US20210139576A1 - Treatment of ophthalmologic diseases - Google Patents

Treatment of ophthalmologic diseases Download PDF

Info

Publication number
US20210139576A1
US20210139576A1 US16/985,477 US202016985477A US2021139576A1 US 20210139576 A1 US20210139576 A1 US 20210139576A1 US 202016985477 A US202016985477 A US 202016985477A US 2021139576 A1 US2021139576 A1 US 2021139576A1
Authority
US
United States
Prior art keywords
weeks
seq
vegf
bispecific antibody
treatment
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
US16/985,477
Other languages
English (en)
Inventor
Aaron Osborne
Jayashree Sahni
Robert James Weikert
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.)
Genentech Inc
Hoffmann La Roche Inc
Original Assignee
Genentech Inc
Hoffmann La Roche 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 Genentech Inc, Hoffmann La Roche Inc filed Critical Genentech Inc
Priority to US16/985,477 priority Critical patent/US20210139576A1/en
Publication of US20210139576A1 publication Critical patent/US20210139576A1/en
Priority to US18/166,996 priority patent/US20230416353A1/en
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
    • 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
    • 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
    • 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
    • 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
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • 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
    • C07K2317/71Decreased effector function due to an Fc-modification

Definitions

  • the current invention relates to the use of antibodies which bind to VEGF and ANG2 for the treatment of ophthalmologic diseases.
  • Angiogenesis is implicated in the pathogenesis of a variety of disorders which include solid tumors, intraocular neovascular syndromes such as proliferative retinopathies or age-related macular degeneration (AMD), rheumatoid arthritis, and psoriasis (Folkman, J., et al., J. Biol. Chem. 267 (1992) 10931-10934; Klagsbrun, M., et al., Annu. Rev. Physiol. 53 (1991) 217-239; and Gamer, A., Vascular diseases, in: Pathobiology of ocular disease, A dynamic approach, Garner, A., and Klintworth, G. K. (eds.), 2nd edition, Marcel Dekker, New York (1994), pp. 1625-1710).
  • Ranibizumab (trade name Lucentis®) is a monoclonal antibody fragment derived from the same parent murine antibody as bevacizumab (Avastin®). However, it has been affinity matured to provide stronger binding to VEGF-A (WO 98/45331). It is known that systemic blockade of VEGF-A is associated with an increased risk of certain adverse events, therefore ranibizumab is missing an Fc part in order to reduce systemic exposure and the risk of systemic toxicities. It is an anti-angiogenic agent that has been approved to treat the “wet” type of age-related macular degeneration (neovascular AMD), a common form of age-related vision loss.
  • neovascular AMD neovascular AMD
  • ANG-2 acts as an apoptosis survival factor for endothelial cells during serum deprivation apoptosis through activation of Tie2 via PI-3 Kinase and Akt pathway (Kim, I., et al., Oncogene 19 (2000) 4549-52).
  • Ocular vascular diseases such as “wet” age related macular degeneration (AMD) and proliferative diabetic retinopathy (PDR), are due to abnormal choroidal or retinal neovascularization respectively. Bleeding and leakage from these vessels can cause retinal dysfunction and loss of cision
  • Other retinal vascular disease such as diabetic macular edema (DME) and macular edema secondary to retinal vein occlusion (RVO) are due to abnormal retinal leakage leading to retinal swelling and impairing visual function. These conditions are leading causes of visual loss in industrialized nations.
  • DME diabetic macular edema
  • RVO retinal vein occlusion
  • RP Retinitis Pigmentosa
  • Ischemic retinopathies are characterized by loss or dysfunction of the retinal vasculature which results in a reduction of blood flow and hypoxia.
  • the retina responds to hypoxia by generating signals to grow new blood vessels, but these new vessels are usually fragile and disorganized. It is the growth of these abnormal new vessels that creates most of the threat to vision since they can leak, hemorrhage or lead to scarring that may end in retinal detachment.
  • Current treatments for ischemic retinopathies seek to halt the growth of the pathological vessels but do not address the underlying ischemia that drives their growth.
  • VEGF vascular endothelial growth factor
  • bispecific antibodies for use
  • medicaments or pharmaceutical formulations for the treatment of patients suffering from an ocular vascular disease the method comprising administering to the patient an effective amount of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2),
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • One aspect of the invention is such method, use, bispecific antibody (for use), medicament or pharmaceutical formulation (for use) of/for treating a patient suffering from an ocular vascular disease
  • the method, use, bispecific antibody (for use), medicament or pharmaceutical formulation (for use) comprising administering (intravitreally) to the patient an effective amount of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), wherein the patient gains 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered intravitreally every 8 weeks or less frequently.
  • One embodiment of the invention is a method of treating a patient suffering from an ocular vascular disease the method comprising administering (intravitreally) to the patient an effective amount of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), wherein the patient experiences an improvement in vision subsequent to the administration of the bispecific VEGF/ANG2 antibody as measured by gaining 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • the bispecific antibody is administered intravitreally every 8 weeks or less frequently.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 4 weeks, and/or at 8 weeks, and/or at 12 weeks, and/or at 16 weeks, and/or at 20 weeks, and/or at 24 weeks after treatment start, respectively.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 24 weeks, and/or at 25 weeks, and/or at 26 weeks, and/or at 27 weeks, and/or at 28 weeks, and/or at 29 weeks, and/or at 30 weeks, and/or at 31 weeks, and/or at 32 weeks, and/or at 33 weeks, and/or at 34 weeks, and/or at 35 weeks, and/or at 36 weeks, and/or at 37 weeks, and/or at 38 weeks, and/or at 39 weeks, and/or at 40 weeks, and/or at 41 weeks, and/or at 42 weeks, and/or at 43 weeks, and/or at 44 weeks, and/or at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 30 weeks, and/or
  • the ocular vascular disease is selected from the group of: wet age-related macular degeneration (wet AMD), neovascular AMD, diabetic macular edema (DME), cystoid macular edema (CME), non-proliferative diabetic retinopathy (NPDR), proliferative diabetic retinopathy (PDR), macular edema secondary to central retinal vein occlusion, secondary to hemiretinal vein occlusion or secondary to branch vein occlusion, retinitis, conjunctivitis, uveitis, choroiditis, choroidal neovascularization (CNV) secondary to ocular inflammation including secondary to ocular histoplasmosis or presumed histoplasmosis or choroiditis; myopic choroidal neovascularization (mCNV). And choroidal neovascularization secondary to trauma, retinopathy of prematurity and rubeosis
  • the ocular vascular disease is diabetic macular edema (DME).
  • DME diabetic macular edema
  • the ocular vascular disease is diabetic macular edema (DME) and the gain of letters in the BCVA/ETDRS letter score is measured at about 9 to 15 month (in one embodiment at 9 to 14 month, in one embodiment at 9 to 12 month) after treatment start.
  • DME diabetic macular edema
  • the ocular vascular disease is diabetic macular edema (DME) and the gain of letters in the BCVA/ETDRS letter score is measured at 36 weeks, and/or at 37 weeks, and/or at 38 weeks, and/or at 39 weeks, and/or at 40 weeks, and/or at 41 weeks, and/or at 42 weeks, and/or at 43 weeks, and/or at 44 weeks, and/or at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 57 weeks, and/or at 58 weeks, and/or at 59 weeks, and/or at 60 weeks after treatment start, respectively.
  • DME diabetic macular edema
  • the ocular vascular disease is wet age-related macular degeneration (wet AMD) (, or neovascular age-related macular degeneration (nAMD).
  • wet AMD wet age-related macular degeneration
  • nAMD neovascular age-related macular degeneration
  • the ocular vascular disease is wet age-related macular degeneration (wet AMD) (, or neovascular age-related macular degeneration (nAMD) and the gain of letters in the BCVA/ETDRS letter score is measured at about 9 to 15 month (in one embodiment at 6 to 9 month, in one embodiment at 6 to 12 month) after treatment start.
  • wet AMD wet age-related macular degeneration
  • nAMD neovascular age-related macular degeneration
  • the ocular vascular disease is wet age-related macular degeneration (wet AMD) (, or neovascular age-related macular degeneration (nAMD) and the gain of letters in the BCVA/ETDRS letter score is measured at 24 weeks, and/or at 25 weeks, and/or at 26 weeks, and/or at 27 weeks, and/or at 28 weeks, and/or at 29 weeks, and/or at 30 weeks, and/or at 31 weeks, and/or at 32 weeks, and/or at 33 weeks, and/or at 34 weeks, and/or at 35 weeks, and/or at 36 weeks, and/or at 37 weeks, and/or at 38 weeks, and/or at 39 weeks, and/or at 40 weeks, and/or at 41 weeks, and/or at 42 weeks, and/or at 43 weeks, and/or at 44 weeks, and/or at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at
  • the bispecific antibody which binds to human VEGF and to human ANG2 is a bispecific, bivalent anti-VEGF/ANG2 antibody comprising a first antigen-binding site that specifically binds to human VEGF and a second antigen-binding site that specifically binds to human ANG-2, wherein
  • the patients suffering from an ocular vascular disease have not been previously treated with anti-VEGF treatment (e.g monotherapy)(are treatment na ⁇ ve).
  • anti-VEGF treatment e.g monotherapy
  • the patients suffering from an ocular vascular disease have been previously treated with anti-VEGF treatment (e.g monotherapy).
  • anti-VEGF treatment e.g monotherapy
  • the ocular vascular disease is DME and the treatment of patients suffering from DME includes a fixed every 8 th week (Q8W) dosing schedule following treatment initiation.
  • Q8W 8 th week
  • the ocular vascular disease is DME and the treatment of patients suffering from DME includes a fixed Q12W dosing schedule following treatment initiation. In one embodiment of the invention following the treatment initiation, first one dose cycle of Q8W follows before the fixed Q12W dosing schedule.
  • the ocular vascular disease is DME and the treatment of patients suffering from DME includes following treatment initiation a dosing schedule that extends the administration interval in stable absence of disease, or shortens the interval if there is disease activity.
  • such dosing schedule includes that the patient receives Q4W or Q8W or Q12W or Q16W dosing, dependent on their disease state.
  • the stable absence of disease is determined as
  • the ocular vascular disease is AMD and the treatment of patients suffering from AMD includes following treatment initiation a dosing schedule that extends the administration interval in stable absence of disease, or shortens the interval if there is disease activity.
  • such dosing schedule includes that the patient receives Q4W or Q8W or Q12W or Q16W dosing, dependent on their disease state.
  • the stable absence of disease is determined as
  • FIG. 1 BCVA change of DME patients treated from Baseline over Time to Week 24 (treatment naive patients).
  • VA2 refers to the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose),
  • RBZ refers to ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose))
  • FIG. 2 CST, central subfield thickness measured by SD OCT.
  • CST change of DME patients treated from Baseline over Time to Week 24 (treatment naive patients).
