WO2022201084A1 - Methods for treating ocular diseases - Google Patents

Methods for treating ocular diseases Download PDF

Info

Publication number
WO2022201084A1
WO2022201084A1 PCT/IB2022/052692 IB2022052692W WO2022201084A1 WO 2022201084 A1 WO2022201084 A1 WO 2022201084A1 IB 2022052692 W IB2022052692 W IB 2022052692W WO 2022201084 A1 WO2022201084 A1 WO 2022201084A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
vegf antagonist
phrd
hrd
vhrd
Prior art date
Application number
PCT/IB2022/052692
Other languages
French (fr)
Inventor
Justis P. Ehlers
Kubra SARICI
Sunil Srivastava
Robert ZAHID
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Publication of WO2022201084A1 publication Critical patent/WO2022201084A1/en

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/16Ophthalmology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/50Determining the risk of developing a disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention relates to methods for treating ocular disease with a VEGF antagonist.
  • Age-related macular degeneration is the leading cause of severe vision loss in people affecting 10%-13% of individuals over the age of 65 in North America, Europe, and Australia (Kawasaki 2010, Rein et al., Arch Ophthalmol. 2009;127:533-40, Smith 2001). Genetic, environmental and health factors play an important role in the pathogenesis of the disease. AMD is classified into 2 clinical subtypes: the non-neovascular (atrophic) or dry form and the neovascular (exudative) or wet form (Ferris et al., Arch Ophthalmol. 1984;102:1640-2, Lim et al., Lancet. 2012;379:1728-38, Miller et al., Am J Ophthalmol.
  • Neovascular AMD is characterized by the growth of abnormal new blood vessels (neovascularization) under the retinal pigment epithelium (RPE) or subretinal space from the subjacent choroid, termed choroidal neovascularization (CNV) (Ferris et al., Arch Ophthalmol. 1984;102:1640-2). These newly formed vessels have an increased likelihood to leak blood and serum, damaging the retina by stimulating inflammation and scar tissue formation. This damage to the retina results in progressive, severe, and irreversible vision loss (Shah et al., Am J Ophthalmol. 2007;143:83-89, Shah et al., Am J Ophthalmol. 2009; 116: 1901-07).
  • VEGF anti-vascular endothelial growth factor
  • VEGF has been shown to be elevated in patients with nAMD and is thought to play a key role in the neovascularization process (Spilsbury et al., Am J Pathol. 2000;157:135-44).
  • IVT intravitreal pharmacotherapy targeting VEGF has significantly improved visual outcomes in patients with nAMD (Bloch et al., Am J Ophthalmol. 2012;153:209-13, Campbell et al., Arch Ophthalmol. 2012;130:794-5).
  • Anti-VEGF treatments such as ranibizumab (LUCENTIS ® ), aflibercept (EYLEA ® ), and brolucizumab (Beovu ® ), inhibit VEGF signaling pathways and have been shown to halt the growth of neovascular lesions and resolve retinal edema.
  • AEs ocular adverse events
  • IOI intraocular inflammation
  • Brolucizumab 6mg represents an important treatment option for patients with wet AMD, with an overall favorable benefit-risk profile.
  • FDA Food and Drug Administration
  • brolucizumab 6 mg for the treatment of nAMD by the Food and Drug Administration (FDA) in October 2019, there were reports of retinal vasculitis and/or retinal artery occlusion (RAO) accompanied by IOI with intravitreal injections of brolucizumab (Safety of Beovu® (brolucizumab). https://www.brolucizumab.info/ Last accessed on 19 November 2020. 2020; Witkin et al., J Vitreoretin Dis 2020;4(4):269-79).
  • IOI intraocular inflammation
  • SRC reported overall incidence of intraocular inflammation (IOI) of any form of 4.6% (50/1088), overall incidence of signs of retinal vasculitis of 3.3% (36/1088), overall incidence of concomitant signs of retinal vasculitis (RV) and retinal vascular occlusion (RO) of 2.1% (23/1088), and overall incidence of IOI of any form associated with losing 15 or more letters at the last visit / end of the study was 0.7% (8/1088) (Member Update: Novartis-Appointed Safety Review Committee Reports Initial Brolucizumab Findings. American Society of Retina Specialists. June 4, 2020; Mones et al., American Academy of Ophthalmology, 2020).
  • Intraocular inflammation is an adverse event that may lead to further complications if not identified, including severe vision loss.
  • II Intraocular inflammation
  • nAMD hyperreflective deposit
  • the post-hoc analysis identified that a formation of HRD, and in particular a formation of a HRD in vitreous compartment, e.g., preretinal, e.g., internal limiting membrane (ILM), precedes or is associated with intraocular inflammation (IOI) adverse events in nAMD patients.
  • the reported hyperreflective deposits were in a form of heterogeneity observed on the OCT scan including hyperreflective debris, hyperreflective round- or rod-shaped dots, e.g., stalagmite like preretinal deposits.
  • the stalagmite-like deposits are discrete HRD, e.g., PHRD, that resemble the elevated speleothems.
  • HRD e.g., VHRD, e.g., PHRD
  • stalagmite-like preretinal hyperreflective material may be used as a safety biomarker for the selection of the patients suitable for treatment with a VEGF inhibitor, e.g., brolucizumab, or for monitoring the patients during the course of treatment with a VEGF inhibitor, e.g., brolucizumab.
  • VEGF inhibitor e.g., brolucizumab
  • brolucizumab e.g., brolucizumab
  • Use of this new safety biomarker thus advances management of ophthalmic disorders such as nAMD in patients.
  • the present invention provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • the present invention provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
  • a VEGF antagonist e.g., brolucizumab
  • the present invention provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • VHRD e.g., PHRD
  • the present invention provides a method for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • VEGF antagonist e.g., brolucizumab
  • HRD e.g., VHRD, e.g., PHRD
  • VEGF antagonist e.g., brolucizumab
  • HRD e.g., VHRD, e.g., PHRD
  • the present invention provides a method of predicting the likelihood that a subject having ocular disease, e.g., nAMD, will develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • the method comprises determining by OCT or SD- OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, wherein:
  • the present invention provides a method of monitoring a subject during a course of treatment with a VEGF antagonist for presence of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, e.g., by performing OCT or SD-OCT analysis, and (i) altering or discontinuing the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined to have the HRD, e.g., VHRD, e.g., PHRD, in the treatment eye; or (ii) maintaining the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined not to have the HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
  • a VEGF antagonist for presence of a HRD in a subject’s eye e.g., VHRD, e.g., PHRD
  • a HRD in a subject’s eye e.
  • the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab
  • the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, characterized in that:
  • a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is administered to the subject.
  • the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, wherein the use comprises
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, characterized in that:
  • a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is administered to the subject.
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, characterized in that:
  • a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is administered to the subject.
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • the present invention provides, a method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • a VEGF antagonist e.g., brolucizumab
  • a HRD in a subject’s eye e.g., VHRD, e.g., PHRD;
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • the present invention provides, a method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., nAMD, may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • IOI intraocular inflammation
  • IOI+RV/RO retinal vasculitis/retinal occlusion
  • a VEGF antagonist e.g., brolucizumab
  • IOI+RV/RO a decreased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • Embodiment 1 A method of treating ocular disease comprising selectively administering a VEGF antagonist to a subject in need thereof on the basis of said subject not having a hyperreflective deposit (HRD), e.g., a vitreous hyperreflective deposit (VHRD), e.g., a preretinal hyperreflective deposit (PHRD).
  • HRD hyperreflective deposit
  • VHRD vitreous hyperreflective deposit
  • PHRD preretinal hyperreflective deposit
  • Embodiment 2 A method of treating ocular disease comprising:
  • Embodiment 3 A method of selectively treating a subject having ocular disease the method comprising:
  • a VEGF antagonist e.g., brolucizumab
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • HRD e.g., VHRD, e.g., PHRD
  • Embodiment 4 The method of embodiment 2 or 3 further comprising assessing the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, e.g., prior to the steps (a)-(b) of embodiment 2 or embodiment 3.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 5 The method of any one of embodiments 1 to 4, wherein the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, is assessed by performing OCT, e.g., SD- OCT, analysis on a subject’s eye.
  • HRD e.g., VHRD, e.g., PHRD
  • Embodiment 6 A method for determining whether a subject with ocular disease should be treated with a VEGF antagonist, the method comprising:
  • Embodiment 7 The method of embodiment 6 further comprising treating the subject with the VEGF antagonist if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
  • HRD e.g., VHRD, e.g., PHRD
  • Embodiment 8 The method of any one of the preceding embodiments, wherein the subject is assessed for presence of the HRD, e.g., VHRD, e.g., PHRD, prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 9 The method of embodiment 8, further comprising:
  • Embodiment 10 The method of any one of the preceding embodiments, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 11 The method of any one of the preceding embodiments, wherein the HRD, e.g., VHRD, e.g., PHRD, is hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
  • the HRD e.g., VHRD, e.g., PHRD
  • the HRD is hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
  • Embodiment 12 The method of any one of the preceding embodiments, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
  • the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
  • Embodiment 13 The method of any one of the preceding embodiments, wherein the VEGF antagonist is an anti -VEGF antibody or a fragment thereof.
  • Embodiment 14 The method of embodiment 13, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
  • Embodiment 15 The method of embodiment 13, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
  • Embodiment 16 The method of embodiment 13, wherein the VEGF antagonist is brolucizumab.
  • Embodiment 17 The method of any one of the preceding embodiments, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4- week intervals, and thereafter administering to the subject an additional dose every 8 to 12 weeks.
  • Embodiment 18 The method of embodiment 17, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 12 weeks.
  • Embodiment 19 The method of embodiment 18, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
  • Embodiment 20 The method of embodiment 15, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 weeks.
  • Embodiment 21 The method of embodiment 20, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
  • Embodiment 22 The method of any one of embodiments 14 to 21, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg.
  • Embodiment 23 The method of embodiment 22, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
  • Embodiment 24 The method of any one of the preceding embodiments, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
  • CNV choroidal neovascular
  • Embodiment 25 The method of embodiment 24, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
  • Embodiment 26 A method of predicting the likelihood that a subject having ocular disease will develop intraocular inflammation (IO I), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD , in particular the method comprising determining by OCT or SD-OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, wherein:
  • a HRD e.g., VHRD, e.g., PHRD
  • IOI+RV/RO e.g., IOI+RV/RO
  • Embodiment 27 The method of embodiment 26 further comprising treating the subject with the VEGF antagonist if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
  • HRD e.g., VHRD, e.g., PHRD
  • Embodiment 28 The method of any one of embodiments 26 to 27, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 29 The method of any one of embodiments 26 to 27, wherein the HRD, e.g., VHRD, e.g., PHRD, is a hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
  • the HRD e.g., VHRD, e.g., PHRD
  • the HRD e.g., VHRD, e.g., PHRD
  • stalagmite-like hyperreflective material e.g., stalagmite-like preretinal hyperreflective material.
  • Embodiment 30 The method of any one of embodiments 26 to 29, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
  • the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
  • Embodiment 31 The method of any one of embodiments 26 to 29, wherein the VEGF antagonist is an anti -VEGF antibody or a fragment thereof.
  • Embodiment 32 The method of embodiment 31, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
  • Embodiment 33 The method of embodiment 31, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
  • Embodiment 34 The method of embodiment 31, wherein the VEGF antagonist is brolucizumab.
  • Embodiment 35 The method of any one of embodiments 32 to 34, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4- week intervals, and thereafter administering to the subject an additional dose every 8 to 12 weeks.
  • Embodiment 36 The method of embodiment 35, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 12 weeks.
  • Embodiment 37 The method of embodiment 36, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
  • Embodiment 38 The method of embodiment 35, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 weeks.
  • Embodiment 39 The method of embodiment 38, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
  • Embodiment 40 The method of any one of embodiments 32 to 39, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg.
  • Embodiment 41 The method of embodiment 40, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
  • Embodiment 42 The method of any one of embodiments 26 to 41, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
  • CNV choroidal neovascular
  • Embodiment 43 The method of embodiment 42, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
  • nAMD neovascular age-related macular degeneration
  • Embodiment 44 A VEGF antagonist for the use in treatment of ocular disease in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e g., VHRD, e g., PHRD.
  • a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e g., VHRD, e g., PHRD.
  • Embodiment 45 A VEGF antagonist for the use in treatment of ocular disease in a subject, characterized in that:
  • a subject is selected for treatment with the VEGF antagonist on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • a therapeutically effective amount of the VEGF antagonist is administered to the subject.
  • Embodiment 46 A VEGF antagonist for the use in treatment of ocular disease in a subject, wherein the use comprises
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • Embodiment 47 The VEGF antagonist for use of embodiment 45 or 46, wherein the use further comprises assessing the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, e.g., prior to the steps (a)-(b) of embodiment 45 or embodiment 46.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 48 The VEGF antagonist for use of any one of embodiments 44 to 47, wherein the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, is assessed by performing OCT, e.g., SD-OCT, analysis on a subject’s eye.
  • HRD e.g., VHRD, e.g., PHRD
  • Embodiment 49 A VEGF antagonist for the use in treatment of ocular disease in a subject, wherein the use comprises:
  • a therapeutically effective amount of the VEGF antagonist e.g., brolucizumab, is to be administered to the subject;
  • a therapeutically effective amount of a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • Embodiment 50 The VEGF antagonist for use of embodiment 49, wherein a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • Embodiment 51 The VEGF antagonist for use of any one of embodiments 44 to 50, wherein the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist.
  • Embodiment 52 The VEGF antagonist for use of embodiment 51, wherein the use further comprises:
  • Embodiment 53 The VEGF antagonist for use of any one of embodiments 44 to 52, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
  • the HRD e.g., VHRD, e.g., PHRD
  • Embodiment 54 The VEGF antagonist for use of any one of embodiments 44 to 53, wherein the HRD, e.g., VHRD, e.g., PHRD, is a hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
  • the HRD e.g., VHRD, e.g., PHRD
  • the HRD e.g., VHRD, e.g., PHRD
  • stalagmite-like hyperreflective material e.g., stalagmite-like preretinal hyperreflective material.
  • Embodiment 55 The VEGF antagonist for use of any one of embodiments 44 to 54, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
  • Embodiment 56 The VEGF antagonist for use of any one of embodiments 44 to 55, wherein the VEGF antagonist is an anti-VEGF antibody or a fragment thereof.
  • Embodiment 57 The VEGF antagonist for use of embodiment 56, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
  • Embodiment 58 The VEGF antagonist for use of embodiment 56, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
  • Embodiment 59 The VEGF antagonist for use of embodiment 56, wherein the VEGF antagonist is brolucizumab.
  • Embodiment 60 The VEGF antagonist for use of any one of embodiments 44 to 59, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 8 to 12 weeks.
  • Embodiment 61 The VEGF antagonist for use of embodiment 60, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 12 weeks.
  • Embodiment 62 The VEGF antagonist for use of embodiment 61, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
  • Embodiment 63 The VEGF antagonist for use of embodiment 60, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 8 weeks.
  • Embodiment 64 The VEGF antagonist for use of embodiment 63, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
  • Embodiment 65 The VEGF antagonist for use of any one of embodiments 51 to 58, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg.
  • Embodiment 66 The VEGF antagonist for use of embodiment 65, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
  • Embodiment 67 The VEGF antagonist for use of any one of embodiments 44 to 66, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
  • CNV
  • Embodiment 68 The VEGF antagonist for use of embodiment 67, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
  • nAMD neovascular age-related macular degeneration
  • Embodiment 69 A method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., neovascular age-related macular degeneration (nAMD), should be treated with a VEGF antagonist, the method comprising:
  • a VEGF antagonist e.g., brolucizumab
  • a HRD in a subject’s eye e.g., VHRD, e.g., PHRD;
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
  • Embodiment 70 A method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., neovascular age-related macular degeneration (nAMD), may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • IOI intraocular inflammation
  • IOI+RV/RO retinal vasculitis/retinal occlusion
  • a VEGF antagonist e.g., brolucizumab
  • FIG. Preretinal hyperreflective material.
  • A Baseline OCT, smooth inner retinal boundary.
  • B Pre-IOI event OCT, new appearance of preretinal hyperreflective material (arrow).
  • FIG. 1 Preretinal hyperreflective material.
  • A Baseline OCT: smooth inner retinal boundary; no preretinal hyperreflective material or vitreous debris observed .
  • B V3/Pre-IOI event OCT: early signs of preretinal hyperreflective material (foci) / vitreous debris.
  • C V7/IOI event OCT: vitreous debris with preretinal hyperreflective material (foci).
  • D V3/Pre-IOI event OCT: numerous preretinal hyperreflective material (foci).
  • Figure 5 Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective foci between eyes in the IOI group and the control group, including prior to the IOI reported event.
  • Figure 6 Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective preretinal foci (preretinal deposits) between eyes in the IOI group and the control group, including prior to the IOI reported event.
  • Figure 7 Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective vitreous foci (vitreous debris) between eyes in the IOI group and the control group, including prior to the IOI reported event. DETAILED DESCRIPTION
  • the term “about” includes and describes the value or parameter per se.
  • “about x” includes and describes “x” per se.
  • the term “about” when used in association with a measurement, or used to modify a value, a unit, a constant, or a range of values refers to variations of ⁇ 1-10% in addition to including the value or parameter per se.
  • the term “about” when used in association with a measurement, or used to modify a value, a unit, a constant, or a range of values refers to variations of ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, or ⁇ 10%.
  • VEGF refers to the 165-amino acid vascular endothelial cell growth factor, and related 121-, 189-, and 206-amino acid vascular endothelial cell growth factors, as described by Leung et ah, Science 246:1306 (1989), and Houck et ah, Mol. Endocrin. 5:1806 (1991) together with the naturally occurring allelic and processed forms of those growth factors.
  • VEGF in particular, refers to the human VEGF.
  • VEGF receptor refers to a cellular receptor for VEGF, ordinarily a cell-surface receptor found on vascular endothelial cells, as well as variants thereof retaining the ability to bind hVEGF.
  • a VEGF receptor is the fms-like tyrosine kinase (fit), a transmembrane receptor in the tyrosine kinase family. DeVries et ah, Science 255:989 (1992); Shibuya et al., Oncogene 5:519 (1990).
  • the fit receptor comprises an extracellular domain, a transmembrane domain, and an intracellular domain with tyrosine kinase activity.
  • VEGF receptor Another example of a VEGF receptor is the flk-1 receptor (also referred to as KDR).
  • KDR flk-1 receptor
  • Binding of VEGF to the fit receptor results in the formation of at least two high molecular weight complexes, having an apparent molecular weight of 205,000 and 300,000 Daltons. The 300,000 Dalton complex is believed to be a dimer comprising two receptor molecules bound to a single molecule of VEGF.
  • a “VEGF antagonist” refers to a compound that can diminish or inhibit VEGF activity in vivo.
  • a VEGF antagonist can bind to a VEGF receptor(s) or block VEGF protein(s) from binding to VEGF receptor(s).
  • a VEGF antagonist can be, for example, a small molecule, an anti-VEGF antibody or antigen-binding fragments thereof, fusion protein (such as aflibercept or other such soluble decoy receptor), an aptamer, an antisense nucleic acid molecule, an interfering RNA, receptor proteins, and the like that can bind specifically to one or more VEGF proteins or one or more VEGF receptors.
  • the VEGF antagonist is any licensed anti-VEGF drug such as brolucizumab, ranibizumab or aflibercept.
  • the VEGF antagonist is an anti-VEGF antibody (such as brolucizumab or ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF
  • antibody as used herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”, “antigen binding polypeptide”, or “immunobinder”) or single chain thereof.
  • An “antibody” includes a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • CL complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the structures and locations of antibody variable domains may be defined using well known numbering schemes, e.g., the Rabat numbering scheme, the Chothia numbering scheme, or a combination of Rabat and Chothia (see, e.g., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services (1991), eds. Rabat et ah; A1 Lazikani et ah, (1997) J. Mol. Bio. 273:927 948); Rabat et al., (1991) Sequences of Proteins of Immunological Interest, 5th edit., NIH Publication no. 91-3242 U.S.
  • the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1, also referred to as CDRHl), 50-65 (HCDR2, also referred to as CDRH2), and 95-102 (HCDR3, also referred to as CDRH3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1, also referred to as CDRLl), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3).
  • CDR amino acids in the VH are numbered 26-32 (HCDR1, also referred to as CDRHl), 52-56 (HCDR2, also referred to as CDRH2), and 95-102 (HCDR3, also referred to as CDRH3); and the amino acid residues in VL are numbered 24-34 (LCDR1, also referred to as CDRLl), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3).
  • the CDRs consist of amino acid residues 26-35 (HCDR1, also referred to as CDRHl), 50-65 (HCDR2, also referred to as CDRH2), and 95- 102 (HCDR3, also referred to as CDRH3) in human VH and amino acid residues 24- 34 (LCDR1, also referred to as CDRL1), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3) in human VL.
  • HCDR1 amino acid residues 26-35
  • CDR2 also referred to as CDRH2
  • CDR3 95- 102
  • the CDR amino acid residues in the VH are numbered approximately 26-35 (HCDR1, also referred to as CDRH1), 51-57 (HCDR2, also referred to as CDRH2) and 93-102 (HCDR3, also referred to as CDRH3), and the CDR amino acid residues in the VL are numbered approximately 27-32 (LCDR1, also referred to as CDRLl), 50-52 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3) (numbering according to "Rabat").
  • the CDRs of an antibody can be determined using the program IMGT/DomainGap Align.
