US20160144025A1 - Methods and formulations for treating vascular eye diseases - Google Patents

Methods and formulations for treating vascular eye diseases Download PDF

Info

Publication number
US20160144025A1
US20160144025A1 US14/943,490 US201514943490A US2016144025A1 US 20160144025 A1 US20160144025 A1 US 20160144025A1 US 201514943490 A US201514943490 A US 201514943490A US 2016144025 A1 US2016144025 A1 US 2016144025A1
Authority
US
United States
Prior art keywords
ang
antibody
pharmaceutical composition
inhibitor
vegf antagonist
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/943,490
Other languages
English (en)
Inventor
Robert L. VITTI
Kristine A. ERICKSON
Karen W. Chu
Stanley J. Wiegand
Jingtai Cao
Ivan B. Lobov
Saurabh Wadhwa
Kenneth S. Graham
Daniel Dix
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals 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 Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US14/943,490 priority Critical patent/US20160144025A1/en
Priority to MA041028A priority patent/MA41028A/fr
Priority to KR1020177017473A priority patent/KR20170087950A/ko
Priority to JP2017545861A priority patent/JP2017536414A/ja
Priority to MA40306A priority patent/MA40306A1/fr
Priority to AU2015353838A priority patent/AU2015353838A1/en
Priority to MX2017006129A priority patent/MX2017006129A/es
Priority to BR112017009807A priority patent/BR112017009807A2/pt
Priority to SG11201703609UA priority patent/SG11201703609UA/en
Priority to CA2968522A priority patent/CA2968522A1/en
Priority to PCT/US2015/061543 priority patent/WO2016085750A1/en
Priority to EP15862329.8A priority patent/EP3224278A4/en
Priority to CN201580063632.5A priority patent/CN107001457A/zh
Priority to EA201791168A priority patent/EA201791168A1/ru
Priority to PE2017000901A priority patent/PE20170900A1/es
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GRAHAM, KENNETH S., LOBOV, IVAN B., WIEGAND, STANLEY J., ERICKSON, KRISTINE A., CAO, JINGTAI, CHU, KAREN W., DIX, DANIEL B., VITTI, ROBERT L., WADHWA, SAURABH
Publication of US20160144025A1 publication Critical patent/US20160144025A1/en
Priority to US15/370,896 priority patent/US20170080086A1/en
Priority to PH12017500834A priority patent/PH12017500834A1/en
Priority to IL252159A priority patent/IL252159B/en
Priority to CONC2017/0004715A priority patent/CO2017004715A2/es
Priority to CL2017001188A priority patent/CL2017001188A1/es
Priority to US16/235,221 priority patent/US11071780B2/en
Priority to JP2020201473A priority patent/JP2021046431A/ja
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the present invention relates to methods of treating or ameliorating at least one symptom or indication of a vascular eye disease comprising administering a pharmaceutical formulation comprising an angiopoietin-2 (Ang-2) inhibitor and a vascular endothelial growth factor (VEGF) antagonist to a subject in need thereof.
  • Ang-2 angiopoietin-2
  • VEGF vascular endothelial growth factor
  • Vascular eye diseases are the leading cause of vision loss in today's aging population. These diseases are characterized by abnormal ‘leaky’ blood vessels growing into the retina. Two of the largest contributors to this patient population are diabetic retinopathy and exudative age-related macular degeneration.
  • Diabetic retinopathy is a major cause of visual impairment in the United States (Klein et al 1984, Ophthalmology 91:1464-1474; Moss et al 1998, Ophthalmology 105:998-1003). Diabetic retinopathy results from microvascular decompensation beginning with basement membrane thickening (Ruggiero et al 1997, Diabetes Metab. 23:30-42), and eventually leading to vascular occlusion and neovascularization (Porta et al 2002, Diabetologia. 45:1617-1634). It is estimated that about 28% of patients 40 years and older with diabetes have DR, and 4.4% have vision threatening DR (Zhang et al 2010, JAMA. 304: 649-656).
  • Diabetic macular edema is a manifestation of DR and is the most frequent cause of blindness in young and mid-aged adults (Klein et al 1984, Ophthalmology 91:1464-1474; Moss et al 1998, Ophthalmology 105:998-1003).
  • Age-related macular degeneration is the leading cause of severe visual loss in people aged 50 years or older in the developed world.
  • major advances have been made in the treatment of AMD, with the introduction of anti-angiogenic agents, offering hope of significant visual recovery for patients with neovascular AMD (Keane et al 2012, Surv Ophthalmol. 57: 389-414).
  • VEGF therapy e.g., aflibercept
  • aflibercept Anti-vascular endothelial growth factor (VEGF) therapy
  • VEGF vascular endothelial growth factor
  • Intravitreal (IVT) deliveries of anti-VEGF therapies such as ranibizumab and aflibercept have demonstrated efficacy and safety for chorioretinal diseases.
  • IVT Intravitreal
  • Binding of Ang-2 to its receptor Tie-2 promotes angiogenesis, both during normal vascular development and in conditions characterized by pathological neovascularization. Genetic deletion of Ang-2 in the mouse markedly inhibits both normal retinal vascular development and pathological neovascularization (Hackett et al 2000, J. Cell Physiol. 184: 275-83;hackett et al 2002, J. Cell Physiol. 192: 182-7; Gale et al 2002, Dev. Cell 3: 411-423). Targeting angiopoietin-2 (Ang2) will support inhibition of any of these factors, either alone or in combination, and has the potential to improve upon the success of anti-VEGF therapy alone.
  • Ang2 angiopoietin-2
  • methods for treating, preventing or ameliorating at least one symptom or indication of a vascular eye disease or disorder in a subject.
  • the methods according to this aspect of the invention comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an angiopoietin-2 (Ang-2) inhibitor to a subject in need thereof.
  • Ang-2 inhibitor is administered in combination with a vascular endothelial growth factor (VEGF) antagonist.
  • VEGF vascular endothelial growth factor
  • methods for inhibiting retinal angiogenesis in a subject.
  • the methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to the subject in need thereof.
  • combined administration results in reduction of the retinal vascular area by at least 65% as compared to the administration of either Ang-2 inhibitor or the VEGF antagonist alone.
  • the retinal angiogenesis is associated with a vascular eye disease or disorder.
  • the present invention provides for methods for inhibiting retinal neovascularization in a subject with an eye disease or disorder associated with angiogenesis, the methods comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to the subject in need thereof.
  • methods for inhibiting vascular leak in a subject with an eye disease or disorder.
  • the methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to a subject in need thereof.
  • the present invention provides methods for suppressing vascular leak in a subject with an eye disease or disorder associated with angiogenesis, wherein the methods comprise administering a single dose of a VEGF antagonist followed by one or more doses of a pharmaceutical composition comprising an Ang-2 inhibitor to the subject in need thereof.
  • the vascular leak is inhibited for at least 3 weeks, more than 3 weeks, more than 4 weeks, more than 8 weeks, or more than 10 weeks as compared to a subject who has been administered the VEGF antagonist alone.
  • the present invention provides for methods for inhibiting choroidal neovascularization comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor to a subject in need thereof.
  • methods for reducing the dependence and treatment burden of frequent intravitreal injections in a subject with a vascular eye disease or disorder.
  • the methods comprise sequentially administering an initial dose followed by one or more secondary doses of a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to the subject in need thereof; wherein the administration of the pharmaceutical composition is reduced to once every 9 weeks as compared to a subject who has been administered the VEGF antagonist alone.
  • the present invention provides method for treating a vascular eye disease, the methods comprising administering one or more doses of a pharmaceutical composition comprising a therapeutically active amount of an anti-Ang-2 inhibitor and a therapeutically active amount of a VEGF antagonist to a subject in need thereof.
  • the methods comprise administering an initial dose of the pharmaceutical composition followed by one or more secondary doses.
  • each secondary dose is administered 1 to 4 weeks after the immediately preceding dose.
  • the methods further comprise administration of one or more tertiary doses to the subject.
  • each tertiary dose is administered 5 to 12 weeks after the immediately preceding dose.
  • each secondary dose is administered 4 weeks after the immediately preceding dose.
  • each tertiary dose is administered 8 weeks or 12 weeks after the immediately preceding dose.
  • the pharmaceutical composition comprises about 10 mg/mL to about 120 mg/mL of the anti-Ang-2 inhibitor.
  • the pharmaceutical composition comprises about 40 mg/mL of the VEGF antagonist.
  • the pharmaceutical composition comprises about 10 mg/mL to about 120 mg/mL of the anti-Ang-2 inhibitor and about 40 mg/mL of the VEGF antagonist.
  • the pharmaceutical composition is intravitreally administered to the subject.
  • each dose of the pharmaceutical composition comprises about 0.5 mg to about 6 mg of the anti-Ang-2 inhibitor and about 2 mg of the VEGF antagonist.
  • each dose of the pharmaceutical composition comprises about 3 mg of the anti-Ang-2 inhibitor and about 2 mg of the VEGF antagonist. In one embodiment, each dose of the pharmaceutical composition comprises about 6 mg of the anti-Ang-2 inhibitor and about 2 mg of the VEGF antagonist.
  • the eye disease or disorder is selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • the Ang-2 inhibitor is administered as a co-formulation with a VEGF antagonist.
  • the Ang-2 inhibitor alone or in combination with the VEGF antagonist is intravitreally administered to a subject in need thereof.
  • the Ang-2 inhibitor is administered intravenously or subcutaneously.
  • the Ang-2 inhibitor is administered intravenously or subcutaneously in combination with the VEGF antagonist, wherein the VEGF antagonist is administered intravitreally.
  • an Ang-2 inhibitor and a VEGF antagonist are co-administered topically or intraocularly (e.g., intravitreally).
  • the Ang-2 inhibitor is administered at a dose of from 0.05 mg to 10 mg to a subject in need thereof. In certain embodiments, the VEGF antagonist is administered at a dose of from 0.01 mg to 5 mg to a subject in need thereof. In some embodiments, the Ang-2 inhibitor is administered at a dose of about 1-50 mg/kg of the subject's body weight.
  • one or more secondary doses of the pharmaceutical composition comprising the Ang-2 inhibitor are administered to a subject in need thereof.
  • the one or more doses comprise at least 2 secondary doses of the pharmaceutical composition.
  • each secondary dose is administered 1 to 4 weeks after the immediately preceding dose.
  • Exemplary Ang-2 inhibitors that can be used in the context of the methods of the present invention include, e.g., small molecule chemical inhibitors of Ang-2, or biological agents that target Ang-2.
  • the Ang-2 inhibitor is an antibody or antigen binding protein that binds the Ang-2 ligand and blocks Tie2 signaling.
  • the anti-Ang-2 antibody or antigen-binding protein comprises the heavy chain complementarity determining regions (HCDRs) of a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO: 1 and the light chain CDRs of a light chain variable region (LCVR) comprising the amino acid sequence of SEQ ID NO: 2.
  • VEGF Traps VEGF-inhibiting fusion proteins
  • An example of a VEGF antagonist that may be used in combination with the anti-Ang-2 antibodies in the methods of treatment of the present invention is aflibercept, a VEGF-inhibiting fusion protein (see U.S. Pat. No. 7,087,411).
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of an Ang-2 inhibitor and a pharmaceutically acceptable carrier or diluent.
  • the pharmaceutical composition further comprises a VEGF antagonist.
  • the present invention provides use of an anti-Ang-2 antibody or antigen-binding fragment thereof of the invention in the manufacture of a medicament to treat or prevent or ameliorate at least a symptom or indication of an eye disease or disorder in a subject, including humans.
  • the present invention provides use of an Ang-2 inhibitor of the invention in conjunction with a VEGF antagonist in the manufacture of a medicament to treat an eye disease or disorder in a subject, including humans.
  • a stable liquid pharmaceutical formulation comprising: (i) a VEGF antagonist; (ii) an antibody or antigen-binding fragment thereof that specifically binds to Ang-2; (iii) a buffer; (iv) a non-ionic detergent; (v) a tonicity agent; and (vi) a stabilizer.
  • a stable ophthalmic formulation comprising: (i) a VEGF antagonist; (ii) an antibody or antigen-binding fragment thereof that specifically binds to Ang-2; (iii) a buffer; (iv) a non-ionic detergent; (v) a tonicity agent; and (vi) a stabilizer.
  • the VEGF antagonist is provided at a concentration of from 5 mg/mL ⁇ 0.75 mg/mL to about 100 mg/mL ⁇ 15 mg/mL and the anti-Ang-2 antibody is provided at a concentration of from 10 ⁇ 1.5 mg/mL to 120 ⁇ 18.0 mg/mL. In some embodiments, the VEGF antagonist is provided at a concentration of from 10 mg/mL ⁇ 1.5 mg/mL to 80 mg/mL ⁇ 12 mg/mL, or from 20 mg/mL ⁇ 3.0 mg/mL to 60 mg/mL ⁇ 9.0 mg/mL. In one embodiment, the VEGF antagonist is provided at a concentration of 40 mg/mL ⁇ 6.0 mg/mL, or about 40 mg/mL.
  • the antibody is provided at a concentration of 10 mg/ml ⁇ 1.5 mg/mL, or about 10 mg/mL. In another embodiment, the antibody is provided at a concentration of 20 mg/mL ⁇ 3.0 mg/mL, or about 20 mg/mL. In another embodiment, the antibody is provided at a concentration of 60 mg/mL ⁇ 9.0 mg/mL, or about 60 mg/mL. In another embodiment, the antibody is provided at a concentration of 120 mg/mL ⁇ 18.0 mg/mL, or about 120 mg/mL.
  • the pH of the liquid formulation is from about pH 5.5 to about pH 6.5. In some embodiments, the pH of the liquid formulation is pH 6.2 ⁇ 0.3, pH 6.2 ⁇ 0.25, pH 6.2 ⁇ 0.2, pH 6.2 ⁇ 0.15, pH 6.2 ⁇ 0.1, pH 6.2 ⁇ 0.05, pH 6.2 ⁇ 0.01, or pH 6.2. In one embodiment, the pH of the liquid formulation is pH 6.2 ⁇ 0.3, or about pH 6.2.
  • the buffer is sodium phosphate.
  • the sodium phosphate is at a concentration of from 5 mM ⁇ 0.75 mM to 50 mM ⁇ 7.5 mM, 5 mM ⁇ 0.75 mM to 40 mM ⁇ 6.0 mM, or 5 mM ⁇ 0.75 mM to 25 mM ⁇ 3.75 mM.
  • the sodium phosphate is at a concentration of 10 mM ⁇ 1.5 mM or about 10 mM.
  • the non-ionic detergent is a nonionic polymer containing a polyoxyethylene moiety. In some embodiments, the non-ionic detergent is any one or more of polysorbate 20, poloxamer 188 and polyethylene glycol 3350. In one embodiment, the detergent is polysorbate 20. In one embodiment, the detergent is polysorbate 80.
  • the non-ionic detergent is polysorbate 20, which is at a concentration of from about 0.01% ⁇ 0.0045% to about 0.05% ⁇ 0.0045% w/v.
  • the non-ionic detergent is polysorbate 20, which is at a concentration of 0.03% ⁇ 0.0045% w/v or about 0.03% w/v.
  • the tonicity agent is sodium chloride or potassium chloride. In one embodiment, the tonicity agent is sodium chloride. In some embodiments, the tonicity agent is sodium chloride at a concentration of from 10 mM ⁇ 1.5 mM to 75 mM ⁇ 11.25 mM, 20 mM ⁇ 3.0 mM to 60 mM ⁇ 9.0 mM, or 30 mM ⁇ 4.5 mM to 50 mM ⁇ 7.5 mM. In one embodiment, the sodium chloride is at a concentration of 40 mM ⁇ 6.0 mM, or about 40 mM.
  • the stabilizer is a sugar.
  • the sugar is selected from the group consisting of sucrose, mannitol and trehalose.
  • the stabilizer is sucrose.
  • the stabilizer is at a concentration of from 1% ⁇ 0.15% w/v to 20% ⁇ 3% w/v. In some embodiments, the stabilizer is sucrose at a concentration of from 1% ⁇ 0.15% w/v to 15% ⁇ 2.25% w/v, or from 1% ⁇ 0.15% w/v to 10% ⁇ 1.5% w/v. In one embodiment, the stabilizer is sucrose at a concentration of 5% ⁇ 0.15% w/v or about 5% w/v. In another embodiment, the stabilizer is sucrose at a concentration of 7.5% ⁇ 1.125% w/v or about 7.5% w/v. In another embodiment, the stabilizer is sucrose at a concentration of 10% ⁇ 1.5% w/v or about 10% w/v.
  • the stabilizer is sucrose at a concentration of 12.5% ⁇ 1.875% w/v or about 12.5% w/v. In another embodiment, the stabilizer is sucrose at a concentration of 15% ⁇ 2.25% w/v or about 15% w/v. In another embodiment, the stabilizer is sucrose at a concentration of 20% ⁇ 3% w/v or about 20% w/v.
  • a stable liquid pharmaceutical formulation comprising: (i) from 5 ⁇ 0.75 mg/mL to 100 ⁇ 15.0 mg/mL of a VEGF antagonist; (ii) from 5 ⁇ 0.75 mg/ml to 150 ⁇ 22.5 mg/ml of a human antibody that specifically binds to human Ang-2; (iii) from 5 mM ⁇ 0.75 mM to 50 mM ⁇ 7.5 mM sodium phosphate; (iv) from 0.01% ⁇ 0.0015% to 0.1% ⁇ 0.015% (w/v) polysorbate 20; (v) from 10 mM ⁇ 1.5 mM to 100 mM ⁇ 15 mM sodium chloride; and (vi) from 1% ⁇ 0.15% to 20% ⁇ 3% (w/v) sucrose, at a pH of from about 5.5 to about 6.5.
  • the pharmaceutical formulation comprises (i) 40 mg/mL ⁇ 6.0 mg/mL of aflibercept; (ii) 10 ⁇ 1.5 mg/mL of anti-Ang-2 antibody; (iii) 10 ⁇ 1.5 mM sodium phosphate; (iv) 0.03% ⁇ 0.0045% (w/v) polysorbate 20; (v) 40 mM ⁇ 6.0 mM sodium chloride; and (vi) 5% ⁇ 0.75% (w/v) sucrose, at a pH of 6.2 ⁇ 0.3.
  • the pharmaceutical formulation comprises (i) 40 mg/mL ⁇ 6.0 mg/mL of aflibercept; (ii) 20 ⁇ 3.0 mg/mL of anti-Ang-2 antibody; (iii) 10 ⁇ 1.5 mM sodium phosphate; (iv) 0.03% ⁇ 0.0045% (w/v) polysorbate 20; (v) 40 mM ⁇ 6.0 mM sodium chloride; and (vi) 5% ⁇ 0.75% (w/v) sucrose, at a pH of 6.2 ⁇ 0.3.
  • the pharmaceutical formulation comprises (i) 40 mg/mL ⁇ 6.0 mg/mL of aflibercept; (ii) 60 ⁇ 9.0 mg/mL of anti-Ang-2 antibody; (iii) 10 ⁇ 1.5 mM sodium phosphate; (iv) 0.03% ⁇ 0.0045% (w/v) polysorbate 20; (v) 40 mM ⁇ 6.0 mM sodium chloride; and (vi) 5% ⁇ 0.75% (w/v) sucrose, at a pH of 6.2 ⁇ 0.3.
  • the pharmaceutical formulation comprises (i) 40 mg/mL ⁇ 6.0 mg/mL of aflibercept; (ii) 120 ⁇ 18.0 mg/mL of anti-Ang-2 antibody; (iii) 10 ⁇ 1.5 mM sodium phosphate; (iv) 0.03% ⁇ 0.0045% (w/v) polysorbate 20; (v) 40 mM ⁇ 6.0 mM sodium chloride; and (vi) 5% ⁇ 0.75% (w/v) sucrose, at a pH of 6.2 ⁇ 0.3.
  • a liquid pharmaceutical formulation of the present invention is provided in a container.
  • the container is a polycarbonate vial.
  • the container is a glass vial.
  • the glass vial is a type 1 borosilicate glass vial with a fluorocarbon-coated butyl rubber stopper.
  • the container is a microinfuser.
  • the container is a syringe.
  • the syringe comprises a fluorocarbon-coated plunger.
  • the syringe is a 2 mL long glass syringe containing less than about 500 parts per billion of tungsten equipped with a 30-G needle, a fluorocarbon-coated butyl rubber stopper, and a latex-free, non-cytotoxic rubber tip cap.
  • the syringe is a NUOVA OMPI 2 mL long glass syringe equipped with a 30-G thin wall needle, a FLUROTEC-coated 4432/50 GRY B2-40 stopper, and a FM 27 rubber tip cap.
  • the syringe is 1 mL, 2 mL or 3 mL plastic syringe fitted with a 27-G needle.
  • the container is a polyvinyl chloride IV bag. In another embodiment, the container is a polyolefin IV bag.
  • the present invention comprises a pre-filled syringe comprising a pharmaceutical formulation of any of the preceding aspects.
  • a kit comprising a pharmaceutical composition of any one of the preceding aspects, a container, and instructions is provided.
  • the container is a prefilled syringe.
  • the container is a borosilicate vial fitted with a FLUROTEC-coated 4432/50 rubber stopper.
  • FIG. 1 shows the percent change in leakage area in rabbits with DL-alpha-aminoadipic acid (DL-alpha-AAA)-induced retinal neovascularization which have been treated with anti-Ang-2 antibody, VEGF antagonist, or a combination of anti-Ang-2 antibody and VEGF antagonist, as described in Example 2 herein.
  • DL-alpha-AAA DL-alpha-aminoadipic acid
  • FIG. 2 shows the percent change in leakage area in rabbits with DL-alpha-AAA-induced retinal neovascularization which have been intravitreally (IVT) treated with VEGF antagonist (VGT) (or control) and intravenously (IV) treated with anti-Ang-2 antibody (or control) according to the following therapeutic regimen, as described in Example 3 herein: (a) control IVT and control IV; (b) VGT IVT and control IV; (c) control IVT and anti-Ang-2 IV; and (d) VGT IVT and anti-Ang-2 antibody IV.
  • FIG. 3 shows quantification of the total subretinal lesion size (A), neovessel volume (B), and vessel density (C) of baseline, control (hFc), and anti-Ang-2 antibody (mAb1) treated groups, as described in Example 5 herein.
  • the present invention includes methods for treating, preventing, or ameliorating at least one symptom or indication of a vascular eye disease or disorder in a subject.
  • the methods according to this aspect of the invention comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor to the subject in need thereof.