  • the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose), was compared to ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)).
  • the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose), was compared to ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)).
  • FIG. 4 Schematic comparison to other treatment options of DME based on published results (Compared agents Lucentis® (ranibizumab), Eylea® (aflibercept), brolucizumab and VA2 (RO6867461/RG7716).
  • FIG. 5 Overview of the study design for the evaluation of the bispecific antibody RO6867461 administered at 12- and 16-week intervals in patients with neovascular age-related macular degeneration (nAMD).
  • nAMD neovascular age-related macular degeneration
  • FIG. 6 BCVA gains from baseline of patients with neovascular age-related macular degeneration (nAMD) comparing the bispecific antibody RO6867461 (comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg) at 12- and 16-week intervals and ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)) at 4-week intervals.
  • RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg) at 12- and 16-week intervals and ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)) at 4-week intervals.
  • FIG. 7 Change from baseline CST (measured via OCT) of patients with neovascular age-related macular degeneration (nAMD) comparing the bispecific antibody RO6867461 (comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg) at 12- and 16-week intervals and ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)) at 4-week intervals.
  • RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg) at 12- and 16-week intervals and ranibizumab (Lucentis®) ((administered intravitreally with a 0.3 mg dose)) at 4-week intervals.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • One embodiment of the invention is a method of treating a patient suffering from a ocular vascular disease the method comprising administering to the patient an effective amount of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), wherein the patient gains 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • One embodiment of the invention is a method of treating a patient suffering from a ocular vascular disease the method comprising administering to the patient an effective amount of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), wherein the patient experiences an improvement in vision subsequent to the administration of the bispecific VEGF/ANG2 antibody as measured by gaining 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • the gain of letters in the BCVA/ETDRS letter score is measured at 4 weeks, and/or at 8 weeks, and/or at 12 weeks, and/or at 16 weeks, and/or at 20 weeks, and/or at 24 weeks after treatment start, respectively.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 24 weeks, and/or at 25 weeks, and/or at 26 weeks, and/or at 27 weeks, and/or at 28 weeks, and/or at 29 weeks, and/or at 30 weeks, and/or at 31 weeks, and/or at 32 weeks, and/or at 33 weeks, and/or at 34 weeks, and/or at 35 weeks, and/or at 36 weeks, and/or at 37 weeks, and/or at 38 weeks, and/or at 39 weeks, and/or at 40 weeks, and/or at 41 weeks, and/or at 42 weeks, and/or at 43 weeks, and/or at 44 weeks, and/or at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 30 weeks, and/or
  • the gain of letters in the BCVA/ETDRS letter score is measured at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 57 weeks, and/or at 58 weeks, and/or at 59 weeks, and/or at 60 weeks after treatment start, respectively.
  • the method is used to prolong the time to retreatment and/or to prolong the time to loss of visual acuity and, wherein the retreatment with the bispecific antibody is administered in case of a disease activity which is determined as
  • One embodiment of the invention is a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of an ocular vascular disease,
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • One embodiment of the invention is a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of a patient suffering from an ocular vascular disease, wherein the patient gains 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • BCVA Best Corrected Visual Acuity
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • One embodiment of the invention is a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of a patient suffering from an ocular vascular disease, wherein the patient experiences an improvement in vision subsequent to the (intravitreal) administration of the bispecific VEGF/ANG2 antibody as measured by gaining 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • the gain of letters in the BCVA/ETDRS letter score is measured at 4 weeks, and/or at 8 weeks, and/or at 12 weeks, and/or at 16 weeks, and/or at 20 weeks, and/or at 24 weeks after treatment start, respectively.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 57 weeks, and/or at 58 weeks, and/or at 59 weeks, and/or at 60 weeks after treatment start, respectively.
  • such bispecific antibody (for use) is used to prolong the time to retreatment and/or to prolong the time to loss of visual acuity and, wherein the retreatment with the bispecific antibody is administered in case of a disease activity which is determined as
  • One embodiment of the invention is a medicament or pharmaceutical formulation comprising a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of an ocular vascular disease,
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • One embodiment of the invention is a medicament or pharmaceutical formulation comprising a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of a patient suffering from an ocular vascular disease, wherein the patient gains 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • One embodiment of the invention is a medicament or pharmaceutical formulation comprising a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for use in the treatment of a patient suffering from an ocular vascular disease, wherein the patient experiences an improvement in vision subsequent to the (intravitreal) administration of the bispecific VEGF/ANG2 antibody as measured by gaining 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • the gain of letters in the BCVA/ETDRS letter score is measured at 4 weeks, and/or at 8 weeks, and/or at 12 weeks, and/or at 16 weeks, and/or at 20 weeks, and/or at 24 weeks after treatment start, respectively.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 57 weeks, and/or at 58 weeks, and/or at 59 weeks, and/or at 60 weeks after treatment start, respectively.
  • such medicament or pharmaceutical formulation is used to prolong the time to retreatment and/or to prolong the time to loss of visual acuity and, wherein the retreatment with the bispecific antibody is administered in case of a disease activity which is determined as
  • One embodiment of the invention is the use of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for the manufacture of a medicament for use in the treatment of an ocular vascular disease,
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • One embodiment of the invention is the use of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for the manufacture of a medicament for use in the treatment of an ocular vascular disease, wherein the patient gains 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • BCVA Best Corrected Visual Acuity
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • One embodiment of the invention is the use of a bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2), for the manufacture of a medicament for use in the treatment of an ocular vascular disease, wherein the patient experiences an improvement in vision subsequent to the (intravitreal) administration of the bispecific VEGF/ANG2 antibody as measured by gaining 12 or more letters (in one embodiment 13 or more letters, in one embodiment 14 or more letters, in one embodiment 15 or more letters) of Best Corrected Visual Acuity (BCVA) measured using Early Treatment Diabetic Retinopathy Study (ETDRS) like charts, compared to the patient's BCVA letter score prior to the dosing of the bispecific VEGF/ANG2 antibody.
  • VEGF vascular endothelial growth factor
  • ANG-2 human angiopoietin-2
  • the bispecific antibody is administered (is to be administered) intravitreally every 8 weeks or less frequently (in one embodiment every 9 weeks or less frequently; in one embodiment every 10 weeks or less frequently; in one embodiment every 11 weeks or less frequently; in one embodiment every 12 weeks or less frequently; in one embodiment every 13 weeks or less frequently; in one embodiment every 14 weeks or less frequently; in one embodiment every 15 weeks or less frequently).
  • the gain of letters in the BCVA/ETDRS letter score is measured at 4 weeks, and/or at 8 weeks, and/or at 12 weeks, and/or at 16 weeks, and/or at 20 weeks, and/or at 24 weeks after treatment start, respectively.
  • the gain of letters in the BCVA/ETDRS letter score is measured at 45 weeks, and/or at 46 weeks, and/or at 47 weeks, and/or at 48 weeks, and/or at 49 weeks, and/or at 50 weeks, and/or at 51 weeks, and/or at 52 weeks, and/or at 53 weeks, and/or at 54 weeks, and/or at 55 weeks, and/or at 56 weeks, and/or at 57 weeks, and/or at 58 weeks, and/or at 59 weeks, and/or at 60 weeks after treatment start, respectively.
  • medicament is used to prolong the time to retreatment and/or to prolong the time to loss of visual acuity and, wherein the retreatment with the bispecific antibody is administered in case of a disease activity which is determined as
  • BCVA determination in such method, use, bispecific antibody (for use), medicament or pharmaceutical formulation is based on the Early Treatment of Diabetic Retinopathy Study (ETDRS) Protocol adapted visual acuity charts and is assessed at a starting distance of 4 meters.
  • EDRS Early Treatment of Diabetic Retinopathy Study
  • Such method, use, bispecific antibody (for use), medicament or pharmaceutical formulation may comprise sequentially administering initial doses (“treatment initiation”) (e.g. 3 to 7 monthly administrations; in one embodiment the treatment initiation includes 3 to 4 monthly administrations, in one embodiment the treatment initiation includes 4 to 5 monthly administrations; in one embodiment the treatment initiation includes 4 to 6 monthly administrations; in one embodiment the treatment initiation includes at least 4 monthly administrations; in one embodiment the treatment initiation includes 5 to 7 monthly administrations, in one embodiment the treatment initiation includes 6 monthly administrations) followed by one or more secondary doses of a therapeutically effective amount of the bispecific antibody, medicament or pharmaceutical formulation.
  • initial doses e.g. 3 to 7 monthly administrations; in one embodiment the treatment initiation includes 3 to 4 monthly administrations, in one embodiment the treatment initiation includes 4 to 5 monthly administrations; in one embodiment the treatment initiation includes 4 to 6 monthly administrations; in one embodiment the treatment initiation includes at least 4 monthly administrations; in one embodiment the treatment initiation includes 5 to 7 monthly administrations, in one embodiment the treatment
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 10 to 12 weeks (following treatment initiation).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 11 to 13 weeks (following treatment initiation).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 12 to 14 weeks (following treatment initiation).
  • the bispecific antibody, medicament or pharmaceutical formulation is is administered every 13 to 15 weeks (following treatment initiation).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 14 to 16 weeks (following treatment initiation).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 10 to 11 weeks, or every 11 to 12 weeks, or every 12 to 13 weeks, or every 13 to 14 weeks, or every 14 to 15 weeks, or every 15 to 16 weeks (following treatment initiation, respectively).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered every 10 weeks, or every 11 weeks, or every 12 weeks, or every 13 weeks, or every 14 weeks, or every 16 weeks (following treatment initiation, respectively).