  • CDRs In the context of the present invention, the numbering system suggested by Honegger and Pluckthun (“AHo) is used (Honegger and Pluckthun, J. Mol. Biol. 309 (2001) 657-670), unless specifically mentioned otherwise. Furthermore, the following residues are defined as CDRs according to AHo numbering scheme: LCDR1 (also referred to as CDRLl): L24-L42; LCDR2 (also referred to as CDRL2): L58-L72; LCDR3 (also referred to as CDRL3): L107- L138; HCDR1 (also referred to as CDRHl): H27-H42; HCDR2 (also referred to as CDRH2): H57-H76; HCDR3 (also referred to as CDRH3): H108-H138.
  • LCDR1 also referred to as CDRLl
  • LCDR2 also referred to as CDRL2
  • LCDR3 also referred to as CDRL3
  • HCDR1 also referred
  • variable domain usually not all positions 1 to 149 will be occupied by an amino acid residue.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g ., effector cells) and the first component (Clq) of the classical complement system.
  • single chain antibody single chain Fv or single chain Fv
  • scFv single chain Fv
  • scFv molecules can have the general structures: NH2-VL-linker-VH-COOH or NH2-VH-linker- VL-COOH.
  • antigen-binding portion of an antibody (or simply “antibody portion” or “antibody fragment”) refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., VEGF).
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a single domain or dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs which may optionally be joined by
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
  • Antibodies can be of different isotype, for example, an IgG (e.g., an IgGl, IgG2, IgG3, or IgG4 subtype), IgAl, IgA2, IgD, IgE, or IgM antibody.
  • homologous refers to the subunit sequence identity between two polymeric molecules, e.g., between two nucleic acid molecules, such as, two DNA molecules or two RNA molecules, or between two polypeptide molecules.
  • two nucleic acid molecules such as, two DNA molecules or two RNA molecules
  • polypeptide molecules between two polypeptide molecules.
  • a subunit position in both of the two molecules is occupied by the same monomeric subunit; e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous or identical at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions; e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two sequences are homologous, the two sequences are 50% homologous; if 90% of the positions (e.g., 9 of 10), are matched or homologous, the two sequences are 90% homologous.
  • Percentage of “sequence identity” can be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of the amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage can be calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity.
  • the output is the percent identity of the subject sequence with respect to the query sequence.
  • a “mammal” includes any animal classified as a mammal, including, but not limited to, humans, domestic animals, farm animals, and companion animals, etc.
  • a subject or patient refers to human and non-human mammals, including but, not limited to, primates, pigs, horses, dogs, cats, sheep, and cows.
  • a subject or patient is a human.
  • a patient is at least 18 years of age.
  • a patient is at least 50 years of age.
  • a patient is at least 65 years of age.
  • treat includes therapeutic treatments, prophylactic treatments and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses the reduction of the symptoms or underlying risk factors.
  • the terms “treat”, “treatment” and “treating” refer to the reduction or amelioration of the progression or severity of an ocular disease or the amelioration of one or more symptoms, suitably of one or more discernible symptoms of an ocular disease.
  • the terms “treat”, “treatment” and “treating” refer to the amelioration of at least one measurable physical parameter of an ocular disease (such as achieve or at least partially achieve a desired effect (e.g.
  • retinal fluid-free status e.g., intraretinal fluid (IRF) and subretinal fluid (SRF), decrease of Central Subfield Thickness (CSFT), improvement in vision, e.g., a change of BCVA > 1, > 2, > 3, > 4 or > 5 letters, or a DRSS score ⁇ 61), wherein the physical parameter is not necessarily discernible by the patient.
  • IRF intraretinal fluid
  • SRF subretinal fluid
  • CSFT Central Subfield Thickness
  • improvement in vision e.g., a change of BCVA > 1, > 2, > 3, > 4 or > 5 letters, or a DRSS score ⁇ 61
  • ocular disease refers to a condition, disease, or disorder associated with ocular neovascularization.
  • An “ocular disease” or “neovascular ocular disease” that can be treated using a method of the disclosure includes, a condition, disease, or disorder associated with ocular neovascularization, including, but not limited to, abnormal angiogenesis, choroidal neovascularization (CNV), choroidal neovascularization (CNV) associated with nAMD, retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), sequela associated with retinal ischemia, Retinal Vein O
  • loading phase refers to the first 2, 3, 4, 5, or 6 doses of a VEGF antagonist administered at q4w or q6w intervals.
  • loading phase refers to the first 2 to 3 doses of a VEGF antagonist administered at q4w.
  • maintenance phase refers to additional doses at 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 week intervals, and can be adjusted as described herein based on Disease Activity Assessments as described herein.
  • maintenance phase refers to additional doses at 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 week intervals, and can be adjusted as described herein based on Disease Activity Assessments as described herein.
  • an administration interval can be referred to as qXw, where the “X” is a number of weeks between administered doses.
  • q6w is an interval of 6 weeks.
  • the term “week” means 7 days ⁇ 1 day.
  • the term “month” means 25 to 31 days.
  • the term “month” means 4 weeks.
  • the terms “effective amount” or “therapeutically effective amount” or “therapeutically effective dose” refer to an amount of a therapy (e.g., a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition provided herein) which is sufficient to reduce and/or ameliorate the severity of a given condition, disorder, or disease and/or a symptom related thereto.
  • a therapy e.g., a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition provided herein
  • the term “effective amount” or “therapeutically effective amount” or “therapeutically effective dose” is defined as an amount sufficient to achieve or at least partially achieve a desired effect (e.g.
  • a therapeutically effective dose is sufficient if it can produce even an incremental change in the symptoms or conditions associated with the disease.
  • the therapeutically effective dose does not have to completely cure the disease or completely eliminate symptoms.
  • a therapeutically effective dose can at least partially arrest the disease and/or its complications in a patient already suffering from the disease.
  • a therapeutic effective dose may involve repeated administration over a period of time. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient’s own immune system.
  • adverse event refers to any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal (investigational) product, whether or not related to the medicinal (investigational) product.
  • adverse events in ocular disease e.g., nAMD
  • patients may include blurred visiona, cataract, conjunctival hemmorage, eye pain, vitreous floaters, intraocular inflammation, e.g., e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO).
  • the term refers to the effect that leads to intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion.
  • the term “intraocular inflammation” encompasses anterior chamber cell, anterior chamber flare, anterior chamber inflammation, chorioretinitis, eye inflammation, iridocyclitis, ulceris, uveitis, vitreous haze, vitritis.
  • the term “hyperreflective deposit” or “HRD” or“hyperreflective material” or “hyperreflective foci” or “hyperreflective foci formation” refers to a region of increased reflectivity on the tomographic scan, e.g., on spectral domain optical coherence tomography (SD-OCT).
  • Hyperreflective material may be classified according to its morphological appearance and location; hyperreflective material (hyperreflective deposit) with high reflectivity whose boundaries are clearly delineated from the surrounding neural components of the retina is classified as well-defined; hyperreflective material (hyperreflective deposit) with low reflectivity and whose borders are less well defined and therefore not easily distinguishable from surrounding neural components is classified as undefined (Casalino et al., Expert Review of Ophthalmology, 2020, Volume 15, No. 2, pp. 83-91).
  • a “hyperreflective deposit” or “HRD” or “hyperreflective material” or “hyperreflective foci formation” is a well-defined (with high reflectivity) hyperreflective material. Morphological appearance of hyperreflective material (hyperreflective deposit) includes a heterogeneity observed on the OCT scan including hyperreflective debris, hyperreflective round- or rod-shaped dots, e.g., stalagmite-like deposit or material.
  • stalagmite-like hyperreflective deposit or “stalagmite like hyperreflective material” or “stalagmite-like hyperreflective foci formation” refers to hyperreflective material that resemble the elevated speleothems, in particular as determined on spectral domain optical coherence tomography (SD-OCT).
  • SD-OCT spectral domain optical coherence tomography
  • the HRD including debris, round- or rod-shaped dots and stalagmite-like HRD, is observed in vitreous compartment - a vitreous hyperreflective deposit (VHRD).
  • the HRD including stalagmite-like hyperreflective material
  • PHRD preretinal hyperreflective deposit
  • ILM internal limiting membrane hyperreflective deposit
  • “likelihood” and “likely” is a measurement of how probable an event is to occur. It may be used interchangeably with “probability”. Likelihood refers to a probability that is more than speculation, but less than certainty. Thus, an event is likely if a reasonable person using common sense, training or experience concludes that, given the circumstances, an event is probable. In some embodiments, once likelihood has been ascertained, the patient may be treated (or treatment continued) with the compound or the treatment may need to be altered or discontinued. In one embodiment, the “likelihood” and “likely” denote a chance in percent of how probable an event is to occur. The phrase “increased likelihood” refers to an increase in the probability that an event will occur.
  • some methods herein allow prediction of whether a patient will display an increased likelihood of developing intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion, in response to the administration of a VEGF antagonist, e.g., brolucizumab .
  • the increased likelihood means that there is more than 50% chance, more than 60 % chance, more than 70 % or more than 80 % chance that an event will occur.
  • a “decreased likelihood” means, that the chance is lower than 50%, lower than 60 %, lower than 70 % or lower than 80 %, respectively, that an event will occur.
  • predicting indicates that the methods described herein provide information to enable a health care provider to determine the likelihood that an individual subjected to a treatment will develop with a higher likelihood intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion. It does not refer to the ability to predict response with 100% accuracy. Instead, the skilled artisan will understand that it refers to an increased probability.
  • the disclosure provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having or having a low level of a hyperreflective deposit (HRD) in a subject’s eye, e.g., a vitreous hyperreflective deposit (VHRD), e.g., a preretinal hyperreflective deposit (PHRD), e.g., internal limiting membrane (ILM) hyperreflective deposit (ILMHRD).
  • a VEGF antagonist e.g., brolucizumab
  • the disclosure provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having or having a low level of a preretinal hyperreflective deposit (PHRD), e.g., internal limiting membrane (ILM) hyperreflective deposit (ILMHRD).
  • a VEGF antagonist e.g., brolucizumab
  • the disclosure provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
  • a VEGF antagonist e.g., brolucizumab
  • the disclosure provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a PHRD or having a low level of a PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
  • a VEGF antagonist e.g., brolucizumab
  • the disclosure n provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.
  • the disclosure n provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a PHRD or having a low level of a PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • the method further comprises assessing the presence or absence of HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • the assessment for the presence or absence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD is performed prior to the steps (a)-(b).
  • the HRD in a subject’s eye e.g., VHRD, e.g., PHRD, e.g., presence or absence or level
  • VHRD e.g., PHRD
  • optical coherence tomography e.g., spectral-domain optical coherence tomography, SD-OCT, analysis on a subject’s eye.
  • the disclosure provides a method for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • a VEGF antagonist e.g., brolucizumab
  • a VEGF antagonist e.g., brolucizumab
  • a VEGF antagonist e.g., brolucizumab
  • a HRD in a subject’s eye e.g., VHRD, preferably PHRD
  • a high level of a HRD in a subject’s eye e.g., VHRD, preferably PHRD
  • the present invention provides a method of predicting the likelihood that a subject having ocular disease, e.g., nAMD, will develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, wherein: (a) the presence of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or the presence of a high level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is indicative of an increased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in
  • the method further comprises treating the subject with the VEGF antagonist if no HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or a low level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is observed in the subject’s eye.
  • the method comprises determining by OCT or SD-OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, preferably PHRD.
  • the subject may be assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist.
  • the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist.
  • the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and prior to an administration of the VEGF antagonist, e.g., on the same day and prior to the administration or 1, 2, 3, 4, 5, 6, or 7 days prior to the administration of the VEGF antagonist or 1, 2, 3, 4, 5, 6, or 7 weeks prior the administration of the VEGF antagonist.
  • VHRD e.g., PHRD
  • the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and after an administration of the VEGF antagonist, e.g., on the same day and after the administration or on the 1 st , 2 nd , 3 rd , 4 th , 5 th , 6 th , or 7 th day after the administration of the VEGF antagonist or 1, 2, 3, 4, 5, 6, or 7 weeks after the administration of the VEGF antagonist.
  • VHRD e.g., PHRD
  • the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and one month after the initial dose of the VEGF antagonist was administered to the subject.
  • a subject’s eye e.g., VHRD, e.g., PHRD
  • a HRD in a subject’s eye e.g., VHRD, e.g., PHRD
  • VHRD e.g., PHRD
  • a VEGF inhibitor e.g., brolucizumab
  • a VEGF inhibitor e.g., brolucizumab
  • the treatment e.g., treatment regimen (frequency and /or amount), choice of medicament
  • the method of the present disclosure further comprises: (a) altering or discontinuing the treatment with the VEGF antagonist, e.g., brolucizumab, if, during the course of treatment, the subject is determined to have a HRD, e.g., VHRD, e.g., PHRD, or a high level of a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye, or (b) maintaining the treatment with the VEGF antagonist, e.g., brolucizumab, if, during the course of treatment, the subject is determined not to have a HRD, e.g., VHRD, e.g., PHRD, or a low level of a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
  • a HRD e.g., VHRD, e.g., PHRD
  • a high level of a HRD
  • the HRD e.g., VHRD, e.g., PHRD
  • the HRD has enough reflectivity to be determined as heterogeneity in comparison to the baseline level or in comparison to the surrounding components of the retina.
  • the HRD, e.g., VHRD, e.g., PHRD suitable for the methods of present invention has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
  • the HRD, e.g., VHRD, e.g., PHRD suitable for the methods of present invention is as a hyperreflective debris or a hyperreflective round- or rod-shaped dot.
  • the HRD e.g., VHRD, e.g., PHRD
  • suitable for the methods of present invention is stalagmite-like hyperreflective material, e.g., stalagmite like preretinal hyperreflective material.
  • the HRD is PHRD having high reflectivity and is round- or rod-shaped dot, e.g., stalagmite-like.
  • the PHRD has high reflectivity and is stalagmite-like.
  • the methods of the present disclosure are suitable for treating ocular disease or for predicting the likelihood that a subject having ocular disease will develop intraocular inflammation (IOI), wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein o
  • ocular disease is nAMD.
  • Age-related macular degeneration is the leading cause of severe vision loss in people affecting 10%-13% of individuals over the age of 65 in North America, Europe, and Australia (Kawasaki 2010, Rein et ah,
  • AMD Arch Ophthalmol. 2009;127:533-40, Smith 2001). Genetic, environmental and health factors play an important role in the pathogenesis of the disease. AMD is classified into 2 clinical subtypes: the non-neovascular (atrophic) or dry form and the neovascular (exudative) or wet form (Ferris et ah, Arch Ophthalmol. 1984;102:1640-2, Lim et ah, Lancet. 2012;379:1728- 38, Miller et ah, Am J Ophthalmol. 2013;155:1-35).
  • VEGF has been shown to be elevated in patients with nAMD and is thought to play a key role in the neovascularization process (Spilsbury et ak, Am J Pathol. 2000;157: 135-44).
  • IVT intravitreal pharmacotherapy targeting VEGF has significantly improved visual outcomes in patients with nAMD (Bloch et ak, Am J Ophthalmol. 2012;153:209-13, Campbell et ak, Arch Ophthalmol. 2012;130:794-5).
  • Anti-VEGF treatments such as ranibizumab (LUCENTIS ® ), aflibercept (EYLEA ® ), and brolucizumab (Beovu ® ), inhibit VEGF signaling pathways and have been shown to halt the growth of neovascular lesions and resolve retinal edema.
  • DM Diabetes mellitus
  • DR Diabetic retinopathy
  • DME diabetic macular edema
  • VA visual acuity
  • ocular disease is DME.
  • DME is a frequent manifestation of DR (Riordan-Eva, 2004, Eye (Lond). 2004, 18:1161-8) and is the major cause of visual loss in patients with DR.
  • DR Raordan-Eva
  • a favorable benefit risk ratio was demonstrated with superior efficacy versus the previous standard of care (laser photocoagulation) in large Phase 3 programs that consequently led to their approval for the treatment of DME.
  • Anti- VEGF treatment led to clinically relevant improvements of BCVA, reduction of fluid accumulation and decreased severity of diabetic retinopathy.
  • ocular disease is DR.
  • DR is the most common cause of vision loss among people with diabetes and the leading cause of vision impairment and blindness in working aged adults. DR occurs when high blood glucose levels cause damage to blood vessels in the retina. These blood vessels can swell and leak or they can close, stopping blood from passing through. Sometimes abnormal new blood vessels can also grow on the retina.
  • Diabetic retinopathy includes both non-proliferative diabetic retinopathy (nPDR) and proliferative diabetic retinopathy (PDR), the more advanced form of the disease.
  • nPDR non-proliferative diabetic retinopathy
  • PDR proliferative diabetic retinopathy
  • DME is a frequent manifestation of DR (Riordan-Eva, 2004, Eye (Lond). 2004, 18:1161-8) and is the major cause of visual loss in patients with DR.
  • DME diabetic macular edema
  • care providers actively monitor mild to moderate nPDR and reserve treatment for severe nPDR and PDR.
  • Both LUCENTIS® and EYLEA® have recently gained approval for DR in the United States.
  • EYLEA® is 2 mg (0.05 mL) administered every 4 weeks for 5 injections followed by an injection once every 8 weeks.
  • the recommended dose of LUCENTIS® is 0.3 mg (0.05 mL) administered once a month.
  • ocular disease is Retinal Vein Occlusion (RVO), e.g., Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO).
  • RVO Retinal Vein Occlusion
  • CRVO Central Retinal Vein Occlusion
  • BRVO Branch Retinal Vein Occlusion
  • Retinal vein occlusion is a blockage of the small veins that carry blood away from the retina.
  • the retina is the layer of tissue at the back of the inner eye that converts light images to nerve signals and sends them to the brain.
  • Retinal vein occlusion is most often caused by hardening of the arteries (atherosclerosis) and the formation of a blood clot.
  • Both CRVO and BRVO are both related to occlusion of the retinal vein, however the cause of the occlusion differs based on location.
  • Central Retinal Vein Occlusion occurs with a thrombus occludes the central retinal vein near the lamina cribosa (Green et ah, Retina. 1:27-55, 1981).
  • Branch Retinal Vein Occlusion occurs when a thrombus occurs at the arteriovenous crossing point secondary to atherosclerosis of the retinal artery causing compression and occlusion of the retinal vein. (Frangieh et ah, Archives of Ophthalmology. 100:1132-40, 1982).
  • Blockage of retinal veins may cause other eye problems, including: (i) glaucoma (high pressure in the eye), caused by new, abnormal blood vessels growing in the front part of the eye; (ii) macular edema, caused by the leakage of fluid in the retina.
  • VEGF has a leading role in RVO pathogenesis and symptoms.
  • Use of anti -VEGF agents by intravitreal injections has become common with the aim to improve the clinical outcomes in these patients.
  • 2 anti-VEGF agents (ranimizumab and aflibercept) have been FDA (Food and Drug Administration) and EMA (European Medicine Agency) approved for the treatment of RVO, while another VEGF inhibitor (bevacizumab) is often used “off-label” in clinical practice.
  • the present invention provides a method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • a VEGF antagonist e.g., brolucizumab
  • a HRD in a subject’s eye e.g., VHRD, e.g., PHRD
  • a low level of a HRD in a subject’s eye e.g., VHRD, e.g., PHRD; or
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • a different ocular disease agent e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab
  • VHRD e.g., PHRD
  • VHRD e.g., PHRD
  • VHRD e.g., PHRD
  • recording the result of the determining step on a tangible or intangible media form for use in transmission e.g., VHRD, e.g., PHRD
  • the present invention provides a method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., nAMD, may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
  • IOI intraocular inflammation
  • a VEGF antagonist e.g., brolucizumab
  • the present disclosure provides methods and uses for treating ocular disease, e.g., nAMD, the method or the use comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
  • a VEGF antagonist e.g., brolucizumab
  • the VEGF antagonist of the disclosure is any licensed anti-VEGF drug such as brolucizumab, ranibizumab or aflibercept.
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody (such as brolucizumab or ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF receptor domains, e.g.,
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody, e.g., a single chain antibody (scFv) or Fab fragment.
  • scFv single chain antibody
  • Fab fragment fragment
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody, particularly anti-VEGF antibodies described in WO 2009/155724, the entire contents of which are hereby incorporated by reference.
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
  • VH SEQ ID NO. 1
  • VL SEQ ID NO. 2
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising the sequence as set forth in SEQ ID NO: 3.
  • the VEGF antagonist of the disclosure is brolucizumab (which comprises the sequence of SEQ ID NO: 3).
  • the sequence of brolucizumab is set forth in SEQ ID NO: 4.
  • a methionine derived from the start codon in an expression vector is present in the final protein in cases where it has not been cleaved posttranslationally as follows.
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising three light chain CDRs (CDRL1, CDRL2, and CDRL3) and three heavy chain CDRs (CDRH1, a CDRH2, a CDRH3) as follows:
  • CDRL1 QASEIIHSWLA SEQ ID NO: 5
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising a heavy chain complementarity determining region 1 (CDRH1) comprising or consisting of the amino acid sequence of SEQ ID NO: 8; a heavy chain complementarity determining region 2 (CDRH2) comprising or consisting of the amino acid sequence of SEQ ID NO: 9; a heavy chain complementarity determining region 3 (CDRH3) comprising or consisting of the amino acid sequence of SEQ ID NO: 10; a light chain complementarity determining region 1 (CDRLl) comprising or consisting of the amino acid sequence of SEQ ID NO: 5; a light chain complementarity determining region 2 (CDRL2) comprising or consisting of the amino acid sequence of SEQ ID NO: 6; and a light chain complementarity determining region 3 (CDRL3) comprising or consisting of the amino acid sequence of SEQ ID NO: 7.
  • CDRH1, CDRH2, CDRH3, CDRLl, CDRL2 and CDRL3 are defined according to
  • variable heavy chain comprising or having the sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 1, and wherein the variable heavy chain comprises CDRH1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8, CDRH2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9, CDRH3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10, and
