 mg/kg) and xylazine hydrochloride (10 mg/kg). Injections were administered using a strict sterile technique, which involved scrubbing the lids with a 5% povidone-iodine solution and covering the field with a sterile eye drape. A sterile lid speculum was used to maintain exposure of the injection site. All injections were performed 2 mm from the limbus through the pars plana, using a 30- gauge needle on a 1 mL tuberculin syringe. After injection, a drop of 2% cyclopentolate and antibiotic ointment was placed in the eye. The animals were examined daily for signs of conjunctival injection, increased intraocular pressure, anterior uveitis vitritis, or endophthalmitis, and were sacrificed on Day 14.
Terminal Procedures
[00225] On Day 14, the pigs were anesthetized with an 8:1 mixture of ketamine and xylazine and perfused through the ear vein with 10 mL PBS containing 3 mg/mL fluorescein- labeled dextran with an average molecular weight 2 x 106 (Sigma, St Louis, MO, USA). The eyes were enucleated and four stab incisions were made at the pars plana followed by fixation in 4% paraformaldehyde for 12 hours at 4 °C. The cornea and the lens were removed, and the neurosensory retina was dissected from the eyecup and four radial cuts were made from the edge of the eyecup to the equator. The choroid-retinal pigment epithelium (RPE) complex was separated from the sclera and flatmounted on a glass slide in Aquamount with the inner surface (RPE) facing upwards. Flat mounts were stained with a monoclonal antibody against elastin (Sigma) and a Cy 3 -conjugated secondary antibody (Sigma) and examined with a confocal microscope (Zeiss LSM510, Thornwood, NY, USA). The vasculature, filled with dextranconjugated fluorescein, stained green and the elastin in the Bruch's membrane stained red. The level of the Bruch's membrane was determined by confocal microscopy using the intense red signal within a series of z-stack images collected at and around the laser spot. The presence of CNV was indicated by the branching linear green signals above the plane of Bruch's membrane. Absence of CNV was defined under very stringent criteria as the total absence of green fluorescence in the vessels in the spot (see Tezel et al., 2007. Ocular Immunol Inflamm 15, pp. 3-10.
Statistical Analysis
[00226] The percentage of laser spots with CNV at different doses of hl-conl or its 100 kD fragment was compared pair wise by a chi-square test. The results were plotted against the hl-conl dose to derive the best-fit curve, which was used to calculate the dose of hl-conl that reduces the fraction of laser spots with CNV by 50% (ED50). A confidence level of p <0.05 was considered to be statistically significant
Results
Effects of Intravitreal Treatment with hl-conl on CNV
[00227] Choroidal neovascularization developed in 71.9 ± 5.8% of the laser spots in control eyes. A single intravitreal injection of hl-conl on Day 10 in pig eyes (n = 2 at each dose) significantly reduced subretinal CNV on Day 14 at all doses tested, i.e., 25-200 pg, Table 4; FIG. 8). The inhibitory effect of hl-conl fit well to a 5-parameter Sigmoidal Weibull curve. The dose causing a 50% decrease in the yield of CNV (ED50) was 13.5 pg.
Figure imgf000058_0001
Figure imgf000059_0001
Effects of Intravitreal Treatment with 100 kD fragment of hi- con 1 on CNV
[00228] Choroidal neovascularization developed in 85.6 ± 4.1% of the laser spots in control eyes. A single intravitreal injection of the 100 kDa fragment of hl-conl on Day 10 in pig eyes (n = 2 at each dose) significantly reduced subretinal CNV on Day 14 at all doses tested, i.e., 25-200 ug. Table 4, FIG.9). The inhibitory effect of hl-conl fit well to a 5-parameter Sigmoidal Weibull curve. The dose causing a 50% decrease in the yield of CNV (ED50) was 16.2 ug.
Figure imgf000059_0002
[00229] Intravitreal injection of hl-conl and its 100 kD fragment at doses from 25-200 μg caused significant regression of pre-existing laser-induced CNV 4 days after the injections were administered. The response of the lesions to the injections was clearly dose-related with EDso doses of 13.5 and 16.2 μg, respectively. These results indicate that the specific activity of the 100 kD fragment of hl-conl is similar to that of the intact molecule. Doses greater than 100 μ§ had very little additional decrease in CNV; thus, the efficacious dose in this model is < 100 pg.
Example 3 - Tissye Cross-Reactivity Study of hl-ςοηΐ yfi\h Normal Human Tissues [00230] In this study, the binding of hl-conl to normal human tissues was assessed using standard immunohistochemistry (IHC) techniques, in a standard tissue cross-reactivity (TCR) study. The study was performed utilizing a single batch of biotinylated hl-conl for IHC staining of normal, as well as positive and negative control human tissues. A positive staining result is indicative of potential toxicities associated with administration of hl-conl to humans in vivo.
[00231] In this model, tissue staining was observed only in the positive control colon carcinoma tumor. All other normal human tissues showed no immunoreactivity. These findings indicate that hl-conl binding is specific to abnormal tissue, with no binding to normal tissues observed.
Example 4 - Evaluation of die Binding of hl-conl to Lipidated Tissue Factors
[00232] To allow for cross species comparison, a Biacore study of the kinetics of binding of hl-conl and hFVUa to human lapidated TF (hTF) and rabbit lipidated TF (rTF) was conducted.