  • the Ang-2 inhibitor is administered subcutaneously or intravenously.
  • the Ang-2 inhibitor is administered in combination with a VEGF antagonist.
  • the Ang-2 inhibitor is intravitreally administered in combination with the VEGF antagonist.
  • the Ang-2 inhibitor is administered as a single combined dosage formulation with the VEGF antagonist.
  • the Ang-2 inhibitor is administered in combination with the VEGF antagonist, wherein the Ang-2 inhibitor is administered intravenously and the VEGF antagonist is administered intravitreally.
  • the VEGF antagonist may be administered before, after or concurrently with the Ang-2 inhibitor.
  • the terms “treat”, “treating”, or the like mean to alleviate symptoms, eliminate the causation of symptoms either on a temporary or permanent basis, or to prevent or slow the appearance of symptoms of a neovascular eye disease.
  • the present methods are useful for treating or ameliorating at least one symptom or indication including, but not limited to, retinal angiogenesis, neovascularization, vascular leak, retinal thickening within 500 ⁇ m of the center of the fovea, hard, yellow exudates within 500 ⁇ m of the center of the fovea with adjacent retinal thickening, and at least 1 disc area of retinal thickening, any part of which is within 1 disc diameter of the center of the fovea, blurry vision, floaters, loss of contrast, double vision, and eventual loss of vision.
  • the term means that, from the initiation of treatment, the patient exhibits gain of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) letters on the Early Treatment Diabetic Retinopathy Study (EDTRS) visual acuity chart.
  • ETRS Early Treatment Diabetic Retinopathy Study
  • the term means that, from initiation of treatment, vision loss of greater than or equal to 15 letters is prevented in the patient.
  • the terms “prevent”, “preventing”, or the like mean to prevent development of a symptom, indication or a complication of a vascular eye disease.
  • the term means, from initiation of treatment, moderate or severe vision loss is prevented in a patient.
  • vascular eye disease or disorder refers to eye disease or disorders that affect blood vessels in the eye.
  • the diseases may be caused due to abnormal angiogenesis (formation of new blood vessels) or occlusion or blockage of blood vessels.
  • the term includes eye diseases or disorders associated with angiogenesis.
  • the term includes, but is not limited to eye disease or disorder selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • the term “neovascular eye disease or disorder” may be used interchangeably with the term “eye disease or disorder associated with angiogenesis.”
  • the present invention includes methods for treating, preventing, or ameliorating at least one symptom or indication of an eye disease or disorder associated with angiogenesis in a subject, wherein the disease or disorder is selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • Diabetic Macular Edema refers to a serious eye condition that affects people with diabetes (type 1 or 2). Macular edema occurs when blood vessels in the retina leak into the macula and fluid and protein deposits collect on or under the macula of the eye (a yellow central area of the retina) and causes it to thicken and swell (edema). The swelling may distort a person's central vision, as the macula is near the center of the retina at the back of the eyeball.
  • the primary symptoms of DME include, but are not limited to, blurry vision, floaters, loss of contrast, double vision, and eventual loss of vision.
  • DME The pathology of DME is characterized by breakdown of the blood-retinal barrier, normally preventing water movement in the retina, thus allowing fluid to accumulate in the retinal tissue, and presence of retinal thickening.
  • DME is presently diagnosed during an eye examination consisting of a visual acuity test, which determines the smallest letters a person can read on a standardized chart, a dilated eye exam to check for signs of the disease, imaging tests such as optical coherence tomography (OCT) or fluorescein angiography (FA) and tonometry, an instrument that measures pressure inside the eye.
  • OCT optical coherence tomography
  • FA fluorescein angiography
  • tonometry an instrument that measures pressure inside the eye.
  • DME can be broadly characterized into two main categories—Focal and Diffuse.
  • Focal DME is characterized by specific areas of separate and distinct leakage in the macula with sufficient macular blood flow.
  • Diffuse DME results from leakage of the entire capillary bed surrounding the macula, resulting from a breakdown of the inner blood-retina barrier of the eye.
  • DME is also categorized based on clinical exam findings into clinically significant macular edema (CSME), non-CSME and CSME with central involvement (CSME-CI), which involves the fovea.
  • CSME clinically significant macular edema
  • CSME-CI central involvement
  • the present invention includes methods to treat the above-mentioned categories of DME.
  • Age-related macular degeneration refers to a serious eye condition when the small central portion of the retina, known as the macula, deteriorates.
  • the wet form of AMD is characterized by the growth of abnormal blood vessels from the choroid underneath the macula. This is called choroidal neovascularization. These blood vessels leak blood and fluid into the retina, causing distortion of vision that makes straight lines look wavy, as well as blind spots and loss of central vision. These abnormal blood vessels eventually scar, leading to permanent loss of central vision.
  • the symptoms of AMD include dark, blurry areas in the center of vision; and diminished or changed color perception. AMD can be detected in a routine eye exam.
  • One of the most common early signs of macular degeneration is the presence of drusen—tiny yellow deposits under the retina—or pigment clumping.
  • a subject in need thereof means a human or non-human mammal that exhibits one or more symptoms or indications of, and/or who has been diagnosed with an eye disease or disorder associated angiogenesis.
  • the term “a subject in need thereof” may also include, e.g., subjects who, prior to treatment, exhibit (or have exhibited) one or more indications of a neovascular eye disease such as, e.g., retinal angiogenesis, neovascularization, vascular leak, retinal thickening within 500 ⁇ m of the center of the fovea, hard, yellow exudates within 500 ⁇ m of the center of the fovea with adjacent retinal thickening, and at least 1 disc area of retinal thickening, any part of which is within 1 disc diameter of the center of the fovea, blurry vision, floaters, loss of contrast, double vision, and eventual loss of vision.
  • a neovascular eye disease such as, e.g., retinal angiogenesis, neovascularization
  • a “subject in need thereof” also includes human or non-human mammal who has a vascular eye disease or disorder selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • a vascular eye disease or disorder selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • a subject in need thereof may include a subset of population which is more susceptible to DME or AMD or may show an elevated level of a DME-associated or an AMD-associated biomarker.
  • a subject in need thereof may include a subject suffering from diabetes for more than 10 years, have frequent high blood sugar levels or high fasting blood glucose levels.
  • the term “a subject in need thereof” includes a subject who, prior to or at the time of administration of the Ang-2 inhibitor and/or VEGF antagonist, has or is diagnosed with diabetes.
  • the term “a subject in need thereof” includes a subject who, prior to or at the time of administration of the Ang-2 inhibitor and/or VEGF antagonist, is more than 50 years old.
  • the term “a subject in need thereof” includes subjects who are smokers, or subjects with high blood pressure or high cholesterol.
  • the present invention includes methods for treating, preventing or reducing the severity of a vascular eye disease comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to a subject in need thereof, wherein the pharmaceutical composition is administered to the subject in multiple doses, e.g., as part of a specific therapeutic dosing regimen.
  • the therapeutic dosing regimen may comprise administering multiple doses of the pharmaceutical composition to the subject at a frequency of about once a day, once every two days, once every three days, once every four days, once every five days, once every six days, once a week, once every two weeks, once every three weeks, once every four weeks, once a month, once every two months, once every three months, once every four months, or less frequently.
  • the therapeutic dosing regimen may comprise administering multiple doses of the pharmaceutical composition to the subject at a frequency of once a day or 2 times a day or more.
  • the methods of the present invention comprise administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist.
  • the Ang-2 inhibitor of the invention may be administered in combination with therapy including laser treatment to stop leakage into the macula.
  • the phrase ‘in combination with” means that the pharmaceutical composition comprising an Ang-2 inhibitor is administered to the subject at the same time as, just before, or just after administration of the VEGF antagonist.
  • the present invention also includes methods for inhibiting or reducing or suppressing vascular leak in a subject.
  • the methods according to this aspect of the invention comprise administering to the subject one or more doses of a pharmaceutical composition comprising an Ang-2 inhibitor to reduce or inhibit vascular leak in the eye of a subject.
  • the methods comprise administering to the subject one or more doses of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist to reduce or inhibit vascular leak in the eye of a subject.
  • the vascular leak is inhibited for more than 3 weeks, more than 4 weeks, more than 8 weeks, or more than 10 weeks than in a subject who has been administered the VEGF antagonist alone.
  • the methods of the present invention comprise administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist.
  • a pharmaceutical composition comprising an Ang-2 inhibitor in combination with a VEGF antagonist.
  • the phrase ‘in combination with” means that the pharmaceutical composition comprising an Ang-2 inhibitor is administered to the subject at the same time as, just before, or just after administration of the VEGF antagonist.
  • the VEGF antagonist is administered as a co-formulation with the Ang-2 inhibitor.
  • the present invention includes methods comprising administering a therapeutically effective amount of a pharmaceutical composition comprising an Ang-2 inhibitor to a subject to provide a greater therapeutic effect or synergistic effect as compared to administration of the VEGF antagonist alone.
  • the subject may be on a therapeutic regimen of intravitreally administered VEGF antagonist.
  • the Ang-2 inhibitor is added to this therapeutic regimen, wherein one or more intravitreal injections of the VEGF antagonist may be reduced or the duration between successive intravitreal injections may be increased.
  • the present invention provides methods to treat a vascular eye disease, the methods comprising administering one or more doses of a pharmaceutical composition comprising therapeutically effective amount of an anti-Ang-2 inhibitor and therapeutically effective amount of a VEGF antagonist to a subject in need thereof.
  • the pharmaceutical composition is intravitreally administered to the subject.
  • the pharmaceutical composition comprises about 10 mg/mL to about 120 mg/mL of the anti-Ang-2 inhibitor and about 40 mg/mL of the VEGF antagonist.
  • the methods comprise administering an initial dose of the pharmaceutical composition, followed by one or more secondary doses, wherein each secondary dose is administered 1 to 4 weeks after the immediately preceding dose.
  • each tertiary dose of the pharmaceutical composition is administered, wherein each tertiary dose is administered 5 to 12 weeks after the immediately preceding dose.
  • each dose of the pharmaceutical composition comprises about 0.5 to about 6 mg of the anti-Ang-2 inhibitor and about 2 mg of the VEGF antagonist.
  • the present invention provides methods to reduce the number of intravitreal injections in a subject with a vascular eye disease, the methods comprising administering a pharmaceutical composition comprising an anti-Ang-2 inhibitor and a VEGF antagonist, wherein the intravitreal administration is reduced to once in 8 weeks as compared to a subject administered with the anti-Ang-2 inhibitor or the VEGF antagonist alone.
  • the methods of the present invention are useful for treating or preventing vascular eye disorders in patients that have been diagnosed with or are at risk of being afflicted with a vascular eye disorder.
  • the methods of the present invention demonstrate efficacy within 36 weeks of the initiation of the treatment regimen (with the initial dose administered at “week 0”), e.g., by the end of week 6, by the end of week 12, by the end of week 18, by the end of week 24, etc.
  • “efficacy” means that, from the initiation of treatment, the patient exhibits a loss of 10 or fewer letters on the Early Treatment Diabetic Retinopathy Study (ETDRS) visual acuity chart.
  • “efficacy” means a gain of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or more) letters on the ETDRS chart from the time of initiation of treatment.
  • Ang-2 or “ANG2” means a human angiopoietin-2, which is generally known as an autocrine antagonist of Tie2 activation.
  • Ang-2 is generally known in the art to “prime” the vascular endothelium to receive the effects of cytokines.
  • Ang-2 is strongly expressed in tumor vasculature, and is generally thought to act synergistically with other cytokines (i.e., vascular endothelial growth factor) to promote angiogenesis and tumor progression.
  • an “Ang-2 inhibitor” (also referred to herein as an “Ang-2 antagonist,” an “Ang-2 blocker,” etc.) is any agent which binds to or interacts with Ang-2, and inhibits or attenuates the normal biological signaling function/activity of Ang-2.
  • Non-limiting examples of categories of Ang-2 inhibitors include small molecule Ang-2 inhibitors, anti-Ang-2 aptamers, peptide-based Ang-2 inhibitors (e.g., “peptibody” molecules), “receptor-bodies” (e.g., engineered molecules comprising the receptor-binding domain of an Ang-2 component), and antibodies or antigen-binding fragments of antibodies that specifically bind human Ang-2.
  • the Ang-2 inhibitor is an antibody or antigen binding fragment thereof as disclosed in e.g., US Patent Application Publication No. US20110027286.
  • the anti-Ang-2 antibody or antigen binding fragment thereof comprises a heavy chain variable region (HCVR) having an amino acid sequence of SEQ ID NO: 1 and a light chain variable region (LCVR) having an amino acid sequence of SEQ ID NO: 2.
  • HCVR heavy chain variable region
  • LCVR light chain variable region
  • the methods of the present invention comprise administering to a subject in need thereof a therapeutic composition comprising an Ang-2 inhibitor.
  • the Ang-2 inhibitor is an anti-Ang-2 antibody or antigen-binding fragment thereof.
  • antibody includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, as well as multimers thereof (e.g., IgM).
  • each heavy chain comprises a heavy chain variable region (abbreviated herein as HCVR or V H ) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, C H 1, C H 2 and C H 3.
  • Each light chain comprises a light chain variable region (abbreviated herein as LCVR or V L ) and a light chain constant region.
  • the light chain constant region comprises one domain (C L 1).
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the FRs of the anti-Ang-2 antibody may be identical to the human germline sequences, or may be naturally or artificially modified.
  • An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs.
  • antibody also includes antigen-binding fragments of full antibody molecules.
  • antigen-binding portion of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • Antigen-binding fragments of an antibody may be derived, e.g., from full antibody molecules using any suitable standard techniques such as proteolytic digestion or recombinant genetic engineering techniques involving the manipulation and expression of DNA encoding antibody variable and optionally constant domains.
  • DNA is known and/or is readily available from, e.g., commercial sources, DNA libraries (including, e.g., phage-antibody libraries), or can be synthesized.
  • the DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc.
  • Non-limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab′)2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide.
  • CDR complementarity determining region
  • engineered molecules such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g. monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression “antigen-binding fragment,” as used herein.
  • SMIPs small modular immunopharmaceuticals
  • an antigen-binding fragment of an antibody will typically comprise at least one variable domain.
  • the variable domain may be of any size or amino acid composition and will generally comprise at least one CDR which is adjacent to or in frame with one or more framework sequences.
  • the V H and V L domains may be situated relative to one another in any suitable arrangement.
  • the variable region may be dimeric and contain V H -V H , V H -V L or V L -V L dimers.
  • the antigen-binding fragment of an antibody may contain a monomeric V H or V L domain.
  • an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain.
  • variable and constant domains that may be found within an antigen-binding fragment of an antibody of the present invention include: (i) V H -C H 1; (ii) V H -C H 2; (iii) V H -C H 3; (iv) V H -C H 1-C H 2; (v) V H -C H 1-C H 2-C H 3, (vi) V H -C H 2-C H 3; (vii) V H -C L ; (viii) V L -C H 1; (ix) V L -C H 2; (x) V L -C H 3; (xi) V L -C H 1-C H 2; (xii) V L -C H 1-C H 2-C H 3, (xiii) V L -C H 2-C H 3; and (xiv) V L -C H 1; (ii) V H -
  • variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region.
  • a hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule.
  • an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric V H or V L domain (e.g., by disulfide bond(s)).
  • antibody also includes multispecific (e.g., bispecific) antibodies.
  • a multispecific antibody or antigen-binding fragment of an antibody will typically comprise at least two different variable domains, wherein each variable domain is capable of specifically binding to a separate antigen or to a different epitope on the same antigen.
  • Any multispecific antibody format may be adapted for use in the context of an antibody or antigen-binding fragment of an antibody of the present invention using routine techniques available in the art.
  • the present invention includes methods comprising the use of bispecific antibodies wherein one arm of an immunoglobulin is specific for IL-4R ⁇ or a fragment thereof, and the other arm of the immunoglobulin is specific for a second therapeutic target or is conjugated to a therapeutic moiety.
  • Exemplary bispecific formats that can be used in the context of the present invention include, without limitation, e.g., scFv-based or diabody bispecific formats, IgG-scFv fusions, dual variable domain (DVD)-Ig, Quadroma, knobs-into-holes, common light chain (e.g., common light chain with knobs-into-holes, etc.), CrossMab, CrossFab, (SEED) body, leucine zipper, Duobody, IgG1/IgG2, dual acting Fab (DAF)-IgG, and Mab 2 bispecific formats (see, e.g., Klein et al.
  • Bispecific antibodies can also be constructed using peptide/nucleic acid conjugation, e.g., wherein unnatural amino acids with orthogonal chemical reactivity are used to generate site-specific antibody-oligonucleotide conjugates which then self-assemble into multimeric complexes with defined composition, valency and geometry. (See, e.g., Kazane et al., J. Am. Chem. Soc . [Epub: Dec. 4, 2012]).
  • the antibodies used in the methods of the present invention may be human antibodies.
  • the term “human antibody,” as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may nonetheless include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term “human antibody,” as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • the antibodies used in the methods of the present invention may be recombinant human antibodies.
  • the term “recombinant human antibody,” as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further below), antibodies isolated from a recombinant, combinatorial human antibody library (described further below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et al. (1992) Nucl. Acids Res.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the V H and V L regions of the recombinant antibodies are sequences that, while derived from and related to human germline V H and V L sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • the antibodies used in the pharmaceutical formulations and methods of the present invention specifically bind Ang-2.