  • variable light chain comprising or having the sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 2, and wherein the variable light chain comprises CDRL1 comprising or consisting of the amino acid sequence of SEQ ID NO: 5, CDRL2 comprising or consisting of the amino acid sequence of SEQ ID NO: 6, and CDRL3 comprising or consisting of the amino acid sequence of SEQ ID NO: 7.
  • CDRL1 comprising or consisting of the amino acid sequence of SEQ ID NO: 5
  • CDRL2 comprising or consisting of the amino acid sequence of SEQ ID NO: 6
  • CDRL3 comprising or consisting of the amino acid sequence of SEQ ID NO: 7.
  • the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3 are defined according to the AHo numbering scheme.
  • the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising or having a sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 3 or SEQ ID NO: 3, and wherein the anti-VEGF antibody comprises CDRH1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8; CDRH2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9; CDRH3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10; CDRL1 comprising or consisting of the amino acid sequence of SEQ ID NO: 5; CDRL2 comprising or consisting of the amino acid sequence of SEQ ID NO: 6; and CDRL3 comprising or consisting of the amino acid sequence of SEQ ID NO: 7.
  • the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3 are defined according to the AHo numbering scheme.
  • Brolucizumab is a humanized single-chain Fv (scFv) antibody fragment inhibitor of VEGF with a molecular weight of ⁇ 26 kDa. It is an inhibitor of VEGF -A and works by binding to the receptor binding site of the VEGF -A molecule, thereby preventing the interaction of VEGF-A with its receptors VEGFRl and VEGFR2 on the surface of endothelial cells. Increased levels of signaling through the VEGF pathway are associated with pathologic ocular angiogenesis and retinal edema. Inhibition of the VEGF pathway has been shown to inhibit the growth of neovascular lesions and resolve retinal edema in patients with nAMD.
  • scFv single-chain Fv
  • the disclosure provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g.,
  • said different ocular disease agent is another VEGF antagonist, such as any licensed anti-VEGF drug, e.g., ranibizumab or aflibercept.
  • said different ocular disease agent is an anti-VEGF antibody (such as ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF receptor domains, e.g., con
  • the VEGF antagonist of the disclosure is brolucizumab and is administered by an injection, e.g., an intravitreal injection.
  • the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of about 1, about 2, about 3, about 4, about 5, or about 6 mg (e.g., about 6 mg/0.05 mL) as an intravitreal injection. In certain embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of 1, 2, 3, 4, 5, or 6 mg (e.g., 6 mg/0.05 mL) as an intravitreal injection. In some embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.5, about 1 or about 2 mg (e.g., about 2 mg/0.05 mL) as an intravitreal injection.
  • the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of 0.5, 1 or 2 mg (e.g., 2 mg/0.05 mL) as an intravitreal injection. In some embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.2, about 0.3, about 0.4 or about 0.5 mg (e.g., about 0.5 mg/0.05 mL) as an intravitreal injection. In certain embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of 0.2, 0.3, 0.4 or 0.5 mg (e.g., 0.5 mg/0.05 mL) as an intravitreal injection.
  • the methods or uses of the disclosure comprise the use of pharmaceutical formulations or pharmaceutical compositions comprising a VEGF antagonist, e.g., an anti-VEGF antibody.
  • a VEGF antagonist e.g., an anti-VEGF antibody.
  • “Pharmaceutically acceptable” excipients are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
  • a “stable” formulation is one in which a therapeutic agent, e.g. a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative thereof, essentially retains its physical stability and/or chemical stability and/or biological activity upon storage.
  • a therapeutic agent e.g. a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative thereof
  • VEGF antagonist e.g., an anti-VEGF antibody or antibody derivative thereof
  • Stability can be measured at a selected temperature for a selected time period.
  • the formulation is stable at room temperature (about 30° C) or at 40° C for at least 1 week and/or stable at about 2-8° C for at least 3 months to 2 years.
  • the formulation is preferably stable following freezing (to, e.g., -70° C) and thawing of the formulation.
  • An antagonist e.g., an antibody or antibody derivative, “retains its physical stability” in a pharmaceutical formulation if it meets the defined release specifications for aggregation, degradation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography, or other suitable art recognized methods.
  • An antagonist e.g., an antibody or antibody derivative, “retains its chemical stability” in a pharmaceutical formulation, if the chemical stability at a given time is such that the compound, e.g., protein, is considered to still retain its biological activity as defined below.
  • Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein.
  • Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF MS), for example.
  • Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange chromatography, for example.
  • An antagonist e.g., an antibody or antibody derivative, “retains its biological activity” in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared as determined in an antigen binding assay, for example.
  • Other “biological activity” assays for antibodies are elaborated herein below.
  • isotonic is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer, for example.
  • a “polyol” is a substance with multiple hydroxyl groups, and includes sugars (reducing and non-reducing sugars), sugar alcohols and sugar acids. Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g. in the range from about 120 to about 400 kD).
  • a “reducing sugar” is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a “non reducing sugar” is one which does not have these properties of a reducing sugar.
  • reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose.
  • Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffmose.
  • Mannitol, xylitol, erythritol, threitol, sorbitol and glycerol are examples of sugar alcohols.
  • sugar acids these include L-gluconate and metallic salts thereof.
  • the polyol is preferably one which does not crystallize at freezing temperatures (e.g.
  • Non-reducing sugars such as sucrose and trehalose are the preferred polyols herein, with trehalose being preferred over sucrose, because of the superior solution stability of trehalose.
  • buffer refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components.
  • the buffer of this disclosure has a pH in the range from about 4.5 to about 8.0; preferably from about 5.5 to about 7.
  • buffers that will control the pH in this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers.
  • the buffer is preferably not phosphate.
  • a “therapeutically effective amount” of a therapeutic agent e.g., a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative
  • a therapeutic agent e.g., a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative
  • a therapeutic agent e.g., a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative
  • an amount effective in the prevention or treatment of a disorder for the treatment of which the antagonist, e.g., antibody or antibody derivative refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antagonist, e.g., antibody or antibody derivative, is effective.
  • a “preservative” is a compound which can be included in the formulation to essentially reduce bacterial action therein, thus facilitating the production of a multi-use formulation, for example.
  • potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride.
  • preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.
  • aromatic alcohols such as phenol, butyl and benzyl alcohol
  • alkyl parabens such as methyl or propyl paraben
  • catechol resorcinol
  • cyclohexanol 3-pentanol
  • m-cresol m-cresol
  • the most preferred preservative herein is benzyl alcohol.
  • the pharmaceutical compositions used in present disclosure comprise a VEGF antagonist A, preferably an anti-VEGF antibody (e.g., an anti-VEGF antibody comprising the variable light chain sequence of SEQ ID NO: 1 and the variable heavy chain sequence of SEQ ID NO: 2, such as brolucizumab), together with at least one physiologically acceptable carrier or ex
  • compositions may comprise, for example, one or more of water, buffers (e.g., neutral buffered saline or phosphate buffered saline), ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g, glucose, mannose, sucrose or dextrans), mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives.
  • buffers e.g., neutral buffered saline or phosphate buffered saline
  • ethanol e.g., mineral oil, vegetable oil, dimethylsulfoxide
  • carbohydrates e.g, glucose, mannose, sucrose or dextrans
  • mannitol e.glycine
  • proteins e.glycine
  • adjuvants e.glycine
  • polypeptides or amino acids such as glycine
  • antioxidants e.
  • a carrier is a substance that may be associated with an antibody or antibody derivative prior to administration to a patient, often for the purpose of controlling stability or bioavailability of the compound.
  • Carriers for use within such formulations are generally biocompatible, and may also be biodegradable.
  • Carriers include, for example, monovalent or multivalent molecules such as serum albumin (e.g, human or bovine), egg albumin, peptides, polylysine and polysaccharides such as aminodextran and polyamidoamines.
  • Carriers also include solid support materials such as beads and microparticles comprising, for example, polylactate polyglycolate, poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose or dextran.
  • a carrier may bear the compounds in a variety of ways, including covalent bonding (either directly or via a linker group), noncovalent interaction or admixture.
  • compositions may be formulated for any appropriate manner of administration, including, for example, topical, intraocular, oral, nasal, rectal or parenteral administration.
  • compositions in a form suitable for intraocular injection such as intravitreal injection, are preferred.
  • Other forms include, for example, pills, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • compositions provided herein may be formulated as a lyophilizate.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g, intravenous), intramuscular, spinal, intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique.
  • the pharmaceutical composition may be prepared as a sterile injectible aqueous or oleaginous suspension in which the active agent (i.e. VEGF antagonist), depending on the vehicle and concentration used, is either suspended or dissolved in the vehicle.
  • the active agent i.e. VEGF antagonist
  • Such a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above.
  • suitable vehicles and solvents that may be employed are water, 1,3-butanediol, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils may be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectible compositions, and adjuvants such as local anesthetics, preservatives and/or buffering agents can be dissolved in the vehicle.
  • aqueous formulation of a VEGF antagonist e.g., an anti-VEGF antibody (e.g., brolucizumab) used in the methods or uses of the disclosure is prepared in a pH-buffered solution.
  • the buffer of such aqueous formulation has a pH in the range from about
  • an aqueous pharmaceutical composition of the disclosure is about
  • 7.0-7.5 or about 7.0-7.4, about 7.0-7.3, about 7.0-7.2, about 7.1-7.6, about 7.2-7.6, about 7.3-
  • an aqueous pharmaceutical composition of the disclosure has a pH of about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5 or about 7.6.
  • the aqueous pharmaceutical composition has a pH of >7.0
  • the aqueous pharmaceutical composition has a pH of about 7.2.
  • the aqueous pharmaceutical composition has a pH of about 7.4.
  • the aqueous pharmaceutical composition has a pH of about 7.6.
  • buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers.
  • the buffer concentration can be from about 1 mM to about 50 mM, preferably from about 5 mM to about 30 mM, depending, for example, on the buffer and the desired isotonicity of the formulation.
  • a polyol which acts as a tonicifier, may be used to stabilize an antibody in an aqueous formulation.
  • the polyol is a non-reducing sugar, such as sucrose or trehalose.
  • the polyol is added to the formulation in an amount that may vary with respect to the desired isotonicity of the formulation.
  • the aqueous formulation is isotonic, in which case suitable concentrations of the polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% w/v, for example.
  • hypertonic or hypotonic formulations may also be suitable.
  • the amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose).
  • a monosaccharide e.g. mannitol
  • a disaccharide such as trehalose
  • a surfactant is also added to an aqueous antibody formulation.
  • exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80 etc) or poloxamers (e.g. poloxamer 188).
  • the amount of surfactant added is such that it reduces aggregation of the formulated antibody/antibody derivative and/or minimizes the formation of particulates in the formulation and/or reduces adsorption.
  • the surfactant may be present in the formulation in an amount from about 0.001% to about 0.5%, preferably from about 0.005% to about 0.2% and most preferably from about 0.01% to about 0.1%.
  • an aqueous antibody formulation used in the methods or uses of the disclosure is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl.
  • a preservative may be included in the formulation, particularly where the formulation is a multidose formulation.
  • the concentration of preservative may be in the range from about 0.1% to about 2%, most preferably from about 0.5% to about 1%.
  • One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 21st edition, Osol, A. Ed. (2006) may be included in the formulation provided that they do not adversely affect the desired characteristics of the formulation.
  • Acceptable carriers, excipients or stabilizers are non-toxic to recipients at the dosages and concentrations employed and include: additional buffering agents, co-solvents, antioxidants including ascorbic acid and methionine, chelating agents such as EDTA, metal complexes (e.g. Zn- protein complexes), biodegradable polymers such as polyesters, and/or salt-forming counterions such as sodium.
  • Formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation.
  • the VEGF antagonist of the disclosure is administered to an eye of a subject in need of treatment in accordance with known methods for ocular delivery.
  • the subject is a human
  • the VEGF antagonist is an anti-VEGF antibody (preferably brolucizumab), and the antibody is administered directly to an eye.
  • Administration to a patient can be accomplished, for example, by intravitreal injection.
  • VEGF antagonist in the methods and uses of the disclosure can be administered as the sole treatment or in conjunction with other drugs or therapies useful in treating the condition in question.
  • a preferred formulation for brolucizumab for intravitreal injection comprises about 4.5% to 11% (w/v) sucrose, 5-20 mM sodium citrate, and 0.001% to 0.05% (w/v) polysorbate 80, wherein the pH of the formulation is about 7.0 to about 7.4.
  • One such formulation comprises 5.9% (w/v) sucrose, 10 mM sodium citrate, 0.02% (w/v) polysorbate 80, pH of 7.2, and 6 mg of brolucizumab.
  • Another such formulation comprises 6.4% (w/v) or 5.8% sucrose, 12 mM or 10 mM sodium citrate, 0.02% (w/v) polysorbate 80, pH of 7.2, and 3 mg of brolucizumab.
  • Preferred concentrations of brolucizumab are about 120 mg/ml and about 60 mg/ml. Doses can be delivered, for example as 6 mg/50 pL and 3 mg/50 pL concentrations.
  • a dose used in the methods or uses of the disclosure is based on the specific disease or condition being treated, and is therapeutically effective dose. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient’s own immune system. The dose amount can be readily determined using known dosage adjustment techniques by a physician having ordinary skill in treatment of the disease or condition.
  • the therapeutically effective amount of a VEGF antagonist used in the methods or uses of the disclosure is determined by taking into account the desired dose volumes and mode(s) of administration, for example.
  • therapeutically effective compositions are administered in a dosage ranging from 0.001 mg/ml to about 200 mg/ml per dose.
  • the VEGF antagonist used in the methods or uses of the disclosure is brolucizumab, and a dosage thereof used in the methods or uses of the disclosure is about 60 mg/ml to about 120 mg/ml (for example, a dosage is 60, 70, 80, 90, 100, 110, or 120 mg/ml). In a preferred embodiment, the dosage of the VEGF antagonist used in the methods or uses of the disclosure is 60 mg/ml or 120 mg/ml.
  • a dose of the VEGF antagonist is administered directly to an eye of a patient.
  • a dose of the VEGF antagonist per eye is at least about 0.5 mg up to about 6 mg.
  • Preferred doses per eye include about 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.2 mg, 1.4 mg, 1.6 mg, 1.8 mg, 2.0 mg, 2.5 mg, 3.0 mg, 3.5 mg, 4.0 mg, 4.5 mg, 5.0 mg, 5.5 mg, and 6.0 mg.
  • a dose per eye is at least about 3 mg up to about 6 mg, e.g., about 3 mg or about 6 mg.
  • Doses can be administered in various volumes suitable for ophthalmic administration, such as 50 pi or 100 m ⁇ , for example, including 3 mg/50 m ⁇ or 6 mg/50 m ⁇ . Smaller volumes can also be used, including 20 m ⁇ or less, for example about 20 m ⁇ , about 10 m ⁇ , or about 8.0 m ⁇ . In certain embodiments, a dose of 2.4 mg/20 m ⁇ , 1.2 mg/10 m ⁇ or 1 mg/8.0 m ⁇ (e.g., 1 mg/8.3 m ⁇ ) is delivered to an eye of a patient for treating or ameliorating one or more of the diseases and disorders described above. Delivery can be, for example, by an injections, e.g., an intravitreal injection.
  • the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of about 1, about 2, about 3, about 4, about 5, or about 6 mg (e.g., about 6 mg/0.05 mL), e.g., 1, 2, 3, 4, 5, or 6 mg (e.g., 6 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
  • the VEGF antagonist of the disclosure is brolucizumab and is administered to the subject in three individual doses at 4-week intervals, and thereafter administered to the subject as an additional dose every 8 to 12 weeks, e.g., every 8 weeks or every 12 weeks.
  • a patient might be on a 12-week interval regimen for some time, and then switch to an 8-week interval, and then switch back to the 12-week interval.
  • patients may not stay on one interval regimen, and may go back and forth depending on assessments according to the criteria set forth herein.
  • a 12 week treatment interval may be switched to an 8 week treatment interval if a disease activity is detected in the subject, or an 8 week treatment interval may be switched to a 12 week treatment interval if no disease activity is detected in the subject.
  • the disease activity may be assessed based on visual function, retinal structure and leakage.
  • An assessment as described herein preferably includes one or more of the following tests to assess activity of a VEGF antagonist (e.g., brolucizumab) on visual function, retinal structure and leakage: (i) best corrected visual acuity (BCVA), e.g., best corrected visual acuity with ETDRS-like chart at 4 meters, (ii) visual acuity (VA), (iii) central subfield thickness (CSFT), (iv) presence of intraretinal cysts/fluid, (v) ETDRS DRSS score based on 7-field stereo Color Fundus Photography (CFP), (vi) anatomical retinal evaluation by Optical Coherence Tomography (OCT), standard or wide-field Fluorescein Angiography (FA), OCT angiography, and/or wide-field CFP/FA, (vii) peripheral visual field assessed by perimetry, (viii) contrast sensitivity, (viii
  • BCVA protocol refraction
  • OCT Optical Coherence Tomography
  • color fundus photography and fluorescein angiography can be assessed according to methods known to those of skill in the art.
  • the CST is the average thickness of circular 1 mm area centered around the fovea measured from retinal pigment epithelium (RPE) to the internal limiting membrane (ILM), inclusively.
  • CST can be measured, for example, using spectral domain Optical Coherence Tomography (SD-OCT).
  • the disease activity may be assessed based on one or more of the following: (i) best corrected visual acuity (BCVA), (ii) visual acuity (VA), (iii) central subfield thickness (CSFT), and (iv) presence of intraretinal cysts/fluid.
  • BCVA best corrected visual acuity
  • VA visual acuity
  • CSFT central subfield thickness
  • the presence of ocular disease activity includes one or more of the following: (i) decrease in Best Corrected Visual Acuity (BCVA), (ii) decrease in Visual Acuity (VA), (iii) increase or lack of reduction in Central Subfield Thickness (CSFT), (iv) new or persistent or recurrent Intraretinal Cysts (IRC) and/or Intraretinal Fluid (IRF) and/or Subretinal Fluid (SRF). Fluid measured in the eye can be intraretinal and/or subretinal fluid.
  • BCVA Best Corrected Visual Acuity
  • VA Visual Acuity
  • CSFT Central Subfield Thickness
  • IRC Intraretinal Cysts
  • IRF Intraretinal Fluid
  • the presence of ocular disease activity includes one or more of the following:
  • BCVA of >4 letters e.g., decrease in BCVA of >5 letters, more particularly wherein:
  • the decrease in BCVA is observed at Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline BCVA, wherein the baseline BCVA was assessed prior to the last administration of the VEGF antagonist, or
  • the decrease in BCVA is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline BCVA, wherein the baseline BCVA was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
  • the decrease in VA is observedat Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline VA, wherein the baseline VA was assessed prior to the last administration of the VEGF antagonist, or
  • the decrease in VA is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline VA, wherein the baseline VA was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
  • CSFT increase >25pm e.g., CSFT increase >50pm, e.g., CSFT increase >75pm, more particularly wherein:
  • the CSFT increase is observed at Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline CSFT, wherein the baseline CSFT was assessed prior to the last administration of the VEGF antagonist, or
  • the CSFT increase is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline CSFT, wherein the baseline CSFT was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
  • IRC intraretinal cysts
  • IRF intraretinal fluid
  • SRF subretinal fluid
  • IRC intraretinal cysts
  • IRF intraretinal fluid
  • SRF subretinal fluid
  • IRC intraretinal cysts
  • IRF intraretinal fluid
  • SRF subretinal fluid
  • the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.5, about 1 or about 2 mg (e.g., about 2 mg/0.05 mL), e.g., 0.5, 1 or 2 mg (e.g., 2 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
  • the VEGF antagonist used in the methods or uses of the disclosure is ranibizumab and is administered at a dose of about 0.2, about 0.3, about 0.4 or about 0.5 mg (e.g., about 0.5 mg/0.05 mL), e.g., 0.2, 0.3, 0.4 or 0.5 mg (e.g., 0.5 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
  • HAWK is a randomized, double-masked, 96-week phase 3 studies comparing brolucizumab to aflibercept in nAMD.
  • the Safety Review Committee (SRC) performed an independent unmasked post hoc review of all cases of investigator-reported IOI, retinal vascular occlusions and endophthalmitis in the phase 3 HAWK and HARRIER studies.
  • SRC Safety Review Committee
  • a higher order comparative discovery analysis was initiated evaluating 34 eyes from the HAWK study identified through the SRC analysis and compared to 34 propensity-matched controls.
  • This analysis includes comparative higher-order OCT, radiomics assessment, and machine learning classification.
  • This report provides an initial assessment of the qualitative comparative discovery analysis of the Safety next Generation imaging study (SAGE). Images were reviewed frame-by-frame for specific features prior to or during the IOI event.
  • Example 2 Predictive Assessment of Radio mic features to identify eyes with Intraocular Inflammation (101) on HAWK-SAGE data
  • radiomics assessment has been performed (Figure 3) to evaluate potential pre- inflammatory event signals within the vitreous compartment. Specifically, the assessment was conducted to identify and characterize vitreous compartment features that might provide early insights into pre-inflammatory event signals.
  • Last timepoint represented the “safety” event or when inflammation develops.
  • the last timepoint vitreous compartment between the Safety group and the Control group was compared.
  • an analysis of the timepoint immediately before the final timepoint was done to assess if we can tell a difference between the groups prior to actual events.
  • assessment at the month one timepoint was conducted to assess whether after a single injection it is possible to identify a difference between those two groups.
  • a 3-fold cross validation was implemented for supervised classification.
  • Different machine learning (ML) classifiers linear discriminant analysis (LDA), quadratic discriminant analysis (QDA), support vector machine (SVM), random forest (RF)
  • LDA linear discriminant analysis
  • QDA quadratic discriminant analysis
  • SVM support vector machine
  • RF random forest
  • K-means Clustering was also conducted. Three clusters were formed: 60% of Sis grouped within Cluster 1; 79% of S2s grouped within Cluster 2; 62% Controls grouped within Cluster 3.
  • the AUC values yielded by LDA are presented in Table 2 on different timepoint images.
  • Table 2 AUC values yielded by LDA in classifying Control and SI group.
  • hyporeflective preretinal foci i.e., preretinal stalagmites
  • preretinal stalagmites appear to be a potential important objective OCT finding of IOI and may provide an important signal for clinicians assessing for underlying posterior inflammation.