[00233] As described in detail below, hl-conl and hFVTia both bound with high and approximately equal affinity to lapidated hTF.
Materials and Methods
[00234] Lipidated rabbit tissue factor (rTF; Product # 4520L; Lot # 051017) purchased from American Diagnostica. Lipidated human tissue factor (hTF; Lot FTL105HO1) supplied by Marin Biological Laboratories, 378 Bel Marin Keys, Novate, CA 94949.
[00235] hl-conl ; 1 ml; 100 μg/ml; MW 157 kDa
[00236] Human FVHa; Lot # A09050525 (Fitzgerald); 1.01 mg/ml; 40 uL/vial; MW 50 kDa
Equipment: Biacore 3000; CM5 Sensor chip
[00237] The GE procedure for proteoliposome immobilization (amine coupling) protocol was used to coat PS/PC/rTF on flow cell 2, and PS/PC/hTF on flow cell 3. Flow cells were equilibrated with running buffer (15 mM Hepes, 150 mM NaCl, 5 mM CaC12, 25 mM Arginine, 0.01% Tween 80, pH7.4) at a flow rate of 5 μί/πιΐη. Kinetic analyses were performed at 37 °C by flowing consecutively increasing concentrations of each analyte (0-10 nM) in the running buffer (15 mM HEPES, 150 mM NaCl, 5 mM CaC12, 25 mM Arginine, 0.01% Tween 80, pH 7.4) over the sensor chip for 5 min followed by a 10 min dissociation period at a flow rate of 30 uL /min in parallel.
[00238] Analyte binding to the lipidated TF was determined by subtracting the RU values noted in the reference flow cell 1 from flow cell 2 and 3. Binding of analytes to the TFs was monitored in real time to obtain on (ka) and off (kd) rates. The equilibrium dissociation constant (KD) was calculated from the observed ka and kd.
[00239] The chips were regenerated with 3 min pulses of 10 mM EDTA in HEPES buffer (20 mM HEPES, 150 mM NaCl, pH 7.4).
[00240] Capture of rTF to the chip - Flow cell 2 was coated with rabbit TF (>Resonance Units [RU] 10,000) by amine coupling. Flow cell 3 was coated with human TF (>RU 8,000) by amine coupling.
Determination of the amount of test lieands (RL) to be captured on the chip
[00241] In this experiment, the desired level of RM¾X for the measurement of ligand- analyte interaction was based on the value determined by a previous experiment where rTF captured at 10,000 Resonance Units ("RU") gave binding of hl-conl with RMSX of 15 RU and hTF captured at 8,000 RU gave binding of hl-conl with RMHX of 10RU. The amount of the analyte to be captured on the chip depended on the molecular weights of the interacting proteins. It is determined by the following formula:
[00242] RMax = MWA / MWL · RL
[00243] MWA is the molecular weight of the analyte (157 kDa for hl-conl, 50 kDa for hFVIIa, and 150 kDa for IgGl).
[00244] MWL is the molecular weight of the ligand, in this assay it is expected to be very large (multiple of 35 kDa). Flow rate of the antibody solution
[00245] The flow rate used for capturing the ligand was 10 μ17πιϊη. [00246] For kinetics analysis, the flow rate of 30 μΐνπΰη. was used. Kinetic analysis
[00247] Based on the saturation concentration of the analyte, binding analysis was performed using saturating analyte concentrations of 0-500 nM for rabbit TF and 0-50 nM for human IF. Chi squared (χ2) analysis was carried out between the actual sensorgram and the calculated on- and off-rates to determine the accuracy of the analysis.
[00248] χ2 value up to 2 is considered significant (accurate) and below 1 is highly significant (highly accurate).
Results
[00249] The Biacore assay results are provided in Table 6 below.
Figure imgf000062_0001
[00250] As shown in Table 7 below, hl-conl and hFVIIa both bound with high, and approximately equal, affinity to lipidated hTF. Bom ligands also bound to lipidated rTF with approximately 10-fold lower affinities.
Figure imgf000062_0002
Figure imgf000063_0001
Example S - Randomized. Double-Masked. Multicenter. Active-Controlled Study
Evaluating ICON-1 in Patients With CNV Secondary to Age-Related Macular
Degeneration
[00251] In this study, the safety and efficacy of intravitreal injections of ICON-1, administered as monotherapy or in combination with ranibizumab (LUCENTIS) compared to ranibizumab (RZB) monotherapy in patients with choroidal neovascularization (CNV) secondary to age-related macular degeneration (AMD) was assessed.
[00252] Additionally, the biological activity and pharmacodynamics effect of ICON-1, administered as monotherapy or in combination with ranibizumab (LUCENTIS) compared to ranibizumab monotherapy was assessed.
[00253] The study presented in this example is a randomized, double-masked, multicenter, active-controlled study. Patients enrolled in this study were naive to treatment for CNV. Patients were randomly assigned to one of the following three treatment arms in the selected study eye in a 1 : 1 : 1 ratio:
• ICON-1 monotherapy (0.3 mg) + sham injection
• ranibizumab monotherapy (0.5 mg) + sham injection
• ICON-1 (0.3 mg) + ranibizumab (0.5 mg) combination therapy
[00254] Randomization was stratified by best-corrected visual acuity (BCVA) letter score in the study eye at baseline (<54 letters versus >55 letters) and by study site.
[00255] Patients received up to two intravitreal injections at each injection visit. In order to maintain the study mask among the treatment arms, a sham injection was employed in patients receiving monotherapy. There were masked and unmasked study personnel. Intravitreal (IVT) injections were administered by unmasked injecting physicians. Masked evaluating physicians or designated masked site staff members performed all study assessments except for post-injection assessments.