  • the bispecific antibody, medicament or pharmaceutical formulation is administered in a dose of about 5 to 7 mg (at each treatment). In one embodiment the bispecific antibody is is administered in a dose of 6 mg+/ ⁇ 10% (at each treatment). In one embodiment the bispecific antibody is is administered in a dose of about 6 mg (at each treatment). (in one embodiment in a dose of 6 mg (at each treatment))
  • the bispecific antibody, medicament or pharmaceutical formulation is administered in a concentration of about 30 mg/ml of the bispecific antibody. In one embodiment of the invention the bispecific antibody, medicament or pharmaceutical formulation is administered in a concentration of about 120 mg/ml of the bispecific antibody.
  • ocular vascular disease and “vascular eye disease” are used interchangeable herein and include, but are not limited to intraocular neovascular syndromes such as diabetic retinopathy, diabetic macular edema, retinopathy of prematurity, neovascular glaucoma, (branch) retinal vein occlusions, central retinal vein occlusions, macular degeneration, age-related macular degeneration, retinitis pigmentosa, retinal angiomatous proliferation, macular telangectasia, ischemic retinopathy, iris neovascularization, intraocular neovascularization, comeal neovascularization, retinal neovascularization, choroidal neovascularization, and retinal degeneration.
  • intraocular neovascular syndromes such as diabetic retinopathy, diabetic macular edema, retinopathy of prematurity, neovascular glaucoma, (branch)
  • ocular vascular disorder refers to any pathological conditions characterized by altered or unregulated proliferation and invasion of new blood vessels into the structures of ocular tissues such as the retina or cornea.
  • the ocular vascular disease is selected from the group consisting of: wet age-related macular degeneration (wet AMD), neovascular AMD (nAMD), diabetic macular edema (DME), cystoid macular edema (CME), non-proliferative diabetic retinopathy (NPDR), proliferative diabetic retinopathy (PDR), macular edema secondary to central retinal vein occlusion, secondary to hemiretinal vein occlusion or secondary to branch vein occlusion, retinitis, conjunctivitis, uveitis, choroiditis, choroidal neovascularization (CNV) secondary to ocular inflammation including secondary to ocular histoplasmosis or presumed histoplasmosis or choroiditis; myopic choroidal neovascularization (mCNV).
  • wet AMD wet age-related macular degeneration
  • nAMD neovascular AMD
  • anti-VEGF/ANG2 bispecific antibodies for use and the methods described herein are useful in the prevention and treatment of wet AMD, nAMD CME, DME, NPDR, PDR, and uveitis, also preferably wet AMD, nAMD, also preferably DME, CME, NPDR and PDR, and also particularly wet AMD.
  • the ocular vascular disease is selected from the group consisting of wet age-related macular degeneration (wet AMD), neovascular age-related macular degeneration (nAMD), (diabetic) macular edema, retinal vein occlusions, retinopathy of prematurity, and diabetic retinopathy.
  • wet AMD wet age-related macular degeneration
  • nAMD neovascular age-related macular degeneration
  • (diabetic) macular edema retinal vein occlusions
  • retinal vein occlusions retinal vein occlusions
  • retinopathy of prematurity retinopathy of prematurity
  • diabetic retinopathy diabetic retinopathy
  • diseases/conditions associated with comeal neovascularization include, but are not limited to, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, sjogrens syndrome, acne rosacea, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections, Kaposi sarcoma, Mooren ulcer, Terrien's marginal degeneration, marginal keratolysis, rheumatoid arthritis, systemic lupus, polyarteritis, trauma, Wegeners sarcoidosis, Scleritis, Steven's Johnson disease, periphigoid radial
  • Diseases/conditions associated with retinal/choroidal neovascularization include, but are not limited to, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum, Pagets disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, mycobacterial infections, Lyme's disease, systemic lupus erythematosis, retinopathy of prematurity, retinitis pigmentosa, retina edema (including macular edema), Eales disease, Bechets disease, infections causing a retinitis or choroiditis, presumed ocular histoplasmosis, Bests disease, myopia, optic (disc) pits, Stargardts disease, pars planitis, chronic retinal detach
  • ROP Retinopathy of prematurity
  • Macular degeneration is a medical condition predominantly found in elderly adults in which the center of the inner lining of the eye, known as the macula area of the retina, suffers thinning, atrophy, and in some cases, bleeding. This can result in loss of central vision, which entails inability to see fine details, to read, or to recognize faces. According to the American Academy of Ophthalmology, it is the leading cause of central vision loss (blindness) in the United States today for those over the age of fifty years. Although some macular dystrophies that affect younger individuals are sometimes referred to as macular degeneration, the term generally refers to age-related macular degeneration (AMD or ARMD).
  • AMD age-related macular degeneration
  • AMD Age-related macular degeneration
  • AMD refers to a serious eye condition when the small central portion of the retina, known as the macula, deteriorates.
  • AMD includes wet AMD and neovascular AMD.
  • the wet form of AMD (wet AMD, wAMD or also called neovascular AMD, nAMD) 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 (below and) into the retina, causing (elevation of the retina and) 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.
  • AMD 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 which are tiny yellow deposits under the retina and pigment clumping.
  • Advanced AMD which is responsible for profound vision loss, has two forms: dry and wet.
  • vitamin supplements with high doses of antioxidants, lutein and zeaxanthin have been demonstrated by the National Eye Institute and others to slow the progression of dry macular degeneration and in some patients, improve visual acuity.
  • Retinitis pigmentosa is a group of genetic eye conditions. In the progression of symptoms for RP, night blindness generally precedes tunnel vision by years or even decades. Many people with RP do not become legally blind until their 40s or 50s and retain some sight all their life. Others go completely blind from RP, in some cases as early as childhood. Progression of RP is different in each case. RP is a type of hereditary retinal dystrophy, a group of inherited disorders in which abnormalities of the photoreceptors (rods and cones) or the retinal pigment epithelium (RPE) of the retina lead to progressive visual loss. Affected individuals first experience defective dark adaptation or nyctalopia (night blindness), followed by reduction of the peripheral visual field (known as tunnel vision) and, sometimes, loss of central vision late in the course of the disease.
  • Macular edema occurs when fluid and protein deposits collect on or under the macula of the eye, the central area of the retina responsible for fine vision, causing it to thicken and swell. 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. This area holds tightly packed cones that provide sharp, clear central vision to enable a person to see form, color, and detail that is directly in the line of sight. Cystoid macular edema is a type of macular edema that includes cyst formation.
  • 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 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 inner the blood-retinal barrier, normally preventing fluid 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.
  • BCVA Best Corrected Visual Acuity
  • Disease activity is determined e.g. via reduction of the BCVA/ETDRs letter score and/or e.g. via the macular thickening by spectral domain optical coherence tomography (SD-OCT) involving the center of the macula as central subfield thickness (CST) (also known as center subfoveal thickness).
  • SD-OCT spectral domain optical coherence tomography
  • Central Subfield Thickness is determined using spectral domain optical coherence tomography (SD-OCT): In one preferred embodiment CST is measured by spectral domain optical coherence tomography (SD-OCT) with a SpectralisTM device; in one preferred embodiment CST is measured by spectral domain optical coherence tomography (SD-OCT) with a CirrusTM device; in one embodiment CST is measured by spectral domain optical coherence tomography (SD-OCT) with a TopconTM device; in one embodiment CST is measured by spectral domain optical coherence tomography (SD-OCT) with a OptovueTM device).
  • SD-OCT spectral domain optical coherence tomography
  • a patient suffering from refers to a human that exhibits one or more symptoms or indications of, and/or who has been diagnosed with an ocular vascular disease as described herein.
  • the term “a patient suffering from” may also include, e.g., subjects who, prior to treatment, exhibit (or have exhibited) one or more indications of a vascular eye disease such as, e.g., retinal angiogenesis, neovascularization, vascular leak, retinal thickening of the center of the fovea, hard, yellow exudates 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 vascular eye disease such as, e.g., retinal angiogenesis, neovascularization, vascular leak, retinal thickening of the center of the fovea, hard,
  • a patient suffering from 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 patient suffering from” includes a subject who, prior to or at the time of administration of the bispecific anti-VEGF/ANG2 antibody, has or is diagnosed with diabetes.
  • the term “a patient suffering from” includes a subject who, prior to or at the time of administration of the anti-VEGF/ANG2 antibody, is more than 50 years old.
  • the term “a patient suffering from” includes subjects who are smokers, or subjects with high blood pressure or high cholesterol.
  • the present invention includes methods or bispecific antibodies (for use), medicaments or pharmaceutical formulations for treating, preventing or reducing the severity of an ocular vascular disease comprising administering a therapeutically effective amount of a bispecific anti-VEGF/ANG2 antibody (or a medicament or pharmaceutical formulation comprising the bispecific anti-VEGF/ANG2 antibody) to a subject in need thereof, wherein the bispecific antibody, medicament or pharmaceutical formulation comprising such bispecific anti-VEGF/ANG2 antibody is administered (intravitreally) to the subject in multiple doses, e.g., as part of a specific therapeutic dosing regimen.
  • One embodiment of the invention is the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein patients suffering from an ocular vascular disease have not been previously treated with anti-VEGF treatment (e.g monotherapy) (are treatment na ⁇ ve).
  • anti-VEGF treatment e.g monotherapy
  • One embodiment of the invention is the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein patients suffering from an ocular vascular disease have been previously treated with anti-VEGF treatment (e.g monotherapy).
  • One embodiment of the invention is the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein the ocular vascular disease is DME and the treatment of patients suffering from DME includes a fixed every 8th week (Q8W) dosing schedule following treatment initiation (In one embodiment the treatment initiation includes 5 to 7 monthly administrations; in one embodiment the treatment initiation includes 6 monthly administrations).
  • Q8W 8th week
  • One embodiment of the invention is the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein the ocular vascular disease is DME and the treatment of patients suffering from DME includes a fixed Q12W dosing schedule following treatment initiation (In one embodiment the treatment initiation includes 5 to 7 monthly administrations; in one embodiment the treatment initiation includes 6 monthly administrations). In one embodiment following the treatment initiation, first one dose cycle of Q8W follows before the fixed Q12W dosing schedule.