Abstract

The invention relates to methods for treating ocular disease with a VEGF antagonist. In particular, the invention relates to methods for treating ocular disease, the method comprising selectively administering a VEGF antagonist to a subject in need thereof on the basis of said subject not having a preretinal or vitreous hyperreflective foci formation. The invention further relates to methods for determining whether a subject with ocular disease should be treated with a VEGF antagonist, methods of predicting the likelihood that a subject having ocular disease will develop intraocular inflammation in response to a treatment with a VEGF antagonist, and methods for producing a transmittable form of information for determining whether a subject with ocular disease should be treated with a VEGF antagonist or for predicting whether a subject with ocular disease may develop intraocular inflammation in response to a treatment with a VEGF antagonist.

Description

METHODS FOR TREATING OCULAR DISEASES
SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on March 22, 2021, is named PAT059060_SEQ_LISTING_ST25.txt and is 8 KB in size.
FIELD
The invention relates to methods for treating ocular disease with a VEGF antagonist.
BACKGROUND
Age-related macular degeneration (AMD) is the leading cause of severe vision loss in people affecting 10%-13% of individuals over the age of 65 in North America, Europe, and Australia (Kawasaki 2010, Rein et al., Arch Ophthalmol. 2009;127:533-40, Smith 2001). Genetic, environmental and health factors play an important role in the pathogenesis of the disease. AMD is classified into 2 clinical subtypes: the non-neovascular (atrophic) or dry form and the neovascular (exudative) or wet form (Ferris et al., Arch Ophthalmol. 1984;102:1640-2, Lim et al., Lancet. 2012;379:1728-38, Miller et al., Am J Ophthalmol. 2013;155:1-35). Neovascular AMD (nAMD) is characterized by the growth of abnormal new blood vessels (neovascularization) under the retinal pigment epithelium (RPE) or subretinal space from the subjacent choroid, termed choroidal neovascularization (CNV) (Ferris et al., Arch Ophthalmol. 1984;102:1640-2). These newly formed vessels have an increased likelihood to leak blood and serum, damaging the retina by stimulating inflammation and scar tissue formation. This damage to the retina results in progressive, severe, and irreversible vision loss (Shah et al., Am J Ophthalmol. 2007;143:83-89, Shah et al., Am J Ophthalmol. 2009; 116: 1901-07). Without treatment, most affected eyes will have poor central vision (20/200) within 12 months (TAP 2003). Although the neovascular form of the disease is only present in about 10% of all AMD cases, it accounted for approximately 90% of the severe vision loss from AMD prior to the introduction of anti-vascular endothelial growth factor (VEGF) treatments (Ferris et al., Am J Ophthalmol. 1983; 118:132-51, Sommer et al., N Engl J Med. 1991;14:1412-17, Wong et al., Ophthalmology. 2008;115:116-26).
VEGF has been shown to be elevated in patients with nAMD and is thought to play a key role in the neovascularization process (Spilsbury et al., Am J Pathol. 2000;157:135-44). The use of intravitreal (IVT) pharmacotherapy targeting VEGF has significantly improved visual outcomes in patients with nAMD (Bloch et al., Am J Ophthalmol. 2012;153:209-13, Campbell et al., Arch Ophthalmol. 2012;130:794-5). Anti-VEGF treatments, such as ranibizumab (LUCENTIS®), aflibercept (EYLEA®), and brolucizumab (Beovu®), inhibit VEGF signaling pathways and have been shown to halt the growth of neovascular lesions and resolve retinal edema.
One of the main ocular adverse events (AEs) associated with these agents is intraocular inflammation (IOI) (Tolentino M., Surv Ophthalmol, 2011;56:95-113; Mones et al., American Academy of Ophthalmology, 2020). In the VIEW studies, for example, in which patients with nAMD were treated with intravitreal ranibizumab or aflibercept, IOI was a predefined AE of interest (Schmidt-Erfurth et al., Ophtalmology, 2014;121:193-201). Between baseline and 96 weeks, this event was reported in 1.5% of patients receiving ranibizumab and 0.5% to 1.1% of patients in the three aflibercept arms (Schmidt-Erfurth et al., Ophtalmology, 2014;121:193-201).
Brolucizumab 6mg represents an important treatment option for patients with wet AMD, with an overall favorable benefit-risk profile. Following approval of brolucizumab 6 mg for the treatment of nAMD by the Food and Drug Administration (FDA) in October 2019, there were reports of retinal vasculitis and/or retinal artery occlusion (RAO) accompanied by IOI with intravitreal injections of brolucizumab (Safety of Beovu® (brolucizumab). https://www.brolucizumab.info/ Last accessed on 19 November 2020. 2020; Witkin et al., J Vitreoretin Dis 2020;4(4):269-79). Based on post-marketing reports, and post hoc review of Hawk & Harrier cases of interest, Novartis initiated internal review of these post-marketing safety case reports, including the establishment of an external Safety Review Committee (SRC) to provide an independent, unmasked post-hoc review of these cases (Beovu® (brolucizumab) Global Safety Information for Healthcare Professionals.
Figure imgf000004_0001
Based on an unmasked post-hoc review, the SRC reported overall incidence of intraocular inflammation (IOI) of any form of 4.6% (50/1088), overall incidence of signs of retinal vasculitis of 3.3% (36/1088), overall incidence of concomitant signs of retinal vasculitis (RV) and retinal vascular occlusion (RO) of 2.1% (23/1088), and overall incidence of IOI of any form associated with losing 15 or more letters at the last visit / end of the study was 0.7% (8/1088) (Member Update: Novartis-Appointed Safety Review Committee Reports Initial Brolucizumab Findings. American Society of Retina Specialists. June 4, 2020; Mones et al., American Academy of Ophthalmology, 2020). Label updates have been approved by several health authorities, including the FDA and the EMA. RV and/or RO have been reported with the use of brolucizumab (Beovu® [US prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals Corp, Oct 2019; Beovu® [summary of product characteristics], Basel, Switzerland, Novartis Pharma AG. Sept 2020). These events may result in severe vision loss and have typically occurred in the presence of IOI (Beovu® (brolucizumab)
Global Safety Information for Healthcare Professionals http s : // zum ab .1 nfo/.
Figure imgf000005_0001
Accessed October 2, 2020).
Intraocular inflammation (IOI) is an adverse event that may lead to further complications if not identified, including severe vision loss. To increase patient’s safety, there is a benefit to identifying susceptible patients before development of intraocular inflammation or at the very early onset of intraocular inflammation and adjust their treatment, for example by discontinuing the treatment with that particular drug that can cause intraocular inflammation or by altering / selecting the treatment (drug and treatment regimen) correspondingly. There is a continuing need in the art for predicting, determining, monitoring delayed drug-induced intraocular inflammation.
SUMMARY
Using advanced imaging methods (spectral-domain optical coherence tomography, SD-OCT) a post hoc analysis was conducted of the HAWK and HARRIER studies, which were 2-year, randomized, double-masked, multicenter, phase 3 trials comparing brolucizumab with aflibercept in treatment-naive patients with nAMD. The post-hoc analysis identified a new biomarker that precedes or is associated with intraocular inflammation (IOI) adverse events in nAMD patients: a hyperreflective deposit (HRD). In particular, the post-hoc analysis identified that a formation of HRD, and in particular a formation of a HRD in vitreous compartment, e.g., preretinal, e.g., internal limiting membrane (ILM), precedes or is associated with intraocular inflammation (IOI) adverse events in nAMD patients. . The reported hyperreflective deposits were in a form of heterogeneity observed on the OCT scan including hyperreflective debris, hyperreflective round- or rod-shaped dots, e.g., stalagmite like preretinal deposits. The stalagmite-like deposits are discrete HRD, e.g., PHRD, that resemble the elevated speleothems. Previously, the stalagmite-like preretinal deposits have been described and associated with vitreous inflammation in patients with vitrectomized eyes and flares of posterior uveitis (Yonekawa et ah, Digital Journal of Ophthalmology, 2017, Vol. 23, pp. 18-22). In accordance specific aspects of the present invention, it has been surprisingly found that there is a correlation between presence of HRD, e.g., VHRD, e.g., PHRD e.g., stalagmite-like preretinal hyperreflective material, with posterior inflammation in nAMD patients treated with a VEGF inhibitor, e.g., brolucizumab. Based on this finding, it is proposed that HRD, e.g., VHRD, e.g., PHRD , in particular stalagmite-like preretinal hyperreflective material, may be used as a safety biomarker for the selection of the patients suitable for treatment with a VEGF inhibitor, e.g., brolucizumab, or for monitoring the patients during the course of treatment with a VEGF inhibitor, e.g., brolucizumab. Use of this new safety biomarker thus advances management of ophthalmic disorders such as nAMD in patients.
In one aspect, the present invention provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
In one aspect, the present invention provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
In a another aspect, the present invention provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD
In a another aspect, the present invention provides a method for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e.g., VHRD, e.g., PHRD; and
(b) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, if no said HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye;
(c) determining that the subject should not be treated with a VEGF antagonist, e.g., brolucizumab, if the HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
In a another aspect, the present invention provides a method of predicting the likelihood that a subject having ocular disease, e.g., nAMD, will develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD. Suitably, the method comprises determining by OCT or SD- OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, wherein:
(a) the presence of the HRD, e.g., VHRD, e.g., PHRD, is indicative of an increased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab; and|
(b) the absence of the HRD, e.g., VHRD, e.g., PHRD, is indicative of a decreased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab. In one aspect, the present invention provides a method of monitoring a subject during a course of treatment with a VEGF antagonist for presence of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, e.g., by performing OCT or SD-OCT analysis, and (i) altering or discontinuing the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined to have the HRD, e.g., VHRD, e.g., PHRD, in the treatment eye; or (ii) maintaining the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined not to have the HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
In one aspect, the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
In a another aspect, the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, characterized in that:
(a) a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) thereafter, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is administered to the subject.
In a another aspect, the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, wherein the use comprises
(a) determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In a another aspect, the present invention provides a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the use in treatment of ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e.g., VHRD, e.g., PHRD; and
(b) determining if the subject has the HRD, e.g., VHRD, e.g., PHRD; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(d) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In one aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, characterized in that:
(a) a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) thereafter, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is administered to the subject.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises
(a) determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and (c) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for the manufacture of a medicament for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e.g., VHRD, e.g., PHRD; and
(b) determining if the subject has a HRD, e.g., VHRD, e.g., PHRD; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(d) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In one aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, characterized in that:
(a) a subject is selected for treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) thereafter, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is administered to the subject.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises
(a) determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In a another aspect, the present invention provides use of a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition comprising said VEGF antagonist for treating ocular disease, e.g., nAMD, in a subject, wherein the use comprises:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(d) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
In one aspect, the present invention provides, a method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining that the subject should be treated with a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission. In one aspect, the present invention provides, a method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., nAMD, may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining an increased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject having HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining a decreased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
Non-limiting embodiments of the present disclosure are described in the following embodiments:
Embodiment 1: A method of treating ocular disease comprising selectively administering a VEGF antagonist to a subject in need thereof on the basis of said subject not having a hyperreflective deposit (HRD), e.g., a vitreous hyperreflective deposit (VHRD), e.g., a preretinal hyperreflective deposit (PHRD).
Embodiment 2: A method of treating ocular disease comprising:
(a) selecting a subject for treatment with a VEGF antagonist on the basis said subject not having a HRD, e.g., VHRD, e.g., PHRD; and
(b) thereafter, selectively administering the VEGF antagonist to the subject.
Embodiment 3: A method of selectively treating a subject having ocular disease the method comprising:
(a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD, e.g., VHRD, e.g., PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD, e.g., VHRD, e.g., PHRD.
Embodiment 4: The method of embodiment 2 or 3 further comprising assessing the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, e.g., prior to the steps (a)-(b) of embodiment 2 or embodiment 3.
Embodiment 5: The method of any one of embodiments 1 to 4, wherein the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, is assessed by performing OCT, e.g., SD- OCT, analysis on a subject’s eye.
Embodiment 6: A method for determining whether a subject with ocular disease should be treated with a VEGF antagonist, the method comprising:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e.g., VHRD, e.g., PHRD; and
(b) determining that the subject should be treated with a VEGF antagonist if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye;
(c) determining that the subject should not be treated with a VEGF antagonist if the HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
Embodiment 7: The method of embodiment 6 further comprising treating the subject with the VEGF antagonist if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
Embodiment 8: The method of any one of the preceding embodiments, wherein the subject is assessed for presence of the HRD, e.g., VHRD, e.g., PHRD, prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist.
Embodiment 9: The method of embodiment 8, further comprising:
(a) altering or discontinuing the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye, or (b) maintaining the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined not to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
Embodiment 10: The method of any one of the preceding embodiments, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
Embodiment 11 : The method of any one of the preceding embodiments, wherein the HRD, e.g., VHRD, e.g., PHRD, is hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
Embodiment 12: The method of any one of the preceding embodiments, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
Embodiment 13: The method of any one of the preceding embodiments, wherein the VEGF antagonist is an anti -VEGF antibody or a fragment thereof.
Embodiment 14: The method of embodiment 13, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
Embodiment 15: The method of embodiment 13, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
Embodiment 16: The method of embodiment 13, wherein the VEGF antagonist is brolucizumab.
Embodiment 17: The method of any one of the preceding embodiments, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4- week intervals, and thereafter administering to the subject an additional dose every 8 to 12 weeks. Embodiment 18: The method of embodiment 17, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 12 weeks.
Embodiment 19: The method of embodiment 18, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
Embodiment 20: The method of embodiment 15, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 weeks.
Embodiment 21 : The method of embodiment 20, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
Embodiment 22: The method of any one of embodiments 14 to 21, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg.
Embodiment 23 : The method of embodiment 22, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
Embodiment 24: The method of any one of the preceding embodiments, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
Embodiment 25: The method of embodiment 24, wherein the ocular disease is neovascular age-related macular degeneration (nAMD). Embodiment 26: A method of predicting the likelihood that a subject having ocular disease will develop intraocular inflammation (IO I), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD , in particular the method comprising determining by OCT or SD-OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, wherein:
(a) the presence of a HRD, e.g., VHRD, e.g., PHRD, is indicative of an increased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist; and
(b) the absence of a HRD, e.g., VHRD, e.g., PHRD, is indicative of a decreased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist.
Embodiment 27: The method of embodiment 26 further comprising treating the subject with the VEGF antagonist if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
Embodiment 28: The method of any one of embodiments 26 to 27, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
Embodiment 29: The method of any one of embodiments 26 to 27, wherein the HRD, e.g., VHRD, e.g., PHRD, is a hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
Embodiment 30: The method of any one of embodiments 26 to 29, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
Embodiment 31 : The method of any one of embodiments 26 to 29, wherein the VEGF antagonist is an anti -VEGF antibody or a fragment thereof. Embodiment 32: The method of embodiment 31, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
Embodiment 33: The method of embodiment 31, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
Embodiment 34: The method of embodiment 31, wherein the VEGF antagonist is brolucizumab.
Embodiment 35: The method of any one of embodiments 32 to 34, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4- week intervals, and thereafter administering to the subject an additional dose every 8 to 12 weeks.
Embodiment 36: The method of embodiment 35, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 12 weeks.
Embodiment 37: The method of embodiment 36, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
Embodiment 38: The method of embodiment 35, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 weeks.
Embodiment 39: The method of embodiment 38, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
Embodiment 40: The method of any one of embodiments 32 to 39, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg. Embodiment 41 : The method of embodiment 40, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
Embodiment 42: The method of any one of embodiments 26 to 41, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
Embodiment 43: The method of embodiment 42, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
Embodiment 44: A VEGF antagonist for the use in treatment of ocular disease in a subject, characterized in that a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e g., VHRD, e g., PHRD.
Embodiment 45: A VEGF antagonist for the use in treatment of ocular disease in a subject, characterized in that:
(a) a subject is selected for treatment with the VEGF antagonist on the basis of the subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) thereafter, a therapeutically effective amount of the VEGF antagonist is administered to the subject.
Embodiment 46: A VEGF antagonist for the use in treatment of ocular disease in a subject, wherein the use comprises
(a) determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and (c) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
Embodiment 47: The VEGF antagonist for use of embodiment 45 or 46, wherein the use further comprises assessing the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, e.g., prior to the steps (a)-(b) of embodiment 45 or embodiment 46.
Embodiment 48: The VEGF antagonist for use of any one of embodiments 44 to 47, wherein the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, is assessed by performing OCT, e.g., SD-OCT, analysis on a subject’s eye.
Embodiment 49: A VEGF antagonist for the use in treatment of ocular disease in a subject, wherein the use comprises:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e.g., VHRD, e.g., PHRD; and
(b) determining if the subject has a HRD, e.g., VHRD, e.g., PHRD; and
(c) if the HRD, e.g., VHRD, e.g., PHRD, is not present, a therapeutically effective amount of the VEGF antagonist, e.g., brolucizumab, is to be administered to the subject; and
(d) if the HRD, e.g., VHRD, e.g., PHRD, is present, a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, is to be administered to the subject.
Embodiment 50: The VEGF antagonist for use of embodiment 49, wherein a therapeutically effective amount of the VEGF antagonist is to be administered to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
Embodiment 51 : The VEGF antagonist for use of any one of embodiments 44 to 50, wherein the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist. Embodiment 52: The VEGF antagonist for use of embodiment 51, wherein the use further comprises:
(a) altering or discontinuing the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye, or
(b) maintaining the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined not to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
Embodiment 53: The VEGF antagonist for use of any one of embodiments 44 to 52, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
Embodiment 54: The VEGF antagonist for use of any one of embodiments 44 to 53, wherein the HRD, e.g., VHRD, e.g., PHRD, is a hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material, e.g., stalagmite-like preretinal hyperreflective material.
Embodiment 55: The VEGF antagonist for use of any one of embodiments 44 to 54, wherein the VEGF antagonist is selected from the group consisting of brolucizumab, aflibercept, ranibizumab, faricimab, conbercept and abicipar.
Embodiment 56: The VEGF antagonist for use of any one of embodiments 44 to 55, wherein the VEGF antagonist is an anti-VEGF antibody or a fragment thereof.
Embodiment 57: The VEGF antagonist for use of embodiment 56, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
Embodiment 58: The VEGF antagonist for use of embodiment 56, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4. Embodiment 59: The VEGF antagonist for use of embodiment 56, wherein the VEGF antagonist is brolucizumab.
Embodiment 60: The VEGF antagonist for use of any one of embodiments 44 to 59, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 8 to 12 weeks.