  • the term “specifically binds,” or the like, means that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Methods for determining whether an antibody specifically binds to an antigen are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like.
  • an antibody that “specifically binds” Ang-2 includes antibodies that bind Ang-2or portion thereof with a K D of less than about 500 nM, less than about 300 nM, less than about 200 nM, less than about 100 nM, less than about 90 nM, less than about 80 nM, less than about 70 nM, less than about 60 nM, less than about 50 nM, less than about 40 nM, less than about 30 nM, less than about 20 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, less than about 1 nM or less than about 0.5 nM, as measured in a surface plasmon resonance assay.
  • An isolated antibody that specifically binds human Ang-2 may, however, have cross-reactivity to other antigens, such as Ang-2 molecules from other (non-human) species.
  • the Ang-2 inhibitor is an anti-Ang-2 antibody, or antigen-binding fragment thereof comprising a heavy chain variable region (HCVR), light chain variable region (LCVR), and/or complementarity determining regions (CDRs) comprising any of the amino acid sequences of the anti-Ang-2 antibodies as set forth in US Patent Application Publication No. US20110027286.
  • HCVR heavy chain variable region
  • LCVR light chain variable region
  • CDRs complementarity determining regions
  • the anti-Ang-2 antibody or antigen-binding fragment thereof comprises three HCDRs (HCDR1, HCDR2 and HCDR3) and three LCDRs (LCDR1, LCDR2 and LCDR3), wherein the HCDR1 comprises the amino acid sequence of SEQ ID NO: 3; the HCDR2 comprises the amino acid sequence of SEQ ID NO: 4; the HCDR3 comprises the amino acid sequence of SEQ ID NO: 5; the LCDR1 comprises the amino acid sequence of SEQ ID NO: 6; the LCDR2 comprises the amino acid sequence of SEQ ID NO: 7; and the LCDR3 comprises the amino acid sequence of SEQ ID NO: 8.
  • the anti-Ang-2 antibody or antigen-binding fragment thereof comprises an HCVR comprising SEQ ID NO: 1 and an LCVR comprising SEQ ID NO: 2.
  • the methods of the present invention comprise the use of an anti-Ang-2 antibody, wherein the antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9.
  • the anti-Ang-2 antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • An exemplary antibody comprising a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and a light chain comprising the amino acid sequence of SEQ ID NO: 10 is the fully human anti-Ang-2 antibody known as nesvacumab.
  • the methods of the present invention comprise the use of nesvacumab, or a bioequivalent thereof.
  • bioequivalent refers to anti-Ang-2 antibodies or Ang-2-binding proteins or fragments thereof that are pharmaceutical equivalents or pharmaceutical alternatives whose rate and/or extent of absorption do not show a significant difference with that of nesvacumab when administered at the same molar dose under similar experimental conditions, either single dose or multiple dose.
  • the term refers to antigen-binding proteins that bind to Ang-2 which do not have clinically meaningful differences with nesvacumab in their safety, purity and/or potency.
  • H1H685P The non-limiting, exemplary antibody used in the Examples herein is referred to as “H1H685P”, as in US 2011/0027286.
  • This antibody comprises an HCVR/LCVR amino acid sequence pair having SEQ ID NOs: 1/2, and HCDR1-HCDR2-HCDR3/LCDR1-LCDR2-LCDR3 domains represented by SEQ ID NOs: 3-4-5/SEQ ID NOs: 6-7-8.
  • the amount of antibody, or antigen-binding fragment thereof, contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used.
  • the pharmaceutical formulations are liquid formulations that may contain 5 ⁇ 0.75 mg/mL to 150 ⁇ 22.5 mg/mL of antibody; 7.5 ⁇ 1.125 mg/mL to 140 ⁇ 21 mg/mL of antibody.
  • the formulations of the present invention may comprise about 10 mg/mL; about 20 mg/mL; about 30 mg/mL; about 50 mg/mL; about 60 mg/mL; about 80 mg/mL; about 100 mg/mL; about 120 mg/mL; or about 150 mg/mL of an antibody or an antigen-binding fragment thereof, that binds specifically to human Ang-2.
  • VEGF antagonist is any agent that binds to or interacts with VEGF, inhibits the binding of VEGF to its receptors (VEGFR1 and VEGFR2), and/or inhibits the biological signaling and activity of VEGF.
  • VEGF antagonists include molecules which interfere with the interaction between VEGF and a natural VEGF receptor, e.g., molecules which bind to VEGF or a VEGF receptor and prevent or otherwise hinder the interaction between VEGF and a VEGF receptor.
  • VEGF antagonists include anti-VEGF antibodies (e.g., ranibizumab [LUCENTIS®]), anti-VEGF receptor antibodies (e.g., anti-VEGFR1 antibodies, anti-VEGFR2 antibodies, etc.), small molecule inhibitors of VEGF (e.g., sunitinib), and VEGF receptor-based chimeric molecules or VEGF-inhibiting fusion proteins (also referred to herein as “VEGF-Traps”).
  • anti-VEGF antibodies e.g., ranibizumab [LUCENTIS®]
  • anti-VEGF receptor antibodies e.g., anti-VEGFR1 antibodies, anti-VEGFR2 antibodies, etc.
  • small molecule inhibitors of VEGF e.g., sunitinib
  • VEGF receptor-based chimeric molecules or VEGF-inhibiting fusion proteins also referred to herein as “VEGF-Traps”.
  • VEGF receptor-based chimeric molecules include chimeric polypeptides which comprise two or more immunoglobulin (Ig)-like domains of a VEGF receptor such as VEGFR1 (also referred to as Flt1) and/or VEGFR2 (also referred to as Flk1 or KDR), and may also contain a multimerizing domain (e.g., an Fc domain which facilitates the multimerization [e.g., dimerization] of two or more chimeric polypeptides).
  • VEGFR1R2-Fc ⁇ C1(a) also known as aflibercept; marketed under the product name EYLEA®.
  • aflibercept is encoded by the amino acid sequence of SEQ ID NO: 11.
  • the amount of the VEGF antagonist contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used.
  • the pharmaceutical formulations are liquid formulations that may contain 5 ⁇ 0.75 mg/mL to 150 ⁇ 22.5 mg/mL of VEGF antagonist; 10 ⁇ 1.5 mg/mL to 100 ⁇ 15.0 mg/mL of VEGF antagonist; 20 ⁇ 3 mg/mL to 80 ⁇ 12 mg/mL of VEGF antagonist; 30 ⁇ 4.5 mg/mL to 70 ⁇ 10.5 mg/mL of VEGF antagonist or 40 ⁇ 6.0 mg/mL of the VEGF antagonist.
  • the formulations of the present invention may comprise about 20 mg/mL; about 30 mg/mL; about 40 mg/mL; about 50 mg/mL; or about 60 mg/mL of a VEGF antagonist.
  • the methods of the present invention comprise administering to the subject a VEGF antagonist in combination with an anti-Ang-2 antibody.
  • a VEGF antagonist in combination with an anti-Ang-2 antibody.
  • the expression “in combination with” means that the VEGF antagonist is administered before, after, or concurrent with the pharmaceutical composition comprising the anti-Ang-2 antibody.
  • the term “in combination with” also includes sequential or concomitant administration of anti-Ang-2 antibody and a VEGF antagonist.
  • the VEGF antagonist when administered “before” the pharmaceutical composition comprising the anti-Ang-2 antibody, may be administered more than 72 hours, about 72 hours, about 60 hours, about 48 hours, about 36 hours, about 24 hours, about 12 hours, about 10 hours, about 8 hours, about 6 hours, about 4 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes or about 10 minutes prior to the administration of the pharmaceutical composition comprising the anti-Ang-2 antibody.
  • the VEGF antagonist When administered “after” the pharmaceutical composition comprising the anti-Ang-2 antibody, the VEGF antagonist may be administered about 10 minutes, about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, about 72 hours, or more than 72 hours after the administration of the pharmaceutical composition comprising the anti-Ang-2 antibody.
  • Administration “concurrent” with the pharmaceutical composition comprising the anti-Ang-2 antibody means that the VEGF antagonist is administered to the subject in a separate dosage form within less than 5 minutes (before, after, or at the same time) of administration of the pharmaceutical composition comprising the anti-Ang-2 antibody, or administered to the subject as a single combined dosage formulation comprising both the VEGF antagonist and the anti-Ang-2 antibody.
  • Combination therapies may include an anti-Ang-2 antibody of the invention and a VEGF antagonist (e.g., aflibercept, a VEGF-Trap, see, e.g., U.S. Pat. No. 7,087,411 (also referred to herein as a “VEGF-inhibiting fusion protein”), anti-VEGF antibody (e.g., ranibizumab), a small molecule kinase inhibitor of VEGF receptor (e.g., sunitinib, sorafenib or pazopanib), etc.
  • a VEGF antagonist e.g., aflibercept, a VEGF-Trap, see, e.g., U.S. Pat. No. 7,087,411 (also referred to herein as a “VEGF-inhibiting fusion protein”
  • anti-VEGF antibody e.g., ranibizumab
  • the methods of the invention comprise administering an anti-Ang-2 antibody in combination with a VEGF antagonist for additive or synergistic activity to treat or ameliorate at least one symptom or indication of an eye disease or disorder selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • a VEGF antagonist for additive or synergistic activity to treat or ameliorate at least one symptom or indication of an eye disease or disorder selected from the group consisting of diabetic retinopathy, diabetic macular edema, age-related macular degeneration, retinal neovascularization, central retinal vein occlusion, branched retinal vein occlusion, polypoidal choroidal vasculopathy, and choroidal neovascularization.
  • the present invention includes methods which comprise administering an Ang-2 inhibitor to a subject wherein the Ang-2 inhibitor is contained within a pharmaceutical composition.
  • the pharmaceutical composition further comprises a VEGF antagonist.
  • the Ang-2 inhibitor and the VEGF antagonist may be in own separate pharmaceutical dosage formulation.
  • the pharmaceutical compositions of the invention may be formulated with suitable carriers, excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • the expression “pharmaceutical formulation” means a combination of at least one active ingredient (e.g., a small molecule, macromolecule, compound, etc. which is capable of exerting a biological effect in a human or non-human animal), and at least one inactive ingredient which, when combined with the active ingredient or one or more additional inactive ingredients, is suitable for therapeutic administration to a human or non-human animal.
  • active ingredient e.g., a small molecule, macromolecule, compound, etc. which is capable of exerting a biological effect in a human or non-human animal
  • inactive ingredient which, when combined with the active ingredient or one or more additional inactive ingredients, is suitable for therapeutic administration to a human or non-human animal.
  • formulation means “pharmaceutical formulation” unless specifically indicated otherwise.
  • the present invention provides pharmaceutical formulations comprising at least one therapeutic polypeptide.
  • the therapeutic polypeptide is an antibody, or an antigen-binding fragment thereof, which binds specifically to human angiopoietin-2 (Ang-2) protein.
  • the present invention provides pharmaceutical formulations comprising more than one therapeutic polypeptide. More specifically, the present invention includes pharmaceutical formulations that comprise: (i) a human antibody that specifically binds to human Ang-2; (ii) a VEGF antagonist; (iii) a sodium phosphate buffer; (iv) an organic co-solvent that is a non-ionic surfactant; (v) a tonicity agent such as sodium chloride; and (vi) a thermal stabilizer that is a carbohydrate. Specific exemplary components and formulations included within the present invention are described in detail below.
  • the amount of antibody, or antigen-binding fragment thereof, contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used.
  • the pharmaceutical formulations are liquid formulations that may contain 5 ⁇ 0.75 mg/mL to 150 ⁇ 22.5 mg/mL of antibody; 7.5 ⁇ 1.125 mg/mL to 140 ⁇ 21 mg/mL of antibody; 10 ⁇ 1.5 mg/mL to 130 ⁇ 19.5 mg/mL of antibody; 10 ⁇ 1.5 mg/mL of antibody; 20 ⁇ 3 mg/mL of antibody; 60 ⁇ 9 mg/mL of antibody; or 120 ⁇ 18 mg/mL of antibody.
  • the formulations of the present invention may comprise about 10 mg/mL; about 20 mg/mL; about 40 mg/mL; about 60 mg/mL; about 80 mg/mL; about 100 mg/mL; about 120 mg/mL; or about 140 mg/mL of an antibody or an antigen-binding fragment thereof that binds specifically to human Ang-2.
  • the pharmaceutical formulations are liquid formulations that may contain 5 ⁇ 0.75 mg/mL to 100 ⁇ 15 mg/mL of a VEGF antagonist.
  • the formulations of the present invention may comprise about 5 mg/mL; about 10 mg/mL; about 15 mg/mL; about 20 mg/mL; about 25 mg/mL; about 30 mg/mL; about 35 mg/mL; about 40 mg/mL; about 50 mg/mL; about 60 mg/mL; about 70 mg/mL; about 80 mg/mL; about 90 mg/mL; or about 100 mg/mL of a VEGF antagonist such as aflibercept.
  • the pharmaceutical formulations are stable liquid co-formulations comprising about 5 mg/mL to about 150 mg/mL of the anti-Ang-2 antibody and about 5 to 100 mg/mL of the VEGF antagonist.
  • the pharmaceutical formulations of the present invention comprise one or more excipients.
  • excipient means any non-therapeutic agent added to the formulation to provide a desired consistency, viscosity or stabilizing effect.
  • the pharmaceutical formulation of the invention comprises at least one organic cosolvent in a type and in an amount that stabilizes the human Ang-2 antibody under conditions of rough handling or agitation, such as, e.g., vortexing.
  • rough handling is vortexing a solution containing the antibody and the organic cosolvent for about 60 minutes or about 120 minutes.
  • the organic cosolvent is a non-ionic surfactant, such as an alkyl poly(ethylene oxide).
  • non-ionic surfactants that can be included in the formulations of the present invention include, e.g., polysorbates such as polysorbate 20, polysorbate 28, polysorbate 40, polysorbate 60, polysorbate 65, polysorbate 80, polysorbate 81, and polysorbate 85; poloxamers such as poloxamer 181, poloxamer 188, poloxamer 407; or polyethylene glycol (PEG).
  • Polysorbate 20 is also known as TWEEN 20, sorbitan monolaurate and polyoxyethylenesorbitan monolaurate. Poloxamer 188 is also known as PLURONIC F68.
  • the amount of non-ionic surfactant contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used. In certain embodiments, the formulations may contain 0.01% ⁇ 0.0015% to 1% ⁇ 0.15% surfactant.
  • the formulations of the present invention may comprise about 0.0085%; about 0.01%; about 0.02%; about 0.03%; about 0.04%; about 0.05%; about 0.06%; about 0.07%; about 0.08%; about 0.09%; about 0.1%; about 0.11%; about 0.12%; about 0.13%; about 0.14%; about 0.15%; about 0.16%; about 0.17%; about 0.18%; about 0.19%; about 0.20%; about 0.21%; about 0.22%; about 0.23%; about 0.24%; about 0.25%; about 0.3%; about 0.4%; about 0.5%; about 0.6%; about 0.7%; about 0.8%; about 0.9%; about 1%; about 1.1%; about 1.15%; or about 1.2% polysorbate 20, polysorbate 80 or poloxamer 188.
  • the pharmaceutical formulations of the present invention may also comprise one or more stabilizers in a type and in an amount that stabilizes the human Ang-2 antibody under conditions of thermal stress.
  • stabilizes is maintaining greater than about 93% of the antibody in a native conformation when the solution containing the antibody and the thermal stabilizer is kept at about 45° C. for up to about 28 days.
  • stabilizes is wherein less than about 4% of the antibody is aggregated when the solution containing the antibody and the thermal stabilizer is kept at about 45° C. for up to about 28 days.
  • what is meant by “stabilizes” is maintaining greater than about 96% of the antibody in a native conformation when the solution containing the antibody and the thermal stabilizer is kept at about 37° C. for up to about 28 days. In some embodiments, what is meant by “stabilizes” is wherein less than about 2% of the antibody is aggregated when the solution containing the antibody and the thermal stabilizer is kept at about 37° C. for up to about 28 days.
  • “native” means the major form of the antibody by size exclusion, which is generally an intact monomer of the antibody.
  • the thermal stabilizer is a sugar or sugar alcohol selected from sucrose, sorbitol, glycerol, trehalose and mannitol, or any combination thereof, the amount of which contained within the formulation can vary depending on the specific circumstances and intended purposes for which the formulation is used.
  • the formulations may contain about 1% to about 20% sugar or sugar alcohol; about 2% to about 18% sugar or sugar alcohol; about 3% to about 15% sugar or sugar alcohol; about 4% to about 10% sugar or sugar alcohol; or about 5% sugar or sugar alcohol.
  • the pharmaceutical formulations of the present invention may comprise 4% ⁇ 0.6%; 5% ⁇ 0.75%; 6% ⁇ 0.9%; 7% ⁇ 1.05%; 8% ⁇ 1.2%; 9% ⁇ 1.35%; 10% ⁇ 1.5%; 11% ⁇ 1.65%; 12% ⁇ 1.8%; 13% ⁇ 1.95%; or about 14% ⁇ 2.1% sugar or sugar alcohol (e.g., sucrose, trehalose or mannitol).
  • sugar or sugar alcohol e.g., sucrose, trehalose or mannitol
  • the pharmaceutical formulations of the present invention comprise a tonicity agent such as sodium chloride or potassium chloride.
  • the tonicity agent is sodium chloride.
  • the sodium chloride is present at a concentration of 5 mM ⁇ 0.75 mM to 100 mM ⁇ 15.0 mM; 10 mM ⁇ 1.5 mM to 50 mM ⁇ 7.5 mM; 40 mM ⁇ 6.0 mM; or about 40 mM.
  • the pharmaceutical formulations of the present invention may also comprise a buffer or buffer system, which serves to maintain a stable pH and to help stabilize the human Ang-2 antibody.
  • a buffer or buffer system which serves to maintain a stable pH and to help stabilize the human Ang-2 antibody.
  • stabilizes is wherein less than 5% ⁇ 0.5% or no more than about 4.3% of the antibody is aggregated when the solution containing the antibody and the buffer is kept at about 45° C. for up to about 28 days.
  • stabilizes is wherein at least 92% ⁇ 0.5% of the antibody is in its native conformation as determined by size exclusion chromatography when the solution containing the antibody and the buffer is kept at about 45° C. for up to about 28 days.
  • the non-aggregated and non-degraded antibody elutes at a fraction that equates to the native antibody, and is generally the main elution fraction. Aggregated antibody elutes at a fraction that indicates a size greater than the native antibody. Degraded antibody elutes at a fraction that indicates a size less than the native antibody.
  • main charge or “main charge form”, what is meant is the fraction of antibody that elutes from an ion exchange resin in the main peak, which is generally flanked by more “basic” peaks on one side and more “acidic” peaks on the other side.
  • the pharmaceutical formulations of the present invention may have a pH of from about 5.5 to about 6.5.
  • the formulations of the present invention may have a pH of about 5.5; about 5.6; about 5.7; about 5.8; about 5.9; about 6.0; about 6.1; about 6.2; about 6.3; about 6.4; or about 6.5.
  • the pH is 6.2 ⁇ 0.3; 6.2 ⁇ 0.2; 6.2 ⁇ 0.1; about 6.2; or 6.2.
  • the buffer or buffer system comprises at least one buffer that has a buffering range that overlaps fully or in part the range of pH 5.5-7.4. In one embodiment, the buffer has a pKa of about 6.2 ⁇ 0.5. In certain embodiments, the buffer comprises a sodium phosphate buffer.
  • the sodium phosphate is present at a concentration of 5 mM ⁇ 0.75 mM to 15 mM ⁇ 2.25 mM; 6 mM ⁇ 0.9 mM to 14 mM ⁇ 2.1 mM; 7 mM ⁇ 1.05 mM to 13 mM ⁇ 1.95 mM; 8 mM ⁇ 1.2 mM to 12 mM ⁇ 1.8 mM; 9 mM ⁇ 1.35 mM to 11 mM ⁇ 1.65 mM; 10 mM ⁇ 1.5 mM; or about 10 mM.
  • the buffer system comprises sodium phosphate at 10 mM ⁇ 1.5 mM, at a pH of 6.2 ⁇ 0.3 or 6.1 ⁇ 0.3.
  • Exemplary formulations comprising a VEGF antagonist that can be used in the context of the present invention are disclosed, e.g., in U.S. Pat. Nos. 7,531,173 and 7,608,261.
  • Exemplary pharmaceutical compositions comprising an anti-Ang-2 antibody that can be used in the context of the present invention are disclosed, e.g., in US Patent Application Publication No. 20130186797.
  • the pharmaceutical formulations of the present invention typically exhibit high levels of stability.
  • stable as used herein in reference to the pharmaceutical formulations, means that the antibodies within the pharmaceutical formulations retain an acceptable degree of chemical structure or biological function after storage under defined conditions. A formulation may be stable even though the antibody contained therein does not maintain 100% of its chemical structure or biological function after storage for a defined amount of time. Under certain circumstances, maintenance of about 90%, about 95%, about 96%, about 97%, about 98% or about 99% of an antibody's structure or function after storage for a defined amount of time may be regarded as “stable”.