  • One embodiment of the invention is the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein the ocular vascular disease is DME and the treatment of patients suffering from DME includes following treatment initiation a dosing schedule that extends the administration interval in stable absence of disease, or shortens the interval if there is disease activity (In one embodiment the treatment initiation includes 3 to 7 monthly administrations; in one embodiment the treatment initiation includes 3 to 5 monthly administrations; in one embodiment the treatment initiation includes at least 4 monthly administrations; in one embodiment the treatment initiation includes 4 to 6 monthly administrations). In one embodiment such dosing schedule includes that the patient receives Q4W or Q8W or Q12W or Q16W dosing, dependent on their disease state. In one embodiment the stable absence of disease is determined as
  • the method of treatment, use, bispecific antibody (for use), medicament or pharmaceutical formulation as described herein wherein the ocular vascular disease is AMD (in one embodiment wet AMD) and the treatment of patients suffering from AMD (in one embodiment wet AMD) includes following treatment initiation a dosing schedule that extends the administration interval in stable absence of disease, or shortens the interval if there is disease activity (In one embodiment the treatment initiation includes 3 to 7 monthly administrations; in one embodiment the treatment initiation includes 3 to 5 monthly administrations; in one embodiment the treatment initiation includes at least 4 monthly administrations; in one embodiment the treatment initiation includes 4 to 6 monthly administrations). In one embodiment such dosing schedule includes that the patient receives Q4W or Q8W or Q12W or Q16W dosing, dependent on their disease state. In one embodiment the stable absence of disease is determined as
  • the vascular ocular disease in such method, use, bispecific antibody (for use), medicament or pharmaceutical formulation is wetAMD (nAMD).
  • antibody refers to a binding protein that comprises antigen-binding sites.
  • binding site or “antigen-binding site” as used herein denotes the region(s) of an antibody molecule to which a ligand actually binds.
  • antigen-binding site comprises an antibody heavy chain variable domains (VH) and an antibody light chain variable domains (VL) (pair of VH/VL).).
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific.
  • “Bispecific antibodies” according to the invention are antibodies which have two different antigen-binding specificities. Antibodies of the present invention are specific for two different antigens, VEGF as first antigen and ANG-2 as second antigen.
  • monospecific antibody denotes an antibody that has one or more binding sites each of which bind to the same epitope of the same antigen.
  • bivalent as used within the current application denotes the presence of a specified number of binding sites in an antibody molecule.
  • the terms “bivalent”, “tetravalent”, and “hexavalent” denote the presence of two binding site, four binding sites, and six binding sites, respectively, in an antibody molecule.
  • the bispecific antibodies according to the invention are preferably “bivalent”.
  • bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2)”, “bispecific anti-VEGF/ANG2 antibody” and bispecific ⁇ VEGF/ANG2> antibody” as used herein are interchangeable and refer to an antibody which has at least two different antigen-binding sites, a first one which binds to VEGF and a second one which binds to ANG2.
  • Bispecific anti-VEGF/ANG2 antibodies are e.g. described in WO2010040508, WO2011/117329, WO2012/131078, WO2015/083978, WO2017/197199, and WO2014/009465.
  • WO2014/009465 describes bispecific anti-VEGF/ANG2 antibodies especially designed for treatment of ocular vascular diseases.
  • the bispecific anti-VEGF/ANG2 antibodies of WO2014/009465 (which is incorporated herein in its entirety) are especially useful in the treatment and treatment schedules of ocular vascular diseases as described herein.
  • the bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2) is a bispecific anti-VEGF/ANG2 antibody comprising a first antigen-binding site that specifically binds to human VEGF and a second antigen-binding site that specifically binds to human ANG-2, wherein
  • such bispecific anti-VEGF/ANG2 antibody is bivalent.
  • such bispecific anti-VEGF/ANG2 antibody is characterized in that
  • such bispecific, bivalent antibody according to the invention is characterized in comprising
  • This bispecific, bivalent antibody format for the bispecific antibody specifically binding to human vascular endothelial growth factor (VEGF) and human angiopoietin-2 (ANG-2) is described in WO 2009/080253 (including Knobs-into-Holes modified CH3 domains).
  • the antibodies based on this bispecific, bivalent antibody format are named CrossMAbs.
  • bispecific, bivalent anti-VEGF/ANG2 antibody is characterized in comprising
  • bispecific, bivalent anti-VEGF/ANG2 antibody is characterized in comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20.
  • the bispecific, bivalent anti-VEGF/ANG2 antibody is faricimab.
  • one embodiment of the invention is a bispecific, bivalent antibody comprising a first antigen-binding site that specifically binds to human VEGF and a second antigen-binding site that specifically binds to human ANG-2, characterized in comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20.
  • the bispecific, bivalent anti-VEGF/ANG2 antibody is faricimab.
  • the CH3 domains of the bispecific, bivalent antibody according to the invention is altered by the “knob-into-holes” technology which is described in detail with several examples in e.g. WO 96/027011, Ridgway J. B., et al., Protein Eng 9 (1996) 617-621; and Merchant, A. M., et al., Nat Biotechnol 16 (1998) 677-681.
  • the interaction surfaces of the two CH3 domains are altered to increase the heterodimerisation of both heavy chains containing these two CH3 domains.
  • Each of the two CH3 domains (of the two heavy chains) can be the “knob”, while the other is the “hole”.
  • the bispecific anti-VEGF/ANG2 antibodies according to the invention are characterized in that the CH3 domain of one heavy chain and the CH3 domain of the other heavy chain each meet at an interface which comprises an original interface between the antibody CH3 domains;
  • said interface is altered to promote the formation of the bispecific antibody, wherein the alteration is characterized in that: a) the CH3 domain of one heavy chain is altered, so that within the original interface the CH3 domain of one heavy chain that meets the original interface of the CH3 domain of the other heavy chain within the bispecific antibody, an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the interface of the CH3 domain of one heavy chain which is positionable in a cavity within the interface of the CH3 domain of the other heavy chain and b) the CH3 domain of the other heavy chain is altered, so that within the original interface of the second CH3 domain that meets the original interface of the first CH3 domain within the bispecific antibody an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the interface of the second CH3 domain within which a protuberance within the interface of the first CH3 domain is positionable.
  • bispecific anti-VEGF/ANG2 antibodies for use described herein are preferably characterized in that
  • amino acid residue having a larger side chain volume is selected from the group consisting of arginine (R), phenylalanine (F), tyrosine (Y), tryptophan (W).
  • amino acid residue having a smaller side chain volume is selected from the group consisting of alanine (A), serine (S), threonine (T), valine (V).
  • both CH3 domains are further altered by the introduction of cysteine (C) as amino acid in the corresponding positions of each CH3 domain such that a disulfide bridge between both CH3 domains can be formed.
  • C cysteine
  • the bispecific antibody comprises a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain”.
  • An additional interchain disulfide bridge between the CH3 domains can also be used (Merchant, A. M, et al., Nature Biotech 16 (1998) 677-681) e.g. by introducing a S354C mutation into one CH3 domain and a Y349C mutation into the other CH3 domain.
  • the bispecific antibody comprises S354C and T366W mutations in one of the two CH3 domains and Y349C, T366S, L368A, Y407V mutations in the other of the two CH3 domains
  • the bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains (the additional Y349C or S354C mutation in one CH3 domain and the additional S354C or Y349C mutation in the other CH3 domain forming a interchain disulfide bridge) (numbering always according to EU index of Kabat (Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • the heterodimerization approach described in EP 1 870 459A1 is used alternatively. This approach is based on the introduction of substitutions/mutations of charged amino acids with the opposite charge at specific amino acid positions of the in the CH3/CH3 domain interface between both heavy chains.
  • One preferred embodiment for said multispecific antibodies are amino acid R409D and K370E mutations in the CH3 domain of one heavy chain and amino acid D399K and E357K mutations in the CH3 domain of the other heavy chain of the multispecific antibody (numberings according to Kabat EU index).
  • said multispecific antibody comprises an amino acid T366W mutation in the CH3 domain of the “knobs chain” and amino acid T366S, L368A and Y407V mutations in the CH3 domain of the “hole chain”; and additionally comprises amino acid R409D and K370E mutations in the CH3 domain of the “knobs chain” and amino acid D399K and E357K mutations in the CH3 domain of the “hole chain”.
  • the heterodimerization approach described in WO2013/157953 is used alternatively.
  • the CH3 domain of one heavy chain comprises an amino acid T366K mutation and the CH3 domain of the other heavy chain comprises an amino acid L351D mutation.
  • the CH3 domain of the one heavy chain further comprises an amino acid L351K mutation.
  • the CH3 domain of the other heavy chain further comprises an amino acid mutation selected from Y349E, Y349D and L368E (in one embodiment L368E).
  • the heterodimerization approach described in WO2012/058768 is used alternatively.
  • the CH3 domain of one heavy chain comprises amino acid L351Y and Y407A mutations and the CH3 domain of the other heavy chain comprises amino acid T366A and K409F mutations.
  • the CH3 domain of the other heavy chain further comprises an amino acid mutation at position T411, D399, 5400, F405, N390 or K392.
  • said amino acid mutation is selected from the group consisting of
  • the CH3 domain of one heavy chain comprises amino acid L351Y and Y407A mutations and the CH3 domain of the other heavy chain comprises amino acid T366V and K409F mutations.
  • the CH3 domain of one heavy chain comprises an amino acid Y407A mutation and the CH3 domain of the other heavy chain comprises amino acid T366A and K409F mutations.
  • the CH3 domain of the other heavy chain further comprises amino acid K392E, T411E, D399R and S400R mutations.
  • heterodimerization approach described in WO2011/143545 is used alternatively.
  • amino acid modification according to WO2011/143545 is introduced in the CH3 domain of the heavy chain at a position selected from the group consisting of 368 and 409.
  • the heterodimerization approach described in WO2011/090762 which also uses the knob-into-hole technology described above is used alternatively.
  • the CH3 domain of one heavy chain comprises an amino acid T366W mutation and the CH3 domain of the other heavy chain comprises an amino acid Y407A mutation.
  • the CH3 domain of one heavy chain comprises an amino acid T366Y mutation and the CH3 domain of the other heavy chain comprises an amino acid Y407T mutation.