[00256] Patients were administered intravitreal injections in the study eye once every four weeks at months 0, 1 and 2. As of Month 3 (at Months 3, 4 and S) patients were retreated according to their assigned treatment arm, based on their individual observed treatment response. The masked investigator used the following retreatment criteria (based on the category of individual patient response) to determine if treatment was required at these visits:
• Loss of >5 letters of BCVA due to AMD compared to the previous scheduled visit.
• Independent of BCVA change, any anatomical evidenced of increased CNV activity (e.g., new or increased fluid and/or leakage, hemorrhage) compared to the previous scheduled visit.
• No BCVA change compared to Baseline (Visit 2), but there is anatomical evidence of persistent CNV activity (e.g., same persistent fluid and CST compared to Baseline.
[00257] Rescue treatment with 0.S mg of ranibizumab was administered to the study eye as an add-on therapy at any time during the 6-month treatment and follow-up period if either of the following conditions occured:
• Loss of > 15 letters of BCVA due to AMD compared to Baseline (Visit 2).
• Loss of > 10 letters from baseline (Visit 2) of BCVA due to AMD that is confirmed at two consecutive visits. Patients with a loss of >10 letters compared to baseline are requested to return within 7 days or as soon as possible for additional follow up at an unscheduled visit.
[00258] The masked physician made a determination if rescue treatment was needed according to the above criteria.
[00259] If rescue treatment was administered to the study eye during a scheduled injection visit, to ensure that the study masking is maintained, the unmasked physician administers rescue treatment and the patient's scheduled study treatment/re-treatment was as follows.
ICON-1 monotherapy arm: ICON-1 (0.3 mg) + rescue therapy (0.5 mg ranibizumab). • ranibizumab monotherapy arm: ranibizumab (0.S mg) + sham injection.
• combination therapy: ICON-1 (0.3 mg) + ranibizumab (0.5 mg).
[00260] If rescue treatment was administered to the study eye at an unscheduled visit, the unmasked physician administered rescue treatment as requested.
[00261] If rescue treatment was administered to the study eye, the patient continued with the study visit schedule for the next visit in accordance with the protocol and continued receiving study treatment according to the assigned randomization arm.
[00262] Safety was evaluated by tracking of adverse events, clinical laboratory tests (serum chemistry, hematology and coagulation), vital signs measurements, abbreviated physical examinations, slit-lamp biomicroscopy, intraocular pressure (IOP) and dilated ophthalmoscopy. Pharmacodynamic and biological activity were measured by means of BCVA by ETDRS visual acuity chart, spectral-domain optical coherence tomography (sdOCT), color fundus photography (CFP), fundus fluorescein angiography (FA), fundus autofluorescence (FAF), contrast sensitivity, and microperimetry. Pharmacokinetic (PK) and immunogenicity was evaluated by means of measuring plasma concentrations of ICON-1 and anti-drug antibodies.
[00263] A total of 88 patients were enrolled and randomized in the study: 30 patients each in the ICON-1 + RZB combination therapy arm and ICON-1 monotherapy arm, and 28 patients in the RZB monotherapy arm. See FIG. 10 for patient distribution across the three treatment arms.
Results
[00264] At baseline, the mean total area of CNV lesion was relatively low in all treatment arms. The lowest mean CNV lesion area at baseline was in the ICON-1 + RZB combination therapy arm (3.69 mm2), while it was 4.74 mm2 for patients in the ICON-1 monotherapy arm and 6.00 mm2 for patients in the RZB monotherapy arm. The mean reduction in the CNV lesion area was higher in the ICON-1 + RZB combination arm at month 6, with the reduction of more than - 0.97 mm2 at month 3 that was maintained at month 6. See FIG. 11 and FIG. 12 for tabulated data pertaining to the baseline CNV lesion area and the mean CNV lesion area change from baseline. FIG. 13 provides a visual representation of the proportion of patients in the three treatment arms as they correspond to shrinkage, growth, or no change in lesion size.
[00265] Reduction of the CNV lesion area was greatest in patients receiving ICON-1 + RZB combination therapy at month 6. Reduction of CST was observed at month 3 and was maintained from months 3 to 6 in all treatment arms (See FIG. 14). Reduction in CST mirrored and supported the outcomes for BCVA over time as signals of biological activity (See FIG. 15 and FIG. 16).
[00266] At baseline, all patients across the 3 treatment arms had fluid/exudative presence on the sdOCT, with the presence of subretinal fluid (SRF), intraretinal fluid 0RF) and/or subretinal pigment epithelium fluid (Sub-RPE). At month 6, the absence of any fluids was observed in a higher proportion of patients in the ICON-1 + RZB combination therapy arm (30.0%) compared to both the ICON-1 monotherapy (3.4%) and RZB monotherapy (11.1%) arms. In the ICON-1 + RZB combination therapy arm, a higher proportion of patients had no fluid present at month 6 (30%) (See FIG. 17 and FIG. 18).