  • Stability can be measured, inter alia, by determining the percentage of native antibody that remains in the formulation after storage for a defined amount of time at a defined temperature.
  • the percentage of native antibody can be determined by, inter alia, size exclusion chromatography (e.g., size exclusion high or ultra performance liquid chromatography [SE-HPLC or SE-UPLC]), such that native means non-aggregated and non-degraded.
  • SE-HPLC or SE-UPLC] size exclusion high or ultra performance liquid chromatography
  • An “acceptable degree of stability”, as that phrase is used herein, means that at least 90% of the native form of the antibody can be detected in the formulation after storage for a defined amount of time at a given temperature.
  • At least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the native form of the antibody can be detected in the formulation after storage for a defined amount of time at a defined temperature.
  • the defined amount of time after which stability is measured can be at least 14 days, at least 28 days, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, or more.
  • the defined temperature at which the pharmaceutical formulation may be stored when assessing stability can be any temperature from about ⁇ 80° C.
  • a pharmaceutical formulation may be deemed stable if after nine months of storage at 5° C., greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC or SE-UPLC.
  • a pharmaceutical formulation may also be deemed stable if after six months of storage at 25° C., greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC or SE-UPLC.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 37° C., greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC or SE-UPLC.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 45° C., greater than about 93%, 94%, 95%, 96%, 97%, 98% or 99% of native antibody is detected by SE-HPLC or SE-UPLC.
  • a pharmaceutical formulation may also be deemed stable if after six months of storage at ⁇ 20° C., greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC.
  • a pharmaceutical formulation may also be deemed stable if after six months of storage at ⁇ 30° C., greater than about greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC or SE-UPLC.
  • a pharmaceutical formulation may also be deemed stable if after six months of storage at ⁇ 80° C., greater than about 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99% or 99.5% of native antibody is detected by SE-HPLC or SE-UPLC.
  • Stability can be measured, inter alia, by determining the percentage of antibody that forms in an aggregate within the formulation after storage for a defined amount of time at a defined temperature, wherein stability is inversely proportional to the percent aggregate that is formed.
  • the percentage of aggregated antibody can be determined by, inter alia, size exclusion chromatography (e.g., size exclusion high performance liquid chromatography [SE-HPLC]).
  • SE-HPLC size exclusion high performance liquid chromatography
  • An “acceptable degree of stability”, as that phrase is used herein, means that at most 6% of the antibody is in an aggregated form detected in the formulation after storage for a defined amount of time at a given temperature.
  • an acceptable degree of stability means that at most about 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the antibody can be detected in an aggregate in the formulation after storage for a defined amount of time at a given temperature.
  • the defined amount of time after which stability is measured can be at least 2 weeks, at least 28 days, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, or more.
  • the temperature at which the pharmaceutical formulation may be stored when assessing stability can be any temperature from about ⁇ 80° C.
  • a pharmaceutical formulation may be deemed stable if after nine months of storage at 5° C., less than about 2%, 1.75%, 1.5%, 1.25%, 1%, 0.75%, 0.5%, 0.25%, or 0.1% of the antibody is detected in an aggregated form.
  • a pharmaceutical formulation may also be deemed stable if after six months of storage at 25° C., less than about 2%, 1.75%, 1.5%, 1.25%, 1%, 0.75%, 0.5%, 0.25%, or 0.1% of the antibody is detected in an aggregated form.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 45° C., less than about 4%, 3.5%, 3%, 2.5%, 2%, 1.5%, 1%, 0.5%, or 0.1% of the antibody is detected in an aggregated form.
  • a pharmaceutical formulation may also be deemed stable if after three months of storage at ⁇ 20° C., ⁇ 30° C., or ⁇ 80° C. less than about 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1%, 0.5%, or 0.1% of the antibody is detected in an aggregated form.
  • Stability can be measured, inter alia, by determining the percentage of antibody that migrates in a more acidic fraction during ion exchange (“acidic form”) than in the main fraction of antibody (“main charge form”), wherein stability is inversely proportional to the fraction of antibody in the acidic form. While not wishing to be bound by theory, deamidation of the antibody may cause the antibody to become more negatively charged and thus more acidic relative to the non-deamidated antibody (see, e.g., Robinson, N., Protein Deamidation, PNAS, Apr. 16, 2002, 99(8):5283-5288). The percentage of “acidified” antibody can be determined by ion exchange chromatography (e.g., cation exchange high performance liquid chromatography [CEX-HPLC]).
  • ion exchange chromatography e.g., cation exchange high performance liquid chromatography [CEX-HPLC]
  • an “acceptable degree of stability”, as that phrase is used herein, means that at most 52% of the antibody is in a more acidic form detected in the formulation after storage for a defined amount of time at a defined temperature.
  • an acceptable degree of stability means that at most about 52%, 50%, 45%, 40%, 35%, 30%, 29%, 28%, 27%, 26%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 4%, 0.5%, or 0.1% of the antibody can be detected in an acidic form in the formulation after storage for a defined amount of time at a given temperature.
  • the defined amount of time after which stability is measured can be at least 2 weeks, at least 28 days, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, at least 18 months, at least 24 months, or more.
  • the temperature at which the pharmaceutical formulation may be stored when assessing stability can be any temperature from about ⁇ 80° C.
  • a pharmaceutical formulation may be deemed stable if after three months of storage at ⁇ 80° C., ⁇ 30° C., or ⁇ 20° C.
  • a pharmaceutical formulation may also be deemed stable if after nine months of storage at 5° C., less than about 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1% of the antibody is in a more acidic form.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 25° C., less than about 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1% of the antibody is in a more acidic form.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 37° C., less than about 37%, 36%, 35%, 34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.50% or 0.1% of the antibody is in a more acidic form.
  • a pharmaceutical formulation may also be deemed stable if after 28 days of storage at 45° C., less than about 52%, 51%, 50%, 49%, 48%, 47%, 46%, 45%, 44%, 43%, 42%, 41%, 40%, 39%, 38%, 37%, 36%, 35%, 34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1% of the antibody can be detected in a more acidic form.
  • Measuring the binding affinity of the antibody to its target may also be used to assess stability.
  • a formulation of the present invention may be regarded as stable if, after storage at e.g., ⁇ 80° C., ⁇ 30° C., ⁇ 20° C., 5° C., 25° C., 37° C., 45° C., etc. for a defined amount of time (e.g., 14 days to 6 months), the anti-Ang-2 antibody contained within the formulation binds to Ang-2 with an affinity that is at least 84%, 90%, 95%, or more of the binding affinity of the antibody prior to said storage. Binding affinity may be determined by any method, such as e.g., ELISA or plasmon resonance.
  • Biological activity may be determined by an Ang-2 activity assay, such as by contacting a cell that expresses Ang-2 with the formulation comprising the anti Ang-2 antibody.
  • the binding of the antibody to such a cell may be measured directly, such as via FACS analysis.
  • the downstream activity of the Ang-2 system may be measured in the presence of the antibody, and compared to the activity of the Ang-2 system in the absence of antibody.
  • the Ang-2 may be endogenous to the cell.
  • the Ang-2 may be ectopically expressed (i.e., heterologous expression) in the cell.
  • compositions of the invention e.g., encapsulation in liposomes, micro-particles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al., 1987, J. Biol. Chem. 262: 4429-4432).
  • Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
  • infusion or bolus injection by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
  • epithelial or mucocutaneous linings e.g., oral mucosa, rectal and intestinal mucosa, etc.
  • the pharmaceutical formulations of the invention may be administered, e.g., by eye drops, subconjunctival injection, subconjunctival implant, intravitreal injection, intravitreal implant, sub-Tenon's injection or sub-Tenon's implant.
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used.
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), 1974, CRC Pres., Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, 1984, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138). Other controlled release systems are discussed in the review by Langer, 1990, Science 249:1527-1533.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by known methods. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the pharmaceutical formulations of the present invention may be contained within any container suitable for storage or administration of medicines and other therapeutic compositions.
  • the pharmaceutical formulations may be contained within a sealed and sterilized plastic or glass container having a defined volume such as a vial, ampule, syringe, cartridge, bottle, or IV bag.
  • a vial e.g., clear and opaque (e.g., amber) glass or plastic vials.
  • any type of syringe can be used to contain or administer the pharmaceutical formulations of the present invention.
  • the pharmaceutical formulations of the present invention may be contained within “normal tungsten” syringes or “low tungsten” syringes.
  • the process of making glass syringes generally involves the use of a hot tungsten rod which functions to pierce the glass thereby creating a hole from which liquids can be drawn and expelled from the syringe. This process results in the deposition of trace amounts of tungsten on the interior surface of the syringe. Subsequent washing and other processing steps can be used to reduce the amount of tungsten in the syringe.
  • normal tungsten means that the syringe contains greater than or equal to 500 parts per billion (ppb) of tungsten.
  • low tungsten means that the syringe contains less than 500 ppb of tungsten.
  • a low tungsten syringe, according to the present invention can contain less than about 490, 480, 470, 460, 450, 440, 430, 420, 410, 390, 350, 300, 250, 200, 150, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10 or fewer ppb of tungsten.
  • the rubber plungers used in syringes, and the rubber stoppers used to close the openings of vials may be coated to prevent contamination of the medicinal contents of the syringe or vial, or to preserve their stability.
  • pharmaceutical formulations of the present invention may be contained within a syringe that comprises a coated plunger, or within a vial that is sealed with a coated rubber stopper.
  • the plunger or stopper may be coated with a fluorocarbon film. Examples of coated stoppers or plungers suitable for use with vials and syringes containing the pharmaceutical formulations of the present invention are mentioned in, e.g., U.S. Pat. Nos.
  • the pharmaceutical formulations may be contained within a low tungsten syringe that comprises a fluorocarbon-coated plunger.
  • the pharmaceutical formulations can be administered to a patient by parenteral routes such as injection (e.g., subcutaneous, intravenous, intramuscular, intraperitoneal, etc.) or percutaneous, mucosal, nasal, pulmonary or oral administration.
  • parenteral routes such as injection (e.g., subcutaneous, intravenous, intramuscular, intraperitoneal, etc.) or percutaneous, mucosal, nasal, pulmonary or oral administration.
  • Numerous reusable pen or autoinjector delivery devices can be used to subcutaneously deliver the pharmaceutical formulations of the present invention.
  • Examples include, but are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Bergdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTM (sanofi-aventis, Frankfurt, Germany).
  • Examples of disposable pen or autoinjector delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but are not limited to the SOLOSTARTM pen (sanofi-aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly), the SURECLICKTM Autoinjector (Amgen, Thousand Oaks, Calif.), the PENLETTM (Haselmeier, Stuttgart, Germany), the EPIPEN (Dey, L.P.), and the HUMIRATM Pen (Abbott Labs, Abbott Park, Ill.).
  • microinfusor means a subcutaneous delivery device designed to slowly administer large volumes (e.g., up to about 2.5 mL or more) of a therapeutic formulation over a prolonged period of time (e.g., about 10, 15, 20, 25, 30 or more minutes). See, e.g., U.S. Pat. No. 6,629,949; U.S. Pat. No. 6,659,982; and Meehan et al., J. Controlled Release 46:107-116 (1996). Microinfusors are particularly useful for the delivery of large doses of therapeutic proteins contained within high concentration (e.g., about 100, 125, 150, 175, 200 or more mg/mL) or viscous solutions.
  • high concentration e.g., about 100, 125, 150, 175, 200 or more mg/mL
  • the pharmaceutical formulation is administered via an IV drip, such that the formulation is diluted in an IV bag containing a physiologically acceptable solution.
  • pharmaceutical composition is a compounded sterile preparation in an intravenous infusion bag, such that a single dose of drug product is diluted into 100 mL, 250 mL (or other like amount suitable for intravenous drip delivery) of a physiological buffer (e.g., 0.9% saline).
  • the infusion bag is made of a polyvinyl chloride (e.g., VIAFLEX, Baxter, Deerfield, Ill.).
  • the infusion bag is made of a polyolefin (EXCEL IV Bags, Braun Medical Inc., Bethlehem, Pa.).
  • the liquid formulation comprising of from 10 mg/mL to 120 mg/mL of anti-Ang-2 antibody is comprised in a prefilled syringe and is administered intravitreally in a volume of approximately upto 100 ⁇ L.
  • the liquid formulation comprising of from 10 mg/mL to 120 mg/mL of anti-Ang-2 antibody and from 10 mg/mL to 100 mg/mL of aflibercept is comprised in a prefilled syringe and is administered intravitreally in a volume of approximately upto 100 ⁇ L.
  • the liquid formulation comprising of from 10 mg/mL to 120 mg/mL of anti-Ang-2 antibody and from 10 mg/mL to 100 mg/mL of aflibercept is in a prefilled syringe and is administered intravitreally in a volume of approximately upto 500 ⁇ L. In certain embodiments, the liquid formulation comprising of from 60 mg/mL to 120 mg/mL of anti-Ang-2 antibody and about 40 mg/mL of aflibercept is in a prefilled syringe and is administered intravitreally in a volume of approximately upto 500 ⁇ L.
  • the syringe is a 2 mL long glass syringe fitted with a 30-gauge thin wall needle, a fluorocarbon coated rubber plunger and a rubber needle shield. In one embodiment, the syringe is a 1 mL long glass syringe fitted with a 30-gauge thin wall needle, a fluorocarbon coated rubber plunger and a rubber needle shield.
  • the present invention includes methods comprising administering to a subject a pharmaceutical composition comprising an anti-Ang-2 antibody at a dosing frequency of about four times a week, twice a week, once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, once every eight weeks, once every twelve weeks, or less frequently so long as a therapeutic response is achieved.
  • the methods involve the administration of a pharmaceutical composition comprising an anti-Ang-2 antibody in combination with a VEGF antagonist at a dosing frequency of about four times a week, twice a week, once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, once every eight weeks, once every nine weeks, once every twelve weeks, or less frequently so long as a therapeutic response is achieved.
  • multiple doses of an anti-Ang-2 antibody may be administered to a subject over a defined time course.
  • the methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of an anti-Ang-2 antibody.
  • sequentially administering means that each dose of anti-Ang-2 antibody is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months).
  • the present invention includes methods which comprise sequentially administering to the patient a single initial dose of an anti-Ang-2 antibody, followed by one or more secondary doses of the anti-Ang-2 antibody, and optionally followed by one or more tertiary doses of the anti-Ang-2 antibody.
  • multiple doses of a co-formulation comprising an anti-Ang-2 antibody and a VEGF antagonist may be administered to a subject over a defined time course.
  • the methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of a co-formulation comprising an anti-Ang-2 antibody and a VEGF antagonist.
  • sequentially administering means that each dose of the anti-Ang-2 antibody in combination with the VEGF antagonist is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months).
  • the present invention includes methods which comprise sequentially administering to the patient a single initial dose of a co-formulation comprising an anti-Ang-2 antibody and a VEGF antagonist, followed by one or more secondary doses of the co-formulated anti-Ang-2 antibody and VEGF antagonist, and optionally followed by one or more tertiary doses of the co-formulated anti-Ang-2 antibody and VEGF antagonist.
  • the terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration.
  • the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”);
  • the “secondary doses” are the doses which are administered after the initial dose;
  • the “tertiary doses” are the doses which are administered after the secondary doses.
  • the initial, secondary, and tertiary doses may all contain the same amount of anti-Ang-2 antibody (or a co-formulation comprising anti-Ang-2 antibody and VEGF antagonist), but generally may differ from one another in terms of frequency of administration.
  • the amount contained in the initial, secondary and/or tertiary doses varies from one another (e.g., adjusted up or down as appropriate) during the course of treatment.
  • one or more (e.g., 1, 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as “loading doses” followed by subsequent doses that are administered on a less frequent basis (e.g., “maintenance doses”).
  • an anti-Ang-2 antibody or a co-formulation comprising anti-Ang-2 antibody and VEGF antagonist
  • each secondary and/or tertiary dose is administered 1 to 14 (e.g., 1, 11 ⁇ 2, 2, 21 ⁇ 2, 3, 31 ⁇ 2, 4, 41 ⁇ 2, 5, 51 ⁇ 2, 6, 61 ⁇ 2, 7, 71 ⁇ 2, 8, 81 ⁇ 2, 9, 91 ⁇ 2, 10, 101 ⁇ 2, 11, 111 ⁇ 2, 12, 121 ⁇ 2, 13, 131 ⁇ 2, 14, 141 ⁇ 2, or more) weeks after the immediately preceding dose.
  • the phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose of anti-Ang-2 antibody (or a co-formulation comprising anti-Ang-2 antibody and VEGF antagonist) which is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.
  • the methods according to this aspect of the invention may comprise administering to a patient any number of secondary and/or tertiary doses of an anti-Ang-2 antibody (or a co-formulation comprising anti-Ang-2 antibody and VEGF antagonist).
  • an anti-Ang-2 antibody or a co-formulation comprising anti-Ang-2 antibody and VEGF antagonist.
  • only a single secondary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient.
  • only a single tertiary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient.
  • each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 2 weeks after the immediately preceding dose.
  • each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 4 weeks after the immediately preceding dose.
  • the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.
  • the present invention includes methods comprising sequential administration of an anti-Ang-2 antibody in combination with a VEGF antagonist, to a patient to treat DME or AMD.
  • the present methods comprise administering one or more doses of an anti-Ang-2 antibody followed by one or more doses of a VEGF antagonist.
  • the present methods comprise administering a single dose of a VEGF antagonist followed by one or more doses of an anti-Ang-2 antibody.