  • the multispecific antibody is of IgG2 isotype and the heterodimerization approach described in WO2010/129304 is used alternatively.
  • the heterodimerization approach described in WO2009/089004 is used alternatively.
  • the CH3 domain of one heavy chain comprises an amino acid substitution of K392 or N392 with a negatively-charged amino acid (in one embodiment glutamic acid (E) or aspartic acid (D); in a further embodiment a K392D or N392D mutation) and the CH3 domain of the other heavy chain comprises an amino acid substitution of D399, E356, D356, or E357 with a positively-charged amino acid (in one embodiment Lysine (K) or arginine (R), in a further embodiment a D399K, E356K, D356K or E357K substitution; and in an even further embodiment a D399K or E356K mutation).
  • the CH3 domain of the one heavy chain further comprises an amino acid substitution of K409 or R409 with a negatively-charged amino acid (in one embodiment glutamic acid (E) or aspartic acid (D); in a further embodiment a K409D or R409D mutation).
  • the CH3 domain of the one heavy chain further or alternatively comprises an amino acid substitution of K439 and/or K370 with a negatively-charged amino acid (in one embodiment glutamic acid (E) or aspartic acid (D)).
  • the heterodimerization approach described in WO2007/147901 is used alternatively.
  • the CH3 domain of one heavy chain comprises amino acid K253E, D282K and K322D mutations and the CH3 domain of the other heavy chain comprises amino acid D239K, E240K and K292D mutations.
  • heterodimerization approach described in WO2007/110205 is used alternatively.
  • the bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2) is a bispecific anti-VEGF/ANG2 antibody comprising a first antigen-binding site that specifically binds to human VEGF and a second antigen-binding site that specifically binds to human ANG-2, wherein
  • the bispecific antibody which binds to human vascular endothelial growth factor (VEGF) and to human angiopoietin-2 (ANG-2) is a bispecific anti-VEGF/ANG2 antibody comprising a first antigen-binding site that specifically binds to human VEGF and a second antigen-binding site that specifically binds to human ANG-2, wherein
  • such bispecific anti-VEGF/ANG2 antibody is bivalent.
  • such bispecific anti-VEGF/ANG2 antibody is characterized in that
  • such bispecific, bivalent antibody according to the invention is characterized in comprising
  • VEGF refers to human vascular endothelial growth factor (VEGF/VEGF-A,) the 165-amino acid human vascular endothelial cell growth factor (amino acid 27-191 of precursor sequence of human VEGF165: SEQ ID NO: 24; amino acids 1-26 represent the signal peptide), and related 121, 189, and 206 vascular endothelial cell growth factor isoforms, as described by Leung, D. W., et al., Science 246 (1989) 1306-9; Houck et al., Mol. Endocrin. 5 (1991) 1806-1814; Keck, P.
  • VEGF vascular endothelial growth factor
  • VEGF is a homodimeric glycoprotein that has been isolated from several sources and includes several isoforms. VEGF shows highly specific mitogenic activity for endothelial cells.
  • a VEGF antagonist/inhibitor inhibits binding of VEGF to its receptor VEGFR.
  • Known VEGF antagonist/inhibitors include bispecific anti-VEGF/ANG2 antibodies as described in WO2014/009465.
  • ANG-2 refers to human angiopoietin-2 (ANG-2) (alternatively abbreviated with ANGPT2 or ANG2) (SEQ ID NO: 25) which is described e.g. in Maisonpierre, P. C., et al, Science 277 (1997) 55-60 and Cheung, A. H., et al., Genomics 48 (1998) 389-91.
  • the angiopoietins-1 (SEQ ID NO: 26) and -2 were discovered as ligands for the Ties, a family of tyrosine kinases that is selectively expressed within the vascular endothelium (Yancopoulos, G.
  • Angiopoietin-3 and -4 may represent widely diverged counterparts of the same gene locus in mouse and man (Kim, I., et al., FEBS Let, 443 (1999) 353-56; Kim, I., et al., J Biol Chem 274 (1999) 26523-28).
  • ANG-1 and ANG-2 were originally identified in tissue culture experiments as agonist and antagonist, respectively (see for ANG-1: Davis, S., et al., Cell 87 (1996) 1161-69; and for ANG-2: Maisonpierre, P.
  • Ang-1 and -2 bind to TIE2 with an affinity of 3 nM (Kd) (Maisonpierre, P. C., et al., Science 277 (1997) 55-60).
  • An ANG2 antagonist/inhibitor inhibits binding of ANG2 to its receptor TIE2.
  • Known ANG2 antagonist/inhibitors include bispecific anti-VEGF/ANG2 antibodies as described in WO2014/009465.
  • An antigen-binding sites of the bispecific antibody of the invention contain six complementarity determining regions (CDRs) which contribute in varying degrees to the affinity of the binding site for antigen. There are three heavy chain variable domain CDRs (CDRH1, CDRH2 and CDRH3) and three light chain variable domain CDRs (CDRL1, CDRL2 and CDRL3). The extent of CDR and framework regions (FRs) is determined by comparison to a compiled database of amino acid sequences in which those regions have been defined according to variability among the sequences.
  • the antibodies of the invention comprise immunoglobulin constant regions derived from human origin of one or more immunoglobulin classes, wherein such immunoglobulin classes include IgG, IgM, IgA, IgD, and IgE classes and, in the case of IgG and IgA, their subclasses, especially IgG1 and IgG4.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition.
  • chimeric antibody refers to an antibody comprising a variable region, i.e., binding region, from one source or species and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising a murine variable region and a human constant region are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those in which the constant region has been modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding. Such chimeric antibodies are also referred to as “class-switched antibodies.”.
  • Chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA segments encoding immunoglobulin constant regions. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art. See, e.g., Morrison, S. L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; U.S. Pat. Nos. 5,202,238 and 5,204,244.
  • humanized antibody refers to antibodies in which the framework or “complementarity determining regions” (CDR) have been modified to comprise the CDR of an immunoglobulin of different specificity as compared to that of the parent immunoglobulin.
  • CDR complementarity determining regions
  • a murine CDR is grafted into the framework region of a human antibody to prepare the “humanized antibody.” See, e.g., Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M. S., et al., Nature 314 (1985) 268-270.
  • Particularly preferred CDRs correspond to those representing sequences recognizing the antigens noted above for chimeric antibodies.
  • humanized antibodies encompassed by the present invention are those in which the constant region has been additionally modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding.
  • FcR Fc receptor
  • human antibody is intended to include antibodies having variable and constant regions derived from human germ line immunoglobulin sequences.
  • Human antibodies are well-known in the state of the art (van Dijk, M. A., and van de Winkel, J. G., Curr. Opin. Chem. Biol. 5 (2001) 368-374).
  • Human antibodies can also be produced in transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire or a selection of human antibodies in the absence of endogenous immunoglobulin production.
  • Human antibodies can also be produced in phage display libraries (Hoogenboom, H. R., and Winter, G., J. Mol. Biol. 227 (1992) 381-388; Marks, J.
  • human antibody as used herein also comprises such antibodies which are modified in the constant region to generate the properties according to the invention, especially in regard to C1q binding and/or FcR binding, e.g. by “class switching” i.e. change or mutation of Fc parts (e.g. from IgG1 to IgG4 and/or IgG1/IgG4 mutation).
  • class switching i.e. change or mutation of Fc parts (e.g. from IgG1 to IgG4 and/or IgG1/IgG4 mutation).
  • recombinant antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NSO or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell.
  • recombinant antibodies have variable and constant regions in a rearranged form.
  • the recombinant antibodies according to the invention have been subjected to in vivo somatic hypermutation.
  • the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germ line VH and VL sequences, may not naturally exist within the human antibody germ line repertoire in vivo.
  • variable domain denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • the domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three “hypervariable regions” (or complementarity determining regions, CDRs).
  • the framework regions adopt a n-sheet conformation and the CDRs may form loops connecting the n-sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • the antibody heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • hypervariable region or “antigen-binding portion of an antibody” when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from the “complementarity determining regions” or “CDRs”.
  • “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. CDRs on each chain are separated by such framework amino acids. Especially, CDR3 of the heavy chain is the region which contributes most to antigen binding.
  • CDR and FR regions are determined according to the standard definition of Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991).
  • full length antibody denotes an antibody consisting of two “full length antibody heavy chains” and two “full length antibody light chains”.
  • a “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3), abbreviated as VH-CH1-HR-CH2-CH3; and optionally an antibody heavy chain constant domain 4 (CH4) in case of an antibody of the subclass IgE.
  • VH antibody heavy chain variable domain
  • CH1 antibody constant heavy chain domain 1
  • HR antibody hinge region
  • CH2 antibody heavy chain constant domain 2
  • CH3 antibody heavy chain constant domain 3
  • the “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of VH, CH1, HR, CH2 and CH3.
  • a “full length antibody light chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL), abbreviated as VL-CL.
  • the antibody light chain constant domain (CL) can be ⁇ (kappa) or ⁇ (lambda).
  • the two full length antibody chains are linked together via inter-polypeptide disulfide bonds between the CL domain and the CH1 domain and between the hinge regions of the full length antibody heavy chains. Examples of typical full length antibodies are natural antibodies like IgG (e.g.
  • the full length antibodies according to the invention can be from a single species e.g. human, or they can be chimerized or humanized antibodies.
  • the full length antibodies according to the invention comprise two antigen binding sites each formed by a pair of VH and VL, which both specifically bind to the same antigen.
  • the C-terminus of the heavy or light chain of said full length antibody denotes the last amino acid at the C-terminus of said heavy or light chain.
  • the N-terminus of the heavy or light chain of said full length antibody denotes the last amino acid at the N-terminus of said heavy or light chain.
  • constant region denotes the sum of the domains of an antibody other than the variable region.
  • the constant region is not involved directly in binding of an antigen, but exhibits various effector functions.
  • antibodies are divided in the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses, such as IgG1, IgG2, IgG3, and IgG4, IgA1 and IgA2.
  • the heavy chain constant regions that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the light chain constant regions which can be found in all five antibody classes are called ⁇ (kappa) and ⁇ (lambda).
  • constant region derived from human origin denotes a constant heavy chain region of a human antibody of the subclass IgG1, IgG2, IgG3, or IgG4 and/or a constant light chain kappa or lambda region.
  • constant regions are well known in the state of the art and e.g. described by Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991) (see also e.g. Johnson, G., and Wu, T. T., Nucleic Acids Res. 28 (2000) 214-218; Kabat, E.
  • EU numbering system according to Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991) is used and referred to as “numbering according to EU Index of Kabat”.
  • the bispecific antibodies according to the invention have a constant region of human IgG1 subclass (derived from human IgG1 subclass).
  • the C-terminal lysine (Lys447), or the C-terminal glycine (Gly446) and the C-terminal lysine (Lys447), of the Fc region may or may not be present.
  • the bispecific antibody as described herein is of IgG1 isotype/subclass and comprises a constant heavy chain domain of SEQ ID NO: 23 or the constant parts of the heavy chain amino acid sequence of SEQ ID NO: 17 and of the heavy chain amino acid sequence of SEQ ID NO: 18.
  • the C-terminal glycine (Gly446) is present.
  • the C-terminal glycine (Gly446) and the C-terminal lysine (Lys447) is present.
  • EU numbering system also called the EU index of Kabat, as described in Kabat, E. A. et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991), NIH Publication 91-3242.
  • the bispecific antibody according to the invention is of human IgG1 subclass with mutations L234A (Leu235Ala), L235A (Leu234Ala) and P329G (Pro329Gly).
  • Such antibody has a reduced FcR binding (especially they show no more binding to FcRgammaI, FcRgammaII and FcRgammaIII). This especially useful to reduce potential side effects like e.g. thrombosis (Meyer, T., et al., J. Thromb. Haemost. 7 (2009) 171-81).
  • Pro329Ala mutation which was described already removes only two third of the FcgammaRIIIa sandwich interaction
  • the Pro329Gly in the antibodies according to the invention fully imparts binding of the Fc part to FcgammaRIII. This is especially useful as the binding to FcgammaRIII is involved in ADCC (antibody-dependent cellular toxicity) which leads to cell death, which may be helpful in the treatment of cancer diseases, but which can cause serious side effect in the antibody based treatment of other vascular or immunological diseases.
  • ADCC antibody-dependent cellular toxicity
  • the antibodies according to the invention of IgG1 subclass with mutations L234A, L235A and P329G and IgG4 subclass with mutations S228P, L235E and P329G are especially useful, as they both show no more binding to FcRgammaI, FcRgammaII and FcRgammaIII.
  • an “effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • the bispecific antibody, medicament or pharmaceutical formulation as described herein is administered via intravitreal application, e.g. via intravitreal injection (is administered “intravitreally”).
  • intravitreal application e.g. via intravitreal injection (is administered “intravitreally”).
  • This can be performed in accordance with standard procedures known in the art. See, e.g., Ritter et al., J. Clin. Invest. 116 (2006) 3266-76; Russelakis-Carneiro et al., Neuropathol. Appl. Neurobiol. 25 (1999) 196-206; and Wray et al., Arch. Neurol. 33 (1976) 183-5.
  • therapeutic kits of the invention can contain one or more doses of the bispecific antibody described present in a medicament or pharmaceutical formulation, a suitable device for intravitreal injection of the medicament or pharmaceutical formulation, and an instruction detailing suitable subjects and protocols for carrying out the injection.
  • the medicament or pharmaceutical formulation are typically administered to the subject in need of treatment via intravitreal injection. This can be performed in accordance with standard procedures known in the art. See, e.g., Ritter et al., J. Clin. Invest. 116 (2006) 3266-76; Russelakis-Cameiro et al., Neuropathol. Appl. Neurobiol. 25 (1999) 196-206; and Wray et al., Arch. Neurol. 33 (1976) 183-5.
  • the bispecific antibody as described herein is formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Example 1A Efficacy and Durability of Treatment of Patients Suffering from Diabetic Macular Edema (DME)
  • this antibody VEGFang2-0016 and its production is also described in detail in WO2014/009465 which is incorporated by reference) compared with an active comparator in treatment na ⁇ ve patients with center-involving diabetic macular edema (CI-DME).
  • Designations of this bispecific anti-VEGF/ANG2 antibody herein are RO6867461 or RG7716 or VEGFang2-0016, or faricimab. Vials of sterile, colorless to brownish, preservative-free solution of RO6867461 for IVT administration of either 1.5 mg or 6 mg dose every 4 weeks were used. The concentration of the bispecific antibody was about 120 mg/ml.
  • Macular edema associated with DR defined as macular thickening by spectral domain optical coherence tomography (SD-OCT) involving the center of the macula: central subfield thickness (CST) of ⁇ 325 ⁇ m with SpectralisTM (Heidelberg) at screening (where SpectralisTM is not available, the following devices and CST thresholds were acceptable: CST ⁇ 315 ⁇ m for CirrusTM, CST ⁇ 315 ⁇ m for Topcon, CST ⁇ 295 ⁇ m for OptovueTM)
  • DM diabetes mellitus
  • ICH International Conference on Harmonisation
  • a legally authorized representative must be able to consent for the patient according to ICH and local regulations.
  • Abstinence is only acceptable if it is in line with the preferred and usual lifestyle of the patient. Periodic abstinence (e.g., calendar, ovulation, symptothermal, or postovulation methods) and withdrawal were not acceptable methods of contraception;
  • contraceptive methods with an expected failure rate of ⁇ 1% per year include male sterilization, hormonal implants, proper use of combined oral or injected hormonal contraceptives, and certain intrauterine devices.
  • two methods e.g., two barrier methods such as a condom and a cervical cap
  • barrier methods must always be supplemented with the use of a spermicide.
  • NVE ⁇ 1 ⁇ 2 disc area within an area equivalent to the standard mydriatic ETDRS 7-field on clinical examination
  • Uncontrolled glaucoma e.g., progressive loss of visual fields or defined as intraocular pressure [IOP] ⁇ 25 mmHg despite treatment with anti-glaucoma medication
  • any current or history of ocular disease other than DME that may confound assessment of the macula or affect central vision e.g., age-related macular degeneration, retinal vein occlusion, uveitis, angioid streaks, histoplasmosis, active or inactive cytomegalovirus, pathological myopia, retinal detachment, macular traction, macular hole, significant cataract
  • central vision e.g., age-related macular degeneration, retinal vein occlusion, uveitis, angioid streaks, histoplasmosis, active or inactive cytomegalovirus, pathological myopia, retinal detachment, macular traction, macular hole, significant cataract
  • Uncontrolled blood pressure BP; defined as systolic >180 mmHg and/or diastolic >100 mmHg while patient at rest). If a patient's initial reading exceeds these values, a second reading may be taken either 30 or more minutes later on the same day or on another day during the screening period. If the patient's BP needs to be controlled by antihypertensive medication, the patient should be taking the same medication continuously for at least 1 month prior to Day 1.
  • the total duration of the study was up to 40 weeks (from screening through study completion) for each enrolled patient as follows:
  • LPLO last patient last observation
  • the primary analysis population was treatment na ⁇ ve patients. Additional analyses may be performed in the overall population and in patients previously treated with IVT anti-VEGF.
  • the primary efficacy outcome measure for this study was the mean change in BCVA (ETDRS letters) from baseline at Week 24 in treatment-na ⁇ ve patients.
  • FFA fundus fluorescein angiography
  • the primary efficacy analyses included all randomized patients, with patients grouped according to the treatment assigned at randomization.
  • the primary efficacy variable was the BCVA change from baseline to Week 24.
  • the primary efficacy analysis was performed using a Mixed Model for Repeated Measurement (MMRM) model.
  • MMRM Mixed Model for Repeated Measurement
  • BCVA at a starting test distance of 4 meters was measured prior to dilating eyes by a trained and certified VA examiner masked to study drug arm assignment.
  • BCVA was measured by using the set of three Precision VisionTM or Lighthouse distance acuity charts (modified ETDRS Charts 1, 2, and R). A VA Manual was provided to the investigators. VA examiner and VA examination room certifications were obtained before any VA examinations were performed.
  • the BCVA examiner was masked to study eye and treatment assignment and will only perform the refraction and BCVA assessment (e.g. Visual Acuity Specification Manual).
  • the BCVA examiner has also been masked to the BCVA letter scores of a patient's previous visits and only knew the patient's refraction data from previous visits.
  • the BCVA examiner was not allowed to perform any other tasks involving direct patient care.
  • FIG. 1 displays the primary efficacy endpoint: BCVA change from Baseline over Time to Week 24 for so far treatment naive patients.
  • VA2 refers to the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose)
  • RBZ refers to ranibizumab (Lucentis®) (administered intravitreally with a 0.3 mg dose).
  • a key secondary endpoint was the change from baseline in CST, central subfield thickness. Results are shown in FIG. 2 .
  • the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose), was compared to ranibizumab (Lucentis®) (administered intravitreally with a 0.3 mg dose).
  • This secondary anatomical endpoint directionally supports BCVA primary outcome
  • BCVA and CST values obtained at week 24 were compared to those obtained at visit week 20. BCVA and CST values obtained at weeks 28, 32 and 36 were compared to those of week 24.
  • the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg or 1.5 mg dose), was compared to ranibizumab (Lucentis®) (administered intravitreally with a 0.3 mg dose).
  • FIG. 4 represents a schematic comparison to other treatment options of DME based on published results (The following agents are compared Lucentis® (ranibizumab), Eylea® (aflibercept), brolucizumab and VA2 (RO6867461/RG7716).
  • Example 1B Efficacy and Durability of Treatment of Patients Suffering from Diabetic Macular Edema (DME)
  • patients suffering from DME are treated with the bispecific antibody that binds to human VEGF and human ANG2 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20.
  • active comparator in treatment e.g. aflibercept and/or ranibizumab and/or brolicuzimab will be used.
  • Patients include anti-VEGF treatment-na ⁇ ve patients (have not been previously treated with anti-VEGF monotherapy with e.g. e.g.
  • aflibercept and/or ranibizumab and/or brolicuzimab) and also a group of patients which have been previously treated with anti-VEGF monotherapy.
  • Designations of the respective bispecific antibody that binds to human VEGF and human ANG2 are RO6867461 or RG7716. Vials of sterile, colorless to brownish, preservative-free solution of RO6867461 for IVT administration of either 1.5 mg or 6 mg dose are used.
  • the disease stability assessment would be based on best-corrected visual acuity (BCVA) and on CST as well as retinal thickness based on Optical coherence tomography (OCT). Outcome measure and results will be evaluated as described e.g. in Example 1A. Primary endpoints will be between 45 and 60 weeks.
  • patients suffering from DME are treatment na ⁇ ve (have not been previously treated with anti-VEGF monotherapy with e.g. aflibercept and/or ranibizumab and/or brolicuzimab)
  • patients suffering from DME have been previously treated with anti-VEGF monotherapy with e.g. aflibercept and/or ranibizumab and/or brolicuzimab.
  • patients suffering from DME will be treated with a fixed Q8W dosing schedule following treatment initiation (e.g. 6 initial monthly injections).
  • patients suffering from DME will be treated with a fixed Q12W dosing (in one embodiment with one cycle of Q8W dosing first), following treatment initiation (e.g. 6 initial monthly injections).
  • patients suffering from DME will be treated following treatment initiation (e.g. with 3-7 initial monthly injections) with a dosing regimen that extends the injection interval in stable absence of disease, or shortens the interval if there is disease activity.
  • a dosing regimen that extends the injection interval in stable absence of disease, or shortens the interval if there is disease activity.
  • such regimen includes that patient receive Q4w/Q8w/Q12w/Q16w dosing, dependent on their disease state.
  • patients suffering from AMD will be treated following treatment initiation (e.g. with 3-4 initial monthly injections) with a dosing regimen that extends the injection interval in stable absence of disease, or shortens the interval if there is disease activity.
  • a dosing regimen that extends the injection interval in stable absence of disease, or shortens the interval if there is disease activity.
  • such regimen includes that patient receive Q4W/Q8W/Q12W/Q16W dosing, dependent on their disease state.
  • Example 2A Efficacy and Durability of Treatment of Patients Suffering from Age-Related Macular Degeneration (AMD)
  • RO6867461 is a bispecific antibody that binds to human VEGF and human ANG2 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (this antibody VEGFang2-0016 and its production is also described in detail in WO2014/009465 which is incorporated by reference). Designations of this bispecific anti-VEGF/ANG2 antibody herein are RO6867461 or RG7716 or VEGFang2-0016 or faricimab.
  • ADA anti-drug antibody
  • Ang-2 angiopoietin-2
  • BCVA best corrected visual acuity
  • CFT central foveal thickness
  • CNV choroidal neovascularization
  • CST central subfield thickness
  • ETDRS Early Treatment Diabetic Retinopathy Study
  • FFA fundus fluorescein angiography
  • IVT intravitreal
  • PK pharmacokinetic
  • SD-OCT spectral domain optical coherence tomography
  • VEGF-A vascular endothelial growth factor A.
  • LPLV last patient last visit
  • RO6867461 Drug Product 120 mg/mL was provided as a sterile, colorless to brownish liquid and contains no preservatives. Vials of sterile, colorless to brownish, preservative-free solution of RO6867461 for IVT administration of 6 mg dose every were used. The concentration of the bispecific antibody was about 120 mg/ml.
  • BCVA must have been conducted before pupil dilation. At screening and Day 1 visits, BCVA could be performed before vital signs and blood sampling to avoid unnecessary investigations in those patients who may be a screen failure as a result of BCVA letter score.
  • IOP mandatory to be performed after all imaging assessments, and the same method should be used throughout the study period
  • BCVA at a starting test distance of 4 meters was measured prior to dilating eyes by a trained and certified visual acuity (VA) examiner masked to study eye treatment assignment.
  • BCVA was measured using the set of three Precision VisionTM or Lighthouse distance acuity charts (modified ETDRS Charts 1, 2, and R).
  • a VA Procedure Manual was provided to the investigators. VA examiner and VA examination room certifications were obtained before any VA examinations were performed.
  • the BCVA examiner was masked to the study eye and treatment assignment and will perform the refraction and BCVA assessments (e.g., VA Specification Manual).
  • the BCVA examiner was also masked to the BCVA letter scores of a patient's previous visits and may only know patient refraction data from previous visits.
  • IOP pressure was conducted prior to study treatment administration and 30 ( ⁇ 15) minutes post-treatment administration in the study eye, and if IOP ⁇ 30 mmHg, IOP should be re-assessed 30 ( ⁇ 15) minutes later. If IOP continued to be elevated, treatment was undertaken at the discretion of the investigator.
  • a post-treatment optic nerve head perfusion was assessed for each patient immediately after study treatment administration (maximum within 15 minutes after treatment administration) by testing finger count vision, hand motion, or light perception as appropriate.
  • the Central Reading Center provided sites with the Central Reading Center Manual and training materials for study-mandated ocular imaging. Before study images were obtained, site personnel and imaging systems (where applicable) was certified by the reading center as specified in the Central Reading Center Manual. All study subject ocular images were obtained only by trained and Central Reading Center certified personnel on certified/registered equipment at the study sites. A copy of all study subject ocular images were transferred to the central reading center for storage and for independent analysis, including for confirmation of eligibility of defined image-related criteria.
  • BCVA Best Corrected Visual Acuity
  • Durability of BCVA Gains Time to Treatment to Maintain BCVA Gain
  • RO6867461 refers to the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg dose either Q12W or Q16W), ranibizumab (Lucentis®) was administered intravitreally with a 0.3 mg dose Q4W.
  • the initial BCVA gains were fully maintained for the RO6867461 Q12W or Q16W groups and in a similar range as the ranibizumab (Lucentis®) Q4W group.
  • a key secondary endpoint was the change from baseline in CST, central subfield thickness. Results are shown in FIG. 7 .
  • the bispecific anti-VEGF/ANG2 antibody RO6867461 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20 (administered intravitreally with a 6.0 mg dose either Q12W or Q16W), was compared to ranibizumab (Lucentis®) (administered intravitreally with a 0.3 mg dose Q4W).
  • This secondary anatomical endpoint directionally supports BCVA primary outcome
  • Example 2B Efficacy and Durability of Treatment of Patients Suffering from Age-Related Macular Degeneration (AMD)
  • patients suffering from AMD are treated with the bispecific antibody that binds to human VEGF and human ANG2 comprising the amino acid sequences of SEQ ID NO: 17, of SEQ ID NO: 18, of SEQ ID NO: 19, and of SEQ ID NO: 20.
  • active comparator in treatment e.g aflibercept and/or ranibizumab and/or brolicuzimab will be used.
  • Patients include anti-VEGF treatment-na ⁇ ve patients (have not been previously treated with anti-VEGF monotherapy with e.g.
  • aflibercept and/or ranibizumab and/or brolicuzimab and also a group of patients which have been previously treated with anti-VEGF monotherapy with e.g. aflibercept and/or ranibizumab and/or brolicuzimab.
  • Designations of the respective bispecific antibody that binds to human VEGF and human ANG2 are RO6867461 or RG7716. Vials of sterile, colorless to brownish, preservative-free solution of RO6867461 for IVT administration of either 1.5 mg or 6 mg dose are used.
  • the disease stability assessment would be based on best-corrected visual acuity (BCVA) and on CST as well as retinal thickness based on Optical coherence tomography (OCT). Outcome measure and results will be evaluated as described e.g. in Example 1A. Primary endpoints will be between 45 and 60 weeks.
  • VEGF Isoforms Kinetic Affinity Including Assessment of Species-Crossreactivity
  • Solution affinity measures the affinity of an interaction by determining the concentration of free interaction partners in an equilibrium mixture.
  • Maximum possible resonance units e.g. 17000 resonance units (RU)
  • the sample and system buffer was HBS-P pH 7.4.
  • Flow cell was set to 25° C. and sample block to 12° C. and primed with running buffer twice.
  • the affinity was calculated by determining the inflection point of this curve.
  • the surface was regenerated by one time 30 sec washing with a 0.85% H3PO4 solution at a flow rate of 30 ⁇ l/min. Bulk refractive index differences were corrected by subtracting the response obtained from a blank-coupled surface. Results are shown in Table 6.
  • FcRn measurement a steady state affinity was used to compare bispecific antibodies against each other.
  • Human FcRn was diluted into coupling buffer (10 ⁇ g/ml, Na-Acetate pH5.0) and immobilized on a C1-Chip (GE Healthcare BR-1005-35) by targeted immobilization procedure using a BIAcoreTM wizard to a final response of 200 RU.
  • Flow cell was set to 25° C. and sample block to 12° C. and primed with running buffer twice.
  • the sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween® 20) pH 6.0.
  • a concentration of 62.5 nM, 125 nM and 250 nM, 500 nM was prepared.
  • Flow rate was set to 30 ⁇ l/min and the different samples were injected consecutively onto the chip surface choosing 180 sec association time.
  • the surface was regenerated by injected PBS-T pH 8 for 60 sec at a flow rate of 30 ⁇ l/min.
  • the method from the Bia-Evaluation software was used. Briefly, the RU values (RU max) were plotted against the analysed concentrations, yielding a dose-response curve.
  • FcgammaRIIIa measurement a direct binding assay was used. Around 3000 resonance units (RU) of the capturing system (1 ⁇ g/ml Penta-His; Qiagen) were coupled on a CMS chip (GE Healthcare BR-1005-30) at pH 5.0 by using an amine coupling kit supplied by the GE Healthcare.
  • the sample and system buffer was HBS-P+ pH 7.4.
  • the flow cell was set to 25° C.—and sample block to 12° C.—and primed with running buffer twice.
  • the FcgammaRIIIa-His-receptor was captured by injecting a 100 nM solution for 60 sec at a flow of 5 ⁇ l/min.
  • Binding was measured by injection of 100 nM of bispecific antibody or monospecific control antibodies (anti-Dig for IgG1 subclass and an IgG4 subclass antibody) for 180 sec at a flow of 30 ⁇ l/. The surface was regenerated by 120 sec washing with Glycine pH 2.5 solution at a flow rate of 30 ⁇ l/min. Because FcgammaRIIIa binding differs from the Langmuir 1:1 model, only binding/no binding was determined with this assay. In a similar manner FcgammaRIa, and FcgammaRIIa binding can be determined. Results are shown in FIG. 6 , where it follows that by introduction of the mutations P329G LALA no more binding to FcgammaRIIIa could be detected.
  • the bispecific antibody was captured by injecting a 10 nM solution for 60 sec at a flow of 5 ⁇ l/min. Independent binding of each ligand to the bispecific antibody was analysed by determining the active binding capacity for each ligand, either added sequentially or simultaneously (flow of 30 ⁇ l/min):
  • the surface was regenerated by 60 sec washing with a 3 mM MgCl2 solution at a flow rate of 30 ⁇ l/min. Bulk refractive index differences were corrected by subtracting the response obtained from a goat anti human IgG surface.
  • VEGF vascular endothelial growth factor
  • the binding response of hAng-2 depends from the amount of the bispecific antibody bound to VEGF and shows simultaneous binding.
  • the surface was regenerated by 60 sec washing with a 0.85% H3PO4 solution at a flow rate of 30 ⁇ l/min. Simultaneous binding is shown by an additional specific binding signal of hAng2 to the previous VEGF bound ⁇ VEGF-ANG-2> bispecific antibodies.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Ophthalmology & Optometry (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Endocrinology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US16/985,477 2018-02-06 2020-08-05 Treatment of ophthalmologic diseases Abandoned US20210139576A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/985,477 US20210139576A1 (en) 2018-02-06 2020-08-05 Treatment of ophthalmologic diseases
US18/166,996 US20230416353A1 (en) 2018-02-06 2023-02-09 Treatment of ophthalmologic diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862627103P 2018-02-06 2018-02-06
US201862729333P 2018-09-10 2018-09-10
PCT/EP2019/052704 WO2019154776A1 (en) 2018-02-06 2019-02-05 Treatment of ophthalmologic diseases
US16/985,477 US20210139576A1 (en) 2018-02-06 2020-08-05 Treatment of ophthalmologic diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/052704 Continuation WO2019154776A1 (en) 2018-02-06 2019-02-05 Treatment of ophthalmologic diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/166,996 Continuation US20230416353A1 (en) 2018-02-06 2023-02-09 Treatment of ophthalmologic diseases