[00267] For the patients in the ICON-1 + RZB combination therapy arm, there were longer treatment-free intervals from months 3 through 5, and more patients received no retreatment (See FIG. 19). The average number of retreatments per patient was 1.0 in the ICON- 1 + RZB combination therapy arm, 1.4 in the RZB monotherapy arm and 2.0 in the ICON-1 monotherapy arm. The mean time from treatment end to first retreatment was longer in patients in the ICON-1 + RZB combination therapy arm (62.8 days) than in the RZB monotherapy arm (51.7 days) and in the ICON-1 monotherapy arm (38.4 days) (See FIG. 20). More patients receiving ICON-1 + RZB combination therapy did not require retreatment (40%) compared with patients receiving RZB monotherapy (14.8%), with a decreased frequency of retreatment and longer treatment-free intervals (See FIG.21).
[00268] The signals of biological activity in both BCVA and CNV lesion changes were achieved, with the lower need for retreatment in patients receiving ICON-1 + RZB combination therapy compared to patients receiving RZB monotherapy, suggestive of a synergistic biological effect of CNV modification. In patients receiving ICON-1 monotherapy, the stable BCVA over 6 months and the reduction in exudation and fluid are supporting and suggestive of a biological signal.
Example 6 - Synergism of ICON- 1 and anti-VEGF Antibodies in Patients With Choroidal Neovascularization. Ocular Neovascularization, and Tumor Neovascularization
[00269] In this study, the synergistic effect of treatment with hl-conl and anti-VEGF antibodies is evaluated in patients with CNV, ocular neovascularization, and tumor neovascularization.
[00270] The study duration lasts for a period of twelve months, which includes a screening visit to determine in patient meet the study parameters, a baseline randomization visit at month 0, followed by monthly visits from months 1 through 12.
[00271] The baseline randomization visit identifies the CNV lesion area and exudation, and measures BCVA.
[00272] The study comprises three different groups: a control group that receives anti- VEGF alone, a group that receives doses of anti-VEGF and ICON-1 (0.3 mg), and a group that receives doses of anti-VEGF and ICON-1 (0.6 mg).
[00273] Patients in each group receive treatments at months 1, 2, and 3, while months 4- 12 are follow up visits in which treatment is administered only if needed, i.e. if CNV activity is observed, based on pre-defined criteria for treatment
[00274] Patients in groups that receive both anti-VEGF and ICON-1 are expected to exhibit an improved best-corrected visual acuity (BCVA) outcome at month 3 and month 12 from the baseline determination, as compared to those receiving only the anti-VEGF treatment. Patients in groups that receive both anti-VEGF and hl-conl are further expected to exhibit a reduction in mean CNV lesion area at month 3 and month 12 from the baseline determination, as compared to those receiving only the anti-VEGF treatment
[00275] Patients receiving both anti-VEGF and hl-conl are expected to exhibit at least a decrease in CNV activity such that there is no increase in the area of the lesion and there is no increase in the CNV-associated exudation. Patients receiving both anti-VEGF and hl-conl are expected to exhibit a reduction of in the area of the lesion and a reduction in CNV-associated exudation. Patients receiving both anti-VEGF and hl-conl are likely to exhibit regression of the lesion and CNV-associated exudation.
[00276] While the study lasts for a period of 12 months, the patients receiving both anti- VEGF and ICON-1 are expected to exhibit a decrease in CNV activity, a reduction of the area of the lesions and a reduction of CNV-associated exudation, and regression of the lesion and CNV- associated exudation earlier in the study than those receiving only anti-VEGF. Furthermore, the durability of treatment in patients receiving both anti-VEGF and hl-conl is expected to be greater than in patients receiving only anti-VEGF.
Example 7 - Dose-dependent Response of ICON-1 and Synergism of ICON-1 and VEGF inhibitor in Laser-induced CNV
[00277] Dose-dependent responses of ICON-1 were determined in ten to twelve week old pigs (Swine/Hampshire Cross) which underwent bilateral laser induction to create ~6 single laser spots in each eye. Each group consisted of 4 animals, for a total of 48 laser spots per group. For efficacy assessments, ICON-1 at 30(^g, 600ug, and 900μ§ was administered intravitreally on day 7 post-laser treatment Eylea (aflibercept), a VEGF inhibitor, was administered on day 0 at a dose of 2mg. Fluorescein Angiography (FA) was used to determine total lesion fluorescence on day 7 as a baseline and day 14 (See FIG. 22). ICON-1 administration was well tolerated with no dose related ocular toxicities and no systemic effects. A dose-dependent reduction in lesion fluorescence, measured by FA, was observed in ICON-1 treated animals at day 14. The largest decrease in lesion fluorescence was the 90C^g group compared to the vehicle-treated group (See FIG. 22). The FA measurements were expressed as Corrected total lesion fluorescence (CTFL); CTFL = integrated density - (area of selected cell x mean fluorescence of background readings).
[00278] The Synergism of ICON-1 and a VEGF inhibitor were determined in in ten to twelve week old pigs (Swine/Hampshire Cross) which underwent bilateral laser induction to crease ~6 single laser spots in each eye. Each group consisted of 4 animals, for a total of 48 laser spots per group. For efficacy assessments, ICON-1 at 600ug was administered intravitreally on day 7 post-laser treatment. Eylea (aflibercept), a VEGF inhibitor, was administered on day 0 at a dose of 2mg. Fluorescein Angiography (FA) was used to determine total lesion fluorescence on day 7 as a baseline and day 14 (See FIG. 23). ICON-1 administration was well tolerated with no dose related ocular toxicities and no systemic effects. Greater reduction in lesion fluorescence was observed in the ICON-1 + Eylea combination treated animals at day 14, compared to the vehicle treated group and to the ICON-1 along and Eylea (aflibercept) alone groups (See FIG. 23). The FA measurements were expressed as Corrected total lesion fluorescence (CTFL); CTFL = integrated density - (area of selected cell x mean fluorescence of background readings)
Example 8 - Pharmacological study evaluating the efficacy of ICON- 1.5 in a model of laser-induced choroidal neovascularization (CN V).
[00279] This study evaluates the efficacy of intra vitreal injections of one-armed FVII-Fc immunoconj ugates, administered as monotherapy or in combination with anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA) compared to anti-VEGF monotherapy in a rabbit model of laser induced choroidal neovascularization (CNV). Four animals per group, and six groups. The groups consist of (1) vehicle, (2) 300 ug, (3) 600 μg, (4) 900 ug, (5) aflibercept 2.0 mg, and (6) aflibercept 2.0 mg + ICON-1 (ICON-1.5 in parallel study) at 600 ug.
[00280] Rabbits are lasered in both eyes (OU) on Day 0 (DO). Test articles and vehicle are dosed bilaterally via intravitreal (IVT) injection on D7. Ranibizumab (LUCENTIS) or aflibercept (EYLEA) are dosed on day 0 immediately after laser (DO). In the combination group (group 7), anti-VEGF agents are injected on DO and one-armed FVII-Fc immunoconj ugates on D7.
[00281] Ocular Examination: Mydriasis for ocular examination is done using topical 1% tropicamide HCL (one drop in each eye IS minutes prior to examination). Complete ocular examination (modified Hackett and McDonald) using a slit lamp biomicroscope and indirect ophthalmoscope to evaluate ocular surface morphology, anterior segment and posterior segment inflammation, cataract formation, and retinal changes are conducted by a veterinary ophthalmologist at baseline and D14.
[00282] Fluorescein angiography (FA): FA is done in both eyes of all animals on D7, D10, and D14 after laser. Mydriasis for FA is done using topical 1% Tropicamide HCL (one drop in each eye 15 minutes prior to examination). Full FA is performed for 1-5 minutes after intravenous sodium fluorescein injection (12 mg kg-1). A reader analyzes the masked images obtained. The area of maximal fluorescein leakage is measured using Image J for each lesion. [00283] All eyes are collected for in situ hybridization (ISH) and flat-mount analysis of choroidal vascularity detected by fluorescein isothiocyanate-dextran staining.
Example 9 - Pharmacological study evaluating the efficacy of ICON-1.5 in a swine model of laser-induced choroidal neovascularization (CN V)
[00284] This study evaluates the efficacy of intravitreal injections of one-armed FVll-Fc immunoconjugates, administered as monotherapy or in combination with anti-VEGF agents such as ranibizumab (LUCENTIS) or aflibercept (EYLEA) compared to anti-VEGF monotherapy in a swine model of laser induced choroidal neovascularization (CNV). Four animals per group, and six groups. The groups consist of (1) vehicle, (2) 300 μg, (3) 600 ug, (4) 900 μg, (5) aflibercept 2.0 mg, and (6) aflibercept 2.0 mg + ICON-1 (ICON-1.5 in parallel study) at 600 μg.
[00285] Pigs are lasered in both eyes (OU) on Day 0 (DO). Test articles and vehicle are dosed bilaterally via intravitreal (IVT) injection on D7. Ranibizumab (LUCENTIS) or aflibercept (EYLEA) are dosed on day 0 immediately after laser (DO). In the combination group (group 7), anti-VEGF agents are injected on DO and one-armed FVH-Fc immunoconjugates on D7.
[00286] Ocular Examination: Mydriasis for ocular examination is done using topical 1% tropicamide HCL (one drop in each eye IS minutes prior to examination). Complete ocular examination (modified Hackett and McDonald) using a slit lamp biomicroscope and indirect ophthalmoscope to evaluate ocular surface morphology, anterior segment and posterior segment inflammation, cataract formation, and retinal changes are conducted by a veterinary ophthalmologist at baseline and D14.
[00287] Fluorescein angiography (FA): FA is done in both eyes of all animals on D7, D10, and D14 after laser. Mydriasis for FA is done using topical 1% Tropicamide HCL (one drop in each eye 15 minutes prior to examination). Full FA is performed for 1-5 minutes after intravenous sodium fluorescein injection (12 mg kg-1). A reader analyzes the masked images obtained. The area of maximal fluorescein leakage is measured using Image J for each lesion.
[00288] All eyes are collected for ISH and flatmount analysis.
* * * * * * * * *
[00289] While the described invention has been described with reference to the specific embodiments thereof it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adopt a particular situation, material, composition of matter, process, process step or steps, to the objective spirit and scope of the described invention. All such modifications are intended to be within the scope of the claims appended hereto.
[00290] Patents, patent applications, patent application publications, journal articles and protocols referenced herein are incorporated by reference in their entireties, for all purposes.

Claims

1. A method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions:
(a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVlIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and
(b) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
2. A method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions, a composition comprising:
(a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and
(b) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
3. A method for reversing tumor neovascularization in an eye of a patient in need thereof, comprising administering to the patient in multiple dosing sessions a composition comprising:
(a) an effective amount of an immunoconjugate dimer, wherein the monomer subunits of the dimer each comprises a mutated human factor Vila (fVIIa) protein conjugated to the human immunoglobulin Gl (IgGl) Fc domain; and (b) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
4. The method of any one of claims 1-3 or 65-67, wherein the VEGF inhibitor comprises an anti-VEGF antibody.
5. The method of any one of claims 1-3 or 65-67, wherein the immunoconj ugate dimer is a homodimer.
6. The method of any one of claims 1-3 or 65-67, wherein the immunoconj ugate dimer is a heterodimer.
7. The method of any one of claims 1-3 or 65-67, wherein the reduction in CNV exudation is a reduction in intraretinal fluid, subretinal fluid, or subretinal pigment epithelium fluid.
8. The method of any one of claims 1-3 or 65-67, wherein the reduction in CNV exudation is a reduction in intraretinal fluid, subretinal fluid, and subretinal pigment epithelium fluid.
9. The method of any one of claims 1-3 or 65-67, wherein at least one of the monomer subunits of the immunoconj ugate comprises a mutated human fVIIa domain comprising a single point mutation at Lys341 or Ser 344.
10. The method of claim 9, wherein the single point mutation is to an Ala residue.
11. The method of claim 10, wherein the single point mutation is Lys341 to Ala341.
12. The method of claim 10, wherein the single point mutation is Ser344 to Ala344.
13. The method of any one of claims 2, 3, 66, or 67, wherein the ocular neovascularization is associated with proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma.
14. The method of any one of claims 2, 3, 66, or 67, wherein the ocular neovascularization is secondary to proliferative diabetic retinopathy, wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP), or neovascular glaucoma.
15. The method of any one of claims 1-3 or 65-67, wherein the ocular neovascularization is choroidal neovascularization.
16. The method of claim 15, wherein the patient has been previously diagnosed with wet age- related macular degeneration (AMD) in the eye.
17. The method of claim 15, wherein the choroidal neovascularization is secondary to wet AMD.
18. The method of claim 16 or 17, wherein the eye of the patient has not been previously treated for choroidal neovascularization or wet AMD.
19. The method of claim 16 or 17, wherein the patient has previously been treated for choroidal vascularization with anti-vascular endothelial growth factor (VEGF) therapy, laser therapy, or surgery.
20. The method of any one of claims 1-3 or 65-67, wherein administering comprises intravitreal injection at each dosing session.
21. The method of any one of claims 1-3 or 65-67, wherein administering comprises suprachoroidal injection at each dosing session.
22. The method of any one of claims 1-3 or 65-67, wherein the multiple dosing sessions comprise two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, fourteen or more, fifteen or more, sixteen or more, seventeen or more, eighteen or more, nineteen or more, or twenty or more dosing sessions.
23. The method of any one of claims 1-22, wherein each dosing session is spaced apart by from about 20 days to about 50 days, or from about 20 days to about 40 days, or from about 20 days to about 30 days.
24. The method of claim 23, wherein the multiple dosing sessions comprise 12 to 24 dosing sessions.
25. The method of any one of claims 1-3 or 65-67, wherein administering comprises intravitreal injection into the eye of the patient once every 28 days, once every 30 days, or once every 35 days.
26. The method of any one of claims 1-3 or 65-67, wherein the immunoconjugate comprises the amino acid sequence of SEQ ID NO: 2 or 3.
27. The method of claim 26, wherein the immunoconjugate comprises the amino acid sequence of SEQ ID NO: 2.
28. The method of claim 26, wherein the immunoconjugate comprises the amino acid sequence of SEQ ID NO: 3.
29. The method of claim 26, wherein the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO:4.
30. The method of claim 26, wherein the immunoconjugate is encoded by a polynucleotide sequence comprising SEQ ID NO: 5.
31. The method of any one of claims 1-3 or 65-67, wherein administering comprises intravenous administration.
32. The method of claim 3, wherein administering comprises intratumoral injection.
33. The method of any one of claims 1-3 or 65-67, wherein the improved reduction in CNV lesion area or CNV exudation is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
34. The method of any one of claims 1-3 or 65-67, wherein the improved durability of treatment is increased by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
35. The method of any one of claims 1-3 or 65-67, wherein subsequent to the multiple dosing sessions, the retinal thickness of the eye of the patient is reduced in the eye of the patient, as compared to the retinal thickness of the eye of patients having been administered the VEGF inhibitor alone.
36. The method of claim 35, wherein the retinal thickness is reduced by at least about 50 μιη, at least about 100 μιη, at least about 150 μητ, at least about 175 μηι, at least about 200 μιη, at least about 225 μιη, or at least about 250 μιη.
37. The method of claim 35, wherein the retinal thickness is reduced by at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%.
38. The method of any one of claims 35-37, wherein the decreased retinal thickness is decreased central retinal subfield thickness (CST), decreased center point thickness (CPT), or decreased central foveal thickness (CFT).
39. The method of any one of claims 1-3 or 65-67, further comprising measuring the intraocular pressure (IOP) in the eye of the patient prior to each dosing session.
40. The method of any one of claims 1-3 or 65-67, further comprising measuring the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about 50 minutes, or about 1 hour after each dosing session.
41. The method of any one of claims 1-3 or 65-67, comprising measuring the IOP in the eye of the patient about 20 minutes, about 30 minutes, about 40 minutes, about 50 minutes, or about 1 hour prior to each dosing session.
42. The method of any one of claims 39-41, wherein the IOP is measured via tonometry.
43. The method of any one of claims 1-3 or 65-67, wherein the VEGF inhibitor is present in the same composition as the immunoconj ugate.
44. The method of any one of claims 1-3 or 65-67, wherein the VEGF inhibitor is present in a different composition than the immunoconj ugate.
45. The method of any one of claims 1-3 or 65-67, wherein the VEGF inhibitor is aflibercept.
46. The method of claim 4, wherein the anti-VEGF antibody is ranibizumab.
47. The method of claim 45, wherein the dosage of ranibizumab is from about 0.2 mg to about 1 mg.
48. The method of claim 46 or 47, wherein the dosage of ranibizumab is 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, or 0.7 mg.
49. The method of any one of claims 1-48, wherein ranibizumab is administered to the eye of the patient via an intravitreal injection.
50. The method of claim 45, wherein the VEGF inhibitor is administered at a dosage from about 0.2 mg to about 0.7 mg.
51. The method of claim 49, wherein the VEGF inhibitor is administered at a dosage of about 0.6 mg.
52. The method of claim 49, wherein the VEGF inhibitor is administered at a dosage of 0.6 mg.
53. The method of claim 49, wherein the VEGF inhibitor is administered at a dosage of about 0.3 mg.
54. The method of claim 49, wherein the VEGF inhibitor is administered at a dosage of 0.3 mg.
55. The method of any one of claims 1-3 or 65-67, wherein the multiple dosing sessions comprise administration once per month.
56. The method of claim 55, wherein the multiple dosing sessions comprise administration once per month for the first three months, followed by monthly treatments in months 4-12 only when CNV activity is observed.
57. The method of claim 55, wherein the multiple dosing sessions comprise administration once per month for the first three months, followed by monthly treatments in months 4-6 only when CNV activity is observed.
58. The method of any one of claims 1-48, wherein the composition comprising the effective amount of the VEGF inhibitor is administered to the eye of the patient via an intravitreal injection.
59. The method of claim 49, wherein the composition comprising the effective amount of the VEGF inhibitor is administered at each of the multiple dosing sessions.
60. The method of any one of claims 1-3 or 65-67, wherein each dosing session comprises the administration of between about 200 μg and about 600 ug of the immunoconjugate dimer.
61. The method of claim 60, wherein the administration is about 300 pg of the immunoconjugate dimer.
62. The method of claim 60, wherein the administration is about 600 ug of the immunoconjugate dimer.
63. The method of any one of claims 1-3 or 65-67, wherein the outcome is measured at least 6 months after beginning treatment.
64. The method of any one of claims 1-63, wherein the patient is a human.
65. A method for preventing, inhibiting, or reversing wet age-related macular degeneration (AMD) in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions:
(a) an effective amount of an immunoconjugate comprising two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers; and
(b) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
66. A method for preventing, inhibiting, or reversing ocular neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions:
(a) an effective amount of an immunoconjugate comprising two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers; and
(b) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
67. A method for preventing, inhibiting, or reversing tumor neovascularization in an eye of a patient in need thereof, comprising, administering to the patient in multiple dosing sessions: (c) an effective amount of an immunoconjugate comprising two dimerized immunoglobulin (Ig) Fc monomers and a mutated factor VII protein, wherein the mutated factor VII protein is fused to only one of the Fc monomers; and
(d) a VEGF inhibitor;
wherein the administration results in an improved best-corrected visual acuity (BCVA) outcome, improved reduction in mean choroidal neovascularization (CNV) lesion area, improved reduction in CNV exudation, or improved durability of treatment compared to patients having been administered the VEGF inhibitor alone.
68. The method of any one of claims 65-67, wherein the mutated factor VII protein exhibits a decreased coagulation response in a mammalian host, as compared to a wild-type factor VII protein.
PCT/US2018/015257 2017-01-25 2018-01-25 Methods for treating disorders associated with angiogenesis and neovascularization WO2018140611A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/480,804 US20190388522A1 (en) 2017-01-25 2018-01-25 Methods for treating disorders associated with angiogenesis and neovascularization
EP18745015.0A EP3573641A4 (en) 2017-01-25 2018-01-25 Methods for treating disorders associated with angiogenesis and neovascularization