  • one or more doses of about 0.05 mg to about 2 mg of a VEGF antagonist may be administered followed by one or more doses of about 0.05 mg to about 10 mg of the Ang-2 inhibitor.
  • one or more doses of about 1 mg/kg to about 15 mg/kg of subject body weight of anti-Ang-2 antibody may be administered after which one or more doses of VEGF antagonist may be administered to treat, alleviate, reduce or ameliorate one or more conditions associated with DME or AMD (e.g., angiogenesis inhibition).
  • the anti-Ang-2 antibody is administered at one or more doses resulting in an improvement in one or more parameters (e.g., retinal thickening, visual acuity) followed by the administration of a VEGF antagonist (e.g., aflibercept) to prevent recurrence or have additive activity.
  • a VEGF antagonist e.g., aflibercept
  • Alternative embodiments of the invention pertain to concomitant administration of anti-Ang-2 antibody and a VEGF antagonist which is administered at a separate dosage at a similar or different frequency relative to the anti-Ang-2 antibody.
  • the VEGF antagonist is administered before, after or concurrently with the anti-Ang-2 antibody.
  • the VEGF antagonist is administered as a single dosage formulation with the anti-Ang-2 antibody.
  • the amount of Ang-2 inhibitor (e.g., anti-Ang-2 antibody) administered to a subject according to the methods of the present invention is, generally, a therapeutically effective amount.
  • therapeutically effective amount means an amount of Ang-2 inhibitor that results in one or more of: (a) a reduction in the severity of retinal vascular leak; (b) a reduction in the area of retinal or choroidal neo-vascularization; (c) change in central retinal thickness; (d) an increase in the duration of suppression of vascular leak in the eye; and (e) a reduction in the number of intravitreal injections in a subject having an eye disease or disorder associated with angiogenesis.
  • a therapeutically effective amount can be from about 0.05 mg to about 100 mg, e.g., about 0.05 mg, about 0.1 mg, about 1.0 mg, about 1.5 mg, about 2.0 mg, about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, or about 100 mg, of the anti-Ang-2 antibody.
  • 0.5 mg, 1.0 mg, 3.0 mg or 6.0 mg of an anti-Ang-2 antibody is administered.
  • the amount of Ang-2 inhibitor contained within the individual doses may be expressed in terms of milligrams of antibody per kilogram of patient body weight (i.e., mg/kg).
  • the Ang-2 inhibitor may be administered to a patient at a dose of about 0.0001 to about 100 mg/kg of patient body weight.
  • 0.5 mg, 1.0 mg, 3.0 mg or 6.0 mg of an anti-Ang-2 antibody is administered in combination with 0.05 mg to about 10 mg of a VEGF antagonist (e.g., aflibercept).
  • a VEGF antagonist e.g., aflibercept
  • VEGF is the key modulator of angiogenesis in normal and pathological angiogenesis.
  • other growth factors are also involved in angiogenesis and are able to mediate blood vessel resistance to anti-VEGF therapies.
  • Angiopoietin-2 (Ang2) was shown to be involved in blood vessel growth and regression in various circumstances in a context-dependent manner.
  • Ang2 retinal vascular development
  • Human anti-Ang-2 antibodies were generated as described in US Patent Application Publication No. US20110027286.
  • the exemplary anti-Ang-2 antibody used in the present and following Examples is the human anti-Ang-2 antibody designated as H1H685 with HCVR/LCVR of SEQ ID NOs: 1/2 (also referred to herein as “mAb1”).
  • anti-Ang-2 antibody alone or in combination with aflibercept was administered intravitreally.
  • P4 was selected as the starting point for treatment with mAb1 to assess the effect of Ang2 and/or VEGF inhibition on RVD.
  • Administration of mAb1 or aflibercept reduced the outgrowth of the superficial retinal vasculature compared to hFc treated controls.
  • mAb1 and aflibercept decreased the mean vascularized area of the retina by 17% and 36%, respectively, relative to hFc controls, while combined treatment with both mAb1 and aflibercept reduced vascular area by 72%, representing complete arrest of retinal vascular development over the treatment period (compared to retinal vascular area at P4).
  • mAb1 and aflibercept also decreased the mean vessel length by 23% and 22%, respectively, compared to hFc controls. Combined treatment with mAb1 and aflibercept had a significant synergistic effect, leading to a dramatic 77% decrease in total vessel length. Total blood vessel length was even smaller by 25% in the combination treated samples compared to P4 retinas.
  • the glial toxin DL-alpha-AAA targets retinal Muller cells and astrocytes and leads to neovascularization and chronic vascular leak lasting at least 12 months (Kato et al 1993; Neuroscience 57: 473).
  • the purpose of this study was to evaluate the effects of IVT injection of co-formulated Anti-Ang2 and aflibercept on RNV induced by DL-alpha-AAA.
  • Group III Co-formulated 125 ⁇ g aflibercept and 500 ⁇ g mAb1/50 ⁇ l, IVT
  • a baseline examination was performed prior to first treatment to balance treatment groups.
  • Follow-up examinations were performed on wk 1, 2, 3, 4, 5, 6, 7, and 8 Post-IVT treatments.
  • Leakage area was quantified using Adobe Photoshop after fluorescein angiography (FA) analysis.
  • FA fluorescein angiography
  • a fluorescent agent was administered intravenously to monitor vessel leak and ocular coherence tomography using light waves was used to monitor retinal structure.
  • the purpose of this study was to evaluate the effects of co-treatment of Anti-Ang2 antibody and aflibercept on RNV induced by DL-alpha-AAA, wherein the intravitreal administration of aflibercept was followed by systemic administrations of mAb1.
  • the rationale of this study was to try and maintain the suppression of leakage over longer periods of time with an initial IVT injection of aflibercept and follow up with systemic injections once in two weeks (q2w) of anti-Ang 2 antibody.
  • retinal neovascularization was induced in male New Zealand rabbits with a single intravitreal injection of DL-alpha-AAA. Stable retinal neovascularization and vascular leak was established 10 weeks post induction. After ten weeks of disease establishment, the subjects were split into four groups with balanced leakage severity and treatment was administered as shown below:
  • Group 1 Control 42 ⁇ g/50 ⁇ l, IVT and Control 5 mg/kg, IV, q2w
  • Group 2 Control 42 ⁇ g/50 ⁇ l, IVT and mAb1 (anti-Ang2 ab) 15 mg/kg, IV, q2w
  • Group 3 Aflibercept 125 ⁇ g/50 ⁇ l, IVT and Control 5 mg/kg, IV, q2w
  • Group 4 Aflibercept 125 ⁇ g/50 ⁇ l, IVT and mAb1 (a-Ang2) 15 mg/kg, IV, q2w
  • FA Fluorescein Angiography
  • FA Baseline fluorescein angiography
  • OCT optic coherent tomography
  • This study is a dose-ranging study on the effect of IVT aflibercept monotherapy on DL-alpha-AAA induced retinal neovascularization (RNV) and vascular leak in rabbit eye.
  • RNV retinal neovascularization
  • DL-alpha-AAA was used to induce RNV in rabbit eyes.
  • the subjects were treated with 4 doses of aflibercept: 50 mcg, 125 mcg, 250 mcg, and 500 mcg.
  • Retinal vascular leak was monitored and quantitated by FA (as disclosed elsewhere herein) at follow-up time points on weeks 1, 2, 3, 4, 5, 6, 8 10 and 12 post-IVT.
  • aflibercept blocked retinal NV leak within 1 week, and leak did not resume until week 12 after treatment.
  • aflibercept treatment leads to suppression of vascular leak and partial regression of neovascularization.
  • Vascular leakage reoccurs but length of suppression is dose dependent.
  • Ang2 is a ligand for the tyrosine kinase receptor Tie-2 and is broadly expressed in the vascular endothelium of developing blood vessels and in vessels undergoing active growth or remodeling in diverse physiological and pathophysiological conditions.
  • mAb1 is a human monoclonal, neutralizing antibody against Ang-2.
  • the study utilized a rat model of choroidal neovascularization (CNV) to assess the inhibitory effects of mAb1-mediated pharmacological inhibition of Ang-2 on CNV.
  • CNV choroidal neovascularization
  • CNV was induced in Sprague Dawley (SD) rats by a subretinal injection of Matrigel on Day 0.
  • SD Sprague Dawley
  • Subretinal lesion and CNV vessel volumes were quantified from 50 ⁇ m sections throughout the entire lesion using the formula
  • Bioanalytical analysis of functional mAb1 and anti-mAb1 antibodies in rat serum samples showed measurable concentrations of functional mAb1 in mAb1-treated animals with levels of 504 ⁇ 107 ⁇ g/mL on terminal date (Day 20) (Table 2). Individual anti-mAb1 responses were negative.
  • Formulation development activities included the screening of buffers, organic co-solvents, and thermal stabilizers in liquid formulations of anti-Ang-2 antibody to identify excipients that enhance the stability of the protein. Buffer conditions were also examined to determine the optimal pH for maximum protein stability. Results generated from these studies were used to develop a stable, liquid formulation suitable for clinical use.
  • the anti-Ang-2 antibody is the human anti-Ang-2 antibody with HCVR/LCVR of SEQ ID NOs: 1/2 and designated as H1H685 in US Patent Application Publication US20110027286 (also referred to herein as “mAb1”).
  • the anti-Ang-2 antibody was formulated at four concentrations:
  • the anti-Ang-2 antibody is formulated in 10 ⁇ 1.5 mM sodium phosphate (pH 6.2 ⁇ 0.3), 0.03% ⁇ 0.0045% polysorbate 20, 40 mM ⁇ 6.0 mM sodium chloride, and 5% ⁇ 0.75% sucrose, in water.
  • Formulated drug substance (FDS) 10 mg/mL mAb1 is stable when stored at ⁇ 20° C. for at least 12 months
  • FDS 120 mg/mL mAb1 is stable when stored at ⁇ 20° C. for at least 12 months
  • FDS 10 mg/mL mAb1 was physically and chemically stable after 56 days of incubation at 5° C. and maintained potency when incubated at 5° C. for 56 days or at 25° C./60% relative humidity (RH) and at 40° C./75% RH for 28 days.
  • FDS 120 mg/mL mAb1 was physically and chemically stable after 56 days of incubation at 5° C. and maintained potency when incubated at 5° C. for 56 days or at 25° C./60% relative humidity (RH) and at 40° C./75% RH for 28 days.
  • 10 mg/mL mAb1 FDS was physically and chemically stable when agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature). No appreciable change in the physical or chemical stability was detected in any of the monitored attributes.
  • mAb1 maintained potency when the 10 mg/mL mAb1 FDS was agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature).
  • 120 mg/mL mAb1 FDS was physically and chemically stable when agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature). No appreciable change in the physical or chemical stability was detected in any of the monitored attributes.
  • mAb1 maintained potency when the 120 mg/mL mAb1 FDS was agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature).
  • Drug product (DP) 10 mg/mL mAb1 is stable when stored at 2-8° C. for at least 9 months
  • DP 120 mg/mL mAb1 is stable when stored at 2-8° C. for at least 9 months.
  • Co-formulation development activities included the screening of buffers, organic co-solvents, and thermal stabilizers in liquid formulations of VEGF antagonist and anti-Ang-2 antibody to identify excipients that enhance the stability of the protein. Buffer conditions were also examined to determine the optimal pH for maximum protein stability. Results generated from these studies were used to develop a stable, liquid co-formulation suitable for clinical use.
  • the VEGF antagonist consists of a dimer of two polypeptides consisting of amino acids 27-457 of SEQ ID NO: 11 (also referred to herein as aflibercept).
  • the anti-Ang-2 antibody is the human anti-Ang-2 antibody with HCVR/LCVR of SEQ ID NOs: 1/2 and designated as H1H685 in US Patent Application Publication US20110027286 (also referred to herein as “mAb1”).
  • the VEGF antagonist was co-formulated with anti-Ang-2 antibody at four concentrations:
  • the anti-Ang-2 antibody and VEGF antagonist are co-formulated in 10 ⁇ 1.5 mM sodium phosphate (pH 6.2 ⁇ 0.3), 0.03% ⁇ 0.0045% polysorbate 20, 40 mM ⁇ 6.0 mM sodium chloride, and 5% ⁇ 0.75% sucrose, in water.
  • the stability of the formulated drug substance and drug product was assessed using assays as described in Example 8 herein. Charge variant analysis was done using imaged capillary isoelectric focusing (iCIEF) for the formulated drug substance and drug product.
  • iCIEF imaged capillary isoelectric focusing
  • Stability studies were initiated to determine the storage, accelerated (temperatures above storage conditions), and stress (agitation and freezing and thawing) stability of research lots of 10 mg/mL: 40 mg/mL and 120 mg/mL: 40 mg/mL (mAb1:aflibercept) FDS. These conditions were chosen to bracket the protein concentrations of FDS that used to manufacture the clinical DP. Evaluation of research lots of FDS under accelerated and stress conditions was performed by subjecting FDS to a variety of tests designed to exceed stresses the FDS may encounter during the manufacture of DP and to elucidate the degradation pathways for mAb1:aflibercept FDS. FDS was filled in 5 mL polycarbonate vials.
  • Formulated drug substance (FDS) 10:40 mg/mL mAb1: aflibercept is stable when stored at ⁇ 20° C. for at least 12 months
  • FDS 120:40 mg/mL mAb1:aflibercept is stable when stored at ⁇ 20° C. for at least 12 months
  • 120 mg/mL: 40 mg/mL mAb1:aflibercept FDS was physically and chemically stable after 56 days of incubation at 5° C. No appreciable change in the physical or chemical stability was detected in any of the monitored attributes.
  • 120 mg/mL: 40 mg/mL FDS was physically stable after 28 days of incubation at 25° C./60% RH. No appreciable change in the physical or chemical stability was detected in any of the other monitored attributes.
  • mAb1 and aflibercept both maintained potency when the 120 mg/mL: 40 mg/mL FDS was incubated at 5° C. for 56 days or at 25° C./60% RH for 28 days.
  • mAb1:aflibercept FDS 10 mg/mL: 40 mg/mL mAb1:aflibercept FDS was physically and chemically stable when agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature). No appreciable change in the physical or chemical stability was detected in any of the monitored attributes.
  • mAb1 and aflibercept maintained potency when the 10 mg/mL: 40 mg/mL FDS was agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature).
  • 120 minutes of vortex agitation is an extreme stress and agitation will be minimized for 120 mg/mL: 40 mg/mL mAb1:aflibercept FDS during the manufacturing process.
  • mAb1 and aflibercept maintained potency when the 10 mg/mL: 40 mg/mL mAb1:aflibercept FDS was agitated (vortexed at ambient temperature) for 120 minutes or when subjected to 8 freeze/thaw cycles (freezing at ⁇ 30° C. and thawing at room temperature).
  • Drug product (DP) 10:40 mg/mL mAb1:aflibercept is stable when stored at 2-8° C. for at least 9 months
  • DP 120:40 mg/mL mAb1:aflibercept is stable when stored at 2-8° C. for at least 9 months.
  • mAb1 and mAb1:aflibercept FDS and DP were assessed using the following assays: Color and appearance by visual inspection; pH; Turbidity measured by increase in Optical Density (OD) at 405 nm; Subvisible particulate analysis on DP by Microflow Imaging (MFI); Protein concentration by reversed-phase high performance liquid chromatography (RP-HPLC).
  • SE-UPLC Size exclusion ultra performance liquid chromatography
  • SDS-PAGE Reduced and non-reduced sodium dodecyl sulfate polyacrylamide gel electrophoresis
  • Cation exchange UPLC CEX-UPLC
  • iCIEF imaged capillary isoelectric focusing
  • Potency was assessed by bioassays performed for mAb1 in the mAb1 formulation; and for both mAb1 and aflibercept in the mAb1:aflibercept formulation.
  • the relative potency of each sample is determined using a bioassay and is defined as: (IC 50 Reference Sample/IC 50 Sample)*100%.
  • the measured potency of storage stability samples must be within 50-150% of the measured potency of the reference standard.
  • the physical stability of a formulation refers to properties such as color, appearance, pH, turbidity and protein concentration.
  • the presence of visible particulates in solution can be detected by visual inspection. A solution passes visual inspection if it is clear to slightly opalescent, essentially free from visible particulates, and colorless to pale yellow.
  • Turbidity measured by an increase in OD at 405 nm, can also be used to detect particulates in solution. An increase in OD at 405 nm may indicate the presence of particulates, an increase in opalescence, or color change of the test articles.
  • MFI is used to measure subvisible particulates that are ⁇ 2 ⁇ m in size. Protein concentration is measured by a RP-HPLC assay and reported as percent protein recovery relative to the starting material.
  • a single mAb1 peak is resolved from a single aflibercept peak following elution from the reversed phase column.
  • the mAb1 concentration is determined by comparing the mAb1 peak area to a calibration curve generated using a mAb1 standard whereas the aflibercept concentration is determined by comparing the aflibercept peak area to a calibration curve generated using an aflibercept standard. Percent recovery is calculated based on the measured mAb1 or aflibercept concentration relative to the starting mAb1 or aflibercept concentration, respectively.
  • Chemical stability refers to the formation of covalently modified forms (e.g. covalent aggregates, cleavage products or charge variant forms) and non-covalently modified forms (e.g. non-covalent aggregates) of protein.
  • Higher and lower molecular weight degradation products can be separated from native molecular weight product using SE-UPLC and SDS-PAGE methods.
  • the percentage of degraded mAb1 in the SE-UPLC method is calculated from the ratio of the area of all non-native peaks to the total area of all mAb1 peaks.
  • mAb1 Purity by non-reduced and reduced SDS-PAGE is calculated from the ratio of main band intensity to the total intensity of all bands.
  • Charge variant forms of mAb1 are resolved using iCIEF and CEX-UPLC. In these methods, peaks that are focused to a pl lower than that of the main peak are labeled “Acidic” peaks, whereas those focused to a pl higher than that of the main peak are labeled “Basic
  • mAb1:aflibercept formulations are characterized using total molecular weight purity (native mAb1+native aflibercept) by SE-UPLC (i.e. molecular weight purity of mAb1 and aflibercept will not be determined individually), because mAb1 and aflibercept native species cannot be fully resolved from each other.
  • mAb1:aflibercept formulations will be characterized using total HMW species (mAb1 HMW+aflibercept HMW) and total LMW species (mAb1 LMW+aflibercept LMW), because mAb1 HMW species cannot be resolved from aflibercept HMW species and mAb1 LMW species cannot be resolved from aflibercept LMW species.
  • the percentage of total HMW species or total LMW species in mAb1:aflibercept, determined using the SE-UPLC method is calculated from the ratio of the area of total HMW species or total LMW species to the total area of all mAb1:aflibercept peaks, respectively. Purity by non-reduced and reduced SDS-PAGE is calculated from the ratio of (aflibercept main band+mAb1 main band) intensity to the total intensity of all bands.
  • Aflibercept is a highly glycosylated protein that contains a high level of sialic acid.
  • the CEX-UPLC method did not have sufficient resolution to separate all charge variant forms and was therefore not used to assay changes in the charge variant profile for the mAb1:aflibercept samples.
  • Charge variant forms of mAb1 and aflibercept that are co-formulated within mAb1:aflibercept were resolved using iCIEF only.
  • peaks that are focused to a pl lower than that of the main peak are labeled “Acidic” peaks, whereas those focused to a pl higher than that of the main peak are labeled “Basic” peaks.
  • peaks 3-8 are the major peaks and are labeled “Main” peaks.
  • Aflibercept peaks that are focused to a pl lower than that of peak 3 are labeled “Acidic” peaks, whereas those focused to a pl higher than that of peak 8 are labeled “Basic” peaks.
  • SE-UPLC results from studies evaluating the storage stability of research formulations of 10 mg/mL mAb1, 120 mg/mL mAb1, and 40 mg/mL aflibercept were compared to results from the mAb1:aflibercept (10 mg/mL: 40 mg/mL) and (120 mg/mL: 40 mg/ml) co-formulations. This evaluation was performed to determine if co-formulating mAb1 and aflibercept resulted in differences in the relative amounts of high molecular weight species formed when compared to the individually formulated solutions of mAb1 and aflibercept (mono-formulations).
  • the DS lots used for these studies are representative of the DS manufactured for clinical use.
  • a summary of the SE-UPLC results for formulations stored at 5° C. containing 10 mg/mL mAb1 alone, 40 mg/mL aflibercept alone, and the mAb1:aflibercept (10 mg/mL: 40 mg/mL) co-formulation is as follows: After 9 months of storage at 5° C., the HMW content in the 10 mg/mL mAb1 formulation did not increase. Over the same assessment period the % HMW in the 40 mg/mL aflibercept formulation increased 0.4%.