Publications (1)

Publication Number Publication Date
US20210139576A1 true US20210139576A1 (en) 2021-05-13

Family

ID=65352014

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/985,477 Abandoned US20210139576A1 (en) 2018-02-06 2020-08-05 Treatment of ophthalmologic diseases
US18/166,996 Pending US20230416353A1 (en) 2018-02-06 2023-02-09 Treatment of ophthalmologic diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/166,996 Pending US20230416353A1 (en) 2018-02-06 2023-02-09 Treatment of ophthalmologic diseases

Country Status (12)

Country Link
US (2) US20210139576A1 (he)
EP (1) EP3749359A1 (he)
JP (3) JP7005772B2 (he)
KR (1) KR20200119274A (he)
CN (1) CN111699004A (he)
AU (1) AU2019219004A1 (he)
CA (1) CA3088355A1 (he)
IL (1) IL276383B2 (he)
MX (1) MX2020007527A (he)
SG (1) SG11202006712XA (he)
TW (1) TWI831764B (he)
WO (1) WO2019154776A1 (he)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
CN108712911A (zh) 2015-12-30 2018-10-26 科达制药股份有限公司 抗体及其缀合物
CN116059318A (zh) 2018-01-26 2023-05-05 加利福尼亚大学董事会 用于使用抗vegf剂治疗血管生成病症的方法和组合物
AU2020326243A1 (en) * 2019-08-06 2022-02-17 F. Hoffmann-La Roche Ag Personalized treatment of ophthalmologic diseases
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder
KR20220104756A (ko) 2019-11-25 2022-07-26 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 안구내 신생혈관형성을 위한 장기지속형 vegf 억제제
IL300616A (he) 2020-09-04 2023-04-01 Hoffmann La Roche נוגדן הנקשר ל-vegf-a ו-ang2 ושיטות שימוש
CN116234574A (zh) * 2020-09-17 2023-06-06 江苏恒瑞医药股份有限公司 特异性结合vegf和ang-2的双特异性抗原结合分子
JP2024512701A (ja) * 2021-03-30 2024-03-19 アブプロ コーポレーション 抗ang2×vegf多重特異性抗体を用いて脈絡膜新生血管形成を治療するための方法
CN117100845A (zh) * 2022-10-24 2023-11-24 景泽生物医药(合肥)股份有限公司 重组人奥克纤溶酶在制备治疗脉络膜血管疾病药物中的用途

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
EP1325932B9 (en) 1997-04-07 2006-07-19 Genentech, Inc. Anti-vegf antibodies
ES2246069T3 (es) 1997-05-02 2006-02-01 Genentech, Inc. Procedimiento de preparacion de anticuerpos multiespecificos que tienen componentes comunes y multimericos.
TWI671403B (zh) 2005-03-31 2019-09-11 中外製藥股份有限公司 控制組裝之多肽的製造方法
PT1999154E (pt) 2006-03-24 2013-01-24 Merck Patent Gmbh Domínios proteicos heterodiméricos modificados
WO2007147901A1 (en) 2006-06-22 2007-12-27 Novo Nordisk A/S Production of bispecific antibodies
US8242247B2 (en) 2007-12-21 2012-08-14 Hoffmann-La Roche Inc. Bivalent, bispecific antibodies
SI2235064T1 (sl) 2008-01-07 2016-04-29 Amgen Inc. Metoda za izdelavo heterodimernih molekul - protitelesa fc z uporabo elektrostatičnih usmerjevalnih učinkov
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
EP2424567B1 (en) 2009-04-27 2018-11-21 OncoMed Pharmaceuticals, Inc. Method for making heteromultimeric molecules
PT2519543T (pt) 2009-12-29 2016-10-07 Emergent Product Dev Seattle Proteínas de ligação de heterodímero e suas utilizações
TWI426920B (zh) 2010-03-26 2014-02-21 Hoffmann La Roche 雙專一性、雙價抗-vegf/抗-ang-2抗體
EP2569337A1 (en) 2010-05-14 2013-03-20 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
CN103429620B (zh) 2010-11-05 2018-03-06 酵活有限公司 在Fc结构域中具有突变的稳定异源二聚的抗体设计
EP4360709A3 (en) * 2011-01-13 2024-07-03 Regeneron Pharmaceuticals, Inc. Use of a vegf antagonist to treat angiogenic eye disorders
US9527925B2 (en) 2011-04-01 2016-12-27 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to VEGF and ANG2
EP2794905B1 (en) 2011-12-20 2020-04-01 MedImmune, LLC Modified polypeptides for bispecific antibody scaffolds
ES2743399T3 (es) 2012-04-20 2020-02-19 Merus Nv Métodos y medios para la producción de moléculas heterodiméricas similares a Ig
HUE056217T2 (hu) * 2012-07-13 2022-02-28 Roche Glycart Ag Bispecifikus anti-VEGF/anti-ANG-2 antitestek és ezek alkalmazása szemészeti érbetegségek kezelésében
KR102177437B1 (ko) * 2012-08-21 2020-11-12 옵코 파마슈티칼스, 엘엘씨 리포좀 제제
EP2992010B1 (en) * 2013-04-29 2021-03-24 F.Hoffmann-La Roche Ag Fc-receptor binding modified asymmetric antibodies and methods of use
KR20150063728A (ko) 2013-12-02 2015-06-10 삼성전자주식회사 항 VEGF-C/항 Ang2 이중 특이 항체
TWI761959B (zh) * 2014-11-07 2022-04-21 瑞士商諾華公司 治療眼部疾病之方法
US20160144025A1 (en) * 2014-11-25 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods and formulations for treating vascular eye diseases
CA2970315C (en) * 2014-12-11 2023-08-22 Bayer Healthcare Llc Use of anti-vegf agents to treat lesions in macular degeneration patients
AR103477A1 (es) * 2015-01-28 2017-05-10 Lilly Co Eli Compuestos de vegfa / ang2
US10654922B2 (en) * 2016-05-13 2020-05-19 Askgene Pharma Inc. Angiopoietin 2, VEGF dual antagonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Clinicaltrials.gov, NCT02484690 v15, Retrieved online: <URL:https://clinicaltrials.gov/ct2/history/NCT02484690?V_15=View#StudyPageTop> [retrieved on 08/04/2022], 24 June 2015. *
Clinicaltrials.gov, NCT0303880 v1, Retrieved online: <URL: https://clinicaltrials.gov/ct2/history/NCT03038880?V_1=View#StudyPageTop> [retrieved on 08/04/2022], January 31, 2017. *

Also Published As

Publication number Publication date
AU2019219004A1 (en) 2020-07-16
SG11202006712XA (en) 2020-08-28
JP2022046738A (ja) 2022-03-23
JP2023099094A (ja) 2023-07-11
KR20200119274A (ko) 2020-10-19
TWI831764B (zh) 2024-02-11
IL276383B1 (he) 2023-09-01
MX2020007527A (es) 2020-09-09
IL276383A (he) 2020-09-30
CN111699004A (zh) 2020-09-22
IL276383B2 (he) 2024-01-01
JP2021506947A (ja) 2021-02-22
TW202000700A (zh) 2020-01-01
EP3749359A1 (en) 2020-12-16
WO2019154776A1 (en) 2019-08-15
JP7005772B2 (ja) 2022-02-10
CA3088355A1 (en) 2019-08-15
US20230416353A1 (en) 2023-12-28
JP7273204B2 (ja) 2023-05-12

Similar Documents

Publication Publication Date Title
US20230416353A1 (en) Treatment of ophthalmologic diseases
JP6796184B2 (ja) 二重特異性抗vegf/抗ang−2抗体及び眼血管疾患の処置におけるそれらの使用
AU2012323849B2 (en) Treatment of ocular disease
US11180551B2 (en) Humanized monoclonal antibodies that target VE-PTP (HPTP-beta)
JP2017536414A (ja) 血管性眼疾患を処置するための方法および製剤
US20220162296A1 (en) Personalized treatment of ophthalmologic diseases
JP2020097567A (ja) 抗体製剤
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
TW202419464A (zh) 眼科疾病之治療
EP4028128A1 (en) Methods for treating ocular diseases
KR20240093772A (ko) 안과 질환의 치료
WO2024129468A1 (en) Methods for the treatment of noninfectious uveitis
NZ623275B2 (en) Treatment of ocular disease

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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