Embodiment 61 : The VEGF antagonist for use of embodiment 60, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 12 weeks.
Embodiment 62: The VEGF antagonist for use of embodiment 61, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
Embodiment 63: The VEGF antagonist for use of embodiment 60, wherein the VEGF antagonist is administered to the subject in three individual doses of at 4-week intervals, and thereafter is administered to the subject as an additional dose every 8 weeks.
Embodiment 64: The VEGF antagonist for use of embodiment 63, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
Embodiment 65: The VEGF antagonist for use of any one of embodiments 51 to 58, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, in particular 6 mg.
Embodiment 66: The VEGF antagonist for use of embodiment 65, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
Embodiment 67: The VEGF antagonist for use of any one of embodiments 44 to 66, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
Embodiment 68: The VEGF antagonist for use of embodiment 67, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
Embodiment 69: A method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., neovascular age-related macular degeneration (nAMD), should be treated with a VEGF antagonist, the method comprising:
(a) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining that the subject should be treated with a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
Embodiment 70: A method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., neovascular age-related macular degeneration (nAMD), may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining an increased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining a decreased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
Specific preferred embodiments of the invention will become evident from the following more detailed description of certain preferred embodiments and the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Preretinal hyperreflective material. (A) Baseline OCT, smooth inner retinal boundary. (B) Pre-IOI event OCT, new appearance of preretinal hyperreflective material (arrow).
Figure 2. Preretinal hyperreflective material. (A) Baseline OCT: smooth inner retinal boundary; no preretinal hyperreflective material or vitreous debris observed . (B) V3/Pre-IOI event OCT: early signs of preretinal hyperreflective material (foci) / vitreous debris. (C) V7/IOI event OCT: vitreous debris with preretinal hyperreflective material (foci). (D) V3/Pre-IOI event OCT: numerous preretinal hyperreflective material (foci).
Figure 3. Radiomics evaluation.
Figure 4. Box and Whisker Plot of the most discriminating baseline feature.
Figure 5. Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective foci between eyes in the IOI group and the control group, including prior to the IOI reported event.
Figure 6. Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective preretinal foci (preretinal deposits) between eyes in the IOI group and the control group, including prior to the IOI reported event.
Figure 7. Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective vitreous foci (vitreous debris) between eyes in the IOI group and the control group, including prior to the IOI reported event. DETAILED DESCRIPTION
Definitions
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which this present disclosure pertains. Any references cited herein, including, e.g., all patents, published patent applications, and non-patent publications, are incorporated by reference in their entirety. To facilitate understanding of the disclosure, several terms and abbreviations as used herein are defined below as follows:
As used herein, all percentages are percentages by weight, unless stated otherwise.
As used herein and unless otherwise indicated, the terms “a” and “an” are taken to mean “one”, “at least one” or “one or more”. Unless otherwise required by context, singular terms used herein shall include pluralities and plural terms shall include the singular.
As used herein, the term “about” includes and describes the value or parameter per se. For example, “about x” includes and describes “x” per se. As used herein, the term “about” when used in association with a measurement, or used to modify a value, a unit, a constant, or a range of values, refers to variations of ±1-10% in addition to including the value or parameter per se. In some embodiments, the term “about” when used in association with a measurement, or used to modify a value, a unit, a constant, or a range of values, refers to variations of ±1, ±2, ±3, ±4, ±5, ±6, ±7, ±8, ±9, or ±10%.
The term “VEGF” refers to the 165-amino acid vascular endothelial cell growth factor, and related 121-, 189-, and 206-amino acid vascular endothelial cell growth factors, as described by Leung et ah, Science 246:1306 (1989), and Houck et ah, Mol. Endocrin. 5:1806 (1991) together with the naturally occurring allelic and processed forms of those growth factors. The term “VEGF”, in particular, refers to the human VEGF.
The term “VEGF receptor” or “VEGFr” refers to a cellular receptor for VEGF, ordinarily a cell-surface receptor found on vascular endothelial cells, as well as variants thereof retaining the ability to bind hVEGF. One example of a VEGF receptor is the fms-like tyrosine kinase (fit), a transmembrane receptor in the tyrosine kinase family. DeVries et ah, Science 255:989 (1992); Shibuya et al., Oncogene 5:519 (1990). The fit receptor comprises an extracellular domain, a transmembrane domain, and an intracellular domain with tyrosine kinase activity. The extracellular domain is involved in the binding of VEGF, whereas the intracellular domain is involved in signal transduction. Another example of a VEGF receptor is the flk-1 receptor (also referred to as KDR). Matthews et al., Proc. Nat. Acad. Sci. 88:9026 (1991); Terman et al., Oncogene 6:1677 (1991); Terman et al., Biochem. Biophys. Res. Commun. 187:1579 (1992). Binding of VEGF to the fit receptor results in the formation of at least two high molecular weight complexes, having an apparent molecular weight of 205,000 and 300,000 Daltons. The 300,000 Dalton complex is believed to be a dimer comprising two receptor molecules bound to a single molecule of VEGF.
As used herein, a “VEGF antagonist” refers to a compound that can diminish or inhibit VEGF activity in vivo. A VEGF antagonist can bind to a VEGF receptor(s) or block VEGF protein(s) from binding to VEGF receptor(s). A VEGF antagonist can be, for example, a small molecule, an anti-VEGF antibody or antigen-binding fragments thereof, fusion protein (such as aflibercept or other such soluble decoy receptor), an aptamer, an antisense nucleic acid molecule, an interfering RNA, receptor proteins, and the like that can bind specifically to one or more VEGF proteins or one or more VEGF receptors. Several VEGF antagonists are described in WO 2006/047325. In one embodiment, the VEGF antagonist is any licensed anti-VEGF drug such as brolucizumab, ranibizumab or aflibercept. In one embodiment, the VEGF antagonist is an anti-VEGF antibody (such as brolucizumab or ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF receptor domains, e.g., conbercept (such as ADVM-022 from Adverum) or any licensed anti-VEGF drug (such as brolucizumab, ranibizumab or aflibercept).
The term “antibody” as used herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”, “antigen binding polypeptide”, or “immunobinder”) or single chain thereof. An “antibody” includes a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The structures and locations of antibody variable domains, e.g., CDRs, may be defined using well known numbering schemes, e.g., the Rabat numbering scheme, the Chothia numbering scheme, or a combination of Rabat and Chothia (see, e.g., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services (1991), eds. Rabat et ah; A1 Lazikani et ah, (1997) J. Mol. Bio. 273:927 948); Rabat et al., (1991) Sequences of Proteins of Immunological Interest, 5th edit., NIH Publication no. 91-3242 U.S. Department of Health and Human Services; Chothia et al., (1987) J. Mol. Biol. 196:901-917; Chothia et al., (1989) Nature 342:877-883; and Al- Lazikani et al., (1997) J. Mai. Biol. 273:927-948), and numbering scheme described in Honegger and Pluckthun, J. Mol. Biol. 309 (2001) 657-670 ("AHo" numbering). For example, for classic formats, under Rabat, the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1, also referred to as CDRHl), 50-65 (HCDR2, also referred to as CDRH2), and 95-102 (HCDR3, also referred to as CDRH3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1, also referred to as CDRLl), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3). Under Chothia the CDR amino acids in the VH are numbered 26-32 (HCDR1, also referred to as CDRHl), 52-56 (HCDR2, also referred to as CDRH2), and 95-102 (HCDR3, also referred to as CDRH3); and the amino acid residues in VL are numbered 24-34 (LCDR1, also referred to as CDRLl), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3). By combining the CDR definitions of both Rabat and Chothia, the CDRs consist of amino acid residues 26-35 (HCDR1, also referred to as CDRHl), 50-65 (HCDR2, also referred to as CDRH2), and 95- 102 (HCDR3, also referred to as CDRH3) in human VH and amino acid residues 24- 34 (LCDR1, also referred to as CDRL1), 50-56 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3) in human VL. Under IMGT the CDR amino acid residues in the VH are numbered approximately 26-35 (HCDR1, also referred to as CDRH1), 51-57 (HCDR2, also referred to as CDRH2) and 93-102 (HCDR3, also referred to as CDRH3), and the CDR amino acid residues in the VL are numbered approximately 27-32 (LCDR1, also referred to as CDRLl), 50-52 (LCDR2, also referred to as CDRL2), and 89-97 (LCDR3, also referred to as CDRL3) (numbering according to "Rabat"). Under IMGT, the CDRs of an antibody can be determined using the program IMGT/DomainGap Align. In the context of the present invention, the numbering system suggested by Honegger and Pluckthun ("AHo) is used (Honegger and Pluckthun, J. Mol. Biol. 309 (2001) 657-670), unless specifically mentioned otherwise. Furthermore, the following residues are defined as CDRs according to AHo numbering scheme: LCDR1 (also referred to as CDRLl): L24-L42; LCDR2 (also referred to as CDRL2): L58-L72; LCDR3 (also referred to as CDRL3): L107- L138; HCDR1 (also referred to as CDRHl): H27-H42; HCDR2 (also referred to as CDRH2): H57-H76; HCDR3 (also referred to as CDRH3): H108-H138. For the sake of clarity, the numbering system according to Honegger and Pluckthun takes the length diversity into account that is found in naturally occurring antibodies, both in the different VH and VL subfamilies and, in particular, in the CDRs, and provides for gaps in the sequences. Thus, in a given antibody variable domain usually not all positions 1 to 149 will be occupied by an amino acid residue. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system ( e.g ., effector cells) and the first component (Clq) of the classical complement system.
The term “single chain antibody”, “single chain Fv” or “scFv” is intended to refer to a molecule comprising an antibody heavy chain variable domain (or region; VH) and an antibody light chain variable domain (or region; VL) connected by a linker. Such scFv molecules can have the general structures: NH2-VL-linker-VH-COOH or NH2-VH-linker- VL-COOH. The term “antigen-binding portion” of an antibody (or simply “antibody portion” or “antibody fragment”) refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., VEGF). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a single domain or dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins. Antibodies can be of different isotype, for example, an IgG (e.g., an IgGl, IgG2, IgG3, or IgG4 subtype), IgAl, IgA2, IgD, IgE, or IgM antibody.
The term “homologous” or “identity” refers to the subunit sequence identity between two polymeric molecules, e.g., between two nucleic acid molecules, such as, two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit; e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous or identical at that position. The homology between two sequences is a direct function of the number of matching or homologous positions; e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two sequences are homologous, the two sequences are 50% homologous; if 90% of the positions (e.g., 9 of 10), are matched or homologous, the two sequences are 90% homologous. Percentage of “sequence identity” can be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of the amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage can be calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity. The output is the percent identity of the subject sequence with respect to the query sequence.
As used herein, a “mammal” includes any animal classified as a mammal, including, but not limited to, humans, domestic animals, farm animals, and companion animals, etc.
As used herein, the term “subject” or “patient” refers to human and non-human mammals, including but, not limited to, primates, pigs, horses, dogs, cats, sheep, and cows. Preferably, a subject or patient is a human. In certain embodiments, a patient is at least 18 years of age. In certain embodiments, a patient is at least 50 years of age. In certain embodiments, a patient is at least 65 years of age.
The term “treat”, “treating” or “treatment” includes therapeutic treatments, prophylactic treatments and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses the reduction of the symptoms or underlying risk factors. As used herein, the terms “treat”, “treatment” and “treating” refer to the reduction or amelioration of the progression or severity of an ocular disease or the amelioration of one or more symptoms, suitably of one or more discernible symptoms of an ocular disease. In specific embodiments, the terms “treat”, “treatment” and “treating” refer to the amelioration of at least one measurable physical parameter of an ocular disease (such as achieve or at least partially achieve a desired effect (e.g. the partial or complete regression of retinal neovascularization, decrease of retinal fluid or achieving retinal fluid-free status, e.g., intraretinal fluid (IRF) and subretinal fluid (SRF), decrease of Central Subfield Thickness (CSFT), improvement in vision, e.g., a change of BCVA > 1, > 2, > 3, > 4 or > 5 letters, or a DRSS score < 61), wherein the physical parameter is not necessarily discernible by the patient.
A term “ocular disease” or “neovascular ocular disease”, as used herein, refers to a condition, disease, or disorder associated with ocular neovascularization. An “ocular disease” or “neovascular ocular disease” that can be treated using a method of the disclosure includes, a condition, disease, or disorder associated with ocular neovascularization, including, but not limited to, abnormal angiogenesis, choroidal neovascularization (CNV), choroidal neovascularization (CNV) associated with nAMD, retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
The term “loading phase” refers to the first 2, 3, 4, 5, or 6 doses of a VEGF antagonist administered at q4w or q6w intervals. Suitably, the term “loading phase” refers to the first 2 to 3 doses of a VEGF antagonist administered at q4w.
The term “maintenance phase” refers to additional doses at 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 week intervals, and can be adjusted as described herein based on Disease Activity Assessments as described herein. Suitably, the term “maintenance phase” refers to additional doses at 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 week intervals, and can be adjusted as described herein based on Disease Activity Assessments as described herein.
As used herein, an administration interval can be referred to as qXw, where the “X” is a number of weeks between administered doses. For example, q6w is an interval of 6 weeks.
As used herein, the term “week” means 7 days ± 1 day. As used herein, the term “month” means 25 to 31 days. Also, as used herein, the term “month” means 4 weeks. As used herein, the terms “effective amount” or “therapeutically effective amount” or “therapeutically effective dose” refer to an amount of a therapy (e.g., a VEGF antagonist, e.g., brolucizumab, or a pharmaceutical composition provided herein) which is sufficient to reduce and/or ameliorate the severity of a given condition, disorder, or disease and/or a symptom related thereto. The term “effective amount” or “therapeutically effective amount” or “therapeutically effective dose” is defined as an amount sufficient to achieve or at least partially achieve a desired effect (e.g. the partial or complete regression of retinal neovascularization, decrease of retinal fluid or achieving retinal fluid-free status, e.g., intraretinal fluid (IRF) and subretinal fluid (SRF), decrease of Central Subfield Thickness (CSFT), improvement in vision, e.g., a change of BCVA > 1, > 2, > 3, > 4 or > 5 letters, or a DRSS score < 61). A therapeutically effective dose is sufficient if it can produce even an incremental change in the symptoms or conditions associated with the disease. The therapeutically effective dose does not have to completely cure the disease or completely eliminate symptoms. Preferably, a therapeutically effective dose can at least partially arrest the disease and/or its complications in a patient already suffering from the disease. In specific embodiments, a therapeutic effective dose may involve repeated administration over a period of time. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient’s own immune system.
As used herein, the term “adverse event” refers to any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal (investigational) product, whether or not related to the medicinal (investigational) product. In particular, adverse events in ocular disease, e.g., nAMD, patients may include blurred visiona, cataract, conjunctival hemmorage, eye pain, vitreous floaters, intraocular inflammation, e.g., e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO). Specifically, the term refers to the effect that leads to intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion.
As used herein, the term “intraocular inflammation” encompasses anterior chamber cell, anterior chamber flare, anterior chamber inflammation, chorioretinitis, eye inflammation, iridocyclitis, iritis, uveitis, vitreous haze, vitritis. As used herein, the term “hyperreflective deposit” or “HRD” or“hyperreflective material” or “hyperreflective foci” or “hyperreflective foci formation” refers to a region of increased reflectivity on the tomographic scan, e.g., on spectral domain optical coherence tomography (SD-OCT). Hyperreflective material (hyperreflective deposit) may be classified according to its morphological appearance and location; hyperreflective material (hyperreflective deposit) with high reflectivity whose boundaries are clearly delineated from the surrounding neural components of the retina is classified as well-defined; hyperreflective material (hyperreflective deposit) with low reflectivity and whose borders are less well defined and therefore not easily distinguishable from surrounding neural components is classified as undefined (Casalino et al., Expert Review of Ophthalmology, 2020, Volume 15, No. 2, pp. 83-91). In one embodiment, a “hyperreflective deposit” or “HRD” or “hyperreflective material” or “hyperreflective foci formation” is a well-defined (with high reflectivity) hyperreflective material. Morphological appearance of hyperreflective material (hyperreflective deposit) includes a heterogeneity observed on the OCT scan including hyperreflective debris, hyperreflective round- or rod-shaped dots, e.g., stalagmite-like deposit or material. As used herein, the term “stalagmite-like hyperreflective deposit” or “stalagmite like hyperreflective material” or “stalagmite-like hyperreflective foci formation” refers to hyperreflective material that resemble the elevated speleothems, in particular as determined on spectral domain optical coherence tomography (SD-OCT). According to the present disclosure, the HRD, including debris, round- or rod-shaped dots and stalagmite-like HRD, is observed in vitreous compartment - a vitreous hyperreflective deposit (VHRD). In particular, according to the present disclosure, the HRD, including stalagmite-like hyperreflective material, is observed in preretinal compartment - a preretinal hyperreflective deposit (PHRD), e.g., internal limiting membrane (ILM) hyperreflective deposit (ILMHRD).
As used herein, “likelihood” and “likely” is a measurement of how probable an event is to occur. It may be used interchangeably with “probability”. Likelihood refers to a probability that is more than speculation, but less than certainty. Thus, an event is likely if a reasonable person using common sense, training or experience concludes that, given the circumstances, an event is probable. In some embodiments, once likelihood has been ascertained, the patient may be treated (or treatment continued) with the compound or the treatment may need to be altered or discontinued. In one embodiment, the “likelihood” and “likely” denote a chance in percent of how probable an event is to occur. The phrase “increased likelihood” refers to an increase in the probability that an event will occur. For example, some methods herein allow prediction of whether a patient will display an increased likelihood of developing intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion, in response to the administration of a VEGF antagonist, e.g., brolucizumab . In one embodiment the increased likelihood means that there is more than 50% chance, more than 60 % chance, more than 70 % or more than 80 % chance that an event will occur. Equally, a “decreased likelihood” means, that the chance is lower than 50%, lower than 60 %, lower than 70 % or lower than 80 %, respectively, that an event will occur.
As used herein, “predicting” indicates that the methods described herein provide information to enable a health care provider to determine the likelihood that an individual subjected to a treatment will develop with a higher likelihood intraocular inflammation, e.g., intraocular inflammation with retinal vasculitis and/or retinal occlusion. It does not refer to the ability to predict response with 100% accuracy. Instead, the skilled artisan will understand that it refers to an increased probability.
Methods of treatment
In one aspect, the disclosure provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having or having a low level of a hyperreflective deposit (HRD) in a subject’s eye, e.g., a vitreous hyperreflective deposit (VHRD), e.g., a preretinal hyperreflective deposit (PHRD), e.g., internal limiting membrane (ILM) hyperreflective deposit (ILMHRD). In a specific embodiment, the disclosure provides a method of treating ocular disease, e.g., neovascular age-related macular degeneration (nAMD), comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having or having a low level of a preretinal hyperreflective deposit (PHRD), e.g., internal limiting membrane (ILM) hyperreflective deposit (ILMHRD).
In one aspect, the disclosure provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject. In a specific embodiment, the disclosure provides a method of treating ocular disease, e.g., nAMD, comprising: (a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a PHRD or having a low level of a PHRD; and (b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
In a another aspect, the disclosure n provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g.,
PHRD. In a specific embodiment, the disclosure n provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a PHRD or having a low level of a PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a PHRD.
In one embodiment, the method further comprises assessing the presence or absence of HRD in a subject’s eye, e.g., VHRD, e.g., PHRD. Suitably, the assessment for the presence or absence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, is performed prior to the steps (a)-(b). Suitably, the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, e.g., presence or absence or level, is assessed by performing optical coherence tomography, e.g., spectral-domain optical coherence tomography, SD-OCT, analysis on a subject’s eye.
In a another aspect, the disclosure provides a method for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence or level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD; and
(b) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, if no HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or a low level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is observed in the subject’s eye;
(c) determining that the subject should not be treated with a VEGF antagonist, e.g., brolucizumab, if a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or a high level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is observed in the subject’s eye.