  • a summary of the SE-UPLC results for formulations stored at 5° C. containing 120 mg/mL mAb1 alone, 40 mg/mL aflibercept alone, and the mAb1:aflibercept (120 mg/mL: 40 mg/mL) co-formulation is as follows: After 9 months of storage at 5° C., the HMW content in the 120 mg/mL mAb1 formulation and the 40 mg/mL aflibercept formulations increased by 0.5% and 0.4%, respectively.
  • IVT Intravitreally
  • This study is a phase I, open-label, dose escalation clinical study designed to evaluate the safety, tolerability, and efficacy of IVT administration of mAb1 alone or in combination with aflibercept in patients with neovascular AMD or DME.
  • the primary objective of the study is to investigate the safety and tolerability of IVT mAb1 and aflibercept, and IVT mAb1 in patients with neovascular AMD, and separately in patients with DME.
  • the secondary objectives of the study are: (i) to characterize the systemic pharmacokinetics (PK) of mAb1 and aflibercept following IVT injection of mAb1 and aflibercept; and (ii) to characterize the presence of anti-mAb1 and anti-aflibercept antibodies following IVT injection.
  • PK systemic pharmacokinetics
  • the primary endpoint is the incidence and severity of ocular and systemic treatment-emergent adverse events (TEAEs) through week 24 in patients treated with IVT mAb1 alone or with IVT co-formulated mAb1 and aflibercept.
  • TEAEs ocular and systemic treatment-emergent adverse events
  • the secondary endpoints are: (i) pharmacokinetics; and (ii) development of anti-drug antibodies (ADA) after IVT injection of the co-formulation.
  • the exploratory endpoints are: (1) change in BCVA from baseline; (2) change in central retinal thickness from baseline (measured by OCT) at week 12 and week 24; and (3) the number of PRN aflibercept injections from week 12 through week 20.
  • Anti-VEGF therapy e.g., aflibercept
  • aflibercept is standard of care treatment for neovascular AMD and DME.
  • Targeting both the VEGF and Angiopoeitin-2 (Ang-2) pathways in neovascular eye disease may result in additional efficacy over treatment with an anti-VEGF therapy alone, and also has the possibility of providing a longer duration of action resulting in a longer treatment interval.
  • Ang-2 Angiopoeitin-2
  • mAb1 the effect of mAb1 on retinal vascular development was compared to that of aflibercept, or to treatment with both mAb1 and aflibercept.
  • Administration of either mAb1 or aflibercept on P3 significantly reduced the mean vascularized area of the retina measured at P6 by 36% and 42%, respectively, relative to hFc controls.
  • the mean vascularized area of the retina was significantly smaller in the animals treated with both mAb1 and aflibercept, compared to animals treated with either agent alone, being reduced by 68%, representing a near complete arrest of retinal vascular development over the treatment period (see details in Example 3).
  • the starting dose is composed of a co-formulation of 0.5 mg mAb1:2 mg aflibercept, administered via IVT injection in the study eye.
  • the 2 mg dose of aflibercept is equivalent to that presently approved and marketed for the treatment of wet AMD and central retinal vein occlusion.
  • the 0.5 mg:2 mg dose of the co-formulation is one-half of the lowest dose administered IVT bilaterally to cynomolgus monkeys during the Good Laboratory Practice toxicology study. Doses of up to 6 mg mAb1 in combination with 2 mg aflibercept, and 6 mg mAb1 alone were well-tolerated in the monkeys.
  • the study consists of a screening period (day-21 to day-1), a baseline visit (day 1), a treatment period (day 1 through day 57), follow-up (day 85 through day 141) and an end of study visit [day 169 (week 24)].
  • a screening period (day-21 to day-1)
  • a baseline visit (day 1)
  • a treatment period (day 1 through day 57)
  • follow-up (day 85 through day 141)
  • an end of study visit [day 169 (week 24)].
  • eligible patients undergo safety assessments prior to receiving the first dose of study drug.
  • Intravitreal aflibercept injection (2 mg) is available to all patients beginning at week 12 for the study eye through week 20. Patients receiving aflibercept at week 12 will continue to receive aflibercept treatment with the possibility of up to monthly injections.
  • the eye with the worse BCVA score is selected as the study eye. If a patient has similar BCVA scores in both eyes, the eye with the clearest media is selected as the study eye. If the ocular media of both eyes are similar in clarity, the patient's non-dominant eye (if identifiable) is selected as the study eye. If neither eye is dominant, the right eye is designated as the study eye.
  • mAb1 and aflibercept 0.5 mg:2 mg, 1 mg:2 mg, 3 mg:2 mg, and 6 mg:2 mg
  • 1 cohort is planned for mAb1 alone (6 mg).
  • Each dose cohort initially consists of 4 patients, 2 DME and 2 AMD. If 1 dose limiting toxicity (DLT) occurs, the cohort can be expanded to include 2 additional DME patients and/or 2 additional AMD patients. Cohorts 4 and 5 can be run in parallel if cohort 4 is reached.
  • DLT dose limiting toxicity
  • the decision to escalate to the next higher dose level of co-formulated mAb1 and aflibercept is based upon safety and tolerability information from both AMD and DME patients evaluated during the ongoing cohort and reviewed.
  • the decision to escalate can take place after the last patient in a cohort (both AMD and DME) is observed for at least 1 week after receiving their first dose of study medication.
  • the mAb1 alone cohort will be enrolled either at the MTD or the highest dose (6 mg), if an MTD is not identified. If an MTD is not identified, enrollment in this cohort will be concurrent with the 6 mg:2 mg mAb1: aflibercept co-formulation cohort.
  • expansion of the cohort may be considered. Expansion of a cohort may involve 2 additional AMD patients and/or 2 additional DME patients.
  • the rationale to increase the number of patients in each cohort beyond 4 following a DLT event is to enhance the ability to differentiate certain AEs that are known to occur sporadically with IVT injections (e.g., hypersensitivity responses or moderate to severe intraocular inflammation) from a true DLT event related to study drug exposure.
  • Cohort expansion will not be automatic. Instead, the findings will be discussed to adjudicate the clinical significance of any potential DLT to determine if cohort expansion or dose escalation to the next higher dose level of the co-formulation will occur.
  • dosing will be suspended until a safety review has been conducted.
  • the outcome of the safety review will be a decision to: continue the study as planned, expand the current dose cohort, or stop dosing at the current dose.
  • the observed toxicity will be considered to be dose-limiting, and the dose below the highest dose administered will be considered the MTD.
  • SAE grade 4 serious adverse event
  • a DLT is defined as the following: A grade 2 or 3 ocular toxicity as determined by the Ocular Toxicity Grading Scale or a grade 3 or 4 toxicity in the Food and Drug Administration (FDA) September 2007 Guidance for Industry, Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials.
  • the MTD is defined as the dose level immediately below the level at which dosing is stopped due to the occurrence of 2 or more DLTs. If the study is not stopped due to the occurrence of a DLT, it will be considered that the MTD has not been determined.
  • the target population are men and women 50 years and older with neovascular AMD, or men and women 18 years and older with clinically significant DME with central involvement.
  • a patient must meet the following criteria to be eligible for inclusion in the study: (1) For patients with AMD: a. active subfoveal choroidal neovascularization (CNV) secondary to AMD, including juxtafoveal lesions that affect the fovea as evidenced by FA or OCT in the study eye, as determined by the investigator; and b. Men or women 50 years and older.
  • CNV active subfoveal choroidal neovascularization
  • DME c. Patients with clinically significant DME with central involvement (300 ⁇ m in the central subfield on spectral domain OCT); and d. Men or women 18 years and older.
  • a patient who meets any of the following criteria is excluded from the study: (1) (a) For patients with neovascular AMD: Evidence of CNV due to any cause other than AMD in either eye; Evidence of DR or DME in either eye; (b) For patients with DME: Evidence of neovascular AMD or CNV due to any cause in either eye; (2) Prior aflibercept in either eye; (3) IVT bevacizumab, ranibizumab, or pegaptanib sodium in the study eye within 8 weeks of day 1 or an AE with any of these previous treatments that would preclude administration of drug in this study; (4) Any prior treatment with angiopoietin inhibitors; (5) Any prior systemic (IV) anti-VEGF administration; (6) History of vitreoretinal surgery in the study eye; (7) Pan retinal laser photocoagulation or macular laser photocoagulation in the study eye within 3 months of the screening visit; (8) Previous use of intraocular or periocular corticosteroids in the study eye within 4 months of screening; (9)
  • Co-formulated mAb1 and aflibercept is a drug product that is composed of mAb1 (anti-Ang2 antibody) and aflibercept. It will be supplied for this study as an aqueous solution in sterile, single-use 2 mL glass vials for IVT administration, in the following concentrations (with progressively higher concentrations to be used for the progressively higher doses in dose escalation): 10:40 mg/mL, 20:40 mg/mL; 60:40 mg/mL; and 120:40 mg/mL (mAb1: aflibercept).
  • the mAb1 alone drug product is also supplied for this study as an aqueous solution in sterile, single-use 2 mL glass vials for IVT administration, at a concentration of 120 mg/mL.
  • the co-formulation and mAb1 is delivered via IVT injection and the injection volume will be 50 ⁇ l (0.05 cc). There will be a 0.050 ml minimum withdrawable content. Each vial contains a withdrawable volume of 0.3 mL of mAb1.
  • Study drug is administered on day 1, day 29 and day 57 by the investigator, or other qualified study personnel. Patients are enrolled in order to receive one of the following co-formulation or mAb1 treatment regimens:
  • Intravitreal aflibercept injections are supplied in sterile, sealed vials with a volume sufficient to prepare a syringe with 50 uL at a concentration of 40 mg/mL. Beginning at week 12 (and continuing through week 20), patients will be eligible to receive monthly aflibercept treatment (2 mg) in the study eye, if any of the re-treatment criteria listed below are satisfied: (1) There is a >50 ⁇ m increase in central retinal thickness on OCT compared to the lowest previous measurement. (2) There are new or persistent cystic retinal changes or subretinal fluid on OCT, or persistent diffuse edema in the central subfield on OCT. (3) A loss of 5 or more letters from the best previous measurement in conjunction with any increase in retinal thickness in the central subfield on OCT. (4) An improvement of BCVA between the current and most recent visit of letters.
  • aflibercept (2 mg) will be made available to patients with AMD or DME in the fellow eye at screening, or who are diagnosed with AMD or DME during the trial.
  • the fellow eye will be assessed for safety during the trial, but will not be considered a study eye.
  • the patient's fellow eye may, at the discretion of the investigator, receive treatment on the same day as the treatment of the study eye. All fellow eye treatments must be recorded on the electronic case report form (CRF) as a procedure for the fellow eye.
  • CRF electronic case report form
  • Patients may not receive any standard or investigational agents for AMD or DME treatment in the study eye other than their assigned study treatment with IVT co-formulation or IVT mAb1, and, if needed, aflibercept, as specified in this protocol.
  • aflibercept (2 mg) may be administered.
  • Other conditions in the fellow eye may be treated with approved therapies; however they must be administered locally.
  • Non-ocular (systemic) standard or investigational treatments for neo-vascular AMD and DME of the study or fellow eye are not permitted.
  • Systemic anti-angiogenic agents will not be permitted during the study.
  • Safety and tolerability is assessed by monitoring/evaluation of treatment-emergent adverse events (TEAEs), physical examinations, vital signs, electrocardiograms (ECGs), and clinical evaluations (hematology, blood chemistry and urinalysis).
  • Ocular safety is assessed by ophthalmic examinations (slit lamp, indirect ophthalmoscopy, intraocular pressure [IOP], spectral domain OCT, BCVA, FAF and information from FP and FA.
  • Serum samples will be collected for assessing mAb1 and plasma PK samples for aflibercept. Serum samples to assess ADA responses will be collected.
  • BCVA Best Corrected Visual Acuity
  • the anatomical state of the retinal vasculature of the study eye and the fellow eye is evaluated by funduscopic examination, FA, and FP at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment. At a minimum, information on the following variables will be collected:
  • DRSS Diabetic Retinopathy Severity Score
  • Retinal and lesion characteristics are evaluated using spectral domain OCT at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment.
  • Images are captured and transmitted at the study site by OCT technicians using spectral domain OCT for the study eye and fellow eye.
  • Optical coherence tomography images are sent to the independent reading center where images for the study eye will be read. All OCTs will be electronically archived at the study sites as part of the source documentation. Optical coherence tomography technicians will be certified by the reading center to ensure consistency and quality in image acquisition and will be masked to patients' dose level of the co-formulation.
  • Anatomic characteristics of the retina are also evaluated using autofluorescence. Certified photographers will perform FAF at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment. Images will be sent to the independent reading center. All images will be archived at the site as part of the source documentation.
  • Intraocular pressure of the study eye is measured at each study visit, using Goldmann applanation tonometry or Tono-PenTM. The same method of IOP measurement must be used in each patient throughout the study. On visits where an aflibercept treatment is given, IOP must be measured pre-treatment (bilateral) and at approximately 30 minutes post-treatment (study eye only).
  • the anterior segment and the anterior vitreous is examined using a slit lamp at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment. Both eyes are examined. Examination of the fundus is also conducted by indirect ophthalmoscopy.
  • Anterior chamber flare and cells are graded.
  • the intensity of the cellular reaction in the anterior chamber is graded according to the number of inflammatory cells seen in a 1 ⁇ 3-mm high-powered beam at full intensity at a 45° to 60° angle.
  • vitreal inflammatory response is graded.
  • Indirect ophthalmoscopy is performed at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment. It will be performed bilaterally predose, and in the study eye immediately after administration of study drug on days when study drug is administered.
  • the appropriate lenses and optics are used, ie, a head mounted or handheld ophthalmoscope for the periphery, and a slit lamp for the central fundus. The pupil is dilated, and examination of the fundus is done on both eyes.
  • Vital signs including temperature, sitting blood pressure, pulse, and respiration is collected, a complete and thorough physical examination, a standard 12-lead ECG as well as standard laboratory testing for hematology, chemistry, urinalysis, and pregnancy testing will be carried out predose at time points each study visit at screening, baseline, and week 1, 4, 6, 8, 12, 16, 20 and 24 after treatment.
  • Safety and tolerability is assessed by monitoring/evaluation of treatment-emergent adverse events (TEAEs), physical examinations, vital signs, electrocardiograms (ECGs), and clinical evaluations (hematology, blood chemistry and urinalysis).
  • TEAEs treatment-emergent adverse events
  • ECGs electrocardiograms
  • clinical evaluations hematology, blood chemistry and urinalysis.
  • An adverse event is any untoward medical occurrence in a patient administered a study drug which may or may not have a causal relationship with the study drug. Therefore, an AE is any unfavorable and unintended sign (including abnormal laboratory finding), symptom, or disease which is temporally associated with the use of a study drug, whether or not considered related to the study drug.
  • a serious adverse event is any untoward medical occurrence that at any dose: results in death, is life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, and/or is an important medical event.
  • Criteria for serious sight-threatening ocular AEs include the following: (1) AE causes a decrease in BCVA of >30 letters (compared with the most recent assessment of BCVA). (2) AE causes a decrease in VA to the level of light perception or worse. (3) AE requires surgical intervention (e.g., vitreous tap or biopsy with IVT injection of anti-infectives, laser or retinal cryopexy with gas) to prevent permanent loss of sight. (4) AE is associated with severe intraocular inflammation (ie, 4+anterior chamber cell/flare or 4+vitritis). (5) In the opinion of the investigator, AE may require medical intervention to prevent permanent loss of sight.
  • Tables 45 and 46 show the baseline demographics of patients with AMD and DME respectively.
  • Results to date demonstrate improvement in vision (increase in visual acuity of up to 20 letters) and retinal morphology (decrease in central subfield thickness) at all dose levels. Duration of effect was extended at higher doses (for the 3 mg:2 mg and 6 mg:2 mg doses).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Endocrinology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Ophthalmology & Optometry (AREA)
  • Inorganic Chemistry (AREA)
  • Toxicology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Emergency Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
US14/943,490 2014-11-25 2015-11-17 Methods and formulations for treating vascular eye diseases Abandoned US20160144025A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
US14/943,490 US20160144025A1 (en) 2014-11-25 2015-11-17 Methods and formulations for treating vascular eye diseases
MA041028A MA41028A (fr) 2014-11-25 2015-11-18 Méthodes et formulations pour le traitement de pathologies oculaires vasculaires
EP15862329.8A EP3224278A4 (en) 2014-11-25 2015-11-19 Methods and formulations for treating vascular eye diseases
EA201791168A EA201791168A1 (ru) 2014-11-25 2015-11-19 Способы и составы для лечения сосудистых заболеваний глаз
MA40306A MA40306A1 (fr) 2014-11-25 2015-11-19 Méthodes et formulations pour le traitement de pathologies oculaires vasculaires
AU2015353838A AU2015353838A1 (en) 2014-11-25 2015-11-19 Methods and formulations for treating vascular eye diseases
MX2017006129A MX2017006129A (es) 2014-11-25 2015-11-19 Métodos y formulaciones para tratar enfermedades vasculares de los ojos.
BR112017009807A BR112017009807A2 (pt) 2014-11-25 2015-11-19 métodos e formulações para tratamento de doenças oculares vasculares
SG11201703609UA SG11201703609UA (en) 2014-11-25 2015-11-19 Methods and formulations for treating vascular eye diseases
CA2968522A CA2968522A1 (en) 2014-11-25 2015-11-19 Methods and formulations for treating vascular eye diseases
PCT/US2015/061543 WO2016085750A1 (en) 2014-11-25 2015-11-19 Methods and formulations for treating vascular eye diseases
KR1020177017473A KR20170087950A (ko) 2014-11-25 2015-11-19 혈관성 안 질환을 치료하기 위한 방법 및 제형
CN201580063632.5A CN107001457A (zh) 2014-11-25 2015-11-19 用于治疗血管性眼病的方法和制剂
JP2017545861A JP2017536414A (ja) 2014-11-25 2015-11-19 血管性眼疾患を処置するための方法および製剤
PE2017000901A PE20170900A1 (es) 2014-11-25 2015-11-19 Metodos y formulaciones para tratar enfermedades vasculares de los ojos
US15/370,896 US20170080086A1 (en) 2014-11-25 2016-12-06 Methods and formulations for treating vascular eye diseases
PH12017500834A PH12017500834A1 (en) 2014-11-25 2017-05-05 Methods and formulations for treating vascular eye diseases
IL252159A IL252159B (en) 2014-11-25 2017-05-08 Methods and formulations for the treatment of diseases of the blood vessels of the eye
CONC2017/0004715A CO2017004715A2 (es) 2014-11-25 2017-05-10 Métodos y formulaciones para tratar enfermedades vasculares de los ojos
CL2017001188A CL2017001188A1 (es) 2014-11-25 2017-05-10 Métodos y formulaciones para tratar enfermedades vasculares de los ojos.
US16/235,221 US11071780B2 (en) 2014-11-25 2018-12-28 Methods and formulations for treating vascular eye diseases using aflibercept and nesvacumab
JP2020201473A JP2021046431A (ja) 2014-11-25 2020-12-04 血管性眼疾患を処置するための方法および製剤