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762450452P 2017-01-25 2017-01-25
US62/450,452 2017-01-25

Publications (1)

Publication Number Publication Date
WO2018140611A1 true WO2018140611A1 (en) 2018-08-02

Family

ID=62979692

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/015257 WO2018140611A1 (en) 2017-01-25 2018-01-25 Methods for treating disorders associated with angiogenesis and neovascularization

Country Status (4)

Country Link
US (1) US20190388522A1 (en)
EP (1) EP3573641A4 (en)
TW (1) TW201836646A (en)
WO (1) WO2018140611A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
EP3397276A4 (en) 2015-12-30 2019-12-18 Kodiak Sciences Inc. Antibodies and conjugates thereof
CN112203679A (en) 2018-03-02 2021-01-08 科达制药股份有限公司 IL-6 antibodies and fusion constructs and conjugates thereof
CA3157509A1 (en) 2019-10-10 2021-04-15 Kodiak Sciences Inc. Methods of treating an eye disorder

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080193441A1 (en) * 2003-11-18 2008-08-14 Iconic Therapeutics, Inc. Homogeneous Preparations of Chimeric Protein
US20120082686A1 (en) * 1999-07-01 2012-04-05 Yale University Neovascular-Targeted Immunoconjugates
US20130216513A1 (en) * 2010-07-09 2013-08-22 Biogen Idec Hemophilia Inc. Chimeric Clotting Factors
US20150139947A1 (en) * 2003-05-06 2015-05-21 Biogen Idec Hemophilia Inc. Immunoglobulin Chimeric Monomer-Dimer Hybrids
US20160101152A1 (en) * 2011-01-13 2016-04-14 Regeneron Pharmaceuticals, Inc. Use of a vegf antagonist to treat angiogenic eye disorders
US20160168240A1 (en) * 2013-07-11 2016-06-16 Sergey AKSENOV Use of vegf antagonist in treating chorioretinal neovascular and permeability disorders in paediatric patients
WO2017015582A1 (en) * 2015-07-22 2017-01-26 Iconic Therapeutics, Inc. Methods for treating disorders associated with angiogenesis and neovascularization

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180355030A1 (en) * 2015-11-13 2018-12-13 Iconic Therapeutics, Inc. Methods and compositions for treating disorders associated with pathological neovascularization
JP2019515904A (en) * 2016-04-14 2019-06-13 アイコニック セラピューティクス,インコーポレイテッド Compositions and methods for treating disorders associated with neovascularization

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120082686A1 (en) * 1999-07-01 2012-04-05 Yale University Neovascular-Targeted Immunoconjugates
US20150139947A1 (en) * 2003-05-06 2015-05-21 Biogen Idec Hemophilia Inc. Immunoglobulin Chimeric Monomer-Dimer Hybrids
US20080193441A1 (en) * 2003-11-18 2008-08-14 Iconic Therapeutics, Inc. Homogeneous Preparations of Chimeric Protein
US20130216513A1 (en) * 2010-07-09 2013-08-22 Biogen Idec Hemophilia Inc. Chimeric Clotting Factors
US20160101152A1 (en) * 2011-01-13 2016-04-14 Regeneron Pharmaceuticals, Inc. Use of a vegf antagonist to treat angiogenic eye disorders
US20160168240A1 (en) * 2013-07-11 2016-06-16 Sergey AKSENOV Use of vegf antagonist in treating chorioretinal neovascular and permeability disorders in paediatric patients
WO2017015582A1 (en) * 2015-07-22 2017-01-26 Iconic Therapeutics, Inc. Methods for treating disorders associated with angiogenesis and neovascularization

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3573641A4 *

Also Published As

Publication number Publication date
US20190388522A1 (en) 2019-12-26
TW201836646A (en) 2018-10-16
EP3573641A4 (en) 2020-11-11
EP3573641A1 (en) 2019-12-04

Similar Documents

Publication Publication Date Title
US20210236649A1 (en) Methods for treating disorders associated with angiogenesis and neovascularization
US10208124B2 (en) Anti-CD160 specific antibodies for the treatment of eye disorders based on neoangiogenesis
US20180194835A1 (en) Use of vegf antagonist in treating chorioretinal neovascular and permeability disorders in paediatric patients
US20180355030A1 (en) Methods and compositions for treating disorders associated with pathological neovascularization
US20190388522A1 (en) Methods for treating disorders associated with angiogenesis and neovascularization
JP2017536414A (en) Methods and formulations for treating vascular ocular diseases
CN101500605A (en) Therapeutic methods for treating vascular eye disorders with Dll4 antagonists
CN109715189A (en) Target the Humanized monoclonal antibodies of VE-PTP (HPTP- β)
US20160130321A1 (en) Use of a vegf antagonist in treating macular edema
US20190153119A1 (en) Compositions and methods for treating disorders associated with neovascularization
Wells et al. A phase 1, open-label, dose-escalation trial to investigate safety and tolerability of single intravitreous injections of ICON-1 targeting tissue factor in wet AMD
JP2023509880A (en) Use of fusion proteins in the treatment of age-related macular degeneration
CN108779173A (en) Inhibit eye rear portion fibrosis by antagonism placenta growth factor
CN113645994A (en) Methods of treating diseases using Pigment Epithelium Derived Factor (PEDF)
TW202116802A (en) Methods for treating ocular diseases
US20220056120A1 (en) Methods of treating ocular pathologies using bifunctional molecules that target growth factors

Legal Events

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

Ref document number: 18745015

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018745015

Country of ref document: EP

Effective date: 20190826