In a another aspect, the present invention provides a method of predicting the likelihood that a subject having ocular disease, e.g., nAMD, will develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, wherein: (a) the presence of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or the presence of a high level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is indicative of an increased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab; and| (b) the absence of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or the presence of a low level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is indicative of a decreased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab. In one embodiment, the method further comprises treating the subject with the VEGF antagonist if no HRD in a subject’s eye, e.g., VHRD, preferably PHRD, or a low level of a HRD in a subject’s eye, e.g., VHRD, preferably PHRD, is observed in the subject’s eye. In a specific embodiment, the method comprises determining by OCT or SD-OCT if the subject has a HRD in a subject’s eye, e.g., VHRD, preferably PHRD.
Suitably, according to the present disclosure, the subject may be assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist. In one embodiment, the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist. In one embodiment, the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and prior to an administration of the VEGF antagonist, e.g., on the same day and prior to the administration or 1, 2, 3, 4, 5, 6, or 7 days prior to the administration of the VEGF antagonist or 1, 2, 3, 4, 5, 6, or 7 weeks prior the administration of the VEGF antagonist. In one embodiment, the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and after an administration of the VEGF antagonist, e.g., on the same day and after the administration or on the 1st, 2nd , 3rd, 4th, 5th, 6th, or 7th day after the administration of the VEGF antagonist or 1, 2, 3, 4, 5, 6, or 7 weeks after the administration of the VEGF antagonist. In one embodiment, the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, during the treatment course with the VEGF antagonist and one month after the initial dose of the VEGF antagonist was administered to the subject.
According to the present disclosure, a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, may be used as a biomarker for the selection of the patients suitable for treatment with a VEGF inhibitor, e.g., brolucizumab, or for monitoring a patient during the course of treatment with a VEGF inhibitor, e.g., brolucizumab, and adjusting, if necessary, the treatment (e.g., treatment regimen (frequency and /or amount), choice of medicament). Thus, in one embodiment, the method of the present disclosure further comprises: (a) altering or discontinuing the treatment with the VEGF antagonist, e.g., brolucizumab, if, during the course of treatment, the subject is determined to have a HRD, e.g., VHRD, e.g., PHRD, or a high level of a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye, or (b) maintaining the treatment with the VEGF antagonist, e.g., brolucizumab, if, during the course of treatment, the subject is determined not to have a HRD, e.g., VHRD, e.g., PHRD, or a low level of a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
In one embodiment, the HRD, e.g., VHRD, e.g., PHRD, suitable for the methods of present invention has enough reflectivity to be determined as heterogeneity in comparison to the baseline level or in comparison to the surrounding components of the retina. In one embodiment, the HRD, e.g., VHRD, e.g., PHRD, suitable for the methods of present invention has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina. In one embodiment, the HRD, e.g., VHRD, e.g., PHRD, suitable for the methods of present invention is as a hyperreflective debris or a hyperreflective round- or rod-shaped dot. In a specific embodiment, the HRD, e.g., VHRD, e.g., PHRD, suitable for the methods of present invention is stalagmite-like hyperreflective material, e.g., stalagmite like preretinal hyperreflective material. In a specific embodiment, the HRD is PHRD having high reflectivity and is round- or rod-shaped dot, e.g., stalagmite-like. In a more specific embodiment, the PHRD has high reflectivity and is stalagmite-like.
Suitably, the methods of the present disclosure are suitable for treating ocular disease or for predicting the likelihood that a subject having ocular disease will develop intraocular inflammation (IOI), wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
In a specific embodiment, ocular disease is nAMD. Age-related macular degeneration (AMD) is the leading cause of severe vision loss in people affecting 10%-13% of individuals over the age of 65 in North America, Europe, and Australia (Kawasaki 2010, Rein et ah,
Arch Ophthalmol. 2009;127:533-40, Smith 2001). Genetic, environmental and health factors play an important role in the pathogenesis of the disease. AMD is classified into 2 clinical subtypes: the non-neovascular (atrophic) or dry form and the neovascular (exudative) or wet form (Ferris et ah, Arch Ophthalmol. 1984;102:1640-2, Lim et ah, Lancet. 2012;379:1728- 38, Miller et ah, Am J Ophthalmol. 2013;155:1-35). VEGF has been shown to be elevated in patients with nAMD and is thought to play a key role in the neovascularization process (Spilsbury et ak, Am J Pathol. 2000;157: 135-44). The use of intravitreal (IVT) pharmacotherapy targeting VEGF has significantly improved visual outcomes in patients with nAMD (Bloch et ak, Am J Ophthalmol. 2012;153:209-13, Campbell et ak, Arch Ophthalmol. 2012;130:794-5). Anti-VEGF treatments, such as ranibizumab (LUCENTIS®), aflibercept (EYLEA®), and brolucizumab (Beovu®), inhibit VEGF signaling pathways and have been shown to halt the growth of neovascular lesions and resolve retinal edema.
Diabetes mellitus (DM) is the most common endocrine disease in developed countries, with prevalence estimates ranging between 2 to 5% of the world population. Diabetic retinopathy (DR) and diabetic macular edema (DME) are common microvascular complications in patients with diabetes and may have a debilitating impact on visual acuity (VA), eventually leading to blindness.
In one embodiment, ocular disease is DME. DME is a frequent manifestation of DR (Riordan-Eva, 2004, Eye (Lond). 2004, 18:1161-8) and is the major cause of visual loss in patients with DR. For anti -VEGF agents like ranibizumab or aflibercept a favorable benefit risk ratio was demonstrated with superior efficacy versus the previous standard of care (laser photocoagulation) in large Phase 3 programs that consequently led to their approval for the treatment of DME. Anti- VEGF treatment led to clinically relevant improvements of BCVA, reduction of fluid accumulation and decreased severity of diabetic retinopathy.
In one embodiment, ocular disease is DR. DR is the most common cause of vision loss among people with diabetes and the leading cause of vision impairment and blindness in working aged adults. DR occurs when high blood glucose levels cause damage to blood vessels in the retina. These blood vessels can swell and leak or they can close, stopping blood from passing through. Sometimes abnormal new blood vessels can also grow on the retina. Diabetic retinopathy includes both non-proliferative diabetic retinopathy (nPDR) and proliferative diabetic retinopathy (PDR), the more advanced form of the disease. DME is a frequent manifestation of DR (Riordan-Eva, 2004, Eye (Lond). 2004, 18:1161-8) and is the major cause of visual loss in patients with DR. While diabetic macular edema (DME) may occur at any stage of DR, it is more likely to manifest following severe nPDR and PDR. Currently, care providers actively monitor mild to moderate nPDR and reserve treatment for severe nPDR and PDR. Both LUCENTIS® and EYLEA® have recently gained approval for DR in the United States. For treating DR, the recommended dose of EYLEA® is 2 mg (0.05 mL) administered every 4 weeks for 5 injections followed by an injection once every 8 weeks. For treating DR, the recommended dose of LUCENTIS® is 0.3 mg (0.05 mL) administered once a month. In one embodiment, ocular disease is Retinal Vein Occlusion (RVO), e.g., Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO). Retinal vein occlusion is a blockage of the small veins that carry blood away from the retina. The retina is the layer of tissue at the back of the inner eye that converts light images to nerve signals and sends them to the brain. Retinal vein occlusion is most often caused by hardening of the arteries (atherosclerosis) and the formation of a blood clot. Both CRVO and BRVO are both related to occlusion of the retinal vein, however the cause of the occlusion differs based on location. Central Retinal Vein Occlusion (CRVO) occurs with a thrombus occludes the central retinal vein near the lamina cribosa (Green et ah, Retina. 1:27-55, 1981). Branch Retinal Vein Occlusion (BRVO) occurs when a thrombus occurs at the arteriovenous crossing point secondary to atherosclerosis of the retinal artery causing compression and occlusion of the retinal vein. (Frangieh et ah, Archives of Ophthalmology. 100:1132-40, 1982). Blockage of retinal veins may cause other eye problems, including: (i) glaucoma (high pressure in the eye), caused by new, abnormal blood vessels growing in the front part of the eye; (ii) macular edema, caused by the leakage of fluid in the retina. VEGF has a leading role in RVO pathogenesis and symptoms. Use of anti -VEGF agents by intravitreal injections has become common with the aim to improve the clinical outcomes in these patients. Currently 2 anti-VEGF agents (ranimizumab and aflibercept) have been FDA (Food and Drug Administration) and EMA (European Medicine Agency) approved for the treatment of RVO, while another VEGF inhibitor (bevacizumab) is often used “off-label” in clinical practice.
In a another aspect, the present invention provides a method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining that the subject should be treated with a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a high level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and (c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
In a another aspect, the present invention provides a method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., nAMD, may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining an increased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a high level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining a decreased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, or having a low level of a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
Anti- VEGF Antagonists
The present disclosure provides methods and uses for treating ocular disease, e.g., nAMD, the method or the use comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
In one embodiment, the VEGF antagonist of the disclosure is any licensed anti-VEGF drug such as brolucizumab, ranibizumab or aflibercept. In one embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody (such as brolucizumab or ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF receptor domains, e.g., conbercept (such as ADVM-022 from Adverum) or any licensed anti-VEGF drug (such as brolucizumab, ranibizumab or aflibercept). In a preferred embodiment, the VEGF antagonist of the disclosure is brolucizumab or ranibizumab or aflibercept. In a more preferred embodiment, the VEGF antagonist of the disclosure is brolucizumab.
In certain embodiments, the VEGF antagonist of the disclosure is an anti-VEGF antibody, e.g., a single chain antibody (scFv) or Fab fragment.
In certain embodiments, the VEGF antagonist of the disclosure is an anti-VEGF antibody, particularly anti-VEGF antibodies described in WO 2009/155724, the entire contents of which are hereby incorporated by reference.
In one embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
VH: SEQ ID NO. 1
EVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGLEWVGFIDPDDDPYYATWAKGRFT
ISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSGWGLDIWGQGTLVTVSS
VL: SEQ ID NO. 2
EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRFSGSGSGA
EFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLG
In another embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising the sequence as set forth in SEQ ID NO: 3.
EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRFSGSGSGA
EFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGGSGGGGSGGGGSEVQLVES
GGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGLEWVGFIDPDDDPYYATWAKGRFTISRDNSK
NTLYLQMNSLRAEDTAVYYCAGGDHNSGWGLDIWGQGTLVTVSS In a preferred embodiment, the VEGF antagonist of the disclosure is brolucizumab (which comprises the sequence of SEQ ID NO: 3). The sequence of brolucizumab is set forth in SEQ ID NO: 4. A methionine derived from the start codon in an expression vector is present in the final protein in cases where it has not been cleaved posttranslationally as follows.
MEIVMTQSPS TLSASVGDRV IITCQASEII HSWLAWYQQK PGKAPKLLIY LASTLASGVP SRFSGSGSGA EFTLTISSLQ PDDFATYYCQ NVYLASTNGA NFGQGTKLTV LGGGGGSGGG GSGGGGSGGG GSEVQLVESG GGLVQPGGSL RLSCTASGFS LTDYYYMTWV RQAPGKGLEW VGFIDPDDDP YYATWAKGRF TISRDNSKNT LYLQMNSLRA EDTAVYYCAG GDHNSGWGLD iwGQGTLVTV ss (SEQ ID N0: 4)
In another embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising three light chain CDRs (CDRL1, CDRL2, and CDRL3) and three heavy chain CDRs (CDRH1, a CDRH2, a CDRH3) as follows:
CDRL1 QASEIIHSWLA SEQ ID NO: 5
CDRL2 LASTLAS SEQ ID NO: 6
CDRL3 QNVYLASTNGAN SEQ ID NO: 7
CDRH1 GFSLTDYYYMT SEQ ID NO: 8
CDRH2 FIDPDDDPYYATWAKG SEQ ID NO: 9
CDRH3 GDHNSGWGLDI SEQ ID NO: 10
In another embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising a heavy chain complementarity determining region 1 (CDRH1) comprising or consisting of the amino acid sequence of SEQ ID NO: 8; a heavy chain complementarity determining region 2 (CDRH2) comprising or consisting of the amino acid sequence of SEQ ID NO: 9; a heavy chain complementarity determining region 3 (CDRH3) comprising or consisting of the amino acid sequence of SEQ ID NO: 10; a light chain complementarity determining region 1 (CDRLl) comprising or consisting of the amino acid sequence of SEQ ID NO: 5; a light chain complementarity determining region 2 (CDRL2) comprising or consisting of the amino acid sequence of SEQ ID NO: 6; and a light chain complementarity determining region 3 (CDRL3) comprising or consisting of the amino acid sequence of SEQ ID NO: 7. In one embodiment, the CDRH1, CDRH2, CDRH3, CDRLl, CDRL2 and CDRL3 are defined according to the AHo numbering scheme. In another embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising:
(i) a variable heavy chain comprising or having the sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 1, and wherein the variable heavy chain comprises CDRH1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8, CDRH2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9, CDRH3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10, and
(ii) a variable light chain comprising or having the sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 2, and wherein the variable light chain comprises CDRL1 comprising or consisting of the amino acid sequence of SEQ ID NO: 5, CDRL2 comprising or consisting of the amino acid sequence of SEQ ID NO: 6, and CDRL3 comprising or consisting of the amino acid sequence of SEQ ID NO: 7. In one embodiment, the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3 are defined according to the AHo numbering scheme.
In another embodiment, the VEGF antagonist of the disclosure is an anti-VEGF antibody comprising or having a sequence having at least 80% identity, e.g., 85%, 90%, 95%, to SEQ ID NO: 3 or SEQ ID NO: 3, and wherein the anti-VEGF antibody comprises CDRH1 comprising or consisting of the amino acid sequence of SEQ ID NO: 8; CDRH2 comprising or consisting of the amino acid sequence of SEQ ID NO: 9; CDRH3 comprising or consisting of the amino acid sequence of SEQ ID NO: 10; CDRL1 comprising or consisting of the amino acid sequence of SEQ ID NO: 5; CDRL2 comprising or consisting of the amino acid sequence of SEQ ID NO: 6; and CDRL3 comprising or consisting of the amino acid sequence of SEQ ID NO: 7. In one embodiment, the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3 are defined according to the AHo numbering scheme.
Brolucizumab, is a humanized single-chain Fv (scFv) antibody fragment inhibitor of VEGF with a molecular weight of ~26 kDa. It is an inhibitor of VEGF -A and works by binding to the receptor binding site of the VEGF -A molecule, thereby preventing the interaction of VEGF-A with its receptors VEGFRl and VEGFR2 on the surface of endothelial cells. Increased levels of signaling through the VEGF pathway are associated with pathologic ocular angiogenesis and retinal edema. Inhibition of the VEGF pathway has been shown to inhibit the growth of neovascular lesions and resolve retinal edema in patients with nAMD.
In one embodiment, the disclosure provides a method of selectively treating a subject having ocular disease, e.g., nAMD, the method comprising: (a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or (b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g.,
PHRD.
In one embodiment, said different ocular disease agent is another VEGF antagonist, such as any licensed anti-VEGF drug, e.g., ranibizumab or aflibercept. In one embodiment, said different ocular disease agent is an anti-VEGF antibody (such as ranibizumab or bevacizumab or a bi-specific antibody such as faricimab) or an anti-VEGF DARPin (such as abicipar) or a soluble VEGF receptor (e.g., a fusion protein composed of the VEGF receptor domains, such as a fusion protein composed of the combination between VEGF receptor domains with the Fc fragment of human immunoglobulin with the Fc fragment of human immunoglobulin, e.g., conbercept, aflibercept) or AAV containing a sequence encoding for an anti-VEGF antibody (such as RGX-314 from Regenxbio), or AAV containing a sequence encoding the VEGF receptor domains, e.g., conbercept (such as ADVM-022 from Adverum) or any licensed anti-VEGF drug (such as ranibizumab or aflibercept). In a preferred embodiment, said different ocular disease agent is ranibizumab or aflibercept or faricimab. In a further embodiment, said different ocular disease agent is not a VEGF antagonist.
In certain embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered by an injection, e.g., an intravitreal injection.
In some embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of about 1, about 2, about 3, about 4, about 5, or about 6 mg (e.g., about 6 mg/0.05 mL) as an intravitreal injection. In certain embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of 1, 2, 3, 4, 5, or 6 mg (e.g., 6 mg/0.05 mL) as an intravitreal injection. In some embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.5, about 1 or about 2 mg (e.g., about 2 mg/0.05 mL) as an intravitreal injection. In certain embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of 0.5, 1 or 2 mg (e.g., 2 mg/0.05 mL) as an intravitreal injection. In some embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.2, about 0.3, about 0.4 or about 0.5 mg (e.g., about 0.5 mg/0.05 mL) as an intravitreal injection. In certain embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of 0.2, 0.3, 0.4 or 0.5 mg (e.g., 0.5 mg/0.05 mL) as an intravitreal injection.
Pharmaceutical Preparations
In one aspect, the methods or uses of the disclosure comprise the use of pharmaceutical formulations or pharmaceutical compositions comprising a VEGF antagonist, e.g., an anti-VEGF antibody. The term “pharmaceutical formulation” or “pharmaceutical composition” refers to preparations which are in such form as to permit the biological activity of the antagonist, e.g., antibody or antibody derivative, to be unequivocally effective, and which contain no additional components which are toxic to the subjects to which the formulation or composition would be administered. “Pharmaceutically acceptable” excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
A “stable” formulation is one in which a therapeutic agent, e.g. a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative thereof, essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10: 29-90 (1993), for example. Stability can be measured at a selected temperature for a selected time period. Preferably, the formulation is stable at room temperature (about 30° C) or at 40° C for at least 1 week and/or stable at about 2-8° C for at least 3 months to 2 years. Furthermore, the formulation is preferably stable following freezing (to, e.g., -70° C) and thawing of the formulation. An antagonist, e.g., an antibody or antibody derivative, “retains its physical stability” in a pharmaceutical formulation if it meets the defined release specifications for aggregation, degradation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography, or other suitable art recognized methods.
An antagonist, e.g., an antibody or antibody derivative, “retains its chemical stability” in a pharmaceutical formulation, if the chemical stability at a given time is such that the compound, e.g., protein, is considered to still retain its biological activity as defined below. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF MS), for example. Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange chromatography, for example.
An antagonist, e.g., an antibody or antibody derivative, “retains its biological activity” in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared as determined in an antigen binding assay, for example. Other “biological activity” assays for antibodies are elaborated herein below.
By “isotonic” is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer, for example.
A “polyol” is a substance with multiple hydroxyl groups, and includes sugars (reducing and non-reducing sugars), sugar alcohols and sugar acids. Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g. in the range from about 120 to about 400 kD). A “reducing sugar” is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a “non reducing sugar” is one which does not have these properties of a reducing sugar. Examples of reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose. Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffmose. Mannitol, xylitol, erythritol, threitol, sorbitol and glycerol are examples of sugar alcohols. As to sugar acids, these include L-gluconate and metallic salts thereof. Where it is desired that the formulation is freeze-thaw stable, the polyol is preferably one which does not crystallize at freezing temperatures (e.g. -20° C) such that it destabilizes the antibody in the formulation. Non-reducing sugars such as sucrose and trehalose are the preferred polyols herein, with trehalose being preferred over sucrose, because of the superior solution stability of trehalose.
As used herein, “buffer” refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components. The buffer of this disclosure has a pH in the range from about 4.5 to about 8.0; preferably from about 5.5 to about 7. Examples of buffers that will control the pH in this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. Where a freeze-thaw stable formulation is desired, the buffer is preferably not phosphate.
In a pharmacological sense, in the context of the present disclosure, a “therapeutically effective amount” of a therapeutic agent, e.g., a VEGF antagonist, e.g., an anti-VEGF antibody or antibody derivative, refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antagonist, e.g., antibody or antibody derivative, is effective. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
A “preservative” is a compound which can be included in the formulation to essentially reduce bacterial action therein, thus facilitating the production of a multi-use formulation, for example. Examples of potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride. Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol. The most preferred preservative herein is benzyl alcohol. The pharmaceutical compositions used in present disclosure comprise a VEGF antagonist A, preferably an anti-VEGF antibody (e.g., an anti-VEGF antibody comprising the variable light chain sequence of SEQ ID NO: 1 and the variable heavy chain sequence of SEQ ID NO: 2, such as brolucizumab), together with at least one physiologically acceptable carrier or excipient. Pharmaceutical compositions may comprise, for example, one or more of water, buffers (e.g., neutral buffered saline or phosphate buffered saline), ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g, glucose, mannose, sucrose or dextrans), mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives. As noted above, other active ingredients may (but need not) be included in the pharmaceutical compositions provided herein.
A carrier is a substance that may be associated with an antibody or antibody derivative prior to administration to a patient, often for the purpose of controlling stability or bioavailability of the compound. Carriers for use within such formulations are generally biocompatible, and may also be biodegradable. Carriers include, for example, monovalent or multivalent molecules such as serum albumin (e.g, human or bovine), egg albumin, peptides, polylysine and polysaccharides such as aminodextran and polyamidoamines. Carriers also include solid support materials such as beads and microparticles comprising, for example, polylactate polyglycolate, poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose or dextran. A carrier may bear the compounds in a variety of ways, including covalent bonding (either directly or via a linker group), noncovalent interaction or admixture.
Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, topical, intraocular, oral, nasal, rectal or parenteral administration. In certain embodiments, compositions in a form suitable for intraocular injection, such as intravitreal injection, are preferred. Other forms include, for example, pills, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Within yet other embodiments, compositions provided herein may be formulated as a lyophilizate. The term parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g, intravenous), intramuscular, spinal, intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique. The pharmaceutical composition may be prepared as a sterile injectible aqueous or oleaginous suspension in which the active agent (i.e. VEGF antagonist), depending on the vehicle and concentration used, is either suspended or dissolved in the vehicle. Such a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above. Among the acceptable vehicles and solvents that may be employed are water, 1,3-butanediol, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectible compositions, and adjuvants such as local anesthetics, preservatives and/or buffering agents can be dissolved in the vehicle.
An aqueous formulation of a VEGF antagonist, e.g., an anti-VEGF antibody (e.g., brolucizumab), used in the methods or uses of the disclosure is prepared in a pH-buffered solution. Preferably, the buffer of such aqueous formulation has a pH in the range from about
4.5 to about 8.0, preferably from about 5.5 to about 7.0, most preferably about 6.75. In one embodiment, the pH of an aqueous pharmaceutical composition of the disclosure is about
7.0-7.5, or about 7.0-7.4, about 7.0-7.3, about 7.0-7.2, about 7.1-7.6, about 7.2-7.6, about 7.3-
7.6 or about 7.4-7.6. In one embodiment, an aqueous pharmaceutical composition of the disclosure has a pH of about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5 or about 7.6. In a preferred embodiment, the aqueous pharmaceutical composition has a pH of >7.0 In a preferred embodiment, the aqueous pharmaceutical composition has a pH of about 7.2. In another preferred embodiment, the aqueous pharmaceutical composition has a pH of about 7.4. In another preferred embodiment, the aqueous pharmaceutical composition has a pH of about 7.6. Examples of buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers. The buffer concentration can be from about 1 mM to about 50 mM, preferably from about 5 mM to about 30 mM, depending, for example, on the buffer and the desired isotonicity of the formulation.
A polyol, which acts as a tonicifier, may be used to stabilize an antibody in an aqueous formulation. In preferred embodiments, the polyol is a non-reducing sugar, such as sucrose or trehalose. If desired, the polyol is added to the formulation in an amount that may vary with respect to the desired isotonicity of the formulation. Preferably the aqueous formulation is isotonic, in which case suitable concentrations of the polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% w/v, for example. However, hypertonic or hypotonic formulations may also be suitable. The amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose).
A surfactant is also added to an aqueous antibody formulation. Exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80 etc) or poloxamers (e.g. poloxamer 188). The amount of surfactant added is such that it reduces aggregation of the formulated antibody/antibody derivative and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. For example, the surfactant may be present in the formulation in an amount from about 0.001% to about 0.5%, preferably from about 0.005% to about 0.2% and most preferably from about 0.01% to about 0.1%.
In one embodiment, an aqueous antibody formulation used in the methods or uses of the disclosure is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl. In another embodiment, a preservative may be included in the formulation, particularly where the formulation is a multidose formulation. The concentration of preservative may be in the range from about 0.1% to about 2%, most preferably from about 0.5% to about 1%. One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 21st edition, Osol, A. Ed. (2006) may be included in the formulation provided that they do not adversely affect the desired characteristics of the formulation. Acceptable carriers, excipients or stabilizers are non-toxic to recipients at the dosages and concentrations employed and include: additional buffering agents, co-solvents, antioxidants including ascorbic acid and methionine, chelating agents such as EDTA, metal complexes (e.g. Zn- protein complexes), biodegradable polymers such as polyesters, and/or salt-forming counterions such as sodium. Formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation.
In one embodiment, the VEGF antagonist of the disclosure is administered to an eye of a subject in need of treatment in accordance with known methods for ocular delivery. Preferably, the subject is a human, the VEGF antagonist is an anti-VEGF antibody (preferably brolucizumab), and the antibody is administered directly to an eye.
Administration to a patient can be accomplished, for example, by intravitreal injection.
The VEGF antagonist in the methods and uses of the disclosure can be administered as the sole treatment or in conjunction with other drugs or therapies useful in treating the condition in question.
A preferred formulation for brolucizumab for intravitreal injection comprises about 4.5% to 11% (w/v) sucrose, 5-20 mM sodium citrate, and 0.001% to 0.05% (w/v) polysorbate 80, wherein the pH of the formulation is about 7.0 to about 7.4. One such formulation comprises 5.9% (w/v) sucrose, 10 mM sodium citrate, 0.02% (w/v) polysorbate 80, pH of 7.2, and 6 mg of brolucizumab. Another such formulation comprises 6.4% (w/v) or 5.8% sucrose, 12 mM or 10 mM sodium citrate, 0.02% (w/v) polysorbate 80, pH of 7.2, and 3 mg of brolucizumab. Preferred concentrations of brolucizumab are about 120 mg/ml and about 60 mg/ml. Doses can be delivered, for example as 6 mg/50 pL and 3 mg/50 pL concentrations.
Dosage and dosing regimen
A dose used in the methods or uses of the disclosure is based on the specific disease or condition being treated, and is therapeutically effective dose. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient’s own immune system. The dose amount can be readily determined using known dosage adjustment techniques by a physician having ordinary skill in treatment of the disease or condition. The therapeutically effective amount of a VEGF antagonist used in the methods or uses of the disclosure is determined by taking into account the desired dose volumes and mode(s) of administration, for example. Typically, therapeutically effective compositions are administered in a dosage ranging from 0.001 mg/ml to about 200 mg/ml per dose.
In one embodiment of the present disclosure, the VEGF antagonist used in the methods or uses of the disclosure is brolucizumab, and a dosage thereof used in the methods or uses of the disclosure is about 60 mg/ml to about 120 mg/ml (for example, a dosage is 60, 70, 80, 90, 100, 110, or 120 mg/ml). In a preferred embodiment, the dosage of the VEGF antagonist used in the methods or uses of the disclosure is 60 mg/ml or 120 mg/ml.
In certain embodiments, a dose of the VEGF antagonist is administered directly to an eye of a patient. In one embodiment, a dose of the VEGF antagonist per eye is at least about 0.5 mg up to about 6 mg. Preferred doses per eye include about 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.2 mg, 1.4 mg, 1.6 mg, 1.8 mg, 2.0 mg, 2.5 mg, 3.0 mg, 3.5 mg, 4.0 mg, 4.5 mg, 5.0 mg, 5.5 mg, and 6.0 mg. In one embodiment, a dose per eye is at least about 3 mg up to about 6 mg, e.g., about 3 mg or about 6 mg. Doses can be administered in various volumes suitable for ophthalmic administration, such as 50 pi or 100 mΐ, for example, including 3 mg/50 mΐ or 6 mg/50 mΐ. Smaller volumes can also be used, including 20 mΐ or less, for example about 20 mΐ, about 10 mΐ, or about 8.0 mΐ. In certain embodiments, a dose of 2.4 mg/20 mΐ, 1.2 mg/10 mΐ or 1 mg/8.0 mΐ (e.g., 1 mg/8.3 mΐ) is delivered to an eye of a patient for treating or ameliorating one or more of the diseases and disorders described above. Delivery can be, for example, by an injections, e.g., an intravitreal injection.
In specific embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered at a dose of about 1, about 2, about 3, about 4, about 5, or about 6 mg (e.g., about 6 mg/0.05 mL), e.g., 1, 2, 3, 4, 5, or 6 mg (e.g., 6 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
In specific embodiments, the VEGF antagonist of the disclosure is brolucizumab and is administered to the subject in three individual doses at 4-week intervals, and thereafter administered to the subject as an additional dose every 8 to 12 weeks, e.g., every 8 weeks or every 12 weeks. In some cases, a patient might be on a 12-week interval regimen for some time, and then switch to an 8-week interval, and then switch back to the 12-week interval. Thus, patients may not stay on one interval regimen, and may go back and forth depending on assessments according to the criteria set forth herein. For example, a 12 week treatment interval may be switched to an 8 week treatment interval if a disease activity is detected in the subject, or an 8 week treatment interval may be switched to a 12 week treatment interval if no disease activity is detected in the subject.
Suitably, the disease activity may be assessed based on visual function, retinal structure and leakage. An assessment as described herein preferably includes one or more of the following tests to assess activity of a VEGF antagonist (e.g., brolucizumab) on visual function, retinal structure and leakage: (i) best corrected visual acuity (BCVA), e.g., best corrected visual acuity with ETDRS-like chart at 4 meters, (ii) visual acuity (VA), (iii) central subfield thickness (CSFT), (iv) presence of intraretinal cysts/fluid, (v) ETDRS DRSS score based on 7-field stereo Color Fundus Photography (CFP), (vi) anatomical retinal evaluation by Optical Coherence Tomography (OCT), standard or wide-field Fluorescein Angiography (FA), OCT angiography, and/or wide-field CFP/FA, (vii) peripheral visual field assessed by perimetry, (viii) contrast sensitivity, (viii) severity of diabetic retinopathy.
Visual acuity can be assessed using best correction determined from protocol refraction (BCVA). BCVA measurements can be taken, for example, in a sitting position using ETDRS-like visual acuity testing charts.
Optical Coherence Tomography (OCT), color fundus photography and fluorescein angiography can be assessed according to methods known to those of skill in the art.
The CST is the average thickness of circular 1 mm area centered around the fovea measured from retinal pigment epithelium (RPE) to the internal limiting membrane (ILM), inclusively. CST can be measured, for example, using spectral domain Optical Coherence Tomography (SD-OCT).
Means of performing the above tests are well understood and commonly used by those skilled in the art.
Suitably, the disease activity may be assessed based on one or more of the following: (i) best corrected visual acuity (BCVA), (ii) visual acuity (VA), (iii) central subfield thickness (CSFT), and (iv) presence of intraretinal cysts/fluid. The presence of ocular disease activity, e.g., nAMD disease activity, includes one or more of the following: (i) decrease in Best Corrected Visual Acuity (BCVA), (ii) decrease in Visual Acuity (VA), (iii) increase or lack of reduction in Central Subfield Thickness (CSFT), (iv) new or persistent or recurrent Intraretinal Cysts (IRC) and/or Intraretinal Fluid (IRF) and/or Subretinal Fluid (SRF). Fluid measured in the eye can be intraretinal and/or subretinal fluid.
In specific embodiments, the presence of ocular disease activity, e.g., nAMD disease activity, includes one or more of the following:
(i) decrease in BCVA of >2 letters, e.g., decrease in BCVA of >3 letters, decrease in
BCVA of >4 letters, e.g., decrease in BCVA of >5 letters, more particularly wherein:
• the decrease in BCVA is observed at Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline BCVA, wherein the baseline BCVA was assessed prior to the last administration of the VEGF antagonist, or
• the decrease in BCVA is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline BCVA, wherein the baseline BCVA was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
(ii) decrease in VA of >1 letters, e.g., decrease in VA of >2 letters, e.g., decrease in VA of >3 letters, more particularly wherein:
• the decrease in VA is observedat Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline VA, wherein the baseline VA was assessed prior to the last administration of the VEGF antagonist, or
• the decrease in VA is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline VA, wherein the baseline VA was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
(iii) CSFT increase >25pm, e.g., CSFT increase >50pm, e.g., CSFT increase >75pm, more particularly wherein:
• the CSFT increase is observed at Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline CSFT, wherein the baseline CSFT was assessed prior to the last administration of the VEGF antagonist, or
• the CSFT increase is observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline CSFT, wherein the baseline CSFT was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist;
(iv) new or persistent or recurrent intraretinal cysts (IRC) and/or intraretinal fluid (IRF) and/or subretinal fluid (SRF), e.g., wherein:
• new or persistent or recurrent intraretinal cysts (IRC) and/or intraretinal fluid (IRF) and/or subretinal fluid (SRF) are observed at at Week 8 or Week 12 after the last administration of the VEGF antagonist (e.g., brolucizumab) compared to a baseline RC and/or IRF and/or SRF, wherein the baseline RC and/or IRF and/or SRF was assessed prior to the last administration of the VEGF antagonist, or
• new or persistent or recurrent intraretinal cysts (IRC) and/or intraretinal fluid (IRF) and/or subretinal fluid (SRF) are observed after 4 months or longer, e.g., after 6 months or longer, of the administration of the VEGF antagonist compared to a baseline IRC and/or IRF and/or SRF, wherein the baseline IRC and/or IRF and/or SRF was assessed 4 months or longer, e.g., 6 months or longer, prior to the last administration of the VEGF antagonist.
Where disease activity is present (for example, loss of letters measured by BCVA, increase in CST, increased fluid accumulation, and or increased severity of ocular disease compared with baseline reading for the patient or compared with any previous assessment), a more frequent dosing interval is prescribed going forward. Where improvement of disease activity is observed, a less frequent dosing interval is prescribed.
In some embodiments, the VEGF antagonist of the disclosure is aflibercept and is administered at a dose of about 0.5, about 1 or about 2 mg (e.g., about 2 mg/0.05 mL), e.g., 0.5, 1 or 2 mg (e.g., 2 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
In some embodiments, the VEGF antagonist used in the methods or uses of the disclosure is ranibizumab and is administered at a dose of about 0.2, about 0.3, about 0.4 or about 0.5 mg (e.g., about 0.5 mg/0.05 mL), e.g., 0.2, 0.3, 0.4 or 0.5 mg (e.g., 0.5 mg/0.05 mL), as an injections, e.g., an intravitreal injection.
The various features, aspects and embodiments of the present invention, referred to in individual sections above apply, as appropriate, to other sections, mutatis mutandis.
Consequently features specified in one section may be combined with features specified in other sections, as appropriate.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific aspects and embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
All references cited herein, including patents, patent applications, papers, publications, text books, and the like, and the references cited therein, to the extent that they are not already, are hereby incorporated herein by reference in their entirety The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLES
Example 1: Safety-specific Next Generation HA WK OCT Analysis
Purpose: To perform a comparative discovery assessment to evaluate the presence of OCT features that may precede or develop in association with intraocular inflammation (IOI), which might serve as OCT biomarkers for IOI.
Methods: HAWK is a randomized, double-masked, 96-week phase 3 studies comparing brolucizumab to aflibercept in nAMD. The Safety Review Committee (SRC) performed an independent unmasked post hoc review of all cases of investigator-reported IOI, retinal vascular occlusions and endophthalmitis in the phase 3 HAWK and HARRIER studies. Following this analysis, a higher order comparative discovery analysis was initiated evaluating 34 eyes from the HAWK study identified through the SRC analysis and compared to 34 propensity-matched controls. This analysis includes comparative higher-order OCT, radiomics assessment, and machine learning classification. This report provides an initial assessment of the qualitative comparative discovery analysis of the Safety next Generation imaging study (SAGE). Images were reviewed frame-by-frame for specific features prior to or during the IOI event.
Results:
Qualitative review of the cases of interest identified a characteristic preretinal hyperreflective deposit, “stalagmite” in 20 of 34 eyes (59%) with IOI compared to 1 of 34 eyes (3%) in the controls (p < 0.001) (Figure 1). This one control subject was noted to have an adverse event secondary to floaters at the time the “stalagmites” were noted on OCT, suggesting a possible underlying IOI event. The presence of the “stalagmites” were identified at the time IOI was initially noted in 11 subjects and was visualized at visits prior to the reported IOI event in 9 subjects. When “stalagmites” were identified at visits prior to the reported IOI event, 77% of these eyes ultimately developed retinal vasculitis and/or vascular occlusive event.
Further qualitative review of the cases of interest identified a characteristic preretinal hyperreflective deposit (PHRD) and / or vitreous debris in eyes with IOI compared to in the controls (Figure 2). Qualitative review of the cases of interest identified a characteristic preretinal hyperreflective deposit or vitreous debris in 20 of 34 eyes (59%) with IOI compared to 1 of 34 eyes (3%) in the controls (p < 0.001). Out of 20 eyes with preretinal hyperreflective deposit or vitreous debris, in 11/20 eyes preretinal hyperreflective deposit and/or vitreous debris were observed at time of the IOI event with 8/11 eyes (72%) defined as having IOI + RV/RO, and in 9/20 eyes preretinal hyperreflective deposit and/or vitreous debris were observed prior to the IOI event with 8/14 eyes (57%) defined as having IOI + RV/RO.
Example 2: Predictive Assessment of Radio mic features to identify eyes with Intraocular Inflammation (101) on HAWK-SAGE data
In addition, radiomics assessment has been performed (Figure 3) to evaluate potential pre- inflammatory event signals within the vitreous compartment. Specifically, the assessment was conducted to identify and characterize vitreous compartment features that might provide early insights into pre-inflammatory event signals.
To evaluate whether radiomics features extracted from vitreous compartment can distinguish eyes (N=68) with intraocular inflammation (IOI). Last timepoint represented the “safety” event or when inflammation develops. The last timepoint vitreous compartment between the Safety group and the Control group was compared. In addition, an analysis of the timepoint immediately before the final timepoint was done to assess if we can tell a difference between the groups prior to actual events. Finally, assessment at the month one timepoint was conducted to assess whether after a single injection it is possible to identify a difference between those two groups.
The Control group was N=34; the Safety group (N=34) was divided into two subgroups: SI (N=20) - with visible OCT findings, and S2 (N=14) - without visible OCT findings.
Supervised Classification for distinsuishins Control and Safety sroups
A 3-fold cross validation was implemented for supervised classification. Different machine learning (ML) classifiers (linear discriminant analysis (LDA), quadratic discriminant analysis (QDA), support vector machine (SVM), random forest (RF)) were trained using the topmost 7 features selected by different feature selection methods (t-test, Wilcoxon, mRmR) on the baseline images. LDA+ttest yielded best AUC of 0.75±0.06 on baseline images. Using the topmost 7 features the same model was evaluated on the other timepoint images, AUC (Area Under the Curve) values shown in Table 1. Table 1: AUC values yielded by LDA in classifying Control and Safety group.
Figure imgf000059_0001
7 most discriminating features:
1. skewness-Gabor CU-Q=1.178, CZ-q=0.000, l=3.261, BW=1 (p<0.001)
2. skewness-Laws E3E3L3(p<0.001)
3. skewness-Gabor CU-Q=2.356, CZ-q=0.000, l=3.261, BW=l(p<0.001)
4. skewness-Gabor CU-Q=0.393, CZ-Q=2.356, l=3.261, BW=l(p<0.001)
5. skewness-Gabor CU-Q=0.785, CZ-q=0.000, l=3.261, BW=l(p<0.001)
6. kurtosis-Gabor CU-Q=0.785, CZ-q=0.000, l=3.261, BW=l(p<0.001)
7. skewness-Laws S3E3S3(p<0.001) 13/20 (65%) eyes of SI were always classified correctly at each of these timepoints. The box and whisker plot of the most discriminating baseline feature significantly distinguishes the two groups of patients (Figure 4).
Feature map for the most discriminating feature at prior to event timepoint (Figure 3) shows that for both the Control and Safety patients the feature expression of the low frequency Gabor feature ‘Gabor XY-0= 1.178, CZ-q=0.000, l=3.261, BW=U (p=2.2186e-l 1) within the vitreous compartment is low. However, higher feature expression is observed (Figure 3e) within the inflammation regions and the surface of ILM and vitreous compartment (Figure 3b, 3c, 3d) for Safety patients. Thus, even if the actual event is late, i.e, when the clinicians actually observe the inflammation in the OCT scans, prior to that the presence of inflammatory safety signal is observed within the texture of the vitreous compartment and surface of ILM and vitreous compartment in the form of heterogeneity. Unsupervised Classification for distinguishing Control and Safety groups
Using Unsupervised Hierechical Clustering on texture-based radiomics features, a heatmap analysis was generated, showing that the patients in S2 and SI aggregated together in two clusters, Cluster 1 and 2, respectively.
K-means Clustering was also conducted. Three clusters were formed: 60% of Sis grouped within Cluster 1; 79% of S2s grouped within Cluster 2; 62% Controls grouped within Cluster 3.
Supervised Classification betweeen Control and SI group
The AUC values yielded by LDA are presented in Table 2 on different timepoint images. Table 2: AUC values yielded by LDA in classifying Control and SI group.
Figure imgf000060_0001
When the subset S2 was excluded from the Safety group, initially at baseline, after 1st injection and prior-to-event timepoint, the AUC values were comparatively less than that of when all Safety group of patients were considered (Table 2). However, the AUC value drastically increased for the last (event) timepoint. Machine learning quantitative characterization of the vitreous compartment identified differences in the number of hyperreflective vitreous and preretinal foci between eyes in the IOI group and the control group, including prior to the IOI reported event (Figures 5 to 7). Conclusion: In this preliminary discovery evaluation for OCT biomarkers for IOI-related events in the HAWK dataset, hyperreflective preretinal foci (i.e., preretinal stalagmites) appear to be a potential important objective OCT finding of IOI and may provide an important signal for clinicians assessing for underlying posterior inflammation.

Claims

1. A method of treating ocular disease comprising selectively administering a VEGF antagonist, e.g., brolucizumab, to a subject in need thereof on the basis of said subject not having a hyperreflective deposit (HRD) in a subject’s eye, e.g., a vitreous hyperreflective deposit (VHRD), e.g., a preretinal hyperreflective deposit (PHRD).
2. A method of treating ocular disease comprising:
(a) selecting a subject for treatment with a VEGF antagonist, e.g., brolucizumab, on the basis said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(b) thereafter, selectively administering the VEGF antagonist, e.g., brolucizumab, to the subject.
3. A method of selectively treating a subject having ocular disease the method comprising:
(a) selectively administering a therapeutically effective amount of a VEGF antagonist, e.g., brolucizumab, to the subject on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) selectively administering a therapeutically effective amount of a different ocular disease agent, e.g., a different VEGF antagonist, e.g., aflibercept, ranibizumab, faricimab, to the subject on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD.
4. The method of claim 2 or 3 further comprising assessing the presence or absence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, e.g., prior to the steps (a)-(b) of claim 2 or claim 3.
5. The method of any one of claims 1 to 4, wherein the presence or absence of the HRD, e.g., VHRD, e.g., PHRD, is assessed by performing OCT, e.g., SD-OCT, analysis on a subject’s eye.
6. A method for determining whether a subject with ocular disease should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising: (a) performing analysis, e.g., OCT, SD-OCT analysis, on a subject’s eye to assess the presence or absence of a HRD, e g., VHRD, e g., PHRD;
(b) determining that the subject should be treated with a VEGF antagonist, e.g., brolucizumab, if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye;
(c) determining that the subject should not be treated with a VEGF antagonist, e.g., brolucizumab, if the HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
7. The method of claim 6 further comprising treating the subject with the VEGF antagonist, e.g., brolucizumab, if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
8. The method of any one of the preceding claims, wherein the subject is assessed for presence of the HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, prior to initiation of the treatment with the VEGF antagonist and / or during the treatment course with the VEGF antagonist.
9. The method of claim 8, further comprising:
(a) altering or discontinuing the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye, or
(b) maintaining the treatment with the VEGF antagonist if, during the course of treatment, the subject is determined not to have a HRD, e.g., VHRD, e.g., PHRD, in the treatment eye.
10. A method of predicting the likelihood that a subject having ocular disease will develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis and / or retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising determining if the subject has a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD, wherein:
(a) the presence of a HRD, e.g., VHRD, e.g., PHRD, is indicative of an increased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab; and
(b) the absence of a HRD, e.g., VHRD, e.g., PHRD, is indicative of a decreased likelihood that the patient will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab.
11. The method of claim 10 further comprising treating the subject with the VEGF antagonist, e.g., brolucizumab, if no HRD, e.g., VHRD, e.g., PHRD, is observed in the subject’s eye.
12. The method of any one of the preceding claims, wherein the HRD, e.g., VHRD, e.g., PHRD, has high reflectivity, e.g., has clearly delineated boundaries from the surrounding components of the retina.
13. The method of any one of the preceding claims, wherein the HRD, e.g., VHRD, e.g., PHRD, is a hyperreflective round- or rod-shaped dot, e.g., stalagmite-like hyperreflective material.
14. The method of any one of the preceding claims, wherein the VEGF antagonist is an anti- VEGF antibody.
15. The method of claim 14, wherein the VEGF antibody comprises a variable heavy chain having the sequence as set forth in SEQ ID NO: 1 and a variable light chain having the sequence as set forth in SEQ ID NO: 2.
16. The method of claim 14, wherein the VEGF antibody comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
17. The method of claim 14, wherein the VEGF antagonist is brolucizumab.
18. The method of any one of claims 15 to 17, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 to 12 weeks.
19. The method of claim 18, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 12 weeks.
20. The method of claim 19, wherein a 12 week treatment interval is switched to an 8 week treatment interval if a disease activity is detected in the subject.
21. The method of claim 18, wherein the method comprises administering to the subject three individual doses of the VEGF antagonist at 4-week intervals, and thereafter administering to the subject an additional dose every 8 weeks.
22. The method of claim 21, wherein an 8 week treatment interval is switched to a 12 week treatment interval if no disease activity is detected in the subject.
23. The method of any one of claims 15 to 22, wherein each dose of the VEGF antagonist is at least 3 mg, e.g., 3 mg or 6 mg, e.g., 6 mg.
24. The method of claim 23, wherein each dose of the VEGF antagonist is administered as a 50 pL intravitreal injection.
25. The method of any one of preceding claims, wherein the ocular disease is selected from the list consisting of abnormal angiogenesis, choroidal neovascularization (CNV), retinal vascular permeability, retinal edema, diabetic retinopathy (particularly proliferative diabetic retinopathy (PDR) and non-proliferative diabetic retinopathy (NPDR)), macular edema (ME), diabetic macular edema (DME), neovascular (exudative) age-related macular degeneration (nAMD), choroidal neovascularization (CNV) associated with nAMD, sequela associated with retinal ischemia, Retinal Vein Occlusion (RVO), Central Retinal Vein Occlusion (CRVO), Branch Retinal Vein Occlusion (BRVO), macular edema following retinal vein occlusion, and posterior segment neovascularization.
26. The method of claim 25, wherein the ocular disease is neovascular age-related macular degeneration (nAMD).
27. A method for producing a transmittable form of information for determining whether a subject with ocular disease, e.g., nAMD, should be treated with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining that the subject should be treated with a VEGF antagonist on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining that the subject should be treated with a different ocular disease agent, e.g., nAMD agent, on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
28. A method for producing a transmittable form of information for predicting whether a subject with ocular disease, e.g., nAMD, may develop intraocular inflammation (IOI), e.g., intraocular inflammation with retinal vasculitis/retinal occlusion (IOI+RV/RO), in response to a treatment with a VEGF antagonist, e.g., brolucizumab, the method comprising:
(a) determining an increased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; or
(b) determining a decreased likelihood that the subject will develop IOI, e.g., IOI+RV/RO, in response to the treatment with the VEGF antagonist, e.g., brolucizumab, on the basis of said subject not having a HRD in a subject’s eye, e.g., VHRD, e.g., PHRD; and
(c) recording the result of the determining step on a tangible or intangible media form for use in transmission.
PCT/IB2022/052692 2021-03-26 2022-03-24 Methods for treating ocular diseases WO2022201084A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163166620P 2021-03-26 2021-03-26
US63/166,620 2021-03-26
US202163252713P 2021-10-06 2021-10-06
US63/252,713 2021-10-06

Publications (1)

Publication Number Publication Date
WO2022201084A1 true WO2022201084A1 (en) 2022-09-29

Family

ID=80999627

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/052692 WO2022201084A1 (en) 2021-03-26 2022-03-24 Methods for treating ocular diseases

Country Status (1)

Country Link
WO (1) WO2022201084A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006047325A1 (en) 2004-10-21 2006-05-04 Genentech, Inc. Method for treating intraocular neovascular diseases
WO2009155724A2 (en) 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting vegf
WO2017025928A2 (en) * 2015-08-12 2017-02-16 Novartis Ag Methods of treating ophthalmic disorders
US20200027209A1 (en) * 2018-07-18 2020-01-23 Case Western Reserve University Predicting response to anti-vascular endothelial growth factor therapy with computer-extracted morphology and spatial arrangement features of leakage patterns on baseline fluorescein angiography in diabetic macular edema

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006047325A1 (en) 2004-10-21 2006-05-04 Genentech, Inc. Method for treating intraocular neovascular diseases
WO2009155724A2 (en) 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting vegf
WO2017025928A2 (en) * 2015-08-12 2017-02-16 Novartis Ag Methods of treating ophthalmic disorders
US20200027209A1 (en) * 2018-07-18 2020-01-23 Case Western Reserve University Predicting response to anti-vascular endothelial growth factor therapy with computer-extracted morphology and spatial arrangement features of leakage patterns on baseline fluorescein angiography in diabetic macular edema

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
"Member Update: Novartis-Appointed Safety Review Committee Reports Initial Brolucizumab Findings", 4 June 2020, AMERICAN SOCIETY OF RETINA SPECIALISTS
"Remington's Pharmaceutical Sciences", 2006
AL-LAZIKANI ET AL., J. MAL. BIOL., vol. 273, 1997, pages 927 - 948
BAUMAL CAROLINE R. ET AL: "Retinal Vasculitis and Intraocular Inflammation after Intravitreal Injection of Brolucizumab", OPHTHALMOLOGY, vol. 127, no. 10, 1 October 2020 (2020-10-01), AMSTERDAM, NL, pages 1345 - 1359, XP055932293, ISSN: 0161-6420, DOI: 10.1016/j.ophtha.2020.04.017 *
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BLOCH ET AL., AM J OPHTHALMOL, vol. 153, 2012, pages 209 - 13
CAMPBELL ET AL., ARCH OPHTHALMOL, vol. 130, 2012, pages 794 - 5
CASALINO ET AL., EXPERT REVIEW OF OPHTHALMOLOGY, vol. 15, no. 2, 2020, pages 83 - 91
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOTHIA ET AL., NATURE, vol. 341, 1989, pages 544 - 546
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; December 2016 (2016-12-01), CHATZIRALLI IRINI P ET AL: "HYPERREFLECTIVE FOCI AS AN INDEPENDENT VISUAL OUTCOME PREDICTOR IN MACULAR EDEMA DUE TO RETINAL VASCULAR DISEASES TREATED WITH INTRAVITREAL DEXAMETHASONE OR RANIBIZUMAB", XP002806833, Database accession no. PREV201700146636 *
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; July 2018 (2018-07-01), KOROT EDWARD ET AL: "Automated Algorithmic Analysis of OCT Scans for Vitreous Hyperreflective Foci in Patients with Retinal Vein Occlusion", XP002806832, Database accession no. PREV201800905258 *
DEVRIES ET AL., SCIENCE, vol. 255, 1992, pages 989
FERRIS ET AL., AM J OPHTHALMOL, vol. 118, 1983, pages 132 - 51
FERRIS ET AL., ARCH OPHTHALMOL, vol. 102, 1984, pages 1640 - 2
FRANGIEH ET AL., ARCHIVES OF OPHTHALMOLOGY, vol. 100, 1982, pages 1132 - 40
GLOBAL SAFETY INFORMATION FOR HEALTHCARE PROFESSIONALS, 2 October 2020 (2020-10-02), Retrieved from the Internet <URL:https://www.brolucizumab.info/>
GREEN ET AL., RETINA, vol. 1, 1981, pages 27 - 55
HONEGGERPLUCKTHUN, J. MOL. BIOL., vol. 309, 2001, pages 657 - 670
HOUCK ET AL., MOL. ENDOCRIN, vol. 5, 1991, pages 1806
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
IYER PRASHANTH G. ET AL: "Brolucizumab-related retinal vasculitis with exacerbation following ranibizumab retreatment: A clinicopathologic case study", AMERICAN JOURNAL OF OPHTHALMOLOGY CASE REPORTS, vol. 20, 1 December 2020 (2020-12-01), pages 100989, XP055932330, ISSN: 2451-9936, DOI: 10.1016/j.ajoc.2020.100989 *
JONES, A, ADV. DRUG DELIVERY REV., vol. 10, 1993, pages 29 - 90
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, pages: 247 - 301
LAZIKANI ET AL., J. MOL. BIO., vol. 273, 1997, pages 927 - 948
LEUNG ET AL., SCIENCE, vol. 246, 1989, pages 1306
LIM ET AL., LANCET, vol. 379, 2012, pages 1728 - 38
MATTHEWS ET AL., PROC. NAT. ACAD. SCI., vol. 88, 1991, pages 9026
MILLER ET AL., AM J OPHTHALMOL, vol. 155, 2013, pages 1 - 35
MONES ET AL., AMERICAN ACADEMY OF OPHTHALMOLOGY, 2020
REIN ET AL., ARCH OPHTHALMOL, vol. 127, 2009, pages 533 - 40
RIORDAN-EVA2004, EYE (LOND, vol. 18, 2004, pages 1161 - 8
SAFETY OF BEOVUG (BROLUCIZUMAB, 19 November 2020 (2020-11-19), Retrieved from the Internet <URL:https://www.brolucizumab.info>
SCHMIDT-ERFURTH ET AL., OPHTALMOLOGY, vol. 121, 2014, pages 193 - 201
SHAH ET AL., AM J OPHTHALMO, vol. 116, 2009, pages 1901 - 07
SHAH ET AL., AM J OPHTHALMOL, vol. 143, 2007, pages 83 - 89
SHIBUYA ET AL., ONCOGENE, vol. 5, 1990, pages 519
SOMMER ET AL., N ENGL J MED, vol. 14, 1991, pages 1412 - 17
SPILSBURY ET AL., AM J PATHOL, vol. 157, 2000, pages 135 - 44
TERMAN ET AL., BIOCHEM. BIOPHYS. RES., vol. 187, 1992, pages 1579
TERMAN ET AL., ONCOGENE, vol. 6, 1991, pages 1677
TOLENTINO M., SURV OPHTHALMOL, vol. 56, 2011, pages 95 - 113
WITKIN ET AL., J VITREORETIN DIS, vol. 4, no. 4, 2020, pages 269 - 79
WONG ET AL., OPHTHALMOLOGY, vol. 115, 2008, pages 116 - 26
YONEKAWA ET AL., DIGITAL JOURNAL OF OPHTHALMOLOGY, vol. 23, 2017, pages 18 - 22

Similar Documents

Publication Publication Date Title
US20210340242A1 (en) Methods for treating ocular diseases
AU2021286278B2 (en) Methods for treating ocular diseases
EP4028128A1 (en) Methods for treating ocular diseases
JP2024510142A (en) Methods for the treatment of thyroid eye disease
WO2022201084A1 (en) Methods for treating ocular diseases
US20230416351A1 (en) Vegf antagonist for use in methods for treating ocular diseases
RU2776850C2 (en) Methods for treatment of eye diseases
WO2023175549A1 (en) Methods for treating neovascular age-related macular degeneration

Legal Events

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

Ref document number: 22714012

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22714012

Country of ref document: EP

Kind code of ref document: A1