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462084003P 2014-11-25 2014-11-25
US201562147232P 2015-04-14 2015-04-14
US14/943,490 US20160144025A1 (en) 2014-11-25 2015-11-17 Methods and formulations for treating vascular eye diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/370,896 Division US20170080086A1 (en) 2014-11-25 2016-12-06 Methods and formulations for treating vascular eye diseases

Publications (1)

Publication Number Publication Date
US20160144025A1 true US20160144025A1 (en) 2016-05-26

Family

ID=56009149

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/943,490 Abandoned US20160144025A1 (en) 2014-11-25 2015-11-17 Methods and formulations for treating vascular eye diseases
US15/370,896 Abandoned US20170080086A1 (en) 2014-11-25 2016-12-06 Methods and formulations for treating vascular eye diseases
US16/235,221 Active 2036-03-21 US11071780B2 (en) 2014-11-25 2018-12-28 Methods and formulations for treating vascular eye diseases using aflibercept and nesvacumab

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/370,896 Abandoned US20170080086A1 (en) 2014-11-25 2016-12-06 Methods and formulations for treating vascular eye diseases
US16/235,221 Active 2036-03-21 US11071780B2 (en) 2014-11-25 2018-12-28 Methods and formulations for treating vascular eye diseases using aflibercept and nesvacumab

Country Status (18)

Country Link
US (3) US20160144025A1 (ja)
EP (1) EP3224278A4 (ja)
JP (2) JP2017536414A (ja)
KR (1) KR20170087950A (ja)
CN (1) CN107001457A (ja)
AU (1) AU2015353838A1 (ja)
BR (1) BR112017009807A2 (ja)
CA (1) CA2968522A1 (ja)
CL (1) CL2017001188A1 (ja)
CO (1) CO2017004715A2 (ja)
EA (1) EA201791168A1 (ja)
IL (1) IL252159B (ja)
MA (2) MA41028A (ja)
MX (1) MX2017006129A (ja)
PE (1) PE20170900A1 (ja)
PH (1) PH12017500834A1 (ja)
SG (1) SG11201703609UA (ja)
WO (1) WO2016085750A1 (ja)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9795594B2 (en) 2006-06-27 2017-10-24 Aerpio Therapeutics, Inc. Human protein tyrosine phosphatase inhibitors and methods of use
US9926367B2 (en) 2006-04-07 2018-03-27 Aerpio Therapeutics, Inc. Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof
US9949956B2 (en) 2009-07-06 2018-04-24 Aerpio Therapeutics, Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
US9994560B2 (en) 2014-03-14 2018-06-12 Aerpio Therapeutics, Inc. HPTP-β inhibitors
US20180186869A1 (en) * 2009-07-29 2018-07-05 Regeneron Pharmaceuticals, Inc. High Affinity Human Antibodies to Human Angiopoietin-2
WO2018208625A1 (en) * 2017-05-06 2018-11-15 Regeneron Pharmaceuticals, Inc. Methods of treating eye disorders with aplnr antagonists and vegf inhibitors
US10150811B2 (en) 2011-10-13 2018-12-11 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
WO2019036604A1 (en) * 2017-08-18 2019-02-21 Regeneron Pharmaceuticals, Inc. IMAGE CAPILLARY ISOELECTRIC FOCUSING FOR ANALYSIS OF PROTEIN VARIANTS IN A SAMPLING MATRIX
US10220048B2 (en) 2013-03-15 2019-03-05 Aerpio Therapeutics, Inc. Compositions and methods for treating ocular diseases
US10253094B2 (en) 2016-07-20 2019-04-09 Aerpio Therapeutics, Inc. Antibodies that target human protein tyrosine phosphatase-beta (HPTP-beta) and methods of use thereof to treat ocular conditions
WO2019173767A1 (en) * 2018-03-08 2019-09-12 Coherus Biosciences Inc. Stable aqueous formulations of aflibercept
US20190300607A1 (en) * 2016-10-12 2019-10-03 Daiichi Sankyo Company, Limited Composition containing anti-robo4 antibody and other agents
US10952992B2 (en) 2015-09-23 2021-03-23 Aerpio Pharmaceuticals, Inc. Methods of treating intraocular pressure with activators of Tie-2
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11103552B2 (en) 2018-05-10 2021-08-31 Regeneron Pharmaceuticals, Inc. High concentration VEGF receptor fusion protein containing formulations
US11104730B2 (en) 2013-11-20 2021-08-31 Regeneren Pharmaceuticals, Inc. Methods of treating eye disorders with APLNR antagonists and VEGF inhibitors
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11253502B2 (en) 2019-04-29 2022-02-22 EyePoint Pharmaceuticals, Inc. Tie-2 activators targeting the Schlemm's canal
EP3836959A4 (en) * 2018-08-17 2022-05-11 Trican Biotechnology Co., Ltd ANTI-ANGIOGENESIS FUSION PROTEIN AND USES ITS
US11426446B2 (en) 2018-03-08 2022-08-30 Coherus Biosciences, Inc. Stable aqueous formulations of aflibercept
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11732024B2 (en) 2006-06-16 2023-08-22 Regeneron Pharmaceuticals, Inc. VEGF antagonist formulations suitable for intravitreal administration
US11806398B2 (en) 2005-03-25 2023-11-07 Regeneron Pharmaceuticals, Inc. Citrate buffered VEGF antagonist formulations
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI738632B (zh) 2014-11-07 2021-09-11 瑞士商諾華公司 穩定的含有高濃度抗vegf抗體之蛋白質溶液調配物
JP2021508687A (ja) * 2017-12-22 2021-03-11 サムスン バイオエピス カンパニー リミテッド Vegfアンタゴニストを含む液体組成物
JP2021511061A (ja) 2018-01-26 2021-05-06 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 抗vegf剤を使用した血管新生障害の治療のための方法および組成物
CA3088355A1 (en) * 2018-02-06 2019-08-15 F. Hoffmann-La Roche Ag Treatment of ophthalmologic diseases
CN108671229B (zh) * 2018-05-08 2022-03-25 华博生物医药技术(上海)有限公司 一种重组人血管内皮生长因子受体-抗体融合蛋白的药物组合制剂
CN109598628B (zh) * 2018-11-30 2022-09-20 平安医疗健康管理股份有限公司 医保欺诈行为的识别方法、装置、设备及可读存储介质
AR117707A1 (es) 2018-12-18 2021-08-25 Novartis Ag Formulación de solución de proteínas que contiene una alta concentración de un anticuerpo anti-vegf
CN110423281B (zh) * 2019-07-31 2021-04-30 成都金唯科生物科技有限公司 用于治疗老年性黄斑变性的融合蛋白、病毒载体和药物
CA3168512A1 (en) 2019-11-25 2021-06-03 Napoleone Ferrara Long-acting vegf inhibitors for intraocular neovascularization
CN113493519B (zh) * 2020-03-19 2022-12-27 浙江道尔生物科技有限公司 一种半衰期显著延长的治疗眼部血管新生疾病的融合蛋白
AU2021294703A1 (en) * 2020-06-22 2023-02-23 Innovent Biologics (Suzhou) Co., Ltd. Anti-ANG-2 antibody and use thereof
WO2022019721A1 (ko) * 2020-07-24 2022-01-27 (주) 팬젠 안과용 액상 조성물

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120189635A1 (en) * 2009-07-29 2012-07-26 Regeneron Pharmaceuticals, Inc. Methods for Treating or Preventing Malaria by Administering an Antibody that Specifically Binds Angiopoietin-2 (Ang-2)
US9062105B1 (en) * 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
WO2005072772A1 (en) * 2004-01-30 2005-08-11 Suomen Punainen Risti Veripalvelu Pharmaceutical compositions
AR059066A1 (es) * 2006-01-27 2008-03-12 Amgen Inc Combinaciones del inhibidor de la angiopoyetina -2 (ang2) y el inhibidor del factor de crecimiento endotelial vascular (vegf)
KR101406811B1 (ko) * 2006-06-16 2014-06-12 리제너론 파마슈티칼스 인코포레이티드 유리체내 투여에 적당한 vegf 길항제 제형
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
CA2665956A1 (en) * 2009-05-07 2010-11-07 Samir Patel Combination treatment for ocular diseases
EA201190273A1 (ru) 2009-05-28 2012-12-28 Глаксо Груп Лимитед Комбинация антагониста tnf-альфа и антагониста vegf для применения в лечении или предупреждении заболеваний глаз
JO3182B1 (ar) * 2009-07-29 2018-03-08 Regeneron Pharma مضادات حيوية بشرية عالية الالفة مع تولد الاوعية البشرية - 2
US8980268B2 (en) * 2009-07-29 2015-03-17 Regeneron Pharamceuticals, Inc. Methods for treating cancer by administering an anti-Ang-2 antibody
AR080794A1 (es) * 2010-03-26 2012-05-09 Hoffmann La Roche Anticuerpos bivalentes biespecificos anti- vegf/ anti-ang-2
WO2012097019A1 (en) * 2011-01-13 2012-07-19 Regeneron Pharmaceuticals, Inc. Use of a vegf antagonist to treat angiogenic eye disorders
KR102063028B1 (ko) 2012-01-23 2020-01-07 리제너론 파아마슈티컬스, 인크. 항-ang2 항체를 함유하는 안정화된 제형
IN2014DN11157A (ja) * 2012-07-13 2015-10-02 Roche Glycart Ag
AP2015008365A0 (en) * 2012-12-05 2015-04-30 Novartis Ag Compositions and methods for antibodies targeting epo
JO3405B1 (ar) * 2013-01-09 2019-10-20 Regeneron Pharma الأجسام المضادة لمضاد مستقبل عامل النمو المشتق من الصفائح الدموية - بيتا واستخداماتها
AU2014288847A1 (en) * 2013-07-11 2016-01-28 Novartis Ag Use of a VEGF antagonist in treating retinopathy of prematurity
DE112014005747T5 (de) * 2013-12-17 2016-10-06 Kymab Limited Antikörper zur Verwendung bei der Behandlung von Zuständen, die mit spezifischen PCSK9 Varianten in spezifischen Patientenpopulationen in Beziehung stehen

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120189635A1 (en) * 2009-07-29 2012-07-26 Regeneron Pharmaceuticals, Inc. Methods for Treating or Preventing Malaria by Administering an Antibody that Specifically Binds Angiopoietin-2 (Ang-2)
US9062105B1 (en) * 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11806398B2 (en) 2005-03-25 2023-11-07 Regeneron Pharmaceuticals, Inc. Citrate buffered VEGF antagonist formulations
US11814425B2 (en) 2006-04-07 2023-11-14 Eye Point Pharmaceuticals, Inc. Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof
US9926367B2 (en) 2006-04-07 2018-03-27 Aerpio Therapeutics, Inc. Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof
US11732024B2 (en) 2006-06-16 2023-08-22 Regeneron Pharmaceuticals, Inc. VEGF antagonist formulations suitable for intravitreal administration
USRE46592E1 (en) 2006-06-27 2017-10-31 Aerpio Therapeutics, Inc. Human protein tyrosine phosphatase inhibitors and methods of use
US10463650B2 (en) 2006-06-27 2019-11-05 Aerpio Pharmaceuticals, Inc. Human protein tyrosine phosphatase inhibitors and methods of use
US9795594B2 (en) 2006-06-27 2017-10-24 Aerpio Therapeutics, Inc. Human protein tyrosine phosphatase inhibitors and methods of use
US9949956B2 (en) 2009-07-06 2018-04-24 Aerpio Therapeutics, Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
US10875911B2 (en) * 2009-07-29 2020-12-29 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human angiopoietin-2
US20180186869A1 (en) * 2009-07-29 2018-07-05 Regeneron Pharmaceuticals, Inc. High Affinity Human Antibodies to Human Angiopoietin-2
US10815300B2 (en) 2011-10-13 2020-10-27 Aerpio Pharmaceuticals, Inc. Methods for treating vascular leak syndrome and cancer
US10150811B2 (en) 2011-10-13 2018-12-11 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
US10220048B2 (en) 2013-03-15 2019-03-05 Aerpio Therapeutics, Inc. Compositions and methods for treating ocular diseases
US11104730B2 (en) 2013-11-20 2021-08-31 Regeneren Pharmaceuticals, Inc. Methods of treating eye disorders with APLNR antagonists and VEGF inhibitors
US10858354B2 (en) 2014-03-14 2020-12-08 Aerpio Pharmaceuticals, Inc. HPTP-Beta inhibitors
US9994560B2 (en) 2014-03-14 2018-06-12 Aerpio Therapeutics, Inc. HPTP-β inhibitors
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11666558B2 (en) 2015-09-23 2023-06-06 EyePoint Pharmaceuticals, Inc. Methods of treating intraocular pressure with activators of Tie-2
US10952992B2 (en) 2015-09-23 2021-03-23 Aerpio Pharmaceuticals, Inc. Methods of treating intraocular pressure with activators of Tie-2
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US10604569B2 (en) 2016-07-20 2020-03-31 Aerpio Pharmaceuticals, Inc. Humanized monoclonal antibodies that target protein tyrosine phosphatase-beta (HPTP-β/VE-PTP)
US11136389B2 (en) 2016-07-20 2021-10-05 Aerpio Pharmaceuticals, Inc. Humanized monoclonal antibodies that target VE-PTP (HPTP-β)
US10597452B2 (en) 2016-07-20 2020-03-24 Aerpio Pharmaceuticals, Inc. Methods of treating ocular conditions by administering humanized monoclonal antibodies that target VE-PTP (HPTP-beta)
US10253094B2 (en) 2016-07-20 2019-04-09 Aerpio Therapeutics, Inc. Antibodies that target human protein tyrosine phosphatase-beta (HPTP-beta) and methods of use thereof to treat ocular conditions
US11180551B2 (en) 2016-07-20 2021-11-23 EyePoint Pharmaceuticals, Inc. Humanized monoclonal antibodies that target VE-PTP (HPTP-beta)
US20190300607A1 (en) * 2016-10-12 2019-10-03 Daiichi Sankyo Company, Limited Composition containing anti-robo4 antibody and other agents
WO2018208625A1 (en) * 2017-05-06 2018-11-15 Regeneron Pharmaceuticals, Inc. Methods of treating eye disorders with aplnr antagonists and vegf inhibitors
IL270267B1 (en) * 2017-05-06 2024-02-01 Regeneron Pharma Methods for treating eye disorders with APLNR antagonists and VEGF inhibitors
CN110709104A (zh) * 2017-05-06 2020-01-17 瑞泽恩制药公司 用aplnr拮抗剂和vegf抑制剂治疗眼部病症的方法
TWI774827B (zh) * 2017-08-18 2022-08-21 美商雷傑納榮製藥公司 使用影像毛細管等電聚焦以分析樣品基質中蛋白質變異體之方法
KR20200078466A (ko) * 2017-08-18 2020-07-01 리제너론 파마슈티칼스 인코포레이티드 샘플 매트릭스에서 단백질 변이체를 분석하기 위한 이미지 모세관 등전 집속
WO2019036604A1 (en) * 2017-08-18 2019-02-21 Regeneron Pharmaceuticals, Inc. IMAGE CAPILLARY ISOELECTRIC FOCUSING FOR ANALYSIS OF PROTEIN VARIANTS IN A SAMPLING MATRIX
JP7399845B2 (ja) 2017-08-18 2023-12-18 リジェネロン・ファーマシューティカルズ・インコーポレイテッド 試料マトリックスにおいてタンパク質変異体を分析するための画像キャピラリー等電点電気泳動法
EP3668546A4 (en) * 2017-08-18 2021-05-12 Regeneron Pharmaceuticals, Inc. IMAGE CAPILLARY ISOELECTRIC FOCUSING FOR ANALYSIS OF PROTEIN VARIANTS IN A SAMPLING MATRIX
CN111132695A (zh) * 2017-08-18 2020-05-08 里珍纳龙药品有限公司 分析样品基质中蛋白变异体的成像毛细管等电聚焦
US20220317088A1 (en) * 2017-08-18 2022-10-06 Regeneron Pharmaceuticals, Inc. Image capillary isoelectric focusing to analyze protein variants in a sample matrix
KR102584848B1 (ko) * 2017-08-18 2023-10-04 리제너론 파마슈티칼스 인코포레이티드 샘플 매트릭스에서 단백질 변이체를 분석하기 위한 이미지 모세관 등전 집속
JP2020531802A (ja) * 2017-08-18 2020-11-05 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. 試料マトリックスにおいてタンパク質変異体を分析するための画像キャピラリー等電点電気泳動法
US11667702B2 (en) 2018-03-08 2023-06-06 Coherus Biosciences, Inc. Stable aqueous formulations of aflibercept
US11426446B2 (en) 2018-03-08 2022-08-30 Coherus Biosciences, Inc. Stable aqueous formulations of aflibercept
WO2019173767A1 (en) * 2018-03-08 2019-09-12 Coherus Biosciences Inc. Stable aqueous formulations of aflibercept
US11103552B2 (en) 2018-05-10 2021-08-31 Regeneron Pharmaceuticals, Inc. High concentration VEGF receptor fusion protein containing formulations
EP3836959A4 (en) * 2018-08-17 2022-05-11 Trican Biotechnology Co., Ltd ANTI-ANGIOGENESIS FUSION PROTEIN AND USES ITS
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11253502B2 (en) 2019-04-29 2022-02-22 EyePoint Pharmaceuticals, Inc. Tie-2 activators targeting the Schlemm's canal
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Also Published As

Publication number Publication date
KR20170087950A (ko) 2017-07-31
SG11201703609UA (en) 2017-06-29
US11071780B2 (en) 2021-07-27
PH12017500834A1 (en) 2017-10-30
WO2016085750A1 (en) 2016-06-02
EA201791168A1 (ru) 2017-09-29
JP2017536414A (ja) 2017-12-07
EP3224278A4 (en) 2018-07-11
US20190117767A1 (en) 2019-04-25
CA2968522A1 (en) 2016-06-02
MA41028A (fr) 2017-10-03
CN107001457A (zh) 2017-08-01
CL2017001188A1 (es) 2017-11-24
MX2017006129A (es) 2018-04-10
IL252159B (en) 2021-03-25
JP2021046431A (ja) 2021-03-25
IL252159A0 (en) 2017-07-31
BR112017009807A2 (pt) 2018-02-27
AU2015353838A1 (en) 2017-06-08
EP3224278A1 (en) 2017-10-04
CO2017004715A2 (es) 2017-09-20
PE20170900A1 (es) 2017-07-12
MA40306A1 (fr) 2019-03-29
US20170080086A1 (en) 2017-03-23

Similar Documents

Publication Publication Date Title
US11071780B2 (en) Methods and formulations for treating vascular eye diseases using aflibercept and nesvacumab
US10973879B2 (en) Use of a VEGF antagonist to treat angiogenic eye disorders
US20230302085A1 (en) Extended, High Dose VEGF Antagonist Regimens for Treatment of Angiogenic Eye Disorders
US11104730B2 (en) Methods of treating eye disorders with APLNR antagonists and VEGF inhibitors
US20230295266A1 (en) Extended, High Dose VEGF Antagonist Regimens for Treatment of Angiogenic Eye Disorders
JP2017532342A (ja) 眼療法のためのアンジオポエチン−1及びアンジオポエチン−2を対象とする抗体
KR20220062279A (ko) 안질환의 치료 방법
AU2018266324B2 (en) Methods of treating eye disorders with APLNR antagonists and VEGF inhibitors
US20240024420A1 (en) Extended, High Dose VEGF Antagonist Regimens for Treatment of Angiogenic Eye Disorders
KR102667021B1 (ko) Aplnr 길항제 및 vegf 저해제를 이용한, 안 장애의 치료 방법
CA3190726A1 (en) Extended, high dose vegf antagonist regimens for treatment of angiogenic eye disorders
EA043390B1 (ru) Способы лечения заболеваний глаз антагонистами aplnr и ингибиторами vegf
EA046420B1 (ru) Применение антагониста vegf для лечения ангиогенных глазных заболеваний

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VITTI, ROBERT L.;ERICKSON, KRISTINE A.;CHU, KAREN W.;AND OTHERS;SIGNING DATES FROM 20160104 TO 20160330;REEL/FRAME:038438/0854

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION