WO2022229932A1 - Hypoxia inducible factor (hif) inhibitors for treatment of atrophy associated with retinal hypoxia - Google Patents

Hypoxia inducible factor (hif) inhibitors for treatment of atrophy associated with retinal hypoxia Download PDF

Info

Publication number
WO2022229932A1
WO2022229932A1 PCT/IB2022/054013 IB2022054013W WO2022229932A1 WO 2022229932 A1 WO2022229932 A1 WO 2022229932A1 IB 2022054013 W IB2022054013 W IB 2022054013W WO 2022229932 A1 WO2022229932 A1 WO 2022229932A1
Authority
WO
WIPO (PCT)
Prior art keywords
retinal
inhibitor
pharmaceutical composition
hif
atrophy
Prior art date
Application number
PCT/IB2022/054013
Other languages
French (fr)
Inventor
Einar Stefansson
Original Assignee
Einar Stefansson
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 Einar Stefansson filed Critical Einar Stefansson
Publication of WO2022229932A1 publication Critical patent/WO2022229932A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00

Definitions

  • This application relates to compositions, uses, and methods for treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia.
  • the methods include administration to a subject in need of said treatment of a pharmaceutical composition comprising an effective amount of an inhibitor of hypoxia inducible factor (HIF).
  • HIF hypoxia inducible factor
  • New vessel formation, edema, tissue atrophy, and combinations thereof are common features and cause of visual loss in ischemic diseases of the retina and choroid.
  • choroidal neovascularization, bleeding, and retinal atrophy e.g, dry or atrophic age related macular degeneration (AMD) or geographic atrophy
  • AMD age related macular degeneration
  • AMD age related macular degeneration
  • new vessel formation and bleeding can occur, and diabetic macular edema can cause loss of vision.
  • Vision loss also can occur with macular retinal atrophy, sometimes referred to as ischemic atrophy or ischemic maculopathy.
  • ischemic diseases that can cause atrophy include central and branch retinal vein occlusions, retinopathy of prematurity, sickle cell retinopathy, retinal detachment and proliferative vitreoretinopathy.
  • Atrophic AMD is one of the major causes of blindness in developed countries, including the United States. Treatment for atrophic AMD is a primary unmet medical need in eye care. No treatment is currently available, despite considerable research and efforts for more than a decade by mumerous research groups and pharmaceutical companies.
  • An exemplary embodiment of this application is a method of treating, minimizing and/or inhibiting atrophy associated with retinal hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor to a subject having retinal hypoxia.
  • the HIF inhibitor is administered in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia.
  • the atrophy associated with retinal hypoxia can be selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
  • compositions e.g., a pharmaceutical composition
  • an HIF inhibitor in an amount effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
  • FIG. 1 depicts results of ischemia and hypoxia through HIF mechanisms. If the VEGF pathway is blocked, the tissue can mitigate ischemia/hypoxia by the apoptosis/atrophy pathway.
  • FIG. 2 depicts deregulated molecular mechanisms in RPE cells and their association with HIF activation and/or stability.
  • Reductions in proteasomal activity and increased reactive oxygen species (ROS) can lead to an increase in available HIF a, which can subsequently heterodimerize with HIF-Ib and activate the transcription of HIF target genes, such as, for example, one or more of VEGF, GLUT1 and other genes related to neovascularization and metabolic conversion.
  • HIF target genes such as, for example, one or more of VEGF, GLUT1 and other genes related to neovascularization and metabolic conversion.
  • Formation of drusen and a thickening of Bruch’s membrane can induce local hypoxia (1), which can activate HIFa.
  • Mitochondrial dysfunction (2) can lead to increased reactive oxygen species (ROS) production, which can stabilize HIFa.
  • Proteasomal dysfunction (3) can prevent the effective clearance of HIFa.
  • HIFa levels can rise and dimerization with H
  • FIG. 3 shows Cytotoxicity of PX-478 in ARPE-19 cells.
  • Cell viability MTT assay, FIG. 3A
  • cellular toxicity leakage of LDH, Fig. 3B
  • the compound was well tolerated up to a concentration of 10 mM.
  • Ctrl - control * - p ⁇ 0.05; ** - p ⁇ 0.01; *** - p ⁇ 0.001; Mann- Whitney U-test, compared to untreated control.
  • FIG. 4 shows cytotoxicity of hydroquinone in ARPE-19 cells pretreated with PX-478 for 24h.
  • Cell viability MTT assay, FIGS 4A and 4C
  • cellular toxicity leakage of LDH, FIGS 4B and 4D
  • PX-478 was cytoprotective at 5mM and 10 mM concentrations.
  • Atrophy e.g., atrophy associated with retinal hypoxia
  • an angiogenesis inhibitor such as an anti-VEGF or anti-VEGFR therapeutic
  • ischemic retinal disease in which the apoptosis-atrophy pathway is minimized and/or substantially inhibited, or is not activated. Activation of the apoptosis-atrophy pathway can occur during treatment with an anti-VEGF therapeutic, since the VEGF response to ischemia/hypoxia is blocked.
  • Hypoxic conditions can activate the HIF pathway, which can result in effects such as increased VEGF production, increased vascular permeability, edema, new vessel formation, and combinations thereof.
  • bleeding can be selected from among the group consisting of subretinal, intraretinal, in vitreous humour, or any combination thereof.
  • Activation of the HIF pathway can result in apoptosis (e.g., apoptosis of retinal cells). Apoptosis can result in retinal atrophy, which can also lead to visual loss.
  • tissue can mitigate hypoxia by apoptosis (e.g., retinal apoptosis) and atrophy.
  • apoptosis e.g., retinal apoptosis
  • Ischemia and hypoxia have several consequences through HIF mechanisms, including: 1) angiogenesis and edema through a VEGF pathway; 2) apoptosis; and 3) atrophy.
  • treatment of ischemic disease in retina is limited to targeting the VEGF pathway.
  • the apoptosis/atrophy pathway remains for the tissue to “mitigate” ischemia/hypoxia that activates HIF pathway (FIG. 1).
  • subjects who are administered VEGF inhibitors can exhibit considerable retinal atrophy and visual loss.
  • the HIF inhibitor can complement or replace treatment with a VEGF inhibitor or VEGFR inhibitor, to reduce or prevent atrophy that VEGF/VEGFR inhibitors are associated with, when administered alone (e.g., to treat retinal ischemic diseases).
  • an HIF inhibitor can treat, minimize or substantially inhibit atrophy that is an adverse effect of a VEGF or VEGFR inhibitor.
  • Mammalian cells such as RPE cells
  • RPE cells can rely on aerobic metabolism for energy generation, a process that requires sufficient levels of oxygen.
  • oxygen levels drop too low cells become hypoxic and can react by activating the hypoxic response, which is designed to ensure survival. It can, e.g., increase the number of red blood cells that transport oxygen, augment the number of blood vessels available, and switch energy metabolism to anaerobic metabolism that does not use mitochondria (see e.g., Shinojima et al., J. Clin. Med. 2021 Nov 24;10(23):5496).
  • HIFs hypoxia-induced factors
  • HIFs are master regulators of the hypoxic response, controlling hundreds of genes involved in, for example, erythropoiesis, angiogenesis (e.g., VEGF) and metabolic conversion (see, e.g., Shinojima et al., J. Clin. Med. 2021 Nov 24;10(23):5496; Pawlus et al., Cell Signal. 2013 Sep;25(9): 1895-903).
  • angiogenesis e.g., VEGF
  • metabolic conversion see, e.g., Shinojima et al., J. Clin. Med. 2021 Nov 24;10(23):5496; Pawlus et al., Cell Signal. 2013 Sep;25(9): 1895-903).
  • HIFs are heterodimers that can include an O2 sensitive a subunit (e.g., HIF- la, HIF-2a or HIF-3a) and an O2 insensitive subunit (e.g., HIF- 1b) (see, e.g., Albadari et al., Expert Opin Drug Discov. 2019 Jul;14(7):667-682; Prabhakar et al. Physiol Rev. 2012 Jul;92(3):967-1003]
  • HIFs e.g., HIF- l/2a
  • HIFs can be quickly degraded, following poly-ubiquitination and proteasomal degradation.
  • hypoxia can stabilize HIF-l/2a via inhibition of these pathways (see, e.g., Maxwell et al., Nature. 1999 May 20;399(6733):271-5; Semenza et al., Biochem Pharmacol. 2002 Sep;64(5-6):993-8.).
  • ROS reactive oxygen species
  • a current standard of care for AMD is the repeated intra ocular injection of anti-VEGF agents, which can slow the wet form of the disease.
  • prolonged therapy has been linked to progressive retinal atrophy (Rofagha et al. Ophthalmology. 2013;120:2292-2299).
  • HIF is upstream of VEGF activation.
  • VEGF deletion can cause retinal atrophy and dysfunction in a mouse model, but deletion of HIF can have minimal or no adverse effects (see, e.g., Kurihara et al., J. Clin. Investig. 2012;122:4213-4217).
  • HIFs can play a crucial role in the adaptive response of tumor cells to changes in oxygen availability through transcriptional activation of one or more downstream genes selected from more than one hundred known downstream genes.
  • HIF-1 can help hypoxic tumor cells shift glucose metabolism from oxidative phosphorylation to the less efficient glycolytic pathway through the induction of enzymes involved in the glycolysis pathway and overexpression of glucose transporters (GFUTs) which can increase glucose import into tumor cells (see, e.g., Masoud et al., Acta Pharm Sin B. 2015 Sep,5(5):378-89; Denko et al, Nat Rev Cancer. 2008;8:705-713; Weinhouse et al., Science. 1956;124:267- 272.).
  • GFUTs glucose transporters
  • HIFs also can cause the transcriptional induction of one or more pro-angiogenic factor(s), such as the vascular endothelial growth factor (VEGF), which in turn can stimulate the development of new blood vessels to enrich tumor cells with oxygen for their growth (see, e.g., Conway et al. 2001;49:507-521).
  • VEGF vascular endothelial growth factor
  • HIF inhibitors Advances in the development of selective inhibitors have led to clinical studies testing the potential of HIF inhibitors in cancer therapy (see, e.g., Albadari et al. Expert Opin Drug Discov. 2019 Jul;14(7):667-682).
  • Belzutifan a selective inhibitor of HIF-2a, has recently been granted FDA approval (www.fda.gov/drugs/resources-information-approved-drugs/fda- approves-belzutifan-cancers-associated-von-hippel-lindau-disease; accessed April 28, 2022).
  • solid carcinomas are not the only targets for the use of HIF inhibitors. Hypoxic conditions have been associated with many age-related diseases, and target genes of HIF, such as, for example VEGF, are implicated in, e.g., age-related macular degeneration (AMD).
  • AMD age-related macular degeneration
  • AMD is the leading cause of vision loss amongst the elderly in the western world (see, e.g., Thomas etal, Med. Clin. North Am., 2021 May;105(3):473-491). It affects an estimated 196 million patients worldwide, a number that is projected to increase to 288 million by 2040 (Wong et al. Fancet Glob Health. 2014 Feb;2(2):el06-16). Only 15% of patients can be treated with anti-VEGF injections, which can be costly and invasive. It was estimated that up to one in three persons over 85 years of age is suffering from AMD in Europe and North America (see, e.g., Wong et al. Lancet Glob Health. 2014 Feb;2(2):el06- 16).
  • AMD AMD is classically divided into dry and wet subtypes, as well as into early and advanced stages (Thomas et al., Med Clin North Am. 2021 May;105(3):473-491).
  • Wet AMD or choroidal neovascularization, is an advanced form of the disease, characterized by a growth of blood vessels from the choroid through Bruch’s membrane and into the subretinal space. Bleeding, swelling and possible scar formation are the consequence, leading to a rapid and drastic loss of vision.
  • Thomas et al., Med Clin North Am. 2021 May;105(3):473-491 Aberrant VEGF production can underlie rapid vision loss in wet AMD (Thomas et al. Med Clin North Am.
  • VEGF levels are often increased in patients suffering from wet AMD, and anti -VEGF treatment via intra ocular injection has proven successful at slowing down disease progression in this subtype of AMD cases.
  • repeated intra ocular injections can place a large strain on healthcare providers, practitioners and patients alike, and real-world data shows a steady decline in adherence, and consequently outcome, with prolonged treatment times (Sobolewska et al. Clin Ophthalmol. 2021 ; 15:4317- 4326).
  • RPE retinal pigment epithelium
  • RPE cells also secrete a number of growth factors and cytokines, aimed at maintaining tissue homeostasis. Deregulated, these factors can cause choroidal neovascularization and uncontrolled, chronic inflammation that will inevitably lead to cell death (see, e.g., Kauppinen et ak, Cell Mol Life Sci. 2016 May;73(9): 1765-86).
  • HIF is involved or linked to these processes in multiple ways (FIG. 2).
  • One or more factors selected from among the group consisting of increased oxidative stress, e.g. ROS produced by dysfunctional mitochondria, as well as reduced proteasomal clearance, and active inflammation through NF-kB, the master regulator of the innate immune response can lead to an increase in available HIF protein levels (Aqamaa et ak, Ageing Res Rev. 2009 Oct;8(4):349-58; Frede et ak (2007) Methods Enzymoh 435, 405-419).
  • HIF has been located in human choroidal neovascular membranes, which are associated with AMD and in drusen (Inoue et ak (2007) Br. J. Ophthalmol. 91:1720-1721; Shimada et ak (2007), Graefes Arch Clin Exp Ophthalmol. 245(2):295-300).
  • CRISPR-mediated knock-out of HIF-Ia or VEGF in a mouse model of wet AMD can reduce the volume of choroidal neovascularisation with the same efficiency as the anti-VEGF agent aflibercept (Shinojima etal. (2021) J. Clin. Med. 10(23):5496; Koo et ak (2016) Nat.
  • HIF is not required for retinal homeostasis in the steady state and might therefore be a better therapeutic target than VEGF (Shinojima et al. (2021) J. Clin. Med. 10(23):5496).
  • HIF inhibitors that can inhibit the HIF pathway.
  • HIF inhibitors can mitigate and block HIF mediated response to hypoxia, which can result in one or more effects selected from the group consisting of limiting new vessel formation, inhibiting edema, preventing or reducing apoptosis (e.g., retinal apoptosis), preventing or reducing atrophy, and any combination thereof.
  • HIF induced apoptosis is reduced, minimized, substantially inhibited, and/or prevented.
  • reduction, minimization, inhibition, and/or prevention of HIF induced apoptosis results in reduction, minimization, inhibition, and/or prevention of one or more effects selected from the group consisting of retinal atrophy, choroidal atrophy, vision loss, combinations thereof and the like.
  • reduction, minimization, inhibition and/or prevention of HIF induced apoptosis results in reduction, minimization, inhibition and/or prevention of one or more adverse effects of treatment selected from the group consisting of retinal atrophy, choroidal atrophy, vision loss, combinations thereof and the like.
  • Atrophy associated with retinal hypoxia refers to atrophy that results from retinal hypoxia and/or activation of the HIF pathway.
  • Hypoxia and/or activation of the HIF pathway can occur, for example from ischemia (e.g., ischemic retinal disease) and/or retinal detachment.
  • Hypoxia can activate HIF, which can activate mitigating responses that counter the hypoxic state.
  • ischemia e.g., ischemic retinal disease
  • hypoxia can activate HIF, which can activate mitigating responses that counter the hypoxic state.
  • VEGF induced angiogenesis and edema include VEGF induced angiogenesis and edema, and apoptosis/atrophy, combinations thereof, and the like.
  • retinal hypoxia “associated with” a disease or condition refers to a correlation between the condition and the retinal hypoxia and/or activation of the HIF pathway.
  • retinal hypoxia associated with ischemic disease or retinal detachment refers to retinal hypoxia that results from ischemic disease or retinal detachment.
  • treating includes curing a condition, treating a condition, minimizing and/or inhibiting and/or substantially inhibiting a condition, preventing or substantially preventing a condition, treating, minimizing and/or inhibiting one or more symptoms of a condition, curing symptoms of a condition, ameliorating, reducing and/or minimizing symptoms of a condition, treating effects of a condition, ameliorating, reducing and/or minimizing effects of a condition, and preventing and/or substantially preventing results of a condition.
  • the term “substantially” means completely or almost completely.
  • “substantially preventing a condition” means that the condition is completely prevented or is almost completely prevented.
  • treating atrophy associated with retinal hypoxia can result in the atrophy being partially or totally alleviated, or remaining static as a result of treatment.
  • treatment encompasses prevention, prophylaxis, therapy and/or cure.
  • Prophylaxis refers to prevention of a potential atrophy and/or a prevention of worsening of symptoms or progression of atrophy.
  • pharmaceutical composition refers to a composition comprising one or more active ingredients with other components such as, for example, pharmaceutically-acceptable ingredients and/or excipients, such as a pharmaceutically- acceptable carrier. The purpose of a pharmaceutical composition is to facilitate administration of an active ingredient to a subject.
  • the terms “pharmaceutically active agent” or “active agent” or “active pharmaceutical ingredient” are interchangeable and mean the ingredient is a pharmaceutical drug, which is biologically- and/or chemically-active and is regulatory-approved or appro vable as such.
  • the term “ingredient” refers to a pharmaceutically-acceptable ingredient, which is included or is amenable to be included in The FDA’s Inactive Ingredient (IIG) database. Inactive ingredients can sometimes exhibit some therapeutic effects, although they are not drugs.
  • a compound or “at least one compound” can include a plurality of compounds, including combinations and/or mixtures thereof.
  • the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, technical and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • hypoxia-inducible factor is a transcription factor in the hypoxia-inducible factor (HIF) pathway that responds to hypoxic conditions.
  • HIF- la HIF-Ib
  • HIF-2a HIF-2
  • ARNT2 HIF-2
  • HIF- 3alpha HIF-3
  • HIF-3 HIF-3a/HIR-2b
  • HIF-a refers to an HIF-a transcription factor (e.g., HIF-la, HIF-2a, or HIF-3a, or a combination thereof).
  • HIF-b refers to an HIF-b transcription factor (e.g., HIF-Ib, or HIR-2b, or the combination thereof).
  • HIF inhibitor is an inhibitor of the hypoxia-inducible factor (HIF) pathway. It is understood that recitation of an HIF inhibitor includes pharmaceutically-acceptable salts thereof, as well as prodrugs thereof. It also is understood that description of an HIF inhibitor as inhibiting a part of the HIF pathway does not preclude the inhibitor from inhibiting another part of the HIF pathway.
  • An “HIF inhibitor” includes pharmaceutically-acceptable salts thereof and analogs thereof.
  • a pharmaceutically-acceptable salt of an HIF inhibitor is a biologically-compatible salt that can be used as a drug, which salts can be derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, combinations thereof and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate, combinations thereof and the like.
  • the pharmaceutically- acceptable salt is an acid addition salt.
  • Pharmaceutically-acceptable acid addition salts are those salts that retain the biological effectiveness of the free bases while formed by acid partners that are not biologically or otherwise undesirable, e.g., inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like
  • organic acids such as acetic acid,
  • the salt is formed with an ion selected from the group consisting of ammonium, lithium, sodium, potassium, cesium; alkaline earth metals to include calcium, magnesium, aluminium; zinc, barium; or quaternary ammoniums; or organic salts such as arginine, organic amines to include aliphatic organic amines, aromatic amines, t-butylamines, (N-benzylphenethylamine), dicyclohexylamines, dimethylamines, diethanolamines, ethanolamines, ethylenediamines, imidazoles, lysines, methylamines, N-methyl-D-glucamines, N,N'-dibenzylethylenediatnines, pyridines, picolinates, piperazines, tris(hydroxymethyl)methylamines, triethylamines, triethanolamines, trimethylamines, or ureas.
  • organic salts such as arginine, organic amines to include alipha
  • HIF target gene is a gene regulated by HIF.
  • ischemic retinal disease or “ischemic retinopathy” refers to diseases of the retina where new vessel formation, edema or atrophy are prominent features.
  • the ischemia can result from a variety of mechanisms. In age related macular degeneration (AMD) this mechanism can include immunologic mechanisms involving complement factor H and more, resulting in choroidal ischemia. Similar features can apply to other diseases with choroidal neovascularization, including myopic choroidal neovascularization, Polypoidal Choroidal Vasculopathy and choroidal inflammation such as VKH disease and Bechets disease.
  • AMD age related macular degeneration
  • hyperglycemia can contribute to damage of retinal capillaries and capillary non-perfusion, which is ischemia.
  • the abnormal red blood cells can block retinal capillaries and cause ischemia.
  • retinal capillaries can be damaged and nonperfusion can occur, similar to diabetic retinopathy.
  • retinopathy of prematurity the retinal vasculature does not develop fully as a consequence of premature birth and relative hyperoxia.
  • ischemia can be caused by venous occlusion.
  • central or branch retinal artery occlusion and anterior ischemic optic neuropathy the ischemia can be caused by arterial occlusion, that can be embolic or inflammatory.
  • hypoxia can be caused by increased distance between the retina and the choroidal source of oxygen, rather than vascular ischemia of the diseases listed above.
  • Retinal ischemia can be evaluated by methods described herein, such as retinal fluorescein angiography, OCT angiography, clinical examination, combinations thereof, and the like.
  • Choroidal ischemia can be evaluated by indocyanin green angiography.
  • Retinal hypoxia can be measured by retinal oximetry and other methods to measure retinal oxygenation (Stefansson etal. (2019) Prog. Retin. Eye Res. 70:1-22).
  • Diabetic retinopathy is a diabetes complication caused by damage to blood vessels of the retina.
  • Diabetic retinopathy includes non-proliferative diabetic retinopathy (NPDR), diabetic macular edema and proliferative diabetic retinopathy (PDR).
  • NPDR non-proliferative diabetic retinopathy
  • PDR proliferative diabetic retinopathy
  • Non-proliferative diabetic retinopathy includes mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, and severe non-proliferative diabetic retinopathy.
  • Examples of a symptom of an ischemic retinal disease include angiogenesis, edema, vision impairment, blindness, retinal apoptosis, retinal atrophy, choroidal atrophy, combinations thereof and the like.
  • an effective amount of an HIF inhibitor for treating atrophy associated with retinal hypoxia is an amount that is sufficient to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia.
  • Such an amount can be administered as a single dosage or can be administered according to a regimen, whereby it is effective.
  • the amount can cure the atrophy associated with retinal hypoxia but, typically, is administered in order to ameliorate or prevent one or more symptoms of the atrophy. Repeated administration can be required to achieve the desired amelioration or prevention of symptoms.
  • administering the HIF inhibitor comprises administration hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months.
  • VEGFR inhibitor can inhibit activity and/or expression of a VEGF receptor.
  • VEGFR inhibitors include, but are not limited to, antibodies, such as, for example an antibody selected from the group consisting of cediranib, cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • a VEGFR inhibitor for example, can block binding of a VEGFR receptor and/or inhibit receptor phosphorylation.
  • a VEGF inhibitor includes an inhibitor of activity and/or expression of a VEGF ligand.
  • VEGF inhibitors include, but are not limted to, antibodies, such as, for example, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof, and the like.
  • a VEGF inhibitor for example, can block binding of a VEGF ligand.
  • An exemplary embodiment of this application is a method of treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor (e.g., an effective amount of an HIF inhibitor) to a subject having retinal hypoxia.
  • an HIF inhibitor e.g., an effective amount of an HIF inhibitor
  • the HIF inhibitor is administered in an amount effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, combinations thereof, and the like.
  • administration of the pharmaceutical composition containing an HIF inhibitor effects treatment of the atrophy associated with the retinal hypoxia.
  • treatment of the atrophy associated with retinal hypoxia comprises preventing, minimizing, slowing, alleviating and/or substantially inhibiting the atrophy. In some exemplary embodiments, treatment of the atrophy associated with retinal hypoxia comprises decreasing the severity, duration, or frequency of occurrence of the atrophy.
  • the method further comprises assessing the atrophy associated with retinal hypoxia.
  • assessing the atrophy associated with retinal hypoxia comprises a method selected from the group consisting of spectral-domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, and combinations thereof.
  • OCT spectral-domain optical coherence tomography
  • EEG electroretinopathy
  • EEG multifocal retinal electroretinopathy
  • retinal perimetry visual field analysis
  • assessing the atrophy associated with retinal hypoxia occurs before administering the pharmaceutical composition comprising the HIF inhibitor. In some exemplary embodiments, assessing the atrophy associated with retinal hypoxia occurs after administering the pharmaceutical composition comprising the HIF inhibitor. In some exemplary embodiments, the atrophy associated with retinal hypoxia is assessed before and after administering the pharmaceutical composition comprising the HIF inhibitor.
  • the method comprises a reduction in retinal apoptosis associated with retinal hypoxia in the subject. In some exemplary embodiments, the method comprises a reduction in retinal apoptosis by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%.
  • the method comprises a reduction in retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
  • the method comprises a reduction in retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
  • the method comprises assessing retinal apoptosis.
  • retinal apoptosis is assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, combinations thereof, and the like.
  • the measuring phosphatidyl extemalization comprises annexin 5 staining.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of macular atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy (glaucomatous atrophy), combinations thereof, and the like.
  • the atrophy associated with retinal hypoxia is glaucomatous atrophy.
  • retinal ganglion cells can undergo apoptosis due to hypoxia.
  • HIF inhibition can protect ganglion cells from cell death in glaucoma.
  • the glaucomatous atrophy is from glaucoma.
  • the glaucoma is selected from the group consisting of chronic open angle glaucoma, closed angle glaucoma, secondary glaucoma, normal tension glaucoma, and combinations thereof.
  • HIF inhibition can provide neuroprotection in glaucoma.
  • the iris atrophy is from anterior segment ischemia.
  • the ciliary body atrophy is from anterior segment ischemia.
  • the optic nerve atrophy is from vascular ischemia.
  • the vascular ischemia comprises a condition selected from the group consisting of giant cell arteritis, embolisms, and a combination thereof.
  • the optic nerve atrophy comprises anterior ischemia optic neuropathy.
  • the atrophy (e.g., retinal atrophy) is macular atrophy or geographic atrophy. In some exemplary embodiments, the atrophy is choroidal atrophy.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), and dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of an angiogenesis inhibitor (e.g., a VEGF or VEGFR inhibitor).
  • an angiogenesis inhibitor e.g., a VEGF or VEGFR inhibitor
  • the retinal detachment is selected from the group consisting of tractional, rhegmatogenous and serous retinal detachment.
  • Retinal detachment can move the retina further away from a choroidal oxygen source. This can cause hypoxia and/or HIF activation.
  • the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
  • the method comprises a reduction in a total area of the atrophy associated with retinal hypoxia.
  • the area of atrophy associated with retinal hypoxia is reduced by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%.
  • the area of atrophy associated with retinal hypoxia is reduced by about 1% to about 10%, by about 10% to about 20%, by about 20% to about 30%, by about 30% to about 40%, by about 40% to about 50%, by about 50% to about 60%, by about 60% to about 70%, by about 70% to about 80%, by about 80% to about 90%, or by about 100%.
  • the area of atrophy associated with retinal hypoxia is reduced by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
  • the method comprises assessing the area of atrophy associated with retinal hypoxia.
  • the area of atrophy associated with retinal hypoxia is assessed by a method selected from among the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like.
  • the area of atrophy associated with retinal hypoxia is assessed by a method selected from the group consisting of spectral -domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, combinations thereof, and the like.
  • OCT spectral -domain optical coherence tomography
  • Atrophy e.g., retinal atrophy
  • atrophy can be assessed by a method selected from among the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, combinations thereof, and the like.
  • the method comprises a reduction in a severity grade of the atrophy associated with retinal hypoxia (e.g., retinal atrophy or choroidal atrophy).
  • retinal hypoxia e.g., retinal atrophy or choroidal atrophy
  • the method comprises an improvement in the Age- Related Eye Disease Study (AREDS) scale.
  • AREDS Age- Related Eye Disease Study
  • the subject is one who is being treated with an angiogenesis inhibitor.
  • the angiogenesis inhibitor is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, a combination thereof, or the like.
  • the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, a combination thereof, and the like.
  • the angiogenesis inhibitor is selected from the group consisting of ranibizumab, bevacizumab, aflibercept, pegaptanib, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • the retinal hypoxia is from a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof.
  • the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, central serous chorioretinopathy and other serous retinal detachments, rhegmatogenous retinal detachment, traction retinal detachment, proliferative vitreoretinopathy (PVR), combinations thereof, and the like.
  • grade A proliferative vitreoretinopathy grade B proliferative vitreoretinopathy
  • grade C P proliferative vitreoretinopathy grade C A proliferative vitreoretinopathy
  • central serous chorioretinopathy and other serous retinal detachments rhegmatogenous retinal detachment
  • traction retinal detachment proliferative vitreoretinopathy (PVR), combinations thereof, and the like.
  • the retinal hypoxia is from an ischemic retinal disease.
  • administration of the pharmaceutical composition containing an HIF inhibitor effects treatment, minimizing and/or substantial inhibition of a symptom associated with the ischemic retinal disease.
  • the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion combinations thereof and the like.
  • the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
  • the ischemic retinal disease is selected from the group consisting of age related macular degeneration (dry atrophic AMD, geographic atrophy), diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy, retinal pigment epithelial detachment, central serous chorioretinopathy, combinations thereof, and the like.
  • age related macular degeneration dry atrophic AMD, geographic atrophy
  • diabetic retinopathy retinal vein occlusion
  • ROP retinopathy of prematurity
  • sickle cell retinopathy retinal pigment epithelial detachment
  • central serous chorioretinopathy combinations thereof, and the like.
  • the pharmaceutical composition reduces progression of retinal atrophy in dry atrophic AMD (Geographic atrophy), and/or reduces or prevents atrophy associated with anti VEGF or anti VEGFR treatment for neovascular AMD.
  • the pharmaceutical composition treats, minimizes or substantially inhibits angiogenesis and/or edema in addition to atrophy.
  • the pharmaceutical composition treats, minimizes, substantially inhibits and/or reduces progression of retinal atrophy (e.g., in dry AMD) and/or severe and symptomatic atrophy (Geographic atrophy).
  • the ischemic retinal disease is diabetic macular edema.
  • the ischemic retinal disease is non-proliferative diabetic retinopathy (DR), including DR with macular ischemia and/or macular atrophy
  • DR non-proliferative diabetic retinopathy
  • the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, severe non-proliferative diabetic retinopathy, and traction retinal detachment in DR.
  • the ischemic retinal disease is proliferative diabetic retinopathy. In some exemplary embodiments, the ischemic retinal disease is central retinal vein occlusion. In some exemplary embodiments, the ischemic retinal disease is branch retinal vein occlusion.
  • the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
  • the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
  • administering effects a decrease in expression of an HIF target gene or locus.
  • the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI, TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR Flk-1, plasminogen-activator inhibitor- 1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2, a hexokinase
  • vascular endothelial growth factor is reduced in an eye of the subject.
  • activity of a vascular endothelial growth factor (VEGF) is reduced in an eye of the subject.
  • expression of a vascular endothelial growth factor receptor is reduced in an eye of the subject.
  • activity of a vascular endothelial growth factor receptor is reduced in an eye of the subject.
  • the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v.
  • the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to about 0.1%, about 0.1 % to about 0.5%, about 0.5 % to about 1 %, about 1 % to about 1.5%, about 1.5% to about 2%, about 2% to about 2.5%, about 3% to about 4%, about 4% to about 5%, about 5% to about 6%, about 6% to about 7%, or about 7% to about 8%.
  • the HIF inhibitor is administered at a dose of or about 0.001 mg, about 0.002 mg, about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg, about 4.5
  • the HIF inhibitor is administered at a dose of or about 0.001 mg to about 0.01 mg, about 0.01 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
  • administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the retina of the subject.
  • administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the choroid or to the suprachoroidal space.
  • the HIF inhibitor is selected from the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, combinations thereof and the like.
  • the HIF inhibitor is an HIF-1 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF -2 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor and an HIF-2 inhibitor.
  • the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3-[[(lS,2S,3R)- 2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5- fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro- lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)- camptothecin (NSC-639174), a cardenolide
  • the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK-6482, 3-[[(lS,2S,3R)-2,3-difluoro-l- hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)-5- fluorobenzonitrile), combinations thereof and the like.
  • the HIF inhibitor is MK-6482.
  • the FDA has granted breakthrough therapy designation to MK-6482, an HIF-2-alpha inhibitor, for renal cell carcinoma subtype for the treatment of certain patients with von Hippel-Lindau disease- associated renal cell carcinoma.
  • MK-6482 is under investigation for patients with von Hippel-Lindau disease-associated renal cell carcinoma with nonmetastatic tumors smaller than 3 cm, who do not require immediate surgery.
  • administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition.
  • administering the pharmaceutical composition comprises administration into the vitreous cavity of the eye.
  • administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition into the vitreous cavity of an eye of the subject.
  • administering the pharmaceutical composition comprises injecting the pharmaceutical composition. In some exemplary embodiments, administering the pharmaceutical composition comprises intravitreal injection.
  • administering the pharmaceutical composition comprises implanting the pharmaceutical composition. In some exemplary embodiments, administering the pharmaceutical composition comprises implanting the pharmaceutical composition into the vitreous cavity.
  • administering the pharmaceutical composition comprises administration selected from the group consisting of intravitreal injection, intravitreal implant, administering an eye drop, suprachoroidal injection, oral administration, parenteral injection, combinations thereof, and the like.
  • administering the pharmaceutical composition comprises topical administration of an eye drop.
  • administering the pharmaceutical composition comprises topical administration of an eye drop and delivery to the retina.
  • administering the pharmaceutical composition comprises administration to the suprachoroidal space. In some exemplary embodiments, administering the pharmaceutical composition comprises repeated administration of the pharmaceutical composition.
  • administering the pharmaceutical composition comprises administration of the pharmaceutical composition hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months.
  • administration occurs over a regimen of about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 12 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, or about 1 year.
  • the method comprises administering a second therapeutic agent or treatment to the subject for treatment of an ischemic retinal disease.
  • the pharmaceutical composition is administered before, after or with the second therapeutic agent or treatment.
  • the second therapeutic agent is an angiogenesis inhibitor.
  • the second therapeutic agent is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, combinations thereof, and the like.
  • the second therapeutic agent is selected from among the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, a VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • an anti-VEGF antibody ranibizumab, bevacizumab, aflibercept, pegaptanib, a VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetani
  • the second therapeutic agent or treatment is selected from among the group consisting of a corticosteroid (e.g., a corticosteroid selected from the group consisting of dexamethasone, triamcinolone, a combination thereof, and the like).
  • a corticosteroid e.g., a corticosteroid selected from the group consisting of dexamethasone, triamcinolone, a combination thereof, and the like.
  • the second therapeutic agent is formulated in a second pharmaceutical composition.
  • the second therapeutic treatment is selected from the group consisting of laser photocoagulation, macular laser photocoagulation, panretinal photocoagulation (scatter photocoagulation), laser photocoagulation for retinal tears, oxygen therapy (such as hyperbaric), carotid surgery, combinations thereof and the like.
  • the pharmaceutical composition and the second therapeutic agent are administered as a single composition or as two compositions.
  • a composition e.g., a pharmaceutical composition
  • an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia.
  • the atrophy associated with the retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, combinations thereof and the like.
  • the amount of the HIF inhibitor is effective to treat, minimize and/or inhibit apoptosis (e.g., retinal apoptosis) associated with retinal hypoxia in the subject.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, a combination thereof, and the like.
  • a method selected from the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal flu
  • measuring phosphatidyl extemalization comprises annexin 5 staining.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, combinations thereof, and the like.
  • a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, combinations thereof, and the like.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, macular atrophy, choroidal atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy, glaucomatous atrophy, ganglion cell atrophy, combinations thereof, and the like.
  • the atrophy associated with retinal hypoxia is glaucomatous atrophy.
  • retinal ganglion cells can undergo apoptosis due to hypoxia.
  • HIF inhibition can protect ganglion cells from cell death in glaucoma.
  • the glaucomatous atrophy is from glaucoma.
  • the glaucoma is selected from the group consisting of chronic open angle glaucoma, closed angle glaucoma, secondary glaucoma, normal tension glaucoma, and combinations thereof.
  • HIF inhibition can provide neuroprotection in glaucoma.
  • the iris atrophy is from anterior segment ischemia.
  • the ciliary body atrophy is from anterior segment ischemia.
  • the optic nerve atrophy is from vascular ischemia.
  • the vascular ischemia comprises a condition selected from the group consisting of giant cell arteritis, embolisms, and a combination thereof.
  • the optic nerve atrophy comprises anterior ischemia optic neuropathy.
  • the retinal atrophy is macular atrophy or geographic atrophy. In some exemplary embodiments, the atrophy is choroidal atrophy.
  • the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor.
  • the retinal detachment is selected from the group consisting of tractional, rhegmatogenous and serous retinal detachment.
  • the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor.
  • the administration of a VEGF or VEGFR inhibitor is for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the total area of the atrophy associated with retinal hypoxia.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
  • the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the total area of atrophy as assessed by a method selected from the group consisting of spectral-domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, biomicroscopy, combinations thereof, and the like.
  • OCT spectral-domain optical coherence tomography
  • OCT near-infrared reflectance
  • fundus photography visual acuity testing
  • microperimetry visual field testing
  • biomicroscopy combinations thereof, and the like.
  • the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the total area of retinal atrophy associated with retinal hypoxia as assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, visual acuity testing, near-infrared reflectance, fundus photography, biomicroscopy, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), retinal oximetry, microperimetry, retinal oximetry, combinations thereof, and the like.
  • a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, visual acuity testing, near-infrared reflectance, fundus photography, biomicroscopy, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), retinal oximetry, microperimetry, retinal oximetry, combinations thereof, and the like
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce and/or substantially maintain the severity grade of atrophy associated with retinal hypoxia.
  • the pharmaceutical composition comprises an amount of the HIF inhibitor effective to improve, or substantially maintain, the Age-Related Eye Disease Study (AREDS) scale.
  • AREDS Age-Related Eye Disease Study
  • the amount of the HIF inhibitor is effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia in a subject being treated with an angiogenesis inhibitor.
  • the angiogenesis inhibitor is a VEGF inhibitor and/or a VEGFR inhibitor.
  • the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof and the like).
  • the angiogenesis inhibitor is a VEGFR inhibitor selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • VEGFR inhibitor selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • the retinal hypoxia is associated with a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof.
  • the retinal hypoxia is associated with retinal detachment.
  • delivery of oxygen to the retina can be reduced because of the increased distance from the choroidal source of oxygen. Retinal detachments thus can be hypoxic.
  • the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, serous retinal detachment, rhegmatogenous retinal detachment, tractional retinal detachment, proliferative vitreoretinopathy (PVR), and central serous chorioretinopathy.
  • grade A proliferative vitreoretinopathy grade B proliferative vitreoretinopathy
  • grade C P proliferative vitreoretinopathy grade C A proliferative vitreoretinopathy
  • serous retinal detachment rhegmatogenous retinal detachment
  • tractional retinal detachment proliferative vitreoretinopathy (PVR), and central serous chorioretinopathy.
  • PVR proliferative vitreoreti
  • HIF inhibition can treat prevent, reduce, or substantially inhibit HIF induced apoptosis and/or atrophy (e.g., retinal atrophy).
  • consequent vision loss can be treated, prevented, reduced or substantially inhibited.
  • retinal atrophy and/or thinning can be treated, reduced, prevented, or substantially inhibited with anti HIF treatment.
  • proliferative vitreoretinopathy PVR
  • atrophy in PVR can be treated.
  • traction retinal detachments e.g., proliferative diabetic retinopathy, retinopathy of prematurity, PVR, combinations thereof and the like
  • retinal atrophy and/or thinning can be reduced and/or prevented by an HIF inhibitor.
  • the retinal hypoxia is associated with an ischemic retinal disease.
  • the HIF inhibitor is effective to treat, minimize and/or substantially inhibit a symptom associated with the ischemic retinal disease.
  • the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion, combinations thereof, and the like.
  • the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
  • the ischemic retinal disease is selected from the group consisting of dry atrophic age related macular degeneration (atrophic AMD; geographic atrophy), diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy retinal pigment epithelial detachment, central serous chorioretinopathy, combinations thereof, and the like.
  • atrophic AMD dry atrophic age related macular degeneration
  • ROP retinopathy of prematurity
  • sickle cell retinopathy retinal pigment epithelial detachment central serous chorioretinopathy, combinations thereof, and the like.
  • the HIF inhibitor is present in an amount effective to reduce progression of retinal atrophy in dry AMD and dry atrophic AMD (Geographic atrophy); and/or reduce or prevent atrophy associated with anti VEGF and/or anti VEGFR treatment for neovascular AMD.
  • HIF inhibitor is present in an amount effective to treat, minimize or substantially inhibit angiogenesis and/or edema in addition to atrophy. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to treat, minimize, substantially inhibit and/or reduce progression of retinal atrophy (e.g., in dry AMD) and/or severe and symptomatic atrophy (Geographic atrophy).
  • the ischemic retinal disease is diabetic macular edema.
  • the ischemic retinal disease is diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy).
  • diabetic retinopathy e.g., proliferative or non-proliferative diabetic retinopathy.
  • the ischemic retinal disease is selected from the group consisting of diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy), retinal vein occlusions, sickle cell retinopathy, combinations thereof, and the like.
  • the HIF inhibitor is present in an amount effective to reduce or prevent retinal atrophy associated with anti VEGF and/or anti VEGFR treatment of diabetic macular edema or proliferative diabetic retinopathy, a combination thereof, or the like.
  • the HIF inhibitor is present in an amount effective to treat, minimizes or substantially inhibit angiogenesis and/or edema in addition to atrophy.
  • the HIF inhibitor is present in an amount effective to treat, minimizes or substantially inhibit progression of retinal atrophy (e.g., in patients with macular ischemia) and severe and symptomatic atrophy.
  • the ischemic retinal disease is diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy or macular ischemia in diabetic retinopathy).
  • the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, and severe non-proliferative diabetic retinopathy.
  • the ischemic retinal disease is proliferative diabetic retinopathy. In some exemplary embodiments, the ischemic retinal disease is central retinal vein occlusion. In some exemplary embodiments, the ischemic retinal disease is branch retinal vein occlusion.
  • the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
  • the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
  • the HIF inhibitor is present in an amount effective to decrease expression of an HIF target gene or locus.
  • the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI, TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, plasminogen- activator inhibitor- 1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2,
  • Leptin Leptin
  • Glucose transporter 1 GLUT1
  • Hexokinase 1 and 2 HK1 and 2
  • 6-Phosphofructo-l -kinase L PPKL
  • 6-Phosphofructo-2-kinase Glyceraldehyde-3-P dehydrogenase
  • Glyceraldehyde-3-P dehydrogenase Glyceraldehyde-3-P dehydrogenase
  • APDH Aldolase A
  • ALDC Aldolase C
  • Enolase 1 ENOl
  • Phosphoglycerate kinase-1 PGK1
  • Lactate dehydroxygenase A LDHA
  • Pyruvate kinase M PPM
  • Carbonic anhydrase 9 CA9
  • Adenylate kinase 3 Transglutaminase 2
  • Pro collagen prolyl hydroxylase al Collagen type V (al) Intest
  • the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor (VEGF) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject.
  • VEGF vascular endothelial growth factor
  • wherein the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor (VEGF) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject.
  • the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor receptor (VEGFR) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject.
  • the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor receptor (VEGFR) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject.
  • the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v.
  • the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to 0.1%, about 0.1% to 0.5%, about 0.5% to 1%, about 1% to 1.5%, about 1.5% to 2%, about 2% to 2.5%, about 3% to 4%, about 4% to 5%, about 5% to 6%, about 6% to 7%, or about 7% to about 8%.
  • the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg, about 0.002 mg, about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg,
  • the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg to 0.01 mg, about 0.01 mg to 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
  • the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the retina of the subject. In some exemplary embodiments, the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the choroid of the subject.
  • the HIF inhibitor is selected from among the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, combinations thereof and the like.
  • the HIF inhibitor is an HIF-1 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF -2 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor and an HIF-2 inhibitor.
  • the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3-[[(lS,2S,3R)- 2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5- fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro- lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)- camptothecin (NSC-639174), a cardenolide
  • the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK-6482; 3-[[(lS,2S,3R)-2,3-difluoro-l- hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)-5- fluorobenzonitrile), combinations thereof and the like.
  • the pharmaceutical composition is formulated for administration by injection and/or implantation.
  • the pharmaceutical composition is formulated for administration into the vitreous cavity of the eye.
  • the pharmaceutical composition is formulated for administration by injection and/or implantation into the vitreous cavity of an eye of the subject.
  • the pharmaceutical composition is formulated for administration by injection. In some exemplary embodiments, the pharmaceutical composition is formulated for administration by intravitreal injection.
  • the pharmaceutical composition is formulated for administration by implantation. In some exemplary embodiments, the pharmaceutical composition is formulated for administration by implantation into the vitreous cavity.
  • the pharmaceutical composition is formulated for administration selected from the group consisting of intravitreal injection, intravitreal implant, eye drop, suprachoroidal injection, oral administration, parenteral injection, combinations thereof, and the like.
  • the pharmaceutical composition is formulated for topical administration as an eye drop.
  • the pharmaceutical composition is formulated for delivery to the retina. In some exemplary embodiments, the pharmaceutical composition is formulated for delivery to the choroid. In some exemplary embodiments, the pharmaceutical composition is formulated for administration into the vitreous cavity of the eye. In some exemplary embodiments, the pharmaceutical composition is formulated for implantation into the vitreous cavity. In some exemplary embodiments, the pharmaceutical composition is formulated for intravitreal injection into the vitreous cavity.
  • the pharmaceutical composition is formulated for administration to the suprachoroidal space.
  • the pharmaceutical composition is formulated for repeated administration. In some exemplary embodiments, the pharmaceutical composition is formulated for administration selected from the group consisting of hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months.
  • the pharmaceutical composition is formulated for administration over a regimen of about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 12 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, or about 1 year or more.
  • Another exemplary embodiment of this application is a combination, comprising the pharmaceutical composition described herein, and a second pharmaceutical composition comprising a second therapeutic agent for treatment of an ischemic retinal disease and/or treatment of a retinal detachment, such as ischemic retinal diseases and retinal detachments described herein.
  • the pharmaceutical composition containing an HIF inhibitor is for administration before, after or with the second pharmaceutical composition.
  • the second therapeutic agent is an angiogenesis inhibitor.
  • the second therapeutic agent is selected from the group consisting of a VEGF inhibitor and/or a VEGFR inhibitor.
  • the second therapeutic agent is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
  • the second therapeutic agent is selected from among the group consisting of dexamethasone, triamcinolone, a corticosteroid, combinations thereof, and the like.
  • the pharmaceutical composition containing an HIF inhibitor and the second pharmaceutical composition are formulated for administration as a single composition or as two compositions.
  • the methods, compositions, combinations, and uses described herein for treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia can also be adapted for other forms of atrophy associated with hypoxia in other body systems, organs, and/or tissue.
  • described herein are methods, compositions, combinations, and uses for treating, minimizing and/or substantially inhibiting atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia.
  • the hypoxia is not retinal hypoxia.
  • an exemplary embodiment of this application is a method of treating, minimizing and/or substantially inhibiting atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor (e.g., an HIF inhibitor described herein), such as an effective amount of an HIF inhibitor, to a subject having hypoxia.
  • an HIF inhibitor e.g., an HIF inhibitor described herein
  • the HIF inhibitor is administered (e.g., systemically and/or as described herein) in an amount effective to treat, minimize and/or substantially inhibit atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia.
  • compositions e.g., a pharmaceutical composition
  • an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia.
  • the composition is formulated for administration systemically and/or as described herein.
  • ischemia leads to the atrophy (e.g., tissue and/or organ atrophy).
  • the atrophy associated with hypoxia is selected from among the group consisting of atrophy of limbs (e.g., caused by vascular occlusions); atrophy of heart muscle (e.g., from coronary artery occlusions); ischemic atrophy of the liver, ischemic atrophy of the kidney, ischemic atrophy of the brain, combinations thereof, and the like.
  • hypoxia-inducible factor HIF
  • hypoxia-inducible factors regulate cellular response to low oxygen concentrations.
  • An HIF transcription factor dimer includes one of three oxygen-regulated a-subunits (e.g., HIF-la, HIF-2a, or HIF-3a) and a constitutively expressed beta-subunit (e.g., HIF-Ib or HIF- 2b).
  • An HIF can bind to consensus sequences (hypoxia responsive elements, HRE) in the regulatory regions of target genes.
  • HIF-a e.g., HIF-la or HIF-2a
  • HIF-PD HIF-prolyl hydroxylase
  • pVHL von-Hippel-Lindau tumor suppressor protein
  • Transcriptional activity of a HIF-a subunit can depend on oxygen-dependent hydroxylation of asparagine residues in HIF-a by the asparagyl hydroxylase factor-inhibiting HIF (FIH), preventing the binding of transcriptional coactivators (CBP/p300) and expression of target genes under oxygenated conditions.
  • Transcriptional activity of HIF can be regulated, for example, by the accumulation or turnover of the HIF-a (e.g., HIF-la or HIF-2a) monomer.
  • HIF activity can be inhibited by targeting one or more components that mediate hypoxic response. For example, SUMOylation of p300 can block interaction with HIF-a.
  • HIF-a Phosphorylation of HIF-a can block interaction with HIF-b (e.g., HIF-Ib or HIR-2b).
  • COMMD1 can bind to HIF-a (e.g., HIF-la or HIF-2a) and block interaction with HIF-b (e.g., HIF-Ib or HIR-2b).
  • Cited-2 can bind to HIF-a (e.g., HIF-la or HIF-2a) and block interaction with p300.
  • HIF-PH activity is decreased.
  • HIF-a e.g., HIF-la or HIF-2a
  • HIF-b e.g., HIF-Ib or HIR-2b
  • HIF target genes can include, but are not limited to, erythropoietin (EPO), VEGF, glucose transporter 1 (GLUT1), glycolytic enzymes (e.g, phosphoglycerate kinase 1, lactate dehydrogenase-A, carbonic anhydrase 9, and aldolase), transforming growth factor alpha, and cyclin D.
  • EPO erythropoietin
  • VEGF VEGF
  • GLUT1 glucose transporter 1
  • glycolytic enzymes e.g, phosphoglycerate kinase 1, lactate dehydrogenase-A, carbonic anhydrase 9, and aldolase
  • transforming growth factor alpha cyclin D.
  • Glycolytic enzymes can be regulated by HIF-la, while HIF-2a can regulate gene transcription of EPO, transforming growth factor alpha, and cyclin D.
  • Some target genes including VEGF, GLUT1, and adrenomedullin 1 (ADM-1), can be regulated by HIF-la and HIF-2a.
  • HIF-3a can directly regulate a subset of hypoxia-inducible genes involved in lipolysis (angiopoietin-like 4) and metabolism (angiopoietin-like 3 and pantothenate kinase 1). HIF- 3a can also interact with the promotor region of the EPO gene. Therefore, HIF-l/2a and HIF- 3 a could have synergistic effects on EPO transcription (Tolonen et al. (2020) Cell. Mol. Life Sci. 77:3627-3642).
  • HIF target genes or loci include genes involved in angiogenic signalling (e.g, Angiopoietin-1, Angiopoietin-2, Angiopoietin-4, Angiopoietin-like Protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-beta 1, TGF-beta 3, VEGF, Endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, Plasminogen-activator inhibitor-1 (PAI1), and Urokinase plasminogen activator receptor (UPAR)), in metabolism (e.g., GAPDH, Glutl, Glut3, Hexokinase 1, Hexokinase 1/2, Hexokinase 2, Hexokinase Activators, Lactate Dehydrogenase A
  • HIF inhibitor can be used in the compositions, uses, and methods described herein, if the HIF inhibitor can inhibit the HIF pathway.
  • Inhibiting the HIF pathway can include inhibiting one or more of any of the components of the HIF pathway.
  • an HIF inhibitor can inhibit HIF, or can target components of the HIF pathway that mediate hypoxic response (e.g., PHDs, pVHL, FIH and CBP/p300).
  • HIF inhibitor can be injected and/or implanted into the vitreous humor or formulated for injection and/or implantation into the vitreous humor.
  • HIF inhibitors can target one or more selected from the group consisting of inhibiting transcription of HIF mRNA, inhibiting HIF protein synthesis, interfering with stabilization of HIF, decreasing transcription of HIF target genes, activating prolyl -4-hydroxylase domain (PHD), interfering with interactions between HIF-a’s to von Hippel-Lindau tumor suppressor protein (pVHL), combinations thereof and the like.
  • the HIF inhibitors can inhibit the HIF pathway, for example, by inhibiting transcription and/or translation, HIF stabilization, HIF-a/b dimerization, transcription complex formation, combinations thereof and the like.
  • the HIF inhibitor disrupts heterodimerization (e.g., heterodimerization selected from the group consisting of HIF-2a/HIF-i , HIF-la/HIF-Ib, HIF-2a/HIF ⁇ , HIF-la/HIF ⁇ , and combinations thereof).
  • heterodimerization selected from the group consisting of HIF-2a/HIF-i , HIF-la/HIF-Ib, HIF-2a/HIF ⁇ , HIF-la/HIF ⁇ , and combinations thereof.
  • the HIF inhibitor is belzutifan (MK-6482; 3-[[(lS,2S,3R)-2,3-difluoro-l-hydroxy-7- methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), an inhibitor of HIF- 2a/HIR-1b heterodimerization. (Courtney et al. (2018) J. Clin. Oncol. 36(9):867-874).
  • the HIF inhibitor blocks binding of HIF to DNA.
  • the HIF inhibitor is doxorubicin. Doxorubicin can inhibit HIF-1 transcriptional activity of blocking binding of HIF- 1 to DNA (Duyndam et al. (2007) Biochem. Pharmacol. 74(2): 191-201; Lee et al. (2009) Proc. Natl. Acad. Sci. USA. 106:2353-8).
  • the HIF inhibitor is a topoisomerase inhibitor. In some further embodiments, the HIF inhibitor is a topoisomerase-I inhibitor. In some further embodiments, the HIF inhibitor is camptothecin or an analog of camptothecin. In some further embodiments, the HIF inhibitor is selected from the group consisting of topotecan (NSC-609699), camptothecin 20-ester(S) (NSC- 606985), and 9-glycineamido-20(S)-camptothecin or an HC1 salt thereof (NSC-639174).
  • the HIF inhibitor is the topoisomerase-I inhibitor topotecan, which inhibits HIF-1 transcriptional activity.
  • the HIF inhibitor is a cardenolide.
  • the HIF inhibitor is a cardenolide that transcriptionally inhibits HIF-1.
  • the HIF inhibitor is SN38 (7-Ethyl-lO-hydroxy-camptothecin). Kami yam a el al. (2005) J. Cancer Res. Clin. Oncol. 131:205-213.
  • the HIF inhibitor is a Ca 2+ channel blocker.
  • the HIF inhibitor is the Ca 2+ channel blocker NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH- benzimidazol-2-yl)propyl]methylamino] ethyl] -6-fluoro- 1 ,2,3,4-tetrahydro- 1 -( 1 -methylethyl)- 2-naphthalenyl ester, dihydrochloride; CAS No.
  • the HIF inhibitor is PX-478 (,S'-2-amino-3-
  • the HIF inhibitor blocks the PI3K/Akt/TOR and/or MAPK pathway.
  • the HIF inhibitor is bortezomib (PS-341), which can represses HIF- la on transcriptional and translational levels, and inhibit recruitment of the coactivator p300, blocking the PI3K Akt/TOR and MAPK pathway.
  • the HIF inhibitor is selected from among resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, and sorafenib.
  • the HIF inhibitor inhibits the PI3K-AKT pathway.
  • the HIF inhibitor is LY294002, wortmannin, or nelfmavir. (Jiang et al. (2001) Cell Growth Differ. 12(7):363-9; Pore et al. (2006) Cancer Res. 66(18):9252-9).
  • the HIF inhibitor inhibits the Pi3K/AKT/mTOR pathway. In some embodiments, the HIF inhibitor decreases HSP90 binding. In some embodiments, the HIF inhibitor is a glyceollin (i.e., a soybean-derived phytoalexin), which can block HIF-la translation via inhibition of the Pi3K/AKT/mTOR pathway and decrease HIF-Ia stability by decreasing Hsp90 binding. (Lee et al. (2015) J. Cell. Physiol. 230:853-862).
  • the HIF inhibitor for use in the compositions, uses, and methods described herein is an HIF-Ia inhibitor.
  • the HIF inhibitor is PX-478.
  • Other HIF inhibitors for use in the compositions, uses, and methods described herein is an HIF-Ia inhibitor selected from among the group consisting of Bortezomib (Velcade®), Glyceollins, NNC55-0396, PX-478, Aminoflavone, Benzopyranyl 1,2,3-trizole, BIX01294, Bortezomib (Velcade®), Cardenolides (e.g.
  • EZN-2208 (PEG-SN38)), CRLX-101, Digoxin, Erotinib, Everolimus, EZN-2968, Gefitinib, Genistein, Glyceollins, IDF-1174, Kresoxim- methyl analoges, LBH589 (Panobinostat), LY294002, MPTOG1S7, Nelfmavir, NNC55- 0396, PD98059, Rapamycin, Resveratrol, RNA interference, Silibinin, Sorafenib, Temsirolimus, Tetrathiomolybdate, Topotecan, and Wortmannin.
  • the HIF inhibitor is a melphalan derivative. In some embodiments, the HIF inhibitor is an alkylating agent. In some embodiments, the HIF inhibitor is a nitrogen mustard or nitrogen mustard derivative. In some embodiments, the HIF inhibitor is a nitrogen mustard N-oxide or a nitrogen mustard N-oxide derivative.
  • the HIF inhibitor increases HIF-a (e.g., HIF-la or HIF-2a) protein degradation.
  • HIF inhibitor can increase pVHL activity.
  • the HIF inhibitor upregulates pVHL expression.
  • the HIF inhibitor is IDF- 11774 (2-(4-((3r,5r,7r)-adamantan- 1 -yl)phenoxy)- 1 -(4-methylpiperazin- 1 -yl)ethan- 1 - one), which can upregulate pVHL expression, resulting in increased degradation of HIF-1.
  • the HIF inhibitor is a histone deacetylase (HDAC) inhibitor.
  • HDAC histone deacetylase
  • the HIF inhibitor is panobinostat (LBH589; (E)-N-hydroxy-3-[4- [[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop-2-enamide), a HDAC inhibitor that can disrupt the Hsp90/HDAC6 complex (Kovacs et al. (2005 )Mol. Cell 18:601-607). Hsp90 complexing with HIF-Ia, and also acetylation of HIF-Ia, can prevent degradation through the proteasome/pVHL pathway complex.
  • histone deacetylase (HDAC) inhibitors such as panobinostat can reduce HIF-Ia protein.
  • the HIF inhibitor is the indole-3 -ethylsulfamoylphenylacrylamide compound MPT0G157.
  • MPT0G157 can inhibit multiple histone deacetylases (1, 2, 3, and 6) and decrease levels of HIF-Ia protein. (Huang et al. (2015) Oncotarget 6: 18590).
  • the HIF inhibitor can increase PHD2 and/or pVHL expression.
  • the HIF inhibitor is a diazepinquinazolin-amine derivate.
  • the HIF inhibitor is BIX01294 (N-(l-benzylpiperidin-4-yl)-6,7- dimethoxy-2-(4-methyl-l,4-diazepan-l-yl)quinazolin-4-amine), which can increase PHD2 and pVHL expression and can reduce HIF-Ia protein levels. (Oh et al. (2015) Mol. Cells 38, 528).
  • the HIF inhibitor can induce HIF (e.g.
  • the HIF inhibitor is a benzopyranyl 1,2,3-triazole.
  • the HIF inhibitor is 4-(4-methoxyphenyl)-l-((2-methyl-6-nitro-2H-chromen-2- yl)methyl)-lH-l, 2, 3-triazole. (Park, (2017) Oncotarget 8:7801).
  • the HIF inhibitor can increase oxygen tension. In some further embodiments the HIF inhibitor can promote proteasomal degradation of HIF-a (e.g., HIF-la) via increased oxygen tension. In some further embodiments, the HIF inhibitor is Kresoxim- methyl or an analog of Kresoxim -methyl. (Lee et al. (2017) Bioorg. Med. Chem. Lett. 27:3026-3029).
  • the HIF inhibitor is a nanoparticle or nanoparticle conjugate. In some further embodiments, the HIF inhibitor is a nanoparticle of an active compound conjugated to a cyclodextrin-based polymer (e.g., a linear cyclodextrin-based polymer). In some further embodiments, the HIF inhibitor is camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer. In some further embodiments, the HIF inhibitor is CRLX-101, and can suppress HIF-a (e.g., HIF-la) protein translation and stability. (Pham et al. (2015) Clin. Cancer Res. 21:808-818).
  • HIF-a e.g., HIF-la
  • the HIF inhibitor can inhibit one or more components of the HIF pathway selected from among the group consisting of HIF-a/b dimerization, transcription complex formation, and combinations thereof.
  • HIF-a/b dimerization is a part of the pathway in which HIF complex induces expression of HIF target genes.
  • the HIF inhibitor is an HIF -2a inhibitor.
  • the HIF inhibitor is selected from among the group consisting of PT2385, PT2399, and PT2977. (Cho et al. (2016) Nature 2016, 539, 107-111; Wallace et al. Cancer Res. 2016, 76, 5491- 5500; and Courtney et al. (2016) J. Clin. Oncol. 36:867).
  • the HIF inhibitor can disrupt HIF heterodimer formation.
  • the HIF inhibitor is the compound 0X3 (N-(3-Chloro-5-fluorophenyl)-4- nitrobenzo[c][l,2,5]oxadiazol-5-amine). (Scheuermann et al. (2013) Nat. Chem. Biol. 9:271).
  • the HIF inhibitor can bind to the PAS-B domain of HIF-la or HIF-2a and block heterodimerization with HIF-b (e.g., HIF-Ib).
  • HIF-b e.g., HIF-Ib
  • the HIF inhibitor is acriflavine (ACF), which can bind to the PAS-B domain of HIF-la and HIF-2a.
  • ACF acriflavine
  • the HIF inhibitor inhibits a transcriptional coactivator of HIF (e.g., CBP or p300).
  • the HIF inhibitor is selected from the group consisting ofCG13250, CCS1477 ((S)-l-(3,4-Difluorophenyl)-6-(5-(3,5- dimethylisoxazol-4-yl)- 1 -(( lr,4S)-4-methoxycyclohexyl)- lH-benzo[d]imidazol-2- yl)piperidin-2-one), bortezomib ([(1 R)-3 -methyl- 1 - [[(2S)-3 -phenyl-2-(pyrazine-2- carbonylamino)propanoyl]amino]butyl]boronic acid) and chetomin (Shin et al.
  • the HIF inhibitor is a receptor tyrosine kinase inhibitor, such as an EGFR tyrosine kinase inhibitor, including, for example, an inhibitor selected from the group consisting of Erotinib, Gefitinib, and Genistein. (Pore et al. (2006) Cancer Res.
  • the HIF inhibitor is selected from the group consisting of apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, and pleurotin (Fang et al. (2007) Carcinogenesis 28(4):858-64; Kim et al. (2009) Cancer Res. 2009;69(4): 1624-32; Alqawi et al. (2006) Prostate Cancer Prostatic Dis.
  • Table 1 below sets forth exemplary HIF inhibitors for use in the compositions, uses, and methods described herein. This is not an exhaustive list. Description in the table is not meant to be limiting. It is understood that description of an inhibition mechanism or pathway does not exclude other inhibition mechanisms or pathways. Table 1. Exemplary HIF Inhibitors
  • Hypoxic conditions can result from different diseases and conditions.
  • ischemia e.g., ischemic retinal disease
  • retinal detachment can result in hypoxic conditions, which can activate HIF.
  • HIF can trigger neovascularization, edema, and apoptosis. Apoptosis can result in atrophy.
  • HIF inhibitors can inhibit hypoxia induced apoptosis and/or atrophy (.e.g, in an ischemic retinal disease and in retinal hypoxic conditions from other causes (for example retinal detachments)).
  • HIF inhibitors can inhibit retinal apoptosis/atrophy that accompanies administration of an angiogenesis inhibitor, for example for treatment of an ischemic retinal disease.
  • Exemplary diseases and conditions that can result in retinal hypoxia include, but are not limited to, age related macular degeneration (AMD), geographic atrophy (also known as dry atrophic age related macular degeneration (atrophic AMD)), dry AMD, diabetic retinopathy (e.g., proliferative diabetic retinopathy or non-proliferative diabetic retinopathy or diabetic macular edema or macular ischemia in diabetic retinopathy), retinal vein occlusion (e.g., central retinal vein occlusion or branch retinal vein occlusion), retinopathy of prematurity (ROP), sickle cell retinopathy, rhegmatogenous or tractional retinal detachment and proliferative vitreoretinopathy (PVR), retinal pigment epithelial detachment, central serous chorioretinopathy and other serous retinal detachments.
  • AMD age related macular degeneration
  • atrophic AMD dry atroph
  • the ischemic disease is atrophic AMD or diabetic retinopathy.
  • the HIF inhibitor prevents and/or substantially inhibits one or more symptoms selected from the group consisting of apoptosis (e.g., retinal apoptosis), retinal atrophy and choroidal atrophy associated with atrophic AMD or diabetic retinopathy.
  • the retinopathy of prematurity is selected from the group consisting of stage 1 ROP (e.g., mildly abnormal blood vessel growth), stage 2 ROP (e.g., moderately abnormal blood vessel growth), stage 3 ROP (e.g., severely abnormal blood vessel growth), stage 4 ROP (e.g., partially detached retina), or stage 5 ROP (e.g., completely detached retina).
  • stage 1 ROP e.g., mildly abnormal blood vessel growth
  • stage 2 ROP e.g., moderately abnormal blood vessel growth
  • stage 3 ROP e.g., severely abnormal blood vessel growth
  • stage 4 ROP e.g., partially detached retina
  • stage 5 ROP e.g., completely detached retina
  • the diabetic retinopathy is selected from among the group consisting of stage 1 diabetic retinopathy (e.g., mild nonproliferative retinopathy), stage 2 diabetic retinopathy (e.g., moderate nonproliferative retinopathy), stage 3 diabetic retinopathy (e.g., severe nonproliferative retinopathy), stage 4 diabetic retinopathy (e.g., proliferative retinopathy) and diabetic macular edema.
  • stage 1 diabetic retinopathy e.g., mild nonproliferative retinopathy
  • stage 2 diabetic retinopathy e.g., moderate nonproliferative retinopathy
  • stage 3 diabetic retinopathy e.g., severe nonproliferative retinopathy
  • stage 4 diabetic retinopathy e.g., proliferative retinopathy
  • diabetic macular edema edema
  • the sickle cell retinopathy is selected from among the group consisting of stage 1 sickle cell retinopathy (e.g., peripheral arterial occlusion), stage 2 sickle cell retinopathy (e.g., Peripheral arteriovenous anastomoses, representing dilated pre-existing capillaries (hairpin loop)), stage 3 sickle cell retinopathy (e.g., neovascular and fibrous proliferation (sea fan)), stage 4 sickle cell retinopathy (e.g., vitreous hemorrhage), and stage 5 sickle cell retinopathy (e.g., tractional retinal detachment).
  • stage 1 sickle cell retinopathy e.g., peripheral arterial occlusion
  • stage 2 sickle cell retinopathy e.g., Peripheral arteriovenous anastomoses, representing dilated pre-existing capillaries (hairpin loop)
  • stage 3 sickle cell retinopathy
  • central serous chorioretinopathy and other serous retinal detachments are treated with HIF inhibitor to prevent or reduce long term retinal atrophy.
  • rhegmatogenous or tractional retinal detachments or proliferative vitreoretinopathy are treated with HIF inhibitor to prevent or reduce long term retinal atrophy.
  • HIF inhibition complements or replaces anti VEGF or anti VEGFR treatment for neovascular AMD, proliferative diabetic retinopathy, diabetic macular edema or neovascularization/edema in retinal vein occlusion, in order to reduce the retinal atrophy that otherwise accompanies VEGF inhibition.
  • the methods can include administration of an HIF inhibitor, alone or in combination with another agent or treatment for treating, minimizing and/or substantially inhibiting an ischemic retinal disease or condition described herein.
  • the methods can include administration of a composition containing an HIF inhibitor as described herein.
  • the methods can include selection of subjects for treatment, e.g., prior to treatment of the subject.
  • Subjects can be determined to have one or more selected from the group consisting of an ischemic retinal disease or condition, retinal hypoxia, retinal detachment (or a related condition), retinal neovascularization, and activated HIF.
  • the method further includes treatment, for example, administration of an HIF inhibitor alone or in combination with one or more other treatments.
  • the method further includes determining reduction in a symptom after treatment. In some examples, the method further includes determining reduction in an adverse effect of treatment. In some examples, the symptom or adverse effect is selected from among the group consisting of apoptosis (e.g., retinal apoptosis), and atrophy (e.g., retinal atrophy and/or choroidal atrophy).
  • apoptosis e.g., retinal apoptosis
  • atrophy e.g., retinal atrophy and/or choroidal atrophy
  • Assays to determine reduction in a symptom or adverse effect can include, for example, assays described herein, such as, for example, measuring pupil’s reaction to light, visual acuity, visual field or peripheral vision testing, microperimetry, annexin 5 staining, measuring phosphatidyl extemalization, retinal fundus photography, spectral-domain optical coherence tomography (OCT), near-infrared reflectance, assessment of Age-Related Eye Disease Study (AREDS) scale, diabetic retinopathy grading, combinations thereof, and the like.
  • assays described herein such as, for example, measuring pupil’s reaction to light, visual acuity, visual field or peripheral vision testing, microperimetry, annexin 5 staining, measuring phosphatidyl extemalization, retinal fundus photography, spectral-domain optical coherence tomography (OCT), near-infrared reflectance, assessment of Age-Related Eye Disease Study (AREDS) scale, diabetic reti
  • a subject with elevated HIF activiation in an eye is treated with an HIF inhibitor.
  • treatment includes a reduction in the HIF activation in the eye of the subject.
  • the HIF activation level can be determined prior to and/or after treating the subject, for example, as described herein or known to those of skill in the art.
  • HIF activation can be reduced by, for example about 5% or more, about 10% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 50% or more, or about 75% or more compared to HIF activation prior to treatment.
  • HIF activation can be reduced by, for example about 5% to about 10%, about 10% to about 15%, about 15% to about 20%, about 20% to about 25%, about 25% to about 30%, or about 30% to about 50%.
  • the HIF inhibitor is administered locally, for example, by topical administration of an eye drop or by injection or implantation into the vitreous humor or suprachoroidal space.
  • the HIF inhibitor is delivered to the retina or choroid.
  • the HIF-inhibitor can be administered, for example, by injection or implantation.
  • the HIF inhibitor is adminstered into the vitreous cavity of the eye by intravitreal injection or by implantation into the vitreous cavity.
  • the HIF inhibitor is administered systemically, for example, intravenously (IV) or intramuscularly.
  • the HIF inhibitor can be administered intraocularly, orally, intravenously (IV), subcutaneously, intramuscularly, intraperitoneally, intradermally, topically, transdermally, rectally or sub-epidermally.
  • the methods, uses and compositions and provided herein also can be used to treat atrophy associated with retinal hypoxia by administration of the HIF inhibitor in combination with, for example, simultaneously, prior to, or after, another therapeutic agent or treatment.
  • the other therapeutic agent or treatment can be to treat a disease or condition selected from the group consisting of for an ischemic retinal disease, a retinal detachment (or related condition), atrophy, combinations thereof, and the like.
  • the other therapeutic agent or treatment can be formulated with, or separate from, the HIF inhibitor.
  • the treatment of an ischemic retinal disease can include administration of an angiogenesis inhibitor (e.g., a VEGFR inhibitor (e.g., an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like), and/or a VEGF inhibitor (e.g., an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof and the like)).
  • angiogenesis inhibitor e.g., a VEGFR inhibitor (e.g., an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sun
  • Treatment with an angiogenesis inhibitor can inhibit neovasculazation.
  • HIF activation can result in one or more adverse effects selected from among apoptosis (e.g., retinal apoptosis), retinal atrophy, choroidal atrophy, vision loss and combinations thereof, as neovascularization is blocked.
  • HIF activation can result in several mitigating actions to hypoxia. These include VEGF production for angiogenesis and increased blood flow.
  • angiogenesis inhibitor e.g., VEGF inhibitor or VEGFR inhibitor
  • mitigating mechanisms such as apoptosis, mitigate hypoxia, which can result in retinal atrophy, for example with anti VEGF treatment in neovascular AMD.
  • Inhibiting HIF can decrease one or more effects selected from among apoptosis (e.g., retinal apoptosis), atrophy (e.g., retinal atrophy and/or choroidal atrophy), vision loss, combinations thereof and the like.
  • apoptosis e.g., retinal apoptosis
  • atrophy e.g., retinal atrophy and/or choroidal atrophy
  • vision loss e.g., combinations thereof and the like.
  • apoptosis can be assessed by measuring phosphatidyl extemalization.
  • phosphatidyl extemalization can be measured by annexin 5 staining.
  • apoptosis can be assessed by a method described herein, such as a method selected from the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like.
  • apoptosis can be assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, combinations thereof, and the like.
  • atrophy can be assessed by the Age-Related Eye Disease Study (AREDS) Grading Scale. ( Arch Ophthalmol.
  • the area of atrophy can be assessed before and after administration of a composition described herein to determine if the area of atrophy changes or stays the same.
  • the area of atrophy can be assessed by fundus photography, spectral-domain optical coherence tomography (OCT), visual field examination, microperimetry, biomicroscopy, multifocal electroretinography, near-infrared reflectance, combinations thereof, and the like.
  • OCT spectral-domain optical coherence tomography
  • treatment can result in a reduction in severity grade of atrophy or reduction in the rate of progression of atrophy.
  • compositions of an HIF inhibitor are provided herein.
  • the composition is a pharmaceutical composition.
  • the HIF inhibitor can be formulated into pharmaceutical compositions using techniques and procedures well known in the art (see e.g., Patel etal. (2013) World J. Pharmacol. 2(2):47-64).
  • the mode of formulation can be a function of the route of administration.
  • the HIF inhibitor or a pharmaceutically-acceptable salt thereof can be in a form selected from solid, solution or suspension.
  • the concentration and/or dose of the HIF inhibitor can be adjusted so that administration provides an effective amount to produce the desired pharmacological effect, and can include any concentration or dose described herein.
  • the HIF inhibitor can be provided in a sufficient amount to inhibit a symptom of an ischemic retinal disease described herein (e.g., retinal apoptosis, retinal atrophy, choroidal atrophy, or a combination thereof), or to inhibit an adverse effect of treatment.
  • the exact dose can depend on the age, weight and condition of the patient or animal as is known in the art.
  • the composition e.g., a pharmaceutical composition
  • the anterior segment can include the cornea, conjunctiva, aqueous humor, iris, ciliary body, or lens.
  • the posterior segment can include the sclera, choroid, retinal pigment epithelium, neural retina, optic nerve or vitreous humor.
  • the composition e.g., a pharmaceutical composition
  • a pharamceutical composition is administered by intravitreal injection.
  • a composition e.g., a pharmaceutical composition
  • a composition e.g., a pharmaceutical composition
  • Ocular barriers to transscleral drug delivery can include static barriers (e.g., sclera, choroid and retinal pigment epithelium (RPE)) and dynamic barriers (e.g., lymphatic flow in the conjunctiva and episclera, and the blood flow in conjunctiva and choroid).
  • the composition can be formulated for delivery across occular drug delivery barriers and/or to improve ocular bioavailability.
  • Compositions e.g., a pharmaceutical composition
  • compositions can be formulated for administration by any route known to those of skill in the art.
  • compositions can be formulated as a suspension, emulsion, ointment, aqueous gel, nanomicelle, nanoparticle, liposome, dendrimer, implant, contact lens, nanosuspension, microneedle, or in situ gel (e.g., in situ thermosensitive gel).
  • Emulstions can contain one or more additives selected from among the group consisting of a lipid additive (e.g., soyabean lecithin and/or stearylamine), a mucoadhesive polymer (e.g., chitosan and/or hydroxypropyl methyl cellulose ether), combinations thereof and the like.
  • a lipid additive e.g., soyabean lecithin and/or stearylamine
  • a mucoadhesive polymer e.g., chitosan and/or hydroxypropyl
  • Administration can be local, topical or systemic.
  • the pharmaceutical compositions can be delivered by topical instillation (e.g., as eye drops).
  • the composition e.g., a pharmaceutical composition
  • the composition can be formulated for topical drop instillation into the lower precorneal pocket.
  • the composition (e.g., a pharmaceutical composition) can be formulated as an extended release formulation (e.g., up to about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 2 weeks, about 4 weeks, about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months or more).
  • the composition (e.g., a pharmaceutical composition) can contain additive(s). Additive(s) can improve one or more properties selected from among drug contact time, permeation and ocular bioavailability.
  • Additives can be selected from among the group consisting of viscosity enhancers, permeation enhancers and cyclodextrins.
  • exemplary viscosity enhancers include hydroxy methyl cellulose, hydroxy ethyl cellulose, sodium carboxy methyl cellulose, hydroxypropyl methyl cellulose, polyalcohol, combinations thereof and the like.
  • Cyclodextrins can act as a carrier for hydrophobic drug molecules in aqueous solution.
  • Permeation enhancers can improve corneal uptake by modifying the comeal integrity.
  • exemplary permeation enhancers include chelating agents, preservatives, surface active agents, bile salts, and combinations thereof.
  • a permeation enhancer is selected from among the group consisting of benzalkonium chloride, polyoxyethylene glycol ethers (e.g., lauryl, stearyl and oleyl), ethylenediaminetetra acetic acid sodium salt, sodium taurocholate, saponins, cremophor EL, polycarbophil-cysteine, combinations thereof and the like.
  • the composition can be formulated as a nanocarrier, such as a nanocarrier selected from the group consisting of nanoparticles, nanosuspensions, liposomes, nanomicelles and dendrimers.
  • Nanomicelles can include amphiphilic molecules and can be surfactant or polymeric in nature.
  • Nanoparticles can include lipids, proteins, natural or synthetic polymers such as albumin, sodium alginate, chitosan, poly (lactide-co-glycolide) (PLGA), polylactic acid (PLA), polycaprolactone, combinations thereof and the like.
  • Nanoparticles can be nanocapsules or nanospheres.
  • the HIF inhibitor can be enclosed inside a shell (e.g., a polymeric shell).
  • the HIF-inhibitor can be uniformly distributed throughout a matrix (e.g., a polymeric matrix).
  • Nanoparticles can include a chitosan coating to improve precorneal residence. Nanosuspensions can be stabilized by polymer(s) and/or surfactant(s).
  • Dendrimers can include terminal end amine, hydroxyl or carboxyl functional groups. Examples of dendrimers include Poly (amidoamine) (PAMAM) dendrimers.
  • compositions formulated as liposomes can include small unilamellar vesicles (10- 100 nm), large unilamellar vesicles (100-300 nm) and multilamellar vesicles (contains more than one bilayer).
  • Liposomes can include cationic liposomes or neutral liposomes.
  • Liposomes can be pegylated liposomes, submicron-sized, or a combination thereof. Liposomes can be multilamellar or unilamellar. Liposomes can include a mucoadhesive polymer. Cationic liposomes can include one or more selected from among the group consisting of Didodecyldimethylammonium bromide, stearylamine, and N-[l-(2,3- dioleoyloxy)propyl] -N,N,N -trimethylammonium chloride .
  • compositions can be formulated as in-situ hydrogels and can undergo sol-gel phase transition to form viscoelastic gel in response to environmental stimuli (e.g., changes in temperature, pH and ions, or a combination thereof) or can be induced by UV irradiation.
  • the composition is formulated as a a thermosensitive gel.
  • thermogelling polymers for use in a thermosensitive gel described herein include poloxamers, multiblock copolymers made of polycaprolactone, polyethylene glycol, poly (lactide), poly (glycolide), poly (N-isopropylacrylamide), chitosan and combinations thereof.
  • polymers can be mixed with an HIF inhibitor in the solution state and solution can be administered which forms an in situ gel depot at physiological temperature.
  • Thermosensitive gels can include, for example, a triblock copolymer of PFGA and PEG ((poly-(DF-lactic acid co-glycolic acid) -polyethylene glycol), or cross linked poly (N- isopropylacrylamide) (PNIPAAm)-poly (ethylene glycol) diacrylate, or the triblock polymer PFGA-PEG-PFGA (poly-(DF-lactic acid co-glycolic acid)-polyethylene glycol-poly-(DF- lactic acid co-glycolic acid) as a ocular delivery carrier for an HIF inhbitor.
  • PFGA-PEG-PFGA poly-(DF-lactic acid co-glycolic acid)-polyethylene glycol-poly-(DF- lactic acid co-glycolic acid
  • the pharamceutical composition containing an HIF inhibitor can be formulated for delivery with a contact lens.
  • the HIF inhibitor can have longer residence time in the post-lens tear fdm which can result in higher drug flux through cornea with less drug inflow into the nasolacrimal duct.
  • the HIF inhibitor can be loaded by soaking the contact lens in a drug solution.
  • the contact lens is a particle-laden contact lenses or a molecularly imprinted contact lens.
  • the active agent can be entrapped in vesicles such as liposomes, nanoparticles or microemulsion and dispersed in the contact lens material.
  • the pharmaceutical composition containing an HIF inhibitor can be formulated as an implant, such as an intraocular implant.
  • the intraocular implant can provide localized controlled drug release over an extended period, and can circumvent multiple intraocular injections and associated complications.
  • the implant can be delivered to posterior ocular tissues.
  • Implants can be placed intravitreally.
  • implants can be placed by making incision through minor surgery at pars plana posterior to the lens and anterior to the retina. Administration by implantation can circumvent the blood retina barrier.
  • the composition can be formulated as a biodegradable implant or as a non- biodegradable implant.
  • a non-biodegradable implant can effect long-lasting release with near zero order release kinetics.
  • Non-biodegradable implants can include a polymer selected from among the group consisting of polyvinyl alcohol (PVA), ethylene vinyl acetate (EVA), polysulfone capillary fiber (PCF), and a combination thereof, such as PV A/EVA.
  • the implant is a silicone laminated PVA implant. Implants can be surgically implanted and removed after drug depletion.
  • the compositon is formulated as a biodegradable implants.
  • the biodegradable implants can be formulated for sustained drug release. In some embodiments, it is not necessary to surgically remove the biodegradable implants.
  • Biodegradable implants can include a polymer (e.g., polylactic acid (PLA), polyglycolic acid (PGA), poly [d,l-lactide- co-glycolide] (PLGA) and poly[d,l-lactide-co-caprolactone] (PLC), poly( L -lactide-co- caprolactone-co-glycolide) (PLGA-PCL), hydroxypropyl methylcellulose, polycaprolactones, or a combination thereof).
  • PLA polylactic acid
  • PGA polyglycolic acid
  • PLGA poly [d,l-lactide- co-glycolide]
  • PLA poly[d,l-lactide-co-caprolactone]
  • PLA-PCL poly( L -
  • the compostion can be formulated as an intravitreal implant.
  • an intravitreal implant can contain a PLGA polymer matrix that degrades to lactic acid and glycolic acid over an extended period, allowing prolonged release over up to about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months or longer.
  • the composition (e.g., a pharmaceutical composition) is formulated for administration by a microneedle based technique.
  • the composition can be formulated for delivery to posterior ocular tissues.
  • Microneedle based administration can circumvent blood retinal barrier and deliver therapeutic drug levels to retina/choroid.
  • Microneedles can be designed to penetrate only hundreds of microns into sclera, so that damage to deeper ocular tissues can be avoided.
  • Microneedles can deposit the HIF inhibitor into sclera or into the suprachoroidal space (SCS) between sclera and choroid.
  • SCS suprachoroidal space
  • compositions containing a second agent that is used to treat an ischemic retinal disease or condition.
  • agents include, but are not limited to, an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, dexamethasone, triamcinolone, a corticosteroid, a steroid, hydroxycarbamide, a blood thinner, warfarin, apixaban, dabigatran, edoxaban, fondaparinux, heparin, rivaroxaban, combinations thereof and the like.
  • the second agent is a VEGFR inhibitor (e.g., selected from the group consisting of cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like).
  • HIF inhibitors can be co-formulated or co-administered with pharmaceutical formulations of such second agents.
  • the HIF inhibitors and second agent can be packaged as separate compositions for administration together, sequentially or intermittently.
  • the combinations can be packaged as a kit.
  • Compositions also can be administered with other biologically active agents, either sequentially, intermittently or in the same composition.
  • compositions provided herein have an osmolarity of at or about 100 mOsm/kg, about 120 mOsm/kg, about 140 mOsm/kg, about 160 mOsm/kg, about 180 mOsm/kg, about 200 mOsm/kg, about 220 mOsm/kg, about 240 mOsm/kg, about 260 mOsm/kg, about 280 mOsm/kg, about 300 mOsm/kg, about 320 mOsm/kg, about 340 mOsm/kg, about 360 mOsm/kg, about 380 mOsm/kg, about 400 mOsm/kg, about 420 mOsm/kg, about 440 mOsm/kg, about 460 mOs
  • the pH of the composition is at or about 4, about 5, about 6, about 7, about 7.2, about 7.4, about 7.6, about 7.8 or about 8. In some embodiments, the pH of the compositions is about 7.4. In some embodiments, the pH of the compositions ranges from about 4 to about 5, ranges from about 5 to about 6, ranges from about 6 to about 7, ranges from about 7 to about 8, ranges from about 8 to about 9, or ranges from about 9 to about 10.
  • the HIF inhibitor can exhibit differential solubility (e.g., ionised and non-ionised forms can coexist).
  • the outer layer of the cornea (the epithelium) is lipid-rich.
  • the inner layer of the cornea (the stroma) is predominantly aqueous. Therefore, ionisation of a drug can increase partitioning into this phase.
  • the pH of the formulation can be adjusted to decrease the ionisation of the therapeutic agent.
  • the HIF inhibitor can be administered as part of a combination therapy, by administering the HIF inhibitor and a second agent or treatment described herein, such as for treating a disease or condition selected from the group consisting of retinal detachment, an ischemic retinal disease or condition, a combination thereof, and the like.
  • the HIF inhibitor and second agent or treatment can be co-formulated and administered together.
  • the HIF inhibitor is administered subsequently, intermittently or simultaneously with the second agent or treatment.
  • the HIF inhibitor can be administered prior to, with, or after administration of the second agent or treatment.
  • the HIF inhibitor is administered together with the second agent or treatment.
  • the HIF inhibitor is one that increases cellular survival. In some examples, the HIF inhibitor is one that increases cellular survival in an in vitro assay. In some embodiments, the HIF inhibitor can be one that increases cellular survival in response to oxidative stress. In some embodiments, oxidative stress is stimulated by treatment with hydroquinone.
  • cells are exposed to hydroquinone at a concentration of 10 mM or about 10 mM, 50 pM or about 50 pM, 100 pM or about 100 pM, 125 pM or about 125 pM, 150 pM or about 150 pM, or 200 pM or about 200 pM, 10 pM or about 10 pM to 50 pM or about 50 pM, 50 pM or about 50 pM to 100 pM or about 100 pM, 100 pM or about 100 pM to 125 pM or about 125 pM, 125 pM or about 125 pM to 150 pM or about 150 pM, or 150 pM or about 150 pM to 200 pM or about 200 pM hydroquinone.
  • cellular survival is assessed in an in vitro assay.
  • the HIF inhibitor increases cellular survival in an in vitro model of AMD.
  • the in vitro assay is a cellular in vitro assay.
  • the cells in the cellular in vitro assay are RPE cells.
  • the cells are ARPE-19 cells.
  • cellular survival is assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and/or by measuring metabolic activity of cells.
  • metabolic activity of cells is assessed by MTT ((3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide)) assay.
  • the HIF inhibitor can increase cellular survival by pretreatment of cells with the HIF inhibitor.
  • the pretreatment is before stimulation of oxidative stress in cells.
  • pretreatment occurs 1 hour or about 1 hour, 2 hours or about 2 hours, 4 hours or about 4 hours, 6 hours or about 6 hours, 12 hours or about 12 hours, 24 hours or about 24 hours, 48 hours or about 48 hours, 72 hours or about 72 hours, 1 to 6 hours, about 1 to about 6 hours, 6 to 12 hours, about 6 to about 12 hours, 12 to 24 hours, about 12 to about 24 hours, 24 to 48 hours, about 24 to about 48 hours, 48 to 72 hours, or about 48 to about 72 hours before stimulation of oxidative stress.
  • the HIF inhibitor can increase cellular survival. In some embodiments, the HIF inhibitor can increase cellular survival in an in vitro assay in which cells are exposed to oxidative stress. In some embodiments, the HIF inhibitor can increase cellular survival by at least 5% or at least about 5%, at least 10% or at least about 10%, at least 15% or at least about 15%, at least 20% or at least about 20%, at least 25% or at least about 25%, at least 30% or at least about 30%, at least 35% or at least about 35%, at least 40% or at least about 40%, at least 50% or at least about 50%, at least 60% or at least about 60%, at least 70% or at least about 70%, at least 80% or at least about 80%, at least 90% or at least about 90%.
  • the HIF inhibitor increases cellular survival at a concentration of 500 nmol or about 500 nmol, ImM or about ImM, 5mM or about 5mM, 10mM or about 10mM, 20mM or about 20mM, 30mM or about 30mM, 50mM or about 50mM, 100mM or about 100mM, or 200mM or about 200mM.
  • the HIF inhibitor increases cellular survival at a concentration of 500 nmol to ImM, about 500 nmol to about ImM, ImM to 5mM, about ImM to about 5mM, 5mM to 10mM, about 5mM to about 10mM, 10mM to 20mM, about 10mM to about 20mM, 20mM to 50mM, about 20mM to about 50mM, 50mM to 100mM, about 50mM to about 100mM, 100mM to 200mM, or about 100mM to about 200mM.
  • cellular survival is assessed in an in vitro assay as described herein.
  • the in vitro assay comprises pretreatment of cells with the HIF inhibitor prior to stimulative oxidative stress in the cells as described herein.
  • the cells are RPE cells.
  • the cells are ARPE-19 cells.
  • cellular survival is assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and/or by measuring metabolic activity of cells.
  • metabolic activity of cells is assessed by MTT ((3-(4,5-dimethylthiazol-2-yl)- 2,5 -diphenyl -2H-tetrazolium bromide)) assay.
  • articles of manufacture containing packaging materials, a pharmaceutical composition that is effective for treating, minimizing and/or substantially inhibiting an ischemic retinal disease or condition, and a label that indicates that the composition is to be used for treating, minimizing and/or substantially inhibiting a ischemic retinal disease or condition.
  • the pharmaceutical composition contains the HIF inhibitor, and no second agent or treatment.
  • the article of manufacture contains the HIF inhibitor and a second agent or agents or treatment or treatments.
  • the pharmaceutical compositions of a second agent and an HIF inhibitor can be provided together or separately, for packaging as articles of manufacture.
  • EXAMPLE 1 HIF inhibition increased survival of ARPE-19 cells under conditions of oxidative stress.
  • the HIF-Ia inhibitor PX-478 which is currently undergoing clinical testing for cancer (Shirai et al. (2021) Cancers (Basel) 13(11):2813), is amelphalan derivative that lowers HIF-Ia levels by affecting multiple levels in the HIF-Ia pathway: it can inhibit HIF- 1 a deubiquitination, reduce HIF-Ia mRNA expression, and inhibit HIF-Ia translation.
  • PX- 478 was reported to show prominent selectivity towards inhibition of HIF-Ia (Masoud et al. (2015) Acta Pharm Sin B. 5(5):378-89; Koh et al. (2008) Mol Cancer Ther. 7:90-100).
  • This HIF inhibitor s ability to prevent RPE cell death was analyzed in an in vitro model of AMD.
  • Immortalized human RPE cells of the cell line ARPE-19 were cultured under routine conditions until confluent.
  • HIF- la was inhibited using a range of PX-478 concentrations for 48 hours.
  • the toxicity of PX-478 was assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and by measuring metabolic activity of cells using the MTT assay.
  • LDH lactate dehydrogenase
  • PX-478 was well tolerated by ARPE-19 cells up to a concentration of 10 mM (FIGS. 3 A and 3B).
  • Oxidative stress and cell death were also stimulated in ARPE-19 cells, using the cigarette smoke component hydroquinone as a model of oxidative stress in RPE cells (Bhattarai et al. (2020) Int. J. Mol. Sci., 21(6):2066; Yang et al. (2020) Invest Ophthalmol Vis Sci., 61(10):35; and Pons et al., (2011) PLoS One., 6(2):el6722).
  • a 24h pretreatment with 5 mM or 10 pM PX-478 was cytoprotective, both in conditions of mild (FIGS. 4A and 4B) and significant (FIGS 4C and 4D) cell death induced by hydroquinone.
  • HIF inhibition prevented RPE cell death under conditions of increased oxidative stress and after exposure to a compound of cigarette smoke, one of the biggest environmental risk factors for AMD development. Preventing RPE cell death and regulating HIF signaling could preserve retinal homeostasis and, thereby, patients’ visual acuity. As such, HIF inhibition could be used to treat, for example, AMD and/or atrophy associated with retinal hypoxia.
  • ARPE-19 cells were obtained from the American Type Culture Collection (ATCC, Mannassas, VA, USA) and used for experiments between passage numbers 25 and 30.
  • Cells were routinely maintained in DMEM/F-12 (1:1) medium (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% HyClone fetal bovine serum (FBS; Thermo Fisher Scientific), 100 U/ml penicillin, 100 pg/ml streptomycin and 2 mM L-glutamine (all Lonza, Basel, Switzerland).
  • FBS HyClone fetal bovine serum
  • penicillin 100 pg/ml streptomycin
  • 2 mM L-glutamine all Lonza, Basel, Switzerland.
  • a 3-(4,5-dimethyldiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Merck KGaA) assay was used to assess cellular viability. Briefly, medium samples were removed from wells and replaced with MTT salt at a final concentration of 0.5 mg/ml in serum-free culture medium. Cells were incubated under absence of light for 90 min at 37°C, after which the MTT-containing medium was replaced with DMSO (Merck KGaA). DMSO dissolved the formed formazan crystals during an incubation step of 20 min at room temperature. The optical density of each well was then measured at a wavelength of 562 nm and results were calculated relative to untreated control, or hydroquinone-treated positive control, which was set to 100% viability.
  • the lactate dehydrogenase (FDH) assay (Cytotox 96® non-radioactive cytotoxicity assay, Promega, Madison, WI, USA) was used to determine cellular toxicity. The assay determined the amount of intracellular FDH that has leaked into the medium and thereby estimated the levels of membrane rupture and cell death. The assay was performed according to the manufacturer’s instructions and results were normalized to FDH levels in untreated control, which were set to be 1.
  • PROPHETIC EXAMPLE Efficacy study of pharmaceutical composition containing an HIF inhibitor for treatment of an ischemic retinal disease A subject having an ischemic retinal disease or condition is selected for treatment.
  • the subject can be identified, for example, having, or being at risk of developing retinal atrophy.
  • the subject can also be identified because he/she has retinal/subretinal neovascularization or macular edema and needs anti VEGF treatment that can be accompanied with retinal atrophy.
  • Examples include dry atrophic AMD in which progression of atrophy has been established.
  • Another example is neovascular AMD in which anti VEGF treatment is planned and retinal atrophy is expected or has started.
  • a pharmaceutical composition containing an HIF inhibitor is administered by injection or implantation into the vitreous cavity of the subject at a dose determined by a clinician to be therapeutically effective.
  • the duration of treatment is a period of time sufficient to treat one or more conditions selected from among retinal apoptosis, retinal atrophy and choroidal atrophy, or to otherwise improve the clinical condition of the subject.
  • treatment can last over months to years.
  • intravitreal injections can be repeated monthly or every few months.
  • Intravitreal injections can be administered, for example, daily, weekly, monthly, every few months, or the like.
  • Eye drops can be applied 1-6 times a day, such as, for example, once per day, twice per day, three times per day, four times per day, five times per day, six times per day, or the like.
  • Systemic administration can be administered, for example, 1-3 times per day (e.g., once per day, twice per day, or three times per day), weekly, monthly, every few months, or the like.
  • Improvement is determined by clinical signs or symptoms or by diagnostic tests. For example, assessments are performed to test for visual function (e.g., visual field, visual acuity, microperimetry, contrast sensitivity, color vision, combinations thereof, and the like). Imaging studies, including fundus photography and/or high speed spectral domain optical coherence tomography (SDOCT), are performed to determine reduction in atrophy.
  • visual function e.g., visual field, visual acuity, microperimetry, contrast sensitivity, color vision, combinations thereof, and the like.
  • Imaging studies including fundus photography and/or high speed spectral domain optical coherence tomography (SDOCT), are performed to determine reduction in atrophy.
  • SDOCT high speed spectral domain optical coherence tomography
  • Apoptosis and/or atrophy is assessed by a suitable morphological, functional, electric or metabolic method, or a combination thereof.
  • suitable methods include a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, annexin 5 staining, a combination thereof, and the like.
  • Apoptotic cells are identified before administration to establish a baseline, and at a time after administration determined by a clinician to result in an a therapeutic improvement (e.g., 5 minutes, 30 minutes, 60 minutes, 120 minutes, 7 days, or 30 days, or any combination thereof).
  • fluorescently-labelled annexin 5 is intravenously administered to the subjects at a dose of 0.1-0.5 mg.
  • Retinal imaging is performed to visualize fluorescent cells, which are quantified. Images are acquired with a confocal scanning laser ophthalmoscope (diode laser 786 nm excitation; photodetector with 800 nm barrier filter), after pupillary dilatation (1% tropicamide and 2.5% phenylephrine) .

Abstract

Provided are methods of treating, minimizing and/or inhibiting atrophy associated with retinal hypoxia, comprising administering a pharmaceutical composition comprising an HIF inhibitor to a subject having retinal hypoxia. The HIF inhibitor is administered in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia. 5 The atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof. Also provided, are pharmaceutical compositions and combinations containing an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, 0 choroidal atrophy, and combinations thereof.

Description

HYPOXIA INDUCIBLE FACTOR (HIF) INHIBITORS FOR TREATMENT OF ATROPHY ASSOCIATED WITH RETINAL HYPOXIA
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No. 63/181,611, filed on April 29, 2021, which is hereby incorporated by reference in its entirety.
TECHNICAL FIELD
This application relates to compositions, uses, and methods for treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia. The methods include administration to a subject in need of said treatment of a pharmaceutical composition comprising an effective amount of an inhibitor of hypoxia inducible factor (HIF).
BACKGROUND
New vessel formation, edema, tissue atrophy, and combinations thereof are common features and cause of visual loss in ischemic diseases of the retina and choroid. For example, choroidal neovascularization, bleeding, and retinal atrophy (e.g, dry or atrophic age related macular degeneration (AMD) or geographic atrophy) can cause visual loss in subjects with age related macular degeneration (AMD). In subjects with diabetic retinopathy (e.g., proliferative diabetic retinopathy), new vessel formation and bleeding can occur, and diabetic macular edema can cause loss of vision. Vision loss also can occur with macular retinal atrophy, sometimes referred to as ischemic atrophy or ischemic maculopathy. Other examples of ischemic diseases that can cause atrophy include central and branch retinal vein occlusions, retinopathy of prematurity, sickle cell retinopathy, retinal detachment and proliferative vitreoretinopathy. Retinal hypoxia in retinal detachments, including central serous chorioretinopathy, also cause apoptosis and atrophy through the HIF pathway.
Atrophic AMD is one of the major causes of blindness in developed countries, including the United States. Treatment for atrophic AMD is a primary unmet medical need in eye care. No treatment is currently available, despite considerable research and efforts for more than a decade by mumerous research groups and pharmaceutical companies.
(Stefansson et al. (2011) Prog. Retin. Eye Res. 30(l):72-80; Ammar et al. (2020) Curr. Opin. Ophthalmol. 31(3):215-221 ; Kandasamy et al. (2017) AsiaPac. J. Ophthalmol 6(6):508-513; Grunwald et al., (2017) Clinical Trial Ophthalmology 124(1):97-104; Li et al. (2017) Expert. Opin Investig. Drugs 26(10): 1103-1114; Kim et al. (2021) Drugs Aging 38(1): 17-27; and Girmens et al. (2012) Intractable Rare Dis. 1(3): 103-114) SUMMARY
An exemplary embodiment of this application is a method of treating, minimizing and/or inhibiting atrophy associated with retinal hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor to a subject having retinal hypoxia. In some exemplary embodiments, the HIF inhibitor is administered in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia.
The atrophy associated with retinal hypoxia can be selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
Another exemplary embodiment of this application is a composition (e.g., a pharmaceutical composition), comprising an HIF inhibitor in an amount effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia. In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
Details of other exemplary embodiments of the present disclosure will be included in the following detailed description. It is appreciated that certain features of the exemplary embodiments described in this application, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment.
BRIEF DESCRIPTION OF THE DRAWINGS
In order to understand the disclosure and to see how it can be carried out in practice, embodiments will now be described, by way of non-limiting examples only, with reference to the accompanying drawings, in which:
FIG. 1 depicts results of ischemia and hypoxia through HIF mechanisms. If the VEGF pathway is blocked, the tissue can mitigate ischemia/hypoxia by the apoptosis/atrophy pathway.
FIG. 2 depicts deregulated molecular mechanisms in RPE cells and their association with HIF activation and/or stability. Reductions in proteasomal activity and increased reactive oxygen species (ROS) can lead to an increase in available HIF a, which can subsequently heterodimerize with HIF-Ib and activate the transcription of HIF target genes, such as, for example, one or more of VEGF, GLUT1 and other genes related to neovascularization and metabolic conversion. Formation of drusen and a thickening of Bruch’s membrane can induce local hypoxia (1), which can activate HIFa. Mitochondrial dysfunction (2) can lead to increased reactive oxygen species (ROS) production, which can stabilize HIFa. Proteasomal dysfunction (3) can prevent the effective clearance of HIFa. HIFa levels can rise and dimerization with HIF (4) can activate target gene transcription. Target gene transcription (5) can lead to neovascularization and a metabolic conversion.
FIG. 3 shows Cytotoxicity of PX-478 in ARPE-19 cells. Cell viability (MTT assay, FIG. 3A) or cellular toxicity (leakage of LDH, Fig. 3B) was determined following a 48h incubation with selected concentrations of PX-478. The compound was well tolerated up to a concentration of 10 mM. Ctrl - control; * - p<0.05; ** - p<0.01; *** - p<0.001; Mann- Whitney U-test, compared to untreated control.
FIG. 4 shows cytotoxicity of hydroquinone in ARPE-19 cells pretreated with PX-478 for 24h. Cell viability (MTT assay, FIGS 4A and 4C) and cellular toxicity (leakage of LDH, FIGS 4B and 4D) were determined following a 24h incubation with selected concentrations of PX-478 and a subsequent 24h exposure to toxic levels of hydroquinone. PX-478 was cytoprotective at 5mM and 10 mM concentrations. Ctrl - control; HQ - hydroquinone; * - p<0.05; *** - p<0.001; Mann-Whitney U-test, compared to HQ group.
DETAILED DESCRIPTION
There is a need for a treatment of atrophy (e.g., atrophy associated with retinal hypoxia). There is no treatment available for retinal apoptosis and atrophy associated with retinal hypoxia. There also is a need for alternative treatments of ischemic retinal disease that do not result in atrophy that can occur during or after administration of an angiogenesis inhibitor, such as an anti-VEGF or anti-VEGFR therapeutic. For example, there is a need for treatments of ischemic retinal disease in which the apoptosis-atrophy pathway is minimized and/or substantially inhibited, or is not activated. Activation of the apoptosis-atrophy pathway can occur during treatment with an anti-VEGF therapeutic, since the VEGF response to ischemia/hypoxia is blocked.
Hypoxic conditions can activate the HIF pathway, which can result in effects such as increased VEGF production, increased vascular permeability, edema, new vessel formation, and combinations thereof. As a result, bleeding and/or visual loss can occur. For example, bleeding can be selected from among the group consisting of subretinal, intraretinal, in vitreous humour, or any combination thereof. Activation of the HIF pathway can result in apoptosis (e.g., apoptosis of retinal cells). Apoptosis can result in retinal atrophy, which can also lead to visual loss. When VEGF inhibitors, VEGF receptor blockers, or a combination thereof, block the VEGF arm of the hypoxic response, tissue can mitigate hypoxia by apoptosis (e.g., retinal apoptosis) and atrophy.
Ischemia and hypoxia have several consequences through HIF mechanisms, including: 1) angiogenesis and edema through a VEGF pathway; 2) apoptosis; and 3) atrophy. Currently, treatment of ischemic disease in retina is limited to targeting the VEGF pathway. No treatment options exist for ischemia/hypoxia induced apoptosis and atrophy. When the VEGF pathway is blocked, the apoptosis/atrophy pathway remains for the tissue to “mitigate” ischemia/hypoxia that activates HIF pathway (FIG. 1). Thus, subjects who are administered VEGF inhibitors can exhibit considerable retinal atrophy and visual loss. In some cases the HIF inhibitor can complement or replace treatment with a VEGF inhibitor or VEGFR inhibitor, to reduce or prevent atrophy that VEGF/VEGFR inhibitors are associated with, when administered alone (e.g., to treat retinal ischemic diseases). Thus, an HIF inhibitor can treat, minimize or substantially inhibit atrophy that is an adverse effect of a VEGF or VEGFR inhibitor.
Mammalian cells, such as RPE cells, can rely on aerobic metabolism for energy generation, a process that requires sufficient levels of oxygen. When oxygen levels drop too low, cells become hypoxic and can react by activating the hypoxic response, which is designed to ensure survival. It can, e.g., increase the number of red blood cells that transport oxygen, augment the number of blood vessels available, and switch energy metabolism to anaerobic metabolism that does not use mitochondria (see e.g., Shinojima et al., J. Clin. Med. 2021 Nov 24;10(23):5496). Cells can achieve this by activating hypoxia-induced factors (HIFs).
HIFs are master regulators of the hypoxic response, controlling hundreds of genes involved in, for example, erythropoiesis, angiogenesis (e.g., VEGF) and metabolic conversion (see, e.g., Shinojima et al., J. Clin. Med. 2021 Nov 24;10(23):5496; Pawlus et al., Cell Signal. 2013 Sep;25(9): 1895-903). HIFs are heterodimers that can include an O2 sensitive a subunit (e.g., HIF- la, HIF-2a or HIF-3a) and an O2 insensitive subunit (e.g., HIF- 1b) (see, e.g., Albadari et al., Expert Opin Drug Discov. 2019 Jul;14(7):667-682; Prabhakar et al. Physiol Rev. 2012 Jul;92(3):967-1003] Under normoxic conditions, HIFs (e.g., HIF- l/2a) can be quickly degraded, following poly-ubiquitination and proteasomal degradation. Conversely, hypoxia can stabilize HIF-l/2a via inhibition of these pathways (see, e.g., Maxwell et al., Nature. 1999 May 20;399(6733):271-5; Semenza et al., Biochem Pharmacol. 2002 Sep;64(5-6):993-8.). Reactive oxygen species (ROS) generated by NADPH or dysfunctional mitochondria can stabilize HIFs and/or activate their target genes (see, e.g., Albadari et al., Expert Opin Drug Discov. 2019 Jul;14(7):667-682).
In the RPE of AMD patients a reduction in proteasomal activity, local hypoxia and increased production of reactive oxygen species (ROS) can stabilize HIF-l/2a (FIG. 2). (Maxwell et al. Nature (1999) 399(6733):271-5; Semenza et al. Biochem Pharmacol. 2002 Sep,64(5-6):993-8; Albadari et al. Expert Opin Drug Discov. 2019 Jul,14(7):667-682; Aqamaa et al. Ageing Res Rev. 2009 Oct,8(4):349-58; Stefansson et al. Prog Retin Eye Res. 2011 Jan;30(l):72-80; Kaamiranta et al. Front Biosci (Elite Ed). 2010 Jun 1;2(4): 1374-84).
A current standard of care for AMD is the repeated intra ocular injection of anti-VEGF agents, which can slow the wet form of the disease. However, prolonged therapy has been linked to progressive retinal atrophy (Rofagha et al. Ophthalmology. 2013;120:2292-2299). HIF is upstream of VEGF activation. VEGF deletion can cause retinal atrophy and dysfunction in a mouse model, but deletion of HIF can have minimal or no adverse effects (see, e.g., Kurihara et al., J. Clin. Investig. 2012;122:4213-4217).
Activated HIFs can play a crucial role in the adaptive response of tumor cells to changes in oxygen availability through transcriptional activation of one or more downstream genes selected from more than one hundred known downstream genes. HIF-1 can help hypoxic tumor cells shift glucose metabolism from oxidative phosphorylation to the less efficient glycolytic pathway through the induction of enzymes involved in the glycolysis pathway and overexpression of glucose transporters (GFUTs) which can increase glucose import into tumor cells (see, e.g., Masoud et al., Acta Pharm Sin B. 2015 Sep,5(5):378-89; Denko et al, Nat Rev Cancer. 2008;8:705-713; Weinhouse et al., Science. 1956;124:267- 272.). HIFs also can cause the transcriptional induction of one or more pro-angiogenic factor(s), such as the vascular endothelial growth factor (VEGF), which in turn can stimulate the development of new blood vessels to enrich tumor cells with oxygen for their growth (see, e.g., Conway et al. 2001;49:507-521).
Advances in the development of selective inhibitors have led to clinical studies testing the potential of HIF inhibitors in cancer therapy (see, e.g., Albadari et al. Expert Opin Drug Discov. 2019 Jul;14(7):667-682). Belzutifan, a selective inhibitor of HIF-2a, has recently been granted FDA approval (www.fda.gov/drugs/resources-information-approved-drugs/fda- approves-belzutifan-cancers-associated-von-hippel-lindau-disease; accessed April 28, 2022). But solid carcinomas are not the only targets for the use of HIF inhibitors. Hypoxic conditions have been associated with many age-related diseases, and target genes of HIF, such as, for example VEGF, are implicated in, e.g., age-related macular degeneration (AMD).
AMD is the leading cause of vision loss amongst the elderly in the western world (see, e.g., Thomas etal, Med. Clin. North Am., 2021 May;105(3):473-491). It affects an estimated 196 million patients worldwide, a number that is projected to increase to 288 million by 2040 (Wong et al. Fancet Glob Health. 2014 Feb;2(2):el06-16). Only 15% of patients can be treated with anti-VEGF injections, which can be costly and invasive. It was estimated that up to one in three persons over 85 years of age is suffering from AMD in Europe and North America (see, e.g., Wong et al. Lancet Glob Health. 2014 Feb;2(2):el06- 16). With a decrease in visual acuity and the progressive loss of central vision being the hallmark of AMD, patients with advanced disease forms can no longer recognize faces or read texts. This loss of vision can impact a patient’s quality of life, increasing the risk of fall and depression and could cause a need for longtime care (see, e.g., Thomas et al., Med Clin North Am. 2021 May;105(3):473-491). The direct cost of AMD to the North American health care system was more than 250 billion dollars in 2008 (Thomas et al., Med Clin North Am. 2021 May;105(3):473-491).
AMD is classically divided into dry and wet subtypes, as well as into early and advanced stages (Thomas et al., Med Clin North Am. 2021 May;105(3):473-491). Wet AMD, or choroidal neovascularization, is an advanced form of the disease, characterized by a growth of blood vessels from the choroid through Bruch’s membrane and into the subretinal space. Bleeding, swelling and possible scar formation are the consequence, leading to a rapid and drastic loss of vision. (Thomas et al., Med Clin North Am. 2021 May;105(3):473-491). Aberrant VEGF production can underlie rapid vision loss in wet AMD (Thomas et al. Med Clin North Am. 2021 May;105(3):473-491.) VEGF levels are often increased in patients suffering from wet AMD, and anti -VEGF treatment via intra ocular injection has proven successful at slowing down disease progression in this subtype of AMD cases. However, repeated intra ocular injections can place a large strain on healthcare providers, practitioners and patients alike, and real-world data shows a steady decline in adherence, and consequently outcome, with prolonged treatment times (Sobolewska et al. Clin Ophthalmol. 2021 ; 15:4317- 4326).
Furthermore, some reports suggest a link between prolonged anti-VEGF treatment and the advancement of geographic atrophy (Rofagha et al. Ophthalmology. 2013;120:2292- 2299.). Moreover, about 85% of AMD cases are made up by the so-called dry form, characterized by the formation of drusen and a progressive retinal atrophy. Patients suffering from dry AMD face a slow but steady progression towards advanced dry AMD, called geographic atrophy, which cannot currently be stopped or even slowed (see, e.g., Kauppinen et al. Cell Mol Life Sci. 2016 May;73(9): 1765-86). Thus, new treatment methods are urgently needed.
Development of new drugs for the successful treatment of AMD can be complicated by the multifactorial nature of the disease. Genetic and environmental risk factors can converge to initiate disease formation. Aging, smoking, obesity, hypertension, and hereditary factors are some of the best-known risk factors for AMD (see, e.g., Arjamaa et ak, Ageing Res Rev. 2009 Oct;8(4):349-58). On a cellular level, these factors can cause retinal pigment epithelium (RPE) cell stress and later cell death and retinal atrophy. RPE cells, which are responsible for maintaining the photoreceptor layer in the retina and are a vital part of the blood-retinal barrier, are at the center of AMD onset and progression. Their loss leads secondarily to the death of photoreceptors and a loss in vision. RPE cells also secrete a number of growth factors and cytokines, aimed at maintaining tissue homeostasis. Deregulated, these factors can cause choroidal neovascularization and uncontrolled, chronic inflammation that will inevitably lead to cell death (see, e.g., Kauppinen et ak, Cell Mol Life Sci. 2016 May;73(9): 1765-86).
Reduced proteasomal and autophagic activity, oxidative stress, and dysfunction of mitochondria, leading to an energetic crisis within the retina, can be involved in the dysregulation of RPE cell function and the onset of AMD (see, e.g., Kauppinen et ak, Cell Mol Life Sci. 2016 May;73(9): 1765-86; Kaamiranta et ak, Front Biosci (Elite Ed). 2010 Jun 1;2(4): 1374-84).
HIF is involved or linked to these processes in multiple ways (FIG. 2). One or more factors selected from among the group consisting of increased oxidative stress, e.g. ROS produced by dysfunctional mitochondria, as well as reduced proteasomal clearance, and active inflammation through NF-kB, the master regulator of the innate immune response can lead to an increase in available HIF protein levels (Aqamaa et ak, Ageing Res Rev. 2009 Oct;8(4):349-58; Frede et ak (2007) Methods Enzymoh 435, 405-419). Furthermore, lesions in central serous chorioretinopathy have been identified as being hypoxic, and it has been postulated that the drusen present in AMD along with a thickened Bruch’s membrane and possible retinal detachment or edema can predispose the retina to local hypoxia (Schlingemann, (2004) Graefes Arch. Clin. Exp. Ophthalmol. 242, 91-101; Aqamaa et ak, Ageing Res Rev. 2009 Oct;8(4):349-58; and Stefansson et ak, Prog Retin Eye Res. 2011 Jan;30(l):72-80). Correspondingly, HIF has been located in human choroidal neovascular membranes, which are associated with AMD and in drusen (Inoue et ak (2007) Br. J. Ophthalmol. 91:1720-1721; Shimada et ak (2007), Graefes Arch Clin Exp Ophthalmol. 245(2):295-300). CRISPR-mediated knock-out of HIF-Ia or VEGF in a mouse model of wet AMD can reduce the volume of choroidal neovascularisation with the same efficiency as the anti-VEGF agent aflibercept (Shinojima etal. (2021) J. Clin. Med. 10(23):5496; Koo et ak (2018) Nat. Commun. 9: 1855). However, RPE-specific VEGF knock-out mice show atrophy and loss of function in choriocapillaris and cone cells within a few days after gene deletion, whereas both HIF-la and HIF-2a knock-out mice show no physiological abnormalities (Kurihara et al. (2012) J. Clin. Investig. 122:4213-4217). Similarly, prolonged VEGF treatment in AMD patients has been reported to lead to atrophy of the RPE and photoreceptor cells (Rofagha et al. (2013) Ophthalmology 120:2292-2299). Unlike VEGF, HIF is not required for retinal homeostasis in the steady state and might therefore be a better therapeutic target than VEGF (Shinojima et al. (2021) J. Clin. Med. 10(23):5496).
Provided herein are methods, compositions, combinations, and uses for treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia. The methods, compositions, combinations, and uses provided herein can employ HIF inhibitors that can inhibit the HIF pathway. HIF inhibitors can mitigate and block HIF mediated response to hypoxia, which can result in one or more effects selected from the group consisting of limiting new vessel formation, inhibiting edema, preventing or reducing apoptosis (e.g., retinal apoptosis), preventing or reducing atrophy, and any combination thereof. In some embodiments, HIF induced apoptosis is reduced, minimized, substantially inhibited, and/or prevented. In some embodiments, reduction, minimization, inhibition, and/or prevention of HIF induced apoptosis results in reduction, minimization, inhibition, and/or prevention of one or more effects selected from the group consisting of retinal atrophy, choroidal atrophy, vision loss, combinations thereof and the like. In some embodiments, reduction, minimization, inhibition and/or prevention of HIF induced apoptosis results in reduction, minimization, inhibition and/or prevention of one or more adverse effects of treatment selected from the group consisting of retinal atrophy, choroidal atrophy, vision loss, combinations thereof and the like.
Advantages and features of the present disclosure, and methods for accomplishing the same will be more clearly understood from exemplary embodiments described below with reference to any accompanying drawings. However, the present disclosure is not limited to the following exemplary embodiments and can be implemented in various different forms. The exemplary embodiments are provided only to provide sufficient disclosure of the present discoveries and to fully provide a person having ordinary skill in the art to which the present disclosure pertains within the technical field, and the present disclosure will be defined by any appended claims and combinations thereof.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the invention(s) belong. All patents, patent applications, published applications and publications, GenBank sequences, databases, websites and other published materials referred to throughout the entire disclosure herein, unless noted otherwise, are incorporated by reference in their entirety.
As used herein, like reference numerals generally denote like elements throughout the present specification. Further, in the following description, a detailed explanation of well- known related technologies can be omitted to avoid unnecessarily obscuring the subject matter of the present disclosure.
As used herein, “atrophy associated with retinal hypoxia” refers to atrophy that results from retinal hypoxia and/or activation of the HIF pathway. Hypoxia and/or activation of the HIF pathway can occur, for example from ischemia (e.g., ischemic retinal disease) and/or retinal detachment. Hypoxia can activate HIF, which can activate mitigating responses that counter the hypoxic state. These include VEGF induced angiogenesis and edema, and apoptosis/atrophy, combinations thereof, and the like.
As used herein retinal hypoxia “associated with” a disease or condition refers to a correlation between the condition and the retinal hypoxia and/or activation of the HIF pathway. For example, “retinal hypoxia associated with” ischemic disease or retinal detachment refers to retinal hypoxia that results from ischemic disease or retinal detachment.
As used herein, terms such as "including" and "having" are generally intended to allow other components to be included unless the terms are used in conjunction with the term "only."
As used herein, the term “treating” or “treatment” includes curing a condition, treating a condition, minimizing and/or inhibiting and/or substantially inhibiting a condition, preventing or substantially preventing a condition, treating, minimizing and/or inhibiting one or more symptoms of a condition, curing symptoms of a condition, ameliorating, reducing and/or minimizing symptoms of a condition, treating effects of a condition, ameliorating, reducing and/or minimizing effects of a condition, and preventing and/or substantially preventing results of a condition.
As used herein, the term “substantially” means completely or almost completely. For example, “substantially preventing a condition” means that the condition is completely prevented or is almost completely prevented.
As used herein, “treating” atrophy associated with retinal hypoxia can result in the atrophy being partially or totally alleviated, or remaining static as a result of treatment.
Hence treatment encompasses prevention, prophylaxis, therapy and/or cure. Prophylaxis refers to prevention of a potential atrophy and/or a prevention of worsening of symptoms or progression of atrophy. As used herein, the term “pharmaceutical composition” refers to a composition comprising one or more active ingredients with other components such as, for example, pharmaceutically-acceptable ingredients and/or excipients, such as a pharmaceutically- acceptable carrier. The purpose of a pharmaceutical composition is to facilitate administration of an active ingredient to a subject.
As used herein, the terms “pharmaceutically active agent” or “active agent” or “active pharmaceutical ingredient” are interchangeable and mean the ingredient is a pharmaceutical drug, which is biologically- and/or chemically-active and is regulatory-approved or appro vable as such.
As used herein, the term “ingredient” refers to a pharmaceutically-acceptable ingredient, which is included or is amenable to be included in The FDA’s Inactive Ingredient (IIG) database. Inactive ingredients can sometimes exhibit some therapeutic effects, although they are not drugs.
Whenever a numerical range is indicated herewith, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases “ranging/ranges between” a first indicated number and a second indicated number and “ranging/ranges from” a first indicated number “to” a second indicated number are used herein interchangeable and are meant to include the first and second indicated numbers and all fractional and integral numerals therebetween.
As used herein, numbers and/or numerical ranges preceded by the term “about” should not be considered to be limited to the recited range. Rather, numbers and/or numerical ranges preceded by the term “about” should be understood to include a range accepted by those skilled in the art for any given element in formations according to the subject invention.
As used herein, when a numerical value is preceded by the term “about,” the term “about” is intended to indicate +/- 10%.
As used herein, the singular form “a,” “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a compound” or “at least one compound” can include a plurality of compounds, including combinations and/or mixtures thereof.
As used herein, the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, technical and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
As used herein, the term “hypoxia-inducible factor” or “HIF” is a transcription factor in the hypoxia-inducible factor (HIF) pathway that responds to hypoxic conditions. Members of the human HIF family include HIF- la, HIF-Ib (ARNT), HIF-2a, HIF-2 (ARNT2), HIF- 3alpha, and HIF-3 , as well as heterodimers thereof, such as, for example HIF-la/HIF-Ib, HIF-la/HIF^, HIF-2a/HIF-^, HIR-2a/HIR-2b, HIF-3a/HIF-^, and HIR-3a/HIR-2b.
As used herein, the term “HIF-a” refers to an HIF-a transcription factor (e.g., HIF-la, HIF-2a, or HIF-3a, or a combination thereof).
As used herein, the term “HIF-b” refers to an HIF-b transcription factor (e.g., HIF-Ib, or HIR-2b, or the combination thereof).
As used herein, the term “Hypoxia Inducible Factor inhibitor” or “HIF inhibitor” is an inhibitor of the hypoxia-inducible factor (HIF) pathway. It is understood that recitation of an HIF inhibitor includes pharmaceutically-acceptable salts thereof, as well as prodrugs thereof. It also is understood that description of an HIF inhibitor as inhibiting a part of the HIF pathway does not preclude the inhibitor from inhibiting another part of the HIF pathway. An “HIF inhibitor” includes pharmaceutically-acceptable salts thereof and analogs thereof.
A pharmaceutically-acceptable salt of an HIF inhibitor is a biologically-compatible salt that can be used as a drug, which salts can be derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, combinations thereof and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate, combinations thereof and the like. In some examples disclosed herein, the pharmaceutically- acceptable salt is an acid addition salt. Pharmaceutically-acceptable acid addition salts are those salts that retain the biological effectiveness of the free bases while formed by acid partners that are not biologically or otherwise undesirable, e.g., inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. In some examples, the salt is formed with an ion selected from the group consisting of ammonium, lithium, sodium, potassium, cesium; alkaline earth metals to include calcium, magnesium, aluminium; zinc, barium; or quaternary ammoniums; or organic salts such as arginine, organic amines to include aliphatic organic amines, aromatic amines, t-butylamines, (N-benzylphenethylamine), dicyclohexylamines, dimethylamines, diethanolamines, ethanolamines, ethylenediamines, imidazoles, lysines, methylamines, N-methyl-D-glucamines, N,N'-dibenzylethylenediatnines, pyridines, picolinates, piperazines, tris(hydroxymethyl)methylamines, triethylamines, triethanolamines, trimethylamines, or ureas.
As used herein, the term “HIF target gene” is a gene regulated by HIF.
As used herein, “ischemic retinal disease” or “ischemic retinopathy” refers to diseases of the retina where new vessel formation, edema or atrophy are prominent features. The ischemia can result from a variety of mechanisms. In age related macular degeneration (AMD) this mechanism can include immunologic mechanisms involving complement factor H and more, resulting in choroidal ischemia. Similar features can apply to other diseases with choroidal neovascularization, including myopic choroidal neovascularization, Polypoidal Choroidal Vasculopathy and choroidal inflammation such as VKH disease and Bechets disease. In diabetic retinopathy, hyperglycemia can contribute to damage of retinal capillaries and capillary non-perfusion, which is ischemia. In sickle cell retinopathy, the abnormal red blood cells can block retinal capillaries and cause ischemia. In radiation retinopathy, retinal capillaries can be damaged and nonperfusion can occur, similar to diabetic retinopathy. In retinopathy of prematurity, the retinal vasculature does not develop fully as a consequence of premature birth and relative hyperoxia. In central and branch retinal vein occlusion, ischemia can be caused by venous occlusion. In central or branch retinal artery occlusion and anterior ischemic optic neuropathy, the ischemia can be caused by arterial occlusion, that can be embolic or inflammatory. In retinal detachment, proliferative vitreoretinopathy, and central serous chorioretinopathy, hypoxia can be caused by increased distance between the retina and the choroidal source of oxygen, rather than vascular ischemia of the diseases listed above. Retinal ischemia can be evaluated by methods described herein, such as retinal fluorescein angiography, OCT angiography, clinical examination, combinations thereof, and the like. Choroidal ischemia can be evaluated by indocyanin green angiography. Retinal hypoxia can be measured by retinal oximetry and other methods to measure retinal oxygenation (Stefansson etal. (2019) Prog. Retin. Eye Res. 70:1-22).
As used herein, “diabetic retinopathy” is a diabetes complication caused by damage to blood vessels of the retina. Diabetic retinopathy includes non-proliferative diabetic retinopathy (NPDR), diabetic macular edema and proliferative diabetic retinopathy (PDR). Non-proliferative diabetic retinopathy includes mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, and severe non-proliferative diabetic retinopathy.
Examples of a symptom of an ischemic retinal disease include angiogenesis, edema, vision impairment, blindness, retinal apoptosis, retinal atrophy, choroidal atrophy, combinations thereof and the like.
In some embodiments, an effective amount of an HIF inhibitor for treating atrophy associated with retinal hypoxia is an amount that is sufficient to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia. Such an amount can be administered as a single dosage or can be administered according to a regimen, whereby it is effective. The amount can cure the atrophy associated with retinal hypoxia but, typically, is administered in order to ameliorate or prevent one or more symptoms of the atrophy. Repeated administration can be required to achieve the desired amelioration or prevention of symptoms. In some exemplary embodiments, administering the HIF inhibitor comprises administration hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months.
As used herein a VEGFR inhibitor can inhibit activity and/or expression of a VEGF receptor. VEGFR inhibitors include, but are not limited to, antibodies, such as, for example an antibody selected from the group consisting of cediranib, cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like. A VEGFR inhibitor, for example, can block binding of a VEGFR receptor and/or inhibit receptor phosphorylation.
As used herein, a VEGF inhibitor includes an inhibitor of activity and/or expression of a VEGF ligand. VEGF inhibitors include, but are not limted to, antibodies, such as, for example, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof, and the like. A VEGF inhibitor, for example, can block binding of a VEGF ligand.
It is appreciated that certain features of the exemplary embodiments described herein, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the exemplary embodiments, which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable sub-combination or as suitable in any other described embodiment. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements. An exemplary embodiment of this application is a method of treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor (e.g., an effective amount of an HIF inhibitor) to a subject having retinal hypoxia. In some exemplary embodiments, the HIF inhibitor is administered in an amount effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia. In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, combinations thereof, and the like.
In some exemplary embodiments, administration of the pharmaceutical composition containing an HIF inhibitor effects treatment of the atrophy associated with the retinal hypoxia.
In some exemplary embodiments, treatment of the atrophy associated with retinal hypoxia comprises preventing, minimizing, slowing, alleviating and/or substantially inhibiting the atrophy. In some exemplary embodiments, treatment of the atrophy associated with retinal hypoxia comprises decreasing the severity, duration, or frequency of occurrence of the atrophy.
In some exemplary embodiments, the method further comprises assessing the atrophy associated with retinal hypoxia. In some exemplary embodiments, assessing the atrophy associated with retinal hypoxia comprises a method selected from the group consisting of spectral-domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, and combinations thereof. In some exemplary embodiments, assessing the atrophy associated with retinal hypoxia occurs before administering the pharmaceutical composition comprising the HIF inhibitor. In some exemplary embodiments, assessing the atrophy associated with retinal hypoxia occurs after administering the pharmaceutical composition comprising the HIF inhibitor. In some exemplary embodiments, the atrophy associated with retinal hypoxia is assessed before and after administering the pharmaceutical composition comprising the HIF inhibitor.
In some exemplary embodiments, the method comprises a reduction in retinal apoptosis associated with retinal hypoxia in the subject. In some exemplary embodiments, the method comprises a reduction in retinal apoptosis by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%. In some exemplary embodiments, the method comprises a reduction in retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%. In some exemplary embodiments, the method comprises a reduction in retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
In some exemplary embodiments, the method comprises assessing retinal apoptosis.
In some exemplary embodiments, retinal apoptosis is assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, combinations thereof, and the like. In some exemplary embodiments, the measuring phosphatidyl extemalization comprises annexin 5 staining.
In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of macular atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy (glaucomatous atrophy), combinations thereof, and the like.
In some embodiments, the atrophy associated with retinal hypoxia is glaucomatous atrophy. In glaucomatous atrophy, retinal ganglion cells can undergo apoptosis due to hypoxia. HIF inhibition can protect ganglion cells from cell death in glaucoma. In some embodiments the glaucomatous atrophy is from glaucoma. In some embodiments the glaucoma is selected from the group consisting of chronic open angle glaucoma, closed angle glaucoma, secondary glaucoma, normal tension glaucoma, and combinations thereof. Thus, HIF inhibition can provide neuroprotection in glaucoma.
In some embodiments, the iris atrophy is from anterior segment ischemia. In some embodiments, the ciliary body atrophy is from anterior segment ischemia. In some embodiments, the optic nerve atrophy is from vascular ischemia. In some embodiments, the vascular ischemia comprises a condition selected from the group consisting of giant cell arteritis, embolisms, and a combination thereof. In some embodiments, the optic nerve atrophy comprises anterior ischemia optic neuropathy.
In some exemplary embodiments, the atrophy (e.g., retinal atrophy) is macular atrophy or geographic atrophy. In some exemplary embodiments, the atrophy is choroidal atrophy.
In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), and dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of an angiogenesis inhibitor (e.g., a VEGF or VEGFR inhibitor). In some exemplary embodiments, the retinal detachment is selected from the group consisting of tractional, rhegmatogenous and serous retinal detachment. Retinal detachment can move the retina further away from a choroidal oxygen source. This can cause hypoxia and/or HIF activation. In some exemplary embodiments, the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
In some exemplary embodiments, the method comprises a reduction in a total area of the atrophy associated with retinal hypoxia.
In some exemplary embodiments, the area of atrophy associated with retinal hypoxia is reduced by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%. In some exemplary embodiments, the area of atrophy associated with retinal hypoxia is reduced by about 1% to about 10%, by about 10% to about 20%, by about 20% to about 30%, by about 30% to about 40%, by about 40% to about 50%, by about 50% to about 60%, by about 60% to about 70%, by about 70% to about 80%, by about 80% to about 90%, or by about 100%. In some exemplary embodiments, the area of atrophy associated with retinal hypoxia is reduced by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
In some exemplary embodiments, the method comprises assessing the area of atrophy associated with retinal hypoxia. In some exemplary embodiments, the area of atrophy associated with retinal hypoxia is assessed by a method selected from among the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like. In some exemplary embodiments, the area of atrophy associated with retinal hypoxia is assessed by a method selected from the group consisting of spectral -domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, combinations thereof, and the like. In some exemplary embodiments, atrophy (e.g., retinal atrophy) can be assessed by a method selected from among the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, combinations thereof, and the like.
In some exemplary embodiments, the method comprises a reduction in a severity grade of the atrophy associated with retinal hypoxia (e.g., retinal atrophy or choroidal atrophy).
In some exemplary embodiments, the method comprises an improvement in the Age- Related Eye Disease Study (AREDS) scale.
In some exemplary embodiments, the subject is one who is being treated with an angiogenesis inhibitor. In some exemplary embodiments, the angiogenesis inhibitor is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, a combination thereof, or the like. In some exemplary embodiments the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, a combination thereof, and the like. In some exemplary embodiments the angiogenesis inhibitor is selected from the group consisting of ranibizumab, bevacizumab, aflibercept, pegaptanib, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
In some exemplary embodiments, the retinal hypoxia is from a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof.
In some exemplary embodiments, the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, central serous chorioretinopathy and other serous retinal detachments, rhegmatogenous retinal detachment, traction retinal detachment, proliferative vitreoretinopathy (PVR), combinations thereof, and the like.
In some exemplary embodiments, the retinal hypoxia is from an ischemic retinal disease.
In some exemplary embodiments, administration of the pharmaceutical composition containing an HIF inhibitor effects treatment, minimizing and/or substantial inhibition of a symptom associated with the ischemic retinal disease. In some exemplary embodiments, the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion combinations thereof and the like. In some exemplary embodiments, the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of age related macular degeneration (dry atrophic AMD, geographic atrophy), diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy, retinal pigment epithelial detachment, central serous chorioretinopathy, combinations thereof, and the like.
In some exemplary embodiments, the pharmaceutical composition reduces progression of retinal atrophy in dry atrophic AMD (Geographic atrophy), and/or reduces or prevents atrophy associated with anti VEGF or anti VEGFR treatment for neovascular AMD. In some exemplary embodiments, the pharmaceutical composition treats, minimizes or substantially inhibits angiogenesis and/or edema in addition to atrophy. In some exemplary embodiments, the pharmaceutical composition treats, minimizes, substantially inhibits and/or reduces progression of retinal atrophy (e.g., in dry AMD) and/or severe and symptomatic atrophy (Geographic atrophy).
In some exemplary embodiments, the ischemic retinal disease is diabetic macular edema.
In some exemplary embodiments, the ischemic retinal disease is non-proliferative diabetic retinopathy (DR), including DR with macular ischemia and/or macular atrophy
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, severe non-proliferative diabetic retinopathy, and traction retinal detachment in DR.
In some exemplary embodiments, the ischemic retinal disease is proliferative diabetic retinopathy. In some exemplary embodiments, the ischemic retinal disease is central retinal vein occlusion. In some exemplary embodiments, the ischemic retinal disease is branch retinal vein occlusion.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
In some exemplary embodiments, administration of the pharmaceutical composition effects a decrease in expression of an HIF target gene or locus. In some exemplary embodiments, the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI, TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR Flk-1, plasminogen-activator inhibitor- 1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2, a hexokinase activator, lactate dehydrogenase A/LDHA, a lactate dehydrogenase A/LDHA inhibitor, lactate dehydrogenase B/LDHB, iNOS, perilipin-2, PGK1, PKM2, cathepsin D, CCL2/JE/MCP-1, CTGF/CCN2, CXCR4, HGFR/c-MET, IL-6, IL-8/CXCL8, integrin alpha 5/CD49e, LOX-1/OLR1, LOXL1, lysyl oxidase homolog 2/LOXL2, MKP-1, MMP-1, MMP- 2, osteopontin/OPN, pref-l/DLKl/FAl, SNAI1, TCF-3/E2A, TRKB, TWIST-1, uPAR, ZEB1, KLF4, NANOG, OCT-3/4, OCT-4A, OCT-4B, and SOX2, Adrenomedullin/ADM, Cyclin Dl, Erythropoietin/EPO, IGF-II/IGF2, IGFBP-1, IGFBP-2, IGFBP-3, NOTCH 1, Survivin, TGF-a, keratin 14, keratin 18, keratin 19, vimentin, CXCR4, c-Met, autocrine motility factor (AMF/GPI), LDL receptor related protein 1 (LRP1), Transforming growth factor-a (TGF-a), Transforming growth factor^ (TGF- 3), Insulin-like growth factor 2 (IGF -2), IGF binding protein 1, 2 and 3 (IGF -BP), WAF1, Cyclin G2), Endothelin 1 (ET1), Adrenomedullin (ADM), Tyrosine hydroxylase, alB-adrenergic receptor, Inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), heme oxygenase- 1, atrial natriuretic peptide, insulin-like growth factor binding protein-1, NIP3, NIX, RTP801, Endoglin (ENG), Wilms' tumour suppressor, a-Fetoprotein, and Calcitonin-receptor like receptor), Erythropoetin (EPO), Leptin (LEP)), Glucose transporter 1 (GLUT1), Hexokinase 1 and 2 (HK1 and 2), 6-Phosphofructo-l -kinase L (PFKL), 6-Phosphofructo-2 -kinase, Glyceraldehyde-3-P dehydrogenase (GAPDH), Aldolase A (ALDA), Aldolase C (ALDC), Enolase 1 (ENOl), Phosphoglycerate kinase-1 (PGK1), Lactate dehydroxygenase A (LDHA), Pyruvate kinase M (PKM), Carbonic anhydrase 9 (CA9), Adenylate kinase 3, and Transglutaminase 2), Pro-collagen prolyl hydroxylase al, Collagen type V (al), Intestinal trefoil factor (TFF), Ecto-5 '-nucleotidase, Cathepsin D (CATHD), Fibronectin 1 (FN1), Matrix metalloproteinase 2 (MMP2)), DEC1, DEC2, ETS-1, CITED2/p35sq, and NUR77), Transferrin, Transferrin receptor, Ceruloplasmin, Multidrug resistance P-glycoprotein, halofuginone, a retrotransposon, retrotransposon VL30, combinations thereof and the like.
In some exemplary embodiments, expression of a vascular endothelial growth factor (VEGF) is reduced in an eye of the subject. In some exemplary embodiments, activity of a vascular endothelial growth factor (VEGF) is reduced in an eye of the subject. In some exemplary embodiments, expression of a vascular endothelial growth factor receptor (VEGFR) is reduced in an eye of the subject. In some exemplary embodiments, activity of a vascular endothelial growth factor receptor (VEGFR) is reduced in an eye of the subject.
In some exemplary embodiments, the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v. In some exemplary embodiments, the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to about 0.1%, about 0.1 % to about 0.5%, about 0.5 % to about 1 %, about 1 % to about 1.5%, about 1.5% to about 2%, about 2% to about 2.5%, about 3% to about 4%, about 4% to about 5%, about 5% to about 6%, about 6% to about 7%, or about 7% to about 8%.
In some exemplary embodiments, the HIF inhibitor is administered at a dose of or about 0.001 mg, about 0.002 mg, about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg, about 4.5 mg, about 5 mg, about 10 mg or more. In some exemplary embodiments, the HIF inhibitor is administered at a dose of or about 0.001 mg to about 0.01 mg, about 0.01 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
In some exemplary embodiments, administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the retina of the subject.
In some exemplary embodiments, administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the choroid or to the suprachoroidal space.
In some exemplary embodiments, the HIF inhibitor is selected from the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF -2 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor and an HIF-2 inhibitor.
In some exemplary embodiments, the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3-[[(lS,2S,3R)- 2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5- fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro- lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)- camptothecin (NSC-639174), a cardenolide, EZN-2208 (PEG-SN38), SN38 (7-Ethyl-lO- hydroxy-camptothecin), a Ca2+ channel blocker, NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH-benzimidazol-2-yl)propyl]methylamino]ethyl]-6-fluoro-l,2,3,4- tetrahydro-l-(l-methylethyl)-2-naphthalenyl ester, dihydrochloride, PX-478 (,S'-2-amino-3- [4'-N,N,-bis(chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride), an inhibitor of the PBK/Akt/TOR pathway, an inhibitor of the MAPK pathway, resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, sorafenib, LY294002, wortmannin, nelfmavir, aHSP90 inhibitor, a glyceollin, IDF-11774 (2-(4-((3r,5r,7r)-adamantan-l-yl)phenoxy)-l-(4- methylpiperazin-l-yl)ethan-l-one), a histone deacetylase (HDAC) inhibitor, panobinostat (LBH589, (E)-N-hydroxy-3-[4-[[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop- 2-enamide), the indole-3-ethylsulfamoylphenylacrylamide compound MPT0G157, a diazepinquinazolin-amine derivate, BIX01294 (N-(l-benzylpiperidin-4-yl)-6,7-dimethoxy-2- (4-methyl-l,4-diazepan-l-yl)quinazolin-4-amine), a benzopyranyl 1,2,3-triazole, 4-(4- methoxyphenyl)- 1 -((2-methyl-6-nitro-2H-chromen-2-yl)methyl)- 1H- 1 ,2,3-triazole, Kresoxim-methyl, an analog of Kresoxim -methyl, a nanoparticle or nanoparticle conjugate, camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer, CRLX-101, PT2399, PT2977, 0X3 (N-(3-Chloro-5-fluorophenyl)-4- nitrobenzo[c][l,2,5]oxadiazol-5-amine), acriflavine (ACF), a CBP inhibitor, a p300 inhibitor, CG 13250, CCS 1477 ((L')- 1 -(3 ,4-Difluorophenyl)-6-(5 -(3 ,5 -dimethylisoxazol-4-yl)- 1 - ((lr,4S)-4-methoxycyclohexyl)-lH-benzo[d]imidazol-2-yl)piperidin-2-one), bortezomib ([(1 R)-3 -methyl- 1 -[ [(2S)-3 -phenyl-2-(pyrazine-2- carbonylamino)propanoyl]amino]butyl]boronic acid), chetomin, Erotinib, Gefitinib, Genistein, apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, pleurotin, aminoflavone, belinostat, CG1350, chidamide, cyclo-CLLFVY, digoxin, EZN-2968, glyceollins, IDF-1174, MPTOG1S7, NNC55-0396, romidepsin (Istodax/FK228), siRNA, tetrathiomolybdate, vorinostat (suberanilohydroxamic acid), combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK-6482, 3-[[(lS,2S,3R)-2,3-difluoro-l- hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)-5- fluorobenzonitrile), combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is MK-6482. The FDA has granted breakthrough therapy designation to MK-6482, an HIF-2-alpha inhibitor, for renal cell carcinoma subtype for the treatment of certain patients with von Hippel-Lindau disease- associated renal cell carcinoma. MK-6482 is under investigation for patients with von Hippel-Lindau disease-associated renal cell carcinoma with nonmetastatic tumors smaller than 3 cm, who do not require immediate surgery.
In some exemplary embodiments, administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition.
In some exemplary embodiments, administering the pharmaceutical composition comprises administration into the vitreous cavity of the eye.
In some exemplary embodiments, administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition into the vitreous cavity of an eye of the subject.
In some exemplary embodiments, administering the pharmaceutical composition comprises injecting the pharmaceutical composition. In some exemplary embodiments, administering the pharmaceutical composition comprises intravitreal injection.
In some exemplary embodiments, administering the pharmaceutical composition comprises implanting the pharmaceutical composition. In some exemplary embodiments, administering the pharmaceutical composition comprises implanting the pharmaceutical composition into the vitreous cavity.
In some exemplary embodiments, administering the pharmaceutical composition comprises administration selected from the group consisting of intravitreal injection, intravitreal implant, administering an eye drop, suprachoroidal injection, oral administration, parenteral injection, combinations thereof, and the like.
In some exemplary embodiments, administering the pharmaceutical composition comprises topical administration of an eye drop.
In some exemplary embodiments, administering the pharmaceutical composition comprises topical administration of an eye drop and delivery to the retina.
In some exemplary embodiments, administering the pharmaceutical composition comprises administration to the suprachoroidal space. In some exemplary embodiments, administering the pharmaceutical composition comprises repeated administration of the pharmaceutical composition.
In some exemplary embodiments, administering the pharmaceutical composition comprises administration of the pharmaceutical composition hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months. In some exemplary embodiments, administration occurs over a regimen of about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 12 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, or about 1 year.
In some exemplary embodiments, the method comprises administering a second therapeutic agent or treatment to the subject for treatment of an ischemic retinal disease. In some exemplary embodiments, the pharmaceutical composition is administered before, after or with the second therapeutic agent or treatment. In some exemplary embodiments, the second therapeutic agent is an angiogenesis inhibitor. In some exemplary embodiments, the second therapeutic agent is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, combinations thereof, and the like. In some exemplary embodiments, the second therapeutic agent is selected from among the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, a VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
In some exemplary embodiments, the second therapeutic agent or treatment is selected from among the group consisting of a corticosteroid (e.g., a corticosteroid selected from the group consisting of dexamethasone, triamcinolone, a combination thereof, and the like).
In some exemplary embodiments, the second therapeutic agent is formulated in a second pharmaceutical composition.
In some exemplary embodiments, the second therapeutic treatment is selected from the group consisting of laser photocoagulation, macular laser photocoagulation, panretinal photocoagulation (scatter photocoagulation), laser photocoagulation for retinal tears, oxygen therapy (such as hyperbaric), carotid surgery, combinations thereof and the like.
In some exemplary embodiments, the pharmaceutical composition and the second therapeutic agent are administered as a single composition or as two compositions. Another exemplary embodiment of this application is a composition (e.g., a pharmaceutical composition), comprising an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia. In some exemplary embodiments, the atrophy associated with the retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, combinations thereof and the like.
In some exemplary embodiments, the amount of the HIF inhibitor is effective to treat, minimize and/or inhibit apoptosis (e.g., retinal apoptosis) associated with retinal hypoxia in the subject. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%.
In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, a combination thereof, and the like.
In some exemplary embodiments, measuring phosphatidyl extemalization comprises annexin 5 staining.
In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, combinations thereof, and the like.
In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, macular atrophy, choroidal atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy, glaucomatous atrophy, ganglion cell atrophy, combinations thereof, and the like.
In some embodiments, the atrophy associated with retinal hypoxia is glaucomatous atrophy. In glaucomatous atrophy, retinal ganglion cells can undergo apoptosis due to hypoxia. HIF inhibition can protect ganglion cells from cell death in glaucoma. In some embodiments the glaucomatous atrophy is from glaucoma. In some embodiments the glaucoma is selected from the group consisting of chronic open angle glaucoma, closed angle glaucoma, secondary glaucoma, normal tension glaucoma, and combinations thereof. Thus, HIF inhibition can provide neuroprotection in glaucoma.
In some embodiments, the iris atrophy is from anterior segment ischemia. In some embodiments, the ciliary body atrophy is from anterior segment ischemia. In some embodiments, the optic nerve atrophy is from vascular ischemia. In some embodiments, the vascular ischemia comprises a condition selected from the group consisting of giant cell arteritis, embolisms, and a combination thereof. In some embodiments, the optic nerve atrophy comprises anterior ischemia optic neuropathy.
In some exemplary embodiments, the retinal atrophy is macular atrophy or geographic atrophy. In some exemplary embodiments, the atrophy is choroidal atrophy.
In some exemplary embodiments, the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor.
In some exemplary embodiments, the retinal detachment is selected from the group consisting of tractional, rhegmatogenous and serous retinal detachment.
In some exemplary embodiments the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor. In some exemplary embodiments the administration of a VEGF or VEGFR inhibitor is for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
In some exemplary embodiments the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the total area of the atrophy associated with retinal hypoxia.
In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 12%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more, or by about 100%. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%. In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of atrophy associated with retinal hypoxia by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or by about 100%. In some exemplary embodiments, the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the total area of atrophy as assessed by a method selected from the group consisting of spectral-domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, biomicroscopy, combinations thereof, and the like.
In some exemplary embodiments, the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the total area of retinal atrophy associated with retinal hypoxia as assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, visual acuity testing, near-infrared reflectance, fundus photography, biomicroscopy, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), retinal oximetry, microperimetry, retinal oximetry, combinations thereof, and the like.
In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce and/or substantially maintain the severity grade of atrophy associated with retinal hypoxia.
In some exemplary embodiments, the pharmaceutical composition comprises an amount of the HIF inhibitor effective to improve, or substantially maintain, the Age-Related Eye Disease Study (AREDS) scale.
In some exemplary embodiments the amount of the HIF inhibitor is effective to treat, minimize and/or substantially inhibit atrophy associated with retinal hypoxia in a subject being treated with an angiogenesis inhibitor.
In some exemplary embodiments, the angiogenesis inhibitor is a VEGF inhibitor and/or a VEGFR inhibitor. In some exemplary embodiments, the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof and the like). In some exemplary embodiments, the angiogenesis inhibitor is a VEGFR inhibitor selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
In some exemplary embodiments, the retinal hypoxia is associated with a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof. In some exemplary embodiments, the retinal hypoxia is associated with retinal detachment. In retinal detachments, delivery of oxygen to the retina can be reduced because of the increased distance from the choroidal source of oxygen. Retinal detachments thus can be hypoxic.
In some exemplary embodiments, the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, serous retinal detachment, rhegmatogenous retinal detachment, tractional retinal detachment, proliferative vitreoretinopathy (PVR), and central serous chorioretinopathy.
In some exemplary embodiments, in serous retinal detachments, including central serous chorioretinopathy, in AMD, in posterior uveitis (VKH), or a combination thereof, HIF inhibition can treat prevent, reduce, or substantially inhibit HIF induced apoptosis and/or atrophy (e.g., retinal atrophy). In some exemplary embodiments, consequent vision loss can be treated, prevented, reduced or substantially inhibited.
In some exemplary embodiments, in long standing rhegmatogenous retinal detachments, retinal atrophy and/or thinning can be treated, reduced, prevented, or substantially inhibited with anti HIF treatment. In some exemplary embodiments, proliferative vitreoretinopathy (PVR) can be treated. In some exemplary embodiments, atrophy in PVR can be treated.
In some exemplary embodiments, traction retinal detachments (e.g., proliferative diabetic retinopathy, retinopathy of prematurity, PVR, combinations thereof and the like), retinal atrophy and/or thinning can be reduced and/or prevented by an HIF inhibitor.
In some exemplary embodiments, the retinal hypoxia is associated with an ischemic retinal disease. In some exemplary embodiments, the HIF inhibitor is effective to treat, minimize and/or substantially inhibit a symptom associated with the ischemic retinal disease. In some exemplary embodiments, the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion, combinations thereof, and the like.
In some exemplary embodiments, the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of dry atrophic age related macular degeneration (atrophic AMD; geographic atrophy), diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy retinal pigment epithelial detachment, central serous chorioretinopathy, combinations thereof, and the like.
In some exemplary embodiments, the HIF inhibitor is present in an amount effective to reduce progression of retinal atrophy in dry AMD and dry atrophic AMD (Geographic atrophy); and/or reduce or prevent atrophy associated with anti VEGF and/or anti VEGFR treatment for neovascular AMD.
In some exemplary embodiments, HIF inhibitor is present in an amount effective to treat, minimize or substantially inhibit angiogenesis and/or edema in addition to atrophy. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to treat, minimize, substantially inhibit and/or reduce progression of retinal atrophy (e.g., in dry AMD) and/or severe and symptomatic atrophy (Geographic atrophy).
In some exemplary embodiments, the ischemic retinal disease is diabetic macular edema.
In some exemplary embodiments, the ischemic retinal disease is diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy).
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy), retinal vein occlusions, sickle cell retinopathy, combinations thereof, and the like. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to reduce or prevent retinal atrophy associated with anti VEGF and/or anti VEGFR treatment of diabetic macular edema or proliferative diabetic retinopathy, a combination thereof, or the like. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to treat, minimizes or substantially inhibit angiogenesis and/or edema in addition to atrophy. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to treat, minimizes or substantially inhibit progression of retinal atrophy (e.g., in patients with macular ischemia) and severe and symptomatic atrophy. In some exemplary embodiments, the ischemic retinal disease is diabetic retinopathy (e.g., proliferative or non-proliferative diabetic retinopathy or macular ischemia in diabetic retinopathy).
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, and severe non-proliferative diabetic retinopathy.
In some exemplary embodiments, the ischemic retinal disease is proliferative diabetic retinopathy. In some exemplary embodiments, the ischemic retinal disease is central retinal vein occlusion. In some exemplary embodiments, the ischemic retinal disease is branch retinal vein occlusion.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
In some exemplary embodiments, the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
In some exemplary embodiments, the HIF inhibitor is present in an amount effective to decrease expression of an HIF target gene or locus. In some exemplary embodiments, the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI, TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, plasminogen- activator inhibitor- 1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2, ahexokinase activator, lactate dehydrogenase A/LDHA, a lactate dehydrogenase A/LDHA inhibitor, lactate dehydrogenase B/LDHB, iNOS, perilipin-2, PGK1, PKM2, cathepsin D, CCL2/JE/MCP-1, CTGF/CCN2, CXCR4, HGFR/c-MET, IL-6, IL-8/CXCL8, integrin alpha 5/CD49e, LOX-1/OLR1, LOXL1, lysyl oxidase homolog 2/LOXL2, MKP-1, MMP-1, MMP-2, osteopontin/OPN, pref- 1/DLKl/FAl, SNAI1, TCF-3/E2A, TRKB, TWIST-1, uPAR, ZEB1, KLF4, NANOG, OCT- 3/4, OCT-4A, OCT-4B, and SOX2, Adrenomedullin/ADM, Cyclin Dl, Erythropoietin/EPO, IGF-II/IGF2, IGFBP-1, IGFBP-2, IGFBP-3, NOTCH 1, Survivin, TGF-a, keratin 14, keratin 18, keratin 19, vimentin, CXCR4, c-Met, autocrine motility factor (AMF/GPI), LDL receptor related protein 1 (LRPl), Transforming growth factor-a (TGF-a), Transforming growth factor^3 (TGF^3), Insulin-like growth factor 2 (IGF-2), IGF binding protein 1, 2 and 3 (IGF-BP), WAF1, Cyclin G2), Endothelin 1 (ET1), Adrenomedullin (ADM), Tyrosine hydroxylase, alB-adrenergic receptor, Inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), heme oxygenase- 1, atrial natriuretic peptide, insulin-like growth factor binding protein- 1, NIP3, NIX, RTP801, Endoglin (ENG), Wilms' tumour suppressor, a-Fetoprotein, and Calcitonin-receptor like receptor), Erythropoetin (EPO),
Leptin (LEP)), Glucose transporter 1 (GLUT1), Hexokinase 1 and 2 (HK1 and 2), 6-Phosphofructo-l -kinase L (PFKL), 6-Phosphofructo-2-kinase, Glyceraldehyde-3-P dehydrogenase (GAPDH), Aldolase A (ALDA), Aldolase C (ALDC), Enolase 1 (ENOl), Phosphoglycerate kinase-1 (PGK1), Lactate dehydroxygenase A (LDHA), Pyruvate kinase M (PKM), Carbonic anhydrase 9 (CA9), Adenylate kinase 3, and Transglutaminase 2), Pro collagen prolyl hydroxylase al, Collagen type V (al), Intestinal trefoil factor (TFF), Ecto-5'- nucleotidase, Cathepsin D (CATHD), Fibronectin 1 (FN1), Matrix metalloproteinase 2 (MMP2)), DEC1, DEC2, ETS-1, CITED2/p35sq, and NUR77), Transferrin, Transferrin receptor, Ceruloplasmin, Multidrug resistance P-glycoprotein, halofuginone, a retrotransposon, retrotransposon VL30, combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor (VEGF) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject. In some exemplary embodiments, wherein the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor (VEGF) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor receptor (VEGFR) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject. In some exemplary embodiments, the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor receptor (VEGFR) in an eye (for example, in the retina, iris, choroid, vitreous humor, combinations thereof, and the like) of the subject.
In some exemplary embodiments, the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v.
In some exemplary embodiments, the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to 0.1%, about 0.1% to 0.5%, about 0.5% to 1%, about 1% to 1.5%, about 1.5% to 2%, about 2% to 2.5%, about 3% to 4%, about 4% to 5%, about 5% to 6%, about 6% to 7%, or about 7% to about 8%. In some exemplary embodiments, the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg, about 0.002 mg, about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg, about 4.5 mg, about 5 mg, about 10 mg or more.
In some exemplary embodiments, the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg to 0.01 mg, about 0.01 mg to 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
In some exemplary embodiments, the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the retina of the subject. In some exemplary embodiments, the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the choroid of the subject.
In some exemplary embodiments, the HIF inhibitor is selected from among the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF -2 inhibitor. In some exemplary embodiments, the HIF inhibitor is an HIF-1 inhibitor and an HIF-2 inhibitor.
In some exemplary embodiments, the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3-[[(lS,2S,3R)- 2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5- fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro- lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)- camptothecin (NSC-639174), a cardenolide, EZN-2208 (PEG-SN38), SN38 (7-Ethyl-10- hydroxy-camptothecin), a Ca2+ channel blocker, NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH-benzimidazol-2-yl)propyl]methylamino]ethyl]-6-fluoro-l,2,3,4- tetrahydro-l-(l-methylethyl)-2-naphthalenyl ester, dihydrochloride, PX-478 (,S'-2-amino-3- [4'-N,N,-bis(chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride), an inhibitor of the PBK/Akt/TOR pathway, an inhibitor of the MAPK pathway, resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, sorafenib, LY294002, wortmannin, nelfmavir, aHSP90 inhibitor, a glyceollin, IDF-11774 (2-(4-((3r,5r,7r)-adamantan-l-yl)phenoxy)-l-(4- methylpiperazin-l-yl)ethan-l-one), a histone deacetylase (HDAC) inhibitor, panobinostat (LBH589, (E)-N-hydroxy-3-[4-[[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop- 2-enamide), the indole-3-ethylsulfamoylphenylacrylamide compound MPT0G157, a diazepinquinazolin-amine derivate, BIX01294 (N-(l-benzylpiperidin-4-yl)-6,7-dimethoxy-2- (4-methyl-l,4-diazepan-l-yl)quinazolin-4-amine), a benzopyranyl 1,2,3-triazole, 4-(4- methoxyphenyl)- 1 -((2-methyl-6-nitro-2H-chromen-2-yl)methyl)- 1H- 1 ,2,3-triazole, Kresoxim-methyl, an analog of Kresoxim -methyl, a nanoparticle or nanoparticle conjugate, camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer, CRLX-101, PT2399, PT2977, 0X3 (N-(3-Chloro-5-fluorophenyl)-4- nitrobenzo[c][l,2,5]oxadiazol-5-amine), acriflavine (ACF), a CBP inhibitor, ap300 inhibitor, CG 13250, CCS 1477 ((L')- 1 -(3 ,4-Difluorophenyl)-6-(5 -(3 ,5 -dimethylisoxazol-4-yl)- 1 - ((lr,4S)-4-methoxycyclohexyl)-lH-benzo[d]imidazol-2-yl)piperidin-2-one), bortezomib ([(1 R)-3 -methyl- 1 -[ [(2S)-3 -phenyl-2-(pyrazine-2- carbonylamino)propanoyl]amino]butyl]boronic acid), chetomin, Erotinib, Gefitinib, Genistein, apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, pleurotin, aminoflavone, belinostat, CG1350, chidamide, cyclo-CLLFVY, digoxin, EZN-2968, glyceollins, IDF-1174, MPTOG1S7, NNC55-0396, romidepsin (Istodax/FK228), siRNA, tetrathiomolybdate, vorinostat (suberanilohydroxamic acid), combinations thereof and the like.
In some exemplary embodiments, the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK-6482; 3-[[(lS,2S,3R)-2,3-difluoro-l- hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)-5- fluorobenzonitrile), combinations thereof and the like. In some exemplary embodiments, the pharmaceutical composition is formulated for administration by injection and/or implantation.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration into the vitreous cavity of the eye.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration by injection and/or implantation into the vitreous cavity of an eye of the subject.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration by injection. In some exemplary embodiments, the pharmaceutical composition is formulated for administration by intravitreal injection.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration by implantation. In some exemplary embodiments, the pharmaceutical composition is formulated for administration by implantation into the vitreous cavity.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration selected from the group consisting of intravitreal injection, intravitreal implant, eye drop, suprachoroidal injection, oral administration, parenteral injection, combinations thereof, and the like.
In some exemplary embodiments, the pharmaceutical composition is formulated for topical administration as an eye drop.
In some exemplary embodiments, the pharmaceutical composition is formulated for delivery to the retina. In some exemplary embodiments, the pharmaceutical composition is formulated for delivery to the choroid. In some exemplary embodiments, the pharmaceutical composition is formulated for administration into the vitreous cavity of the eye. In some exemplary embodiments, the pharmaceutical composition is formulated for implantation into the vitreous cavity. In some exemplary embodiments, the pharmaceutical composition is formulated for intravitreal injection into the vitreous cavity.
In some exemplary embodiments, the pharmaceutical composition is formulated for administration to the suprachoroidal space.
In some exemplary embodiments, the pharmaceutical composition is formulated for repeated administration. In some exemplary embodiments, the pharmaceutical composition is formulated for administration selected from the group consisting of hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly, or every few months. In some exemplary embodiments, the pharmaceutical composition is formulated for administration over a regimen of about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 12 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, or about 1 year or more.
Another exemplary embodiment of this application is a combination, comprising the pharmaceutical composition described herein, and a second pharmaceutical composition comprising a second therapeutic agent for treatment of an ischemic retinal disease and/or treatment of a retinal detachment, such as ischemic retinal diseases and retinal detachments described herein.
In some exemplary embodiments, the pharmaceutical composition containing an HIF inhibitor is for administration before, after or with the second pharmaceutical composition.
In some exemplary embodiments, the second therapeutic agent is an angiogenesis inhibitor.
In some exemplary embodiments, the second therapeutic agent is selected from the group consisting of a VEGF inhibitor and/or a VEGFR inhibitor.
In some exemplary embodiments, the second therapeutic agent is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like.
In some exemplary embodiments, the second therapeutic agent is selected from among the group consisting of dexamethasone, triamcinolone, a corticosteroid, combinations thereof, and the like.
In some exemplary embodiments, the pharmaceutical composition containing an HIF inhibitor and the second pharmaceutical composition are formulated for administration as a single composition or as two compositions.
It is understood that the methods, compositions, combinations, and uses described herein for treating, minimizing and/or substantially inhibiting atrophy associated with retinal hypoxia can also be adapted for other forms of atrophy associated with hypoxia in other body systems, organs, and/or tissue. Thus, described herein are methods, compositions, combinations, and uses for treating, minimizing and/or substantially inhibiting atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia. In some embodiments, the hypoxia is not retinal hypoxia. Thus, an exemplary embodiment of this application is a method of treating, minimizing and/or substantially inhibiting atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor (e.g., an HIF inhibitor described herein), such as an effective amount of an HIF inhibitor, to a subject having hypoxia. In some exemplary embodiments, the HIF inhibitor is administered (e.g., systemically and/or as described herein) in an amount effective to treat, minimize and/or substantially inhibit atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia. Another exemplary embodiment of this application is a composition (e.g., a pharmaceutical composition), comprising an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy (e.g., tissue and/or organ atrophy) associated with hypoxia. In some exemplary embodiments, the composition is formulated for administration systemically and/or as described herein. In some exemplary embodiments, ischemia leads to the atrophy (e.g., tissue and/or organ atrophy). In some exemplary embodiments, the atrophy associated with hypoxia is selected from among the group consisting of atrophy of limbs (e.g., caused by vascular occlusions); atrophy of heart muscle (e.g., from coronary artery occlusions); ischemic atrophy of the liver, ischemic atrophy of the kidney, ischemic atrophy of the brain, combinations thereof, and the like.
Hypoxia-inducible factor (HIF)
Hypoxia-inducible factors regulate cellular response to low oxygen concentrations.
An HIF transcription factor dimer includes one of three oxygen-regulated a-subunits (e.g., HIF-la, HIF-2a, or HIF-3a) and a constitutively expressed beta-subunit (e.g., HIF-Ib or HIF- 2b). An HIF can bind to consensus sequences (hypoxia responsive elements, HRE) in the regulatory regions of target genes.
Under normoxic conditions, two proline residues of a HIF-a (e.g., HIF-la or HIF-2a) subunits are hydroxylated by HIF-prolyl hydroxylase (HIF-PD) proteins, and recognized by von-Hippel-Lindau tumor suppressor protein (pVHL) as part of an E3 ubiquitin ligase complex. The monomer is marked for proteasomal degradation. (Schofield et al. (2005) Biochem. Biophys. Res. Commun. 338, 617-626; Fandrey et al. Cardiovasc. Res. 2006, 71, 642-651).
Transcriptional activity of a HIF-a subunit (e.g., HIF-la or HIF-2a) can depend on oxygen-dependent hydroxylation of asparagine residues in HIF-a by the asparagyl hydroxylase factor-inhibiting HIF (FIH), preventing the binding of transcriptional coactivators (CBP/p300) and expression of target genes under oxygenated conditions. Transcriptional activity of HIF can be regulated, for example, by the accumulation or turnover of the HIF-a (e.g., HIF-la or HIF-2a) monomer. HIF activity can be inhibited by targeting one or more components that mediate hypoxic response. For example, SUMOylation of p300 can block interaction with HIF-a. Phosphorylation of HIF-a can block interaction with HIF-b (e.g., HIF-Ib or HIR-2b). COMMD1 can bind to HIF-a (e.g., HIF-la or HIF-2a) and block interaction with HIF-b (e.g., HIF-Ib or HIR-2b). Cited-2 can bind to HIF-a (e.g., HIF-la or HIF-2a) and block interaction with p300.
In hypoxic conditions, HIF-PH activity is decreased. HIF-a (e.g., HIF-la or HIF-2a) accumulates, dimerizes with a HIF-b (e.g., HIF-Ib or HIR-2b), and activates a transcriptional response to hypoxia, activating transcriptional activity of an HIF target gene. HIF target genes can include, but are not limited to, erythropoietin (EPO), VEGF, glucose transporter 1 (GLUT1), glycolytic enzymes (e.g, phosphoglycerate kinase 1, lactate dehydrogenase-A, carbonic anhydrase 9, and aldolase), transforming growth factor alpha, and cyclin D. Glycolytic enzymes can be regulated by HIF-la, while HIF-2a can regulate gene transcription of EPO, transforming growth factor alpha, and cyclin D. Some target genes, including VEGF, GLUT1, and adrenomedullin 1 (ADM-1), can be regulated by HIF-la and HIF-2a.
HIF-3a can directly regulate a subset of hypoxia-inducible genes involved in lipolysis (angiopoietin-like 4) and metabolism (angiopoietin-like 3 and pantothenate kinase 1). HIF- 3a can also interact with the promotor region of the EPO gene. Therefore, HIF-l/2a and HIF- 3 a could have synergistic effects on EPO transcription (Tolonen et al. (2020) Cell. Mol. Life Sci. 77:3627-3642).
Examples of HIF target genes or loci include genes involved in angiogenic signalling (e.g, Angiopoietin-1, Angiopoietin-2, Angiopoietin-4, Angiopoietin-like Protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-beta 1, TGF-beta 3, VEGF, Endothelial gland derived vascular endothelial growth factor (EG- VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, Plasminogen-activator inhibitor-1 (PAI1), and Urokinase plasminogen activator receptor (UPAR)), in metabolism (e.g., GAPDH, Glutl, Glut3, Hexokinase 1, Hexokinase 1/2, Hexokinase 2, Hexokinase Activators, Lactate Dehydrogenase A/LDHA, Lactate Dehydrogenase A/LDHA Inhibitors, Lactate Dehydrogenase B/LDHB, iNOS, Perilipin-2, PGK1, PKM2), in metastasis/cell migration, (e.g., Cathepsin D, CCL2/JE/MCP- 1, CTGF/CCN2, CXCR4, HGFR/c-MET, IL-6, IL-8/CXCL8, Integrin alpha 5/CD49e, LOX- 1/OLRl, LOXL1, Lysyl Oxidase Homolog 2/LOXL2, MKP-1, MMP-1, MMP-2, Osteopontin/OPN, Pref-l/DLKl/FAl, Snail, TCF-3/E2A, TrkB, Twist-1, uPAR, ZEB1), in pluripotency (e.g., KLF4, Nanog, Oct-3/4, Oct-4A, Oct-4B, and SOX2), in Proliferation/Survival (e.g., Adrenomedullin/ADM, Cyclin Dl, Erythropoietin/EPO, IGF- II/IGF2, IGFBP-1, IGFBP-2, IGFBP-3, Notch-1, Survivin, and TGF-a), in cytoskeleton formation (e.g., Keratin 14 (KRT14), Keratin 18 (KRT18), Keratin 19 (KRT19), and Vimentin), in cell migration (e.g., Chemokine receptor CXCR4, c-Met, Autocrine motility factor (AMF/GPI), LDL receptor related protein 1 (LRPl), Transforming growth factor-a (TGF-a), in cell proliferation (e.g., Transforming growth factor-a (TGF-a), Transforming growth factor^ (TGF- 3), Insulin-like growth factor 2 (IGF-2), IGF binding protein 1, 2 and 3 (IGF-BP), WAF1, and Cyclin G2), in vasomotor regulation (e.g., Endothelin 1 (ET1), Adrenomedullin (ADM), Tyrosine hydroxylase, alB-adrenergic receptor, Inducible nitric oxide synthase (iNOS), Endothelial nitric oxide synthase (eNOS), Heme oxygenase- 1, and Atrial natriuretic peptide), in growth and apoptosis (e.g., Insulin-like growth factor binding protein-1, NIP3, NIX, RTP801, Endoglin (ENG), Wilms' tumour suppressor, a-Fetoprotein, and Calcitonin-receptor like receptor), in Hormonal regulation (e.g., Erythropoetin (EPO), and Leptin (LEP)), in Energy Metabolism (e.g., Glucose transporter 1 (GLUT1), Hexokinase 1 and 2 (HK1 and 2), 6-Phosphofructo-l -kinase L (PFKL), 6-Phosphofructo-2 -kinase, Glyceraldehyde-3-P dehydrogenase (GAPDH), Aldolase A (ALDA), Aldolase C (ALDC), Enolase 1 (ENOl), Phosphoglycerate kinase-1 (PGK1), Lactate dehydroxygenase A (LDHA), Pyruvate kinase M (PKM), Carbonic anhydrase 9 (CA9), Adenylate kinase 3, and Transglutaminase 2), in Matrix and barrier functions (e.g., Pro-collagen prolyl hydroxylase al, Collagen type V (al), Intestinal trefoil factor (TFF), Ecto-5'-nucleotidase, Cathepsin D (CATHD), Fibronectin 1 (FN1), and Matrix metalloproteinase 2 (MMP2)), in Transcriptional regulation (e.g., DEC1, DEC2, ETS-1, CITED2/p35sq, and NUR77), in Transport (e.g., Transferrin, Transferrin receptor, Ceruloplasmin, Multidrug resistance P-glycoprotein), and retrotransposons (e.g., Retrotransposon VL30).
HIF Inhibitors
Any HIF inhibitor can be used in the compositions, uses, and methods described herein, if the HIF inhibitor can inhibit the HIF pathway. Inhibiting the HIF pathway can include inhibiting one or more of any of the components of the HIF pathway. For example, an HIF inhibitor can inhibit HIF, or can target components of the HIF pathway that mediate hypoxic response (e.g., PHDs, pVHL, FIH and CBP/p300). In some embodiments, HIF inhibitor can be injected and/or implanted into the vitreous humor or formulated for injection and/or implantation into the vitreous humor. In some embodiments of the compositions, uses, and methods described herein, HIF inhibitors can target one or more selected from the group consisting of inhibiting transcription of HIF mRNA, inhibiting HIF protein synthesis, interfering with stabilization of HIF, decreasing transcription of HIF target genes, activating prolyl -4-hydroxylase domain (PHD), interfering with interactions between HIF-a’s to von Hippel-Lindau tumor suppressor protein (pVHL), combinations thereof and the like. In some embodiments, the HIF inhibitors can inhibit the HIF pathway, for example, by inhibiting transcription and/or translation, HIF stabilization, HIF-a/b dimerization, transcription complex formation, combinations thereof and the like.
In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor disrupts heterodimerization (e.g., heterodimerization selected from the group consisting of HIF-2a/HIF-i , HIF-la/HIF-Ib, HIF-2a/HIF^, HIF-la/HIF^, and combinations thereof). In some examples of the compositions, uses, and methods described herein, the HIF inhibitor is belzutifan (MK-6482; 3-[[(lS,2S,3R)-2,3-difluoro-l-hydroxy-7- methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5-fluorobenzonitrile), an inhibitor of HIF- 2a/HIR-1b heterodimerization. (Courtney et al. (2018) J. Clin. Oncol. 36(9):867-874).
In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor blocks binding of HIF to DNA. In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor is doxorubicin. Doxorubicin can inhibit HIF-1 transcriptional activity of blocking binding of HIF- 1 to DNA (Duyndam et al. (2007) Biochem. Pharmacol. 74(2): 191-201; Lee et al. (2009) Proc. Natl. Acad. Sci. USA. 106:2353-8).
In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor is a topoisomerase inhibitor. In some further embodiments, the HIF inhibitor is a topoisomerase-I inhibitor. In some further embodiments, the HIF inhibitor is camptothecin or an analog of camptothecin. In some further embodiments, the HIF inhibitor is selected from the group consisting of topotecan (NSC-609699), camptothecin 20-ester(S) (NSC- 606985), and 9-glycineamido-20(S)-camptothecin or an HC1 salt thereof (NSC-639174). In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor is the topoisomerase-I inhibitor topotecan, which inhibits HIF-1 transcriptional activity. (Rapisarda et al., 2002, Cancer Res, 62:4316-4324; Rapisarda et al., 2004, Cancer Res, 64: 1475-1482 and Rapisarda et al., 2004, Cancer Res, 64:6845-6848).
In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor is a cardenolide. In some further emodiments, the HIF inhibitor is a cardenolide that transcriptionally inhibits HIF-1. In some further embodiments, the HIF inhibitor is SN38 (7-Ethyl-lO-hydroxy-camptothecin). Kami yam a el al. (2005) J. Cancer Res. Clin. Oncol. 131:205-213.
In some embodiments of the compositions, uses, and methods described herein, the HIF inhibitor is a Ca2+ channel blocker. In some further emodiments, the HIF inhibitor is the Ca2+ channel blocker NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH- benzimidazol-2-yl)propyl]methylamino] ethyl] -6-fluoro- 1 ,2,3,4-tetrahydro- 1 -( 1 -methylethyl)- 2-naphthalenyl ester, dihydrochloride; CAS No. 357400-13-6), which can decrease mitochondrial reactive oxygen species (ROS) production, block HIF-1 activation, increase HIF-Ia protein hydroxylation and degradation, and suppress HIF-Ia de novo synthesis. (Kim et al. (2015) J. Mol. Med. 93:499-509).
In some further embodiments, the HIF inhibitor is PX-478 (,S'-2-amino-3-|4'-N,N,- bis(chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride), which can decrease HIF-Ia mRNA levels, block HIF- la translation, and inhibit deubiquitination, resulting in increased protein degradation. (Koh et al. (2008) Mol. Cancer Ther. 7, 90-100; Koh et al. (2008) Mol Cancer Ther. 7(1):90-100. PMID: 18202012.).
In some embodiments, the HIF inhibitor blocks the PI3K/Akt/TOR and/or MAPK pathway. In some embodiments, the HIF inhibitor is bortezomib (PS-341), which can represses HIF- la on transcriptional and translational levels, and inhibit recruitment of the coactivator p300, blocking the PI3K Akt/TOR and MAPK pathway. (Hideshima et al. (2003) Blood J. Am. Soc. Hematol. 2003, 101, 1530-1534; Befani et al. (2012) J. Mol. Med. 90, 45- 54). In some embodiuments, the HIF inhibitor is selected from among resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, and sorafenib. In some embodiments, the HIF inhibitor inhibits the PI3K-AKT pathway. In some embodiuments, the HIF inhibitor is LY294002, wortmannin, or nelfmavir. (Jiang et al. (2001) Cell Growth Differ. 12(7):363-9; Pore et al. (2006) Cancer Res. 66(18):9252-9).
In some embodiments, the HIF inhibitor inhibits the Pi3K/AKT/mTOR pathway. In some embodiments, the HIF inhibitor decreases HSP90 binding. In some embodiments, the HIF inhibitor is a glyceollin (i.e., a soybean-derived phytoalexin), which can block HIF-la translation via inhibition of the Pi3K/AKT/mTOR pathway and decrease HIF-Ia stability by decreasing Hsp90 binding. (Lee et al. (2015) J. Cell. Physiol. 230:853-862).
In some examples, the HIF inhibitor for use in the compositions, uses, and methods described herein is an HIF-Ia inhibitor. In some examples, the HIF inhibitor is PX-478. Other HIF inhibitors for use in the compositions, uses, and methods described herein is an HIF-Ia inhibitor selected from among the group consisting of Bortezomib (Velcade®), Glyceollins, NNC55-0396, PX-478, Aminoflavone, Benzopyranyl 1,2,3-trizole, BIX01294, Bortezomib (Velcade®), Cardenolides (e.g. EZN-2208 (PEG-SN38)), CRLX-101, Digoxin, Erotinib, Everolimus, EZN-2968, Gefitinib, Genistein, Glyceollins, IDF-1174, Kresoxim- methyl analoges, LBH589 (Panobinostat), LY294002, MPTOG1S7, Nelfmavir, NNC55- 0396, PD98059, Rapamycin, Resveratrol, RNA interference, Silibinin, Sorafenib, Temsirolimus, Tetrathiomolybdate, Topotecan, and Wortmannin.
In some embodiments, the HIF inhibitor is a melphalan derivative. In some embodiments, the HIF inhibitor is an alkylating agent. In some embodiments, the HIF inhibitor is a nitrogen mustard or nitrogen mustard derivative. In some embodiments, the HIF inhibitor is a nitrogen mustard N-oxide or a nitrogen mustard N-oxide derivative.
In some examples, the HIF inhibitor increases HIF-a (e.g., HIF-la or HIF-2a) protein degradation. For example, the HIF inhibitor can increase pVHL activity. In some examples, the HIF inhibitor upregulates pVHL expression. In some embodiments, the HIF inhibitor is IDF- 11774 (2-(4-((3r,5r,7r)-adamantan- 1 -yl)phenoxy)- 1 -(4-methylpiperazin- 1 -yl)ethan- 1 - one), which can upregulate pVHL expression, resulting in increased degradation of HIF-1. (Ban et al. (2017) Cell Death Dis. 8(6):e2843; Lee et al. (2010) Biochem. Pharmacol. 80, 982-989).
In some embodiments, the HIF inhibitor is a histone deacetylase (HDAC) inhibitor. If some further embodiments, the HIF inhibitor is panobinostat (LBH589; (E)-N-hydroxy-3-[4- [[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop-2-enamide), a HDAC inhibitor that can disrupt the Hsp90/HDAC6 complex (Kovacs et al. (2005 )Mol. Cell 18:601-607). Hsp90 complexing with HIF-Ia, and also acetylation of HIF-Ia, can prevent degradation through the proteasome/pVHL pathway complex. Therefore, histone deacetylase (HDAC) inhibitors such as panobinostat can reduce HIF-Ia protein. In some embodiments, the HIF inhibitor is the indole-3 -ethylsulfamoylphenylacrylamide compound MPT0G157.
MPT0G157 can inhibit multiple histone deacetylases (1, 2, 3, and 6) and decrease levels of HIF-Ia protein. (Huang et al. (2015) Oncotarget 6: 18590).
In some embodiments, the HIF inhibitor can increase PHD2 and/or pVHL expression. In some further embodiments, the HIF inhibitor is a diazepinquinazolin-amine derivate. In some further embodiments, the HIF inhibitor is BIX01294 (N-(l-benzylpiperidin-4-yl)-6,7- dimethoxy-2-(4-methyl-l,4-diazepan-l-yl)quinazolin-4-amine), which can increase PHD2 and pVHL expression and can reduce HIF-Ia protein levels. (Oh et al. (2015) Mol. Cells 38, 528). In some embodiments, the HIF inhibitor can induce HIF (e.g. HIF-Ia) hydroxylation and ubiquitination, which can result in increased protein degradation. In some further embodiments, the HIF inhibitor is a benzopyranyl 1,2,3-triazole. In some further embodiments, the HIF inhibitor is 4-(4-methoxyphenyl)-l-((2-methyl-6-nitro-2H-chromen-2- yl)methyl)-lH-l, 2, 3-triazole. (Park, (2017) Oncotarget 8:7801).
In some embodiments the HIF inhibitor can increase oxygen tension. In some further embodiments the HIF inhibitor can promote proteasomal degradation of HIF-a (e.g., HIF-la) via increased oxygen tension. In some further embodiments, the HIF inhibitor is Kresoxim- methyl or an analog of Kresoxim -methyl. (Lee et al. (2017) Bioorg. Med. Chem. Lett. 27:3026-3029).
In some further embodiments, the HIF inhibitor is a nanoparticle or nanoparticle conjugate. In some further embodiments, the HIF inhibitor is a nanoparticle of an active compound conjugated to a cyclodextrin-based polymer (e.g., a linear cyclodextrin-based polymer). In some further embodiments, the HIF inhibitor is camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer. In some further embodiments, the HIF inhibitor is CRLX-101, and can suppress HIF-a (e.g., HIF-la) protein translation and stability. (Pham et al. (2015) Clin. Cancer Res. 21:808-818).
In some embodiments, the HIF inhibitor can inhibit one or more components of the HIF pathway selected from among the group consisting of HIF-a/b dimerization, transcription complex formation, and combinations thereof. HIF-a/b dimerization is a part of the pathway in which HIF complex induces expression of HIF target genes. In some embodiments, the HIF inhibitor is an HIF -2a inhibitor. In some further embodiments, the HIF inhibitor is selected from among the group consisting of PT2385, PT2399, and PT2977. (Cho et al. (2016) Nature 2016, 539, 107-111; Wallace et al. Cancer Res. 2016, 76, 5491- 5500; and Courtney et al. (2018) J. Clin. Oncol. 36:867). In some embodiments, the HIF inhibitor can disrupt HIF heterodimer formation. In some further embodiments, the HIF inhibitor is the compound 0X3 (N-(3-Chloro-5-fluorophenyl)-4- nitrobenzo[c][l,2,5]oxadiazol-5-amine). (Scheuermann et al. (2013) Nat. Chem. Biol. 9:271).
In some embodiments, the HIF inhibitor can bind to the PAS-B domain of HIF-la or HIF-2a and block heterodimerization with HIF-b (e.g., HIF-Ib). In some further embodiments, the HIF inhibitor is acriflavine (ACF), which can bind to the PAS-B domain of HIF-la and HIF-2a. (Lee et al., (2009) Proc. Natl. Acad. Sci. USA 2009, 106, 17910-17915.)
In some embodiments, the HIF inhibitor inhibits a transcriptional coactivator of HIF (e.g., CBP or p300). In some further embodiments, the HIF inhibitor is selected from the group consisting ofCG13250, CCS1477 ((S)-l-(3,4-Difluorophenyl)-6-(5-(3,5- dimethylisoxazol-4-yl)- 1 -(( lr,4S)-4-methoxycyclohexyl)- lH-benzo[d]imidazol-2- yl)piperidin-2-one), bortezomib ([(1 R)-3 -methyl- 1 - [[(2S)-3 -phenyl-2-(pyrazine-2- carbonylamino)propanoyl]amino]butyl]boronic acid) and chetomin (Shin et al. (2008) Blood 111(6):3131—6; Kung et al. AL, (2004) Cancer cell. 6(1):33— 43; Knurowski et al. (2019) Blood 134: 1266; and Imayoshi et al. (2017) Biochem. Biophys. Res. Commun. 2017, 484, 262-268.).
In some embodiments, the HIF inhibitor is a receptor tyrosine kinase inhibitor, such as an EGFR tyrosine kinase inhibitor, including, for example, an inhibitor selected from the group consisting of Erotinib, Gefitinib, and Genistein. (Pore et al. (2006) Cancer Res.
66(6):3197-204; Buchler et al. (2004) Cancer 100(1):201-10).
In some examples, the HIF inhibitor is selected from the group consisting of apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, and pleurotin (Fang et al. (2007) Carcinogenesis 28(4):858-64; Kim et al. (2009) Cancer Res. 2009;69(4): 1624-32; Alqawi et al. (2006) Prostate Cancer Prostatic Dis. 9(2): 126-35; 225. Mie et al. (2003) Biochem Biophys Res Commun. 300(l):241-6; Shankar et al. (2009) Mol. Cancer Ther. 8(6): 1596-605; Yang et al. (2006) J. Exp. Clin. Cancer Res. 25(4):593-9; Newcomb et al. (2005) Neuro. Oncol. 7(3):225-35; Duyndam et al. (2007) 74(2): 191-201; Kong et al. (2005) Cancer Res. 2005;65(19):9047-55; Olenyuk (2004) Proc Natl Acad Sci USA 101(48): 16768-73;
Mabjeesh etal. (2003) Cancer cell. 3(4):363-75; Bae et al. (2006) Oncol. Rep. 15(6): 1557— 62; Maeda et al. (2006) Biol Pharm Bull. 29(7): 1344-8; Shin et al. (2008) Blood 111(6):3131—6; Kung et al. (2004) Cancer cell. 6( 1):33— 43; Yasui et al. (2008) Br. J. Cancer 99(9): 1442-52; Zhao et al. (2007) Pancreas 34(2):242-7; Welsh et al. (2003) Mol. Cancer. Ther. 2(3):235-43.
Table 1 below sets forth exemplary HIF inhibitors for use in the compositions, uses, and methods described herein. This is not an exhaustive list. Description in the table is not meant to be limiting. It is understood that description of an inhibition mechanism or pathway does not exclude other inhibition mechanisms or pathways. Table 1. Exemplary HIF Inhibitors
Figure imgf000047_0001
Figure imgf000048_0001
Retinal Hypoxia
Provided herein are methods, uses, compositions, and combinations for treating, minimizing and/or substantially reducing atrophy associated with retinal hypoxia. Hypoxic conditions can result from different diseases and conditions. For example, ischemia (e.g., ischemic retinal disease) and/or retinal detachment can result in hypoxic conditions, which can activate HIF. HIF can trigger neovascularization, edema, and apoptosis. Apoptosis can result in atrophy. Thus, in some embodiments of the methods, uses, compositions described herein, HIF inhibitors can inhibit hypoxia induced apoptosis and/or atrophy (.e.g, in an ischemic retinal disease and in retinal hypoxic conditions from other causes (for example retinal detachments)). In some embodiments of the methods, uses, and compositions provided herein, HIF inhibitors can inhibit retinal apoptosis/atrophy that accompanies administration of an angiogenesis inhibitor, for example for treatment of an ischemic retinal disease.
Exemplary diseases and conditions that can result in retinal hypoxia include, but are not limited to, age related macular degeneration (AMD), geographic atrophy (also known as dry atrophic age related macular degeneration (atrophic AMD)), dry AMD, diabetic retinopathy (e.g., proliferative diabetic retinopathy or non-proliferative diabetic retinopathy or diabetic macular edema or macular ischemia in diabetic retinopathy), retinal vein occlusion (e.g., central retinal vein occlusion or branch retinal vein occlusion), retinopathy of prematurity (ROP), sickle cell retinopathy, rhegmatogenous or tractional retinal detachment and proliferative vitreoretinopathy (PVR), retinal pigment epithelial detachment, central serous chorioretinopathy and other serous retinal detachments.
In some embodiments, the ischemic disease is atrophic AMD or diabetic retinopathy. In some embodiments, the HIF inhibitor prevents and/or substantially inhibits one or more symptoms selected from the group consisting of apoptosis (e.g., retinal apoptosis), retinal atrophy and choroidal atrophy associated with atrophic AMD or diabetic retinopathy.
In some embodiments, the retinopathy of prematurity (ROP) is selected from the group consisting of stage 1 ROP (e.g., mildly abnormal blood vessel growth), stage 2 ROP (e.g., moderately abnormal blood vessel growth), stage 3 ROP (e.g., severely abnormal blood vessel growth), stage 4 ROP (e.g., partially detached retina), or stage 5 ROP (e.g., completely detached retina). (Parveen et al. (2018) Taiwan J Ophthalmol. 8(4): 205-215).
In some embodiments, the diabetic retinopathy is selected from among the group consisting of stage 1 diabetic retinopathy (e.g., mild nonproliferative retinopathy), stage 2 diabetic retinopathy (e.g., moderate nonproliferative retinopathy), stage 3 diabetic retinopathy (e.g., severe nonproliferative retinopathy), stage 4 diabetic retinopathy (e.g., proliferative retinopathy) and diabetic macular edema.
In some embodiments, the sickle cell retinopathy is selected from among the group consisting of stage 1 sickle cell retinopathy (e.g., peripheral arterial occlusion), stage 2 sickle cell retinopathy (e.g., Peripheral arteriovenous anastomoses, representing dilated pre-existing capillaries (hairpin loop)), stage 3 sickle cell retinopathy (e.g., neovascular and fibrous proliferation (sea fan)), stage 4 sickle cell retinopathy (e.g., vitreous hemorrhage), and stage 5 sickle cell retinopathy (e.g., tractional retinal detachment).
In some embodiments, central serous chorioretinopathy and other serous retinal detachments are treated with HIF inhibitor to prevent or reduce long term retinal atrophy.
In some embodiments, rhegmatogenous or tractional retinal detachments or proliferative vitreoretinopathy are treated with HIF inhibitor to prevent or reduce long term retinal atrophy.
In some embodiments, HIF inhibition complements or replaces anti VEGF or anti VEGFR treatment for neovascular AMD, proliferative diabetic retinopathy, diabetic macular edema or neovascularization/edema in retinal vein occlusion, in order to reduce the retinal atrophy that otherwise accompanies VEGF inhibition.
Methods and Compositions
The methods can include administration of an HIF inhibitor, alone or in combination with another agent or treatment for treating, minimizing and/or substantially inhibiting an ischemic retinal disease or condition described herein. The methods can include administration of a composition containing an HIF inhibitor as described herein. The methods can include selection of subjects for treatment, e.g., prior to treatment of the subject. Subjects can be determined to have one or more selected from the group consisting of an ischemic retinal disease or condition, retinal hypoxia, retinal detachment (or a related condition), retinal neovascularization, and activated HIF. In some examples, the method further includes treatment, for example, administration of an HIF inhibitor alone or in combination with one or more other treatments. In some examples, the method further includes determining reduction in a symptom after treatment. In some examples, the method further includes determining reduction in an adverse effect of treatment. In some examples, the symptom or adverse effect is selected from among the group consisting of apoptosis (e.g., retinal apoptosis), and atrophy (e.g., retinal atrophy and/or choroidal atrophy).
Assays to determine reduction in a symptom or adverse effect can include, for example, assays described herein, such as, for example, measuring pupil’s reaction to light, visual acuity, visual field or peripheral vision testing, microperimetry, annexin 5 staining, measuring phosphatidyl extemalization, retinal fundus photography, spectral-domain optical coherence tomography (OCT), near-infrared reflectance, assessment of Age-Related Eye Disease Study (AREDS) scale, diabetic retinopathy grading, combinations thereof, and the like.
In some examples, a subject with elevated HIF activiation in an eye is treated with an HIF inhibitor. In some further embodiments, treatment includes a reduction in the HIF activation in the eye of the subject. The HIF activation level can be determined prior to and/or after treating the subject, for example, as described herein or known to those of skill in the art. After treatment, HIF activation can be reduced by, for example about 5% or more, about 10% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 50% or more, or about 75% or more compared to HIF activation prior to treatment. After treatment, HIF activation can be reduced by, for example about 5% to about 10%, about 10% to about 15%, about 15% to about 20%, about 20% to about 25%, about 25% to about 30%, or about 30% to about 50%.
In some examples, the HIF inhibitor is administered locally, for example, by topical administration of an eye drop or by injection or implantation into the vitreous humor or suprachoroidal space. In some examples, the HIF inhibitor is delivered to the retina or choroid. The HIF-inhibitor can be administered, for example, by injection or implantation.
In some examples, the HIF inhibitor is adminstered into the vitreous cavity of the eye by intravitreal injection or by implantation into the vitreous cavity. In some examples, the HIF inhibitor is administered systemically, for example, intravenously (IV) or intramuscularly. In some examples, the HIF inhibitor can be administered intraocularly, orally, intravenously (IV), subcutaneously, intramuscularly, intraperitoneally, intradermally, topically, transdermally, rectally or sub-epidermally.
In addition to treatment of atrophy associated with retinal hypoxia with the HIF inhibitor alone, the methods, uses and compositions and provided herein also can be used to treat atrophy associated with retinal hypoxia by administration of the HIF inhibitor in combination with, for example, simultaneously, prior to, or after, another therapeutic agent or treatment. The other therapeutic agent or treatment can be to treat a disease or condition selected from the group consisting of for an ischemic retinal disease, a retinal detachment (or related condition), atrophy, combinations thereof, and the like. The other therapeutic agent or treatment can be formulated with, or separate from, the HIF inhibitor.
In some embodiments, the treatment of an ischemic retinal disease can include administration of an angiogenesis inhibitor (e.g., a VEGFR inhibitor (e.g., an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like), and/or a VEGF inhibitor (e.g., an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, combinations thereof and the like)). Treatment with an angiogenesis inhibitor can inhibit neovasculazation. When angiogenesis is inhibited, HIF activation can result in one or more adverse effects selected from among apoptosis (e.g., retinal apoptosis), retinal atrophy, choroidal atrophy, vision loss and combinations thereof, as neovascularization is blocked. HIF activation can result in several mitigating actions to hypoxia. These include VEGF production for angiogenesis and increased blood flow. If angiogenesis is blocked, for example, by an angiogenesis inhibitor (e.g., VEGF inhibitor or VEGFR inhibitor), and cannot mitigate the hypoxic state, other mitigating mechanisms, such as apoptosis, mitigate hypoxia, which can result in retinal atrophy, for example with anti VEGF treatment in neovascular AMD. (Evans el al. (2020) JAMA Ophthalmol.
138(10): 1043-1051). Inhibiting HIF can decrease one or more effects selected from among apoptosis (e.g., retinal apoptosis), atrophy (e.g., retinal atrophy and/or choroidal atrophy), vision loss, combinations thereof and the like.
The methods described herein can include assessing a symptom or adverse effect. For example, apoptosis can be assessed by measuring phosphatidyl extemalization. In some embodiments, phosphatidyl extemalization can be measured by annexin 5 staining. In some embodiments, apoptosis can be assessed by a method described herein, such as a method selected from the group consisting of morphological, functional, electric and metabolic methods, combinations thereof, and the like. In some exemplary embodiments, apoptosis can be assessed by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, combinations thereof, and the like. In some embodiments, atrophy can be assessed by the Age-Related Eye Disease Study (AREDS) Grading Scale. ( Arch Ophthalmol. 2005;123(11): 1484-1498.) In some embodiments, the area of atrophy can be assessed before and after administration of a composition described herein to determine if the area of atrophy changes or stays the same. For example, the area of atrophy can be assessed by fundus photography, spectral-domain optical coherence tomography (OCT), visual field examination, microperimetry, biomicroscopy, multifocal electroretinography, near-infrared reflectance, combinations thereof, and the like. In some embodiments, treatment can result in a reduction in severity grade of atrophy or reduction in the rate of progression of atrophy.
Compositions of an HIF inhibitor are provided herein. In some embodiments, the composition is a pharmaceutical composition. Typically, the HIF inhibitor can be formulated into pharmaceutical compositions using techniques and procedures well known in the art (see e.g., Patel etal. (2013) World J. Pharmacol. 2(2):47-64). Generally, the mode of formulation can be a function of the route of administration. In some exemplary embodiments, the HIF inhibitor or a pharmaceutically-acceptable salt thereof can be in a form selected from solid, solution or suspension.
The concentration and/or dose of the HIF inhibitor can be adjusted so that administration provides an effective amount to produce the desired pharmacological effect, and can include any concentration or dose described herein. The HIF inhibitor can be provided in a sufficient amount to inhibit a symptom of an ischemic retinal disease described herein (e.g., retinal apoptosis, retinal atrophy, choroidal atrophy, or a combination thereof), or to inhibit an adverse effect of treatment. The exact dose can depend on the age, weight and condition of the patient or animal as is known in the art.
In the compositions, uses and methods provided herein the composition (e.g., a pharmaceutical composition) can be administered, or formulated for administration, to the anterior segment of the eye or to the posterior segment of the eye. The anterior segment can include the cornea, conjunctiva, aqueous humor, iris, ciliary body, or lens. The posterior segment can include the sclera, choroid, retinal pigment epithelium, neural retina, optic nerve or vitreous humor. In the compositions, uses and methods provided herein, the composition (e.g., a pharmaceutical composition) can be adminstered, for formulated for administration, to the posterior segment ocular tissues, for example, by administration selected from the group consisting of intravitreal injection or implantation, suprachoroidal injection, periocular injections, and systemic administration. In some embodiments, a pharamceutical composition is administered by intravitreal injection. In some embodiments, a composition (e.g., a pharmaceutical composition) can be administered by periocular administration. In some further embodiments, a composition (e.g., a pharmaceutical composition) can be administered by transscleral drug delivery with periocular administration route. Also described herein are compositions formulated for administration as described herein.
Delivery of drugs to the targeted ocular tissues can be limited by various precorneal, dynamic and static ocular barriers. Ocular barriers to transscleral drug delivery can include static barriers (e.g., sclera, choroid and retinal pigment epithelium (RPE)) and dynamic barriers (e.g., lymphatic flow in the conjunctiva and episclera, and the blood flow in conjunctiva and choroid). The composition can be formulated for delivery across occular drug delivery barriers and/or to improve ocular bioavailability. Compositions (e.g., a pharmaceutical composition) can contain one or more additives selected from the group consisting of a permeation enhancer, a viscosity enhancer.
Compositions can be formulated for administration by any route known to those of skill in the art. For example, compositions can be formulated as a suspension, emulsion, ointment, aqueous gel, nanomicelle, nanoparticle, liposome, dendrimer, implant, contact lens, nanosuspension, microneedle, or in situ gel (e.g., in situ thermosensitive gel). Emulstions can contain one or more additives selected from among the group consisting of a lipid additive (e.g., soyabean lecithin and/or stearylamine), a mucoadhesive polymer (e.g., chitosan and/or hydroxypropyl methyl cellulose ether), combinations thereof and the like. Administration can be local, topical or systemic. In some examples, the pharmaceutical compositions can be delivered by topical instillation (e.g., as eye drops). In some embodiments, the composition (e.g., a pharmaceutical composition) can be formulated for topical drop instillation into the lower precorneal pocket. In some embodiments, the composition (e.g., a pharmaceutical composition) can be formulated as an extended release formulation (e.g., up to about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 2 weeks, about 4 weeks, about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months or more). The composition (e.g., a pharmaceutical composition) can contain additive(s). Additive(s) can improve one or more properties selected from among drug contact time, permeation and ocular bioavailability. Additives can be selected from among the group consisting of viscosity enhancers, permeation enhancers and cyclodextrins. Exemplary viscosity enhancers include hydroxy methyl cellulose, hydroxy ethyl cellulose, sodium carboxy methyl cellulose, hydroxypropyl methyl cellulose, polyalcohol, combinations thereof and the like. Cyclodextrins can act as a carrier for hydrophobic drug molecules in aqueous solution.
Permeation enhancers can improve corneal uptake by modifying the comeal integrity. Exemplary permeation enhancers include chelating agents, preservatives, surface active agents, bile salts, and combinations thereof. In some embodiments, a permeation enhancer is selected from among the group consisting of benzalkonium chloride, polyoxyethylene glycol ethers (e.g., lauryl, stearyl and oleyl), ethylenediaminetetra acetic acid sodium salt, sodium taurocholate, saponins, cremophor EL, polycarbophil-cysteine, combinations thereof and the like.
In some embodiments, the composition can be formulated as a nanocarrier, such as a nanocarrier selected from the group consisting of nanoparticles, nanosuspensions, liposomes, nanomicelles and dendrimers. Nanomicelles can include amphiphilic molecules and can be surfactant or polymeric in nature. Nanoparticles can include lipids, proteins, natural or synthetic polymers such as albumin, sodium alginate, chitosan, poly (lactide-co-glycolide) (PLGA), polylactic acid (PLA), polycaprolactone, combinations thereof and the like. Nanoparticles can be nanocapsules or nanospheres. As nanocapsules, the HIF inhibitor can be enclosed inside a shell (e.g., a polymeric shell). As nanospheres, the HIF-inhibitor can be uniformly distributed throughout a matrix (e.g., a polymeric matrix). Nanoparticles can include a chitosan coating to improve precorneal residence. Nanosuspensions can be stabilized by polymer(s) and/or surfactant(s). Dendrimers can include terminal end amine, hydroxyl or carboxyl functional groups. Examples of dendrimers include Poly (amidoamine) (PAMAM) dendrimers.
Compositions formulated as liposomes can include small unilamellar vesicles (10- 100 nm), large unilamellar vesicles (100-300 nm) and multilamellar vesicles (contains more than one bilayer). Liposomes can include cationic liposomes or neutral liposomes.
Liposomes can be pegylated liposomes, submicron-sized, or a combination thereof. Liposomes can be multilamellar or unilamellar. Liposomes can include a mucoadhesive polymer. Cationic liposomes can include one or more selected from among the group consisting of Didodecyldimethylammonium bromide, stearylamine, and N-[l-(2,3- dioleoyloxy)propyl] -N,N,N -trimethylammonium chloride .
Compositions can be formulated as in-situ hydrogels and can undergo sol-gel phase transition to form viscoelastic gel in response to environmental stimuli (e.g., changes in temperature, pH and ions, or a combination thereof) or can be induced by UV irradiation. In some embodiments, the composition is formulated as a a thermosensitive gel. Examples of thermogelling polymers for use in a thermosensitive gel described herein include poloxamers, multiblock copolymers made of polycaprolactone, polyethylene glycol, poly (lactide), poly (glycolide), poly (N-isopropylacrylamide), chitosan and combinations thereof. For delivery, polymers can be mixed with an HIF inhibitor in the solution state and solution can be administered which forms an in situ gel depot at physiological temperature.
Thermosensitive gels can include, for example, a triblock copolymer of PFGA and PEG ((poly-(DF-lactic acid co-glycolic acid) -polyethylene glycol), or cross linked poly (N- isopropylacrylamide) (PNIPAAm)-poly (ethylene glycol) diacrylate, or the triblock polymer PFGA-PEG-PFGA (poly-(DF-lactic acid co-glycolic acid)-polyethylene glycol-poly-(DF- lactic acid co-glycolic acid) as a ocular delivery carrier for an HIF inhbitor.
In some embodiments, the pharamceutical composition containing an HIF inhibitor can be formulated for delivery with a contact lens. In presence of contact lens, the HIF inhibitor can have longer residence time in the post-lens tear fdm which can result in higher drug flux through cornea with less drug inflow into the nasolacrimal duct. The HIF inhibitor can be loaded by soaking the contact lens in a drug solution. In some embodiments, the contact lens is a particle-laden contact lenses or a molecularly imprinted contact lens. In a particle-laden contact lenses, the active agent can be entrapped in vesicles such as liposomes, nanoparticles or microemulsion and dispersed in the contact lens material.
In some embodiments, the pharmaceutical composition containing an HIF inhibitor can be formulated as an implant, such as an intraocular implant. The intraocular implant can provide localized controlled drug release over an extended period, and can circumvent multiple intraocular injections and associated complications. In some embodiments, the implant can be delivered to posterior ocular tissues. Implants can be placed intravitreally. In some embodiments, implants can be placed by making incision through minor surgery at pars plana posterior to the lens and anterior to the retina. Administration by implantation can circumvent the blood retina barrier.
The composition can be formulated as a biodegradable implant or as a non- biodegradable implant. A non-biodegradable implant can effect long-lasting release with near zero order release kinetics. Non-biodegradable implants can include a polymer selected from among the group consisting of polyvinyl alcohol (PVA), ethylene vinyl acetate (EVA), polysulfone capillary fiber (PCF), and a combination thereof, such as PV A/EVA. In some embodiments, the implant is a silicone laminated PVA implant. Implants can be surgically implanted and removed after drug depletion.
In some embodiments, the compositon is formulated as a biodegradable implants. The biodegradable implants can be formulated for sustained drug release. In some embodiments, it is not necessary to surgically remove the biodegradable implants. Biodegradable implants can include a polymer (e.g., polylactic acid (PLA), polyglycolic acid (PGA), poly [d,l-lactide- co-glycolide] (PLGA) and poly[d,l-lactide-co-caprolactone] (PLC), poly( L -lactide-co- caprolactone-co-glycolide) (PLGA-PCL), hydroxypropyl methylcellulose, polycaprolactones, or a combination thereof). In some embodiuments, the compostion can be formulated as an intravitreal implant. For example, an intravitreal implant can contain a PLGA polymer matrix that degrades to lactic acid and glycolic acid over an extended period, allowing prolonged release over up to about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months or longer.
In some embodiments, the composition (e.g., a pharmaceutical composition) is formulated for administration by a microneedle based technique. For example, the composition can be formulated for delivery to posterior ocular tissues. Microneedle based administration can circumvent blood retinal barrier and deliver therapeutic drug levels to retina/choroid. Microneedles can be designed to penetrate only hundreds of microns into sclera, so that damage to deeper ocular tissues can be avoided. Microneedles can deposit the HIF inhibitor into sclera or into the suprachoroidal space (SCS) between sclera and choroid.
Also provided are compositions (e.g., pharmaceutical compositions) containing a second agent that is used to treat an ischemic retinal disease or condition. Exemplary of such agents include, but are not limited to, an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, dexamethasone, triamcinolone, a corticosteroid, a steroid, hydroxycarbamide, a blood thinner, warfarin, apixaban, dabigatran, edoxaban, fondaparinux, heparin, rivaroxaban, combinations thereof and the like. In some exemplary embodiments, the second agent is a VEGFR inhibitor (e.g., selected from the group consisting of cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, combinations thereof, and the like). HIF inhibitors can be co-formulated or co-administered with pharmaceutical formulations of such second agents. The HIF inhibitors and second agent can be packaged as separate compositions for administration together, sequentially or intermittently. The combinations can be packaged as a kit. Compositions also can be administered with other biologically active agents, either sequentially, intermittently or in the same composition.
The pH and the osmolarity of the compositions can be adjusted by one of skill in the art to optimize the conditions for the desired activity and stability of the composition. In some examples, the compositions provided herein have an osmolarity of at or about 100 mOsm/kg, about 120 mOsm/kg, about 140 mOsm/kg, about 160 mOsm/kg, about 180 mOsm/kg, about 200 mOsm/kg, about 220 mOsm/kg, about 240 mOsm/kg, about 260 mOsm/kg, about 280 mOsm/kg, about 300 mOsm/kg, about 320 mOsm/kg, about 340 mOsm/kg, about 360 mOsm/kg, about 380 mOsm/kg, about 400 mOsm/kg, about 420 mOsm/kg, about 440 mOsm/kg, about 460 mOsm/kg, about 500 mOsm/kg or more. In some embodiments, the pH of the composition is at or about 4, about 5, about 6, about 7, about 7.2, about 7.4, about 7.6, about 7.8 or about 8. In some embodiments, the pH of the compositions is about 7.4. In some embodiments, the pH of the compositions ranges from about 4 to about 5, ranges from about 5 to about 6, ranges from about 6 to about 7, ranges from about 7 to about 8, ranges from about 8 to about 9, or ranges from about 9 to about 10.
If drug absorption across the cornea is desired, the HIF inhibitor can exhibit differential solubility (e.g., ionised and non-ionised forms can coexist). The outer layer of the cornea (the epithelium) is lipid-rich. The inner layer of the cornea (the stroma) is predominantly aqueous. Therefore, ionisation of a drug can increase partitioning into this phase. The pH of the formulation can be adjusted to decrease the ionisation of the therapeutic agent.
In one embodiment, the HIF inhibitor, can be administered as part of a combination therapy, by administering the HIF inhibitor and a second agent or treatment described herein, such as for treating a disease or condition selected from the group consisting of retinal detachment, an ischemic retinal disease or condition, a combination thereof, and the like. In one embodiment, the HIF inhibitor and second agent or treatment can be co-formulated and administered together. In another embodiment, the HIF inhibitor, is administered subsequently, intermittently or simultaneously with the second agent or treatment. The HIF inhibitor can be administered prior to, with, or after administration of the second agent or treatment. In some embodiments, the HIF inhibitor is administered together with the second agent or treatment.
In some examples of the methods, uses, compositions, and combinations provided herein, the HIF inhibitor is one that increases cellular survival. In some examples, the HIF inhibitor is one that increases cellular survival in an in vitro assay. In some embodiments, the HIF inhibitor can be one that increases cellular survival in response to oxidative stress. In some embodiments, oxidative stress is stimulated by treatment with hydroquinone. In some embodiments, cells are exposed to hydroquinone at a concentration of 10 mM or about 10 mM, 50 pM or about 50 pM, 100 pM or about 100 pM, 125 pM or about 125 pM, 150 pM or about 150 pM, or 200 pM or about 200 pM, 10 pM or about 10 pM to 50 pM or about 50 pM, 50 pM or about 50 pM to 100 pM or about 100 pM, 100 pM or about 100 pM to 125 pM or about 125 pM, 125 pM or about 125 pM to 150 pM or about 150 pM, or 150 pM or about 150 pM to 200 pM or about 200 pM hydroquinone. In some embodiments, cellular survival is assessed in an in vitro assay. In some examples of the methods, uses, compositions, and combinations provided herein, the HIF inhibitor increases cellular survival in an in vitro model of AMD. In some embodiments, the in vitro assay is a cellular in vitro assay. In some embodiments, the cells in the cellular in vitro assay are RPE cells. In some embodiments, the cells are ARPE-19 cells. In some embodiments, cellular survival is assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and/or by measuring metabolic activity of cells. In some embodiments, metabolic activity of cells is assessed by MTT ((3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide)) assay.
In some embodiments, the HIF inhibitor can increase cellular survival by pretreatment of cells with the HIF inhibitor. In some embodiments, the pretreatment is before stimulation of oxidative stress in cells. In some embodiments, pretreatment occurs 1 hour or about 1 hour, 2 hours or about 2 hours, 4 hours or about 4 hours, 6 hours or about 6 hours, 12 hours or about 12 hours, 24 hours or about 24 hours, 48 hours or about 48 hours, 72 hours or about 72 hours, 1 to 6 hours, about 1 to about 6 hours, 6 to 12 hours, about 6 to about 12 hours, 12 to 24 hours, about 12 to about 24 hours, 24 to 48 hours, about 24 to about 48 hours, 48 to 72 hours, or about 48 to about 72 hours before stimulation of oxidative stress.
In some embodiments, the HIF inhibitor can increase cellular survival. In some embodiments, the HIF inhibitor can increase cellular survival in an in vitro assay in which cells are exposed to oxidative stress. In some embodiments, the HIF inhibitor can increase cellular survival by at least 5% or at least about 5%, at least 10% or at least about 10%, at least 15% or at least about 15%, at least 20% or at least about 20%, at least 25% or at least about 25%, at least 30% or at least about 30%, at least 35% or at least about 35%, at least 40% or at least about 40%, at least 50% or at least about 50%, at least 60% or at least about 60%, at least 70% or at least about 70%, at least 80% or at least about 80%, at least 90% or at least about 90%.
In some embodiments, the HIF inhibitor increases cellular survival at a concentration of 500 nmol or about 500 nmol, ImM or about ImM, 5mM or about 5mM, 10mM or about 10mM, 20mM or about 20mM, 30mM or about 30mM, 50mM or about 50mM, 100mM or about 100mM, or 200mM or about 200mM. In some embodiments, the HIF inhibitor increases cellular survival at a concentration of 500 nmol to ImM, about 500 nmol to about ImM, ImM to 5mM, about ImM to about 5mM, 5mM to 10mM, about 5mM to about 10mM, 10mM to 20mM, about 10mM to about 20mM, 20mM to 50mM, about 20mM to about 50mM, 50mM to 100mM, about 50mM to about 100mM, 100mM to 200mM, or about 100mM to about 200mM. In some embodiments, cellular survival is assessed in an in vitro assay as described herein.
In some embodiments, the in vitro assay comprises pretreatment of cells with the HIF inhibitor prior to stimulative oxidative stress in the cells as described herein. In some embodiments, the cells are RPE cells. In some embodiments, the cells are ARPE-19 cells. In some embodiments, cellular survival is assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and/or by measuring metabolic activity of cells. In some embodiments, metabolic activity of cells is assessed by MTT ((3-(4,5-dimethylthiazol-2-yl)- 2,5 -diphenyl -2H-tetrazolium bromide)) assay.
Also provided, are articles of manufacture containing packaging materials, a pharmaceutical composition that is effective for treating, minimizing and/or substantially inhibiting an ischemic retinal disease or condition, and a label that indicates that the composition is to be used for treating, minimizing and/or substantially inhibiting a ischemic retinal disease or condition. In one example, the pharmaceutical composition contains the HIF inhibitor, and no second agent or treatment. In another example, the article of manufacture contains the HIF inhibitor and a second agent or agents or treatment or treatments. In this example, the pharmaceutical compositions of a second agent and an HIF inhibitor, can be provided together or separately, for packaging as articles of manufacture.
EXEMPLARY EMBODIMENTS
EXAMPLE 1: HIF inhibition increased survival of ARPE-19 cells under conditions of oxidative stress.
The HIF-Ia inhibitor PX-478, which is currently undergoing clinical testing for cancer (Shirai et al. (2021) Cancers (Basel) 13(11):2813), is amelphalan derivative that lowers HIF-Ia levels by affecting multiple levels in the HIF-Ia pathway: it can inhibit HIF- 1 a deubiquitination, reduce HIF-Ia mRNA expression, and inhibit HIF-Ia translation. PX- 478 was reported to show prominent selectivity towards inhibition of HIF-Ia (Masoud et al. (2015) Acta Pharm Sin B. 5(5):378-89; Koh et al. (2008) Mol Cancer Ther. 7:90-100).
This HIF inhibitor’s ability to prevent RPE cell death was analyzed in an in vitro model of AMD. Immortalized human RPE cells of the cell line ARPE-19 were cultured under routine conditions until confluent. HIF- la was inhibited using a range of PX-478 concentrations for 48 hours. The toxicity of PX-478 was assessed by determining the leakage of intracellular lactate dehydrogenase (LDH) and by measuring metabolic activity of cells using the MTT assay. PX-478 was well tolerated by ARPE-19 cells up to a concentration of 10 mM (FIGS. 3 A and 3B).
Oxidative stress and cell death were also stimulated in ARPE-19 cells, using the cigarette smoke component hydroquinone as a model of oxidative stress in RPE cells (Bhattarai et al. (2020) Int. J. Mol. Sci., 21(6):2066; Yang et al. (2020) Invest Ophthalmol Vis Sci., 61(10):35; and Pons et al., (2011) PLoS One., 6(2):el6722). A 24h pretreatment with 5 mM or 10 pM PX-478 was cytoprotective, both in conditions of mild (FIGS. 4A and 4B) and significant (FIGS 4C and 4D) cell death induced by hydroquinone. As measured by the MTT assay, pretreatment with 10 pM PX-478 increased cellular survival by 24.5% and 30.4% compared to cells exposed only to 100 pM or 125 pM hydroquinone, respectively (FIGS. 4A and 4C). Microscopic observations confirmed that PX-478 pretreatment helped to preserve a healthy cellular phenotype.
HIF inhibition prevented RPE cell death under conditions of increased oxidative stress and after exposure to a compound of cigarette smoke, one of the biggest environmental risk factors for AMD development. Preventing RPE cell death and regulating HIF signaling could preserve retinal homeostasis and, thereby, patients’ visual acuity. As such, HIF inhibition could be used to treat, for example, AMD and/or atrophy associated with retinal hypoxia.
Materials and Methods
Cell Culture Conditions and Compounds
ARPE-19 cells were obtained from the American Type Culture Collection (ATCC, Mannassas, VA, USA) and used for experiments between passage numbers 25 and 30. Cells were routinely maintained in DMEM/F-12 (1:1) medium (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% HyClone fetal bovine serum (FBS; Thermo Fisher Scientific), 100 U/ml penicillin, 100 pg/ml streptomycin and 2 mM L-glutamine (all Lonza, Basel, Switzerland). For routine maintenance and experiments, cells were kept at 37°C in an incubator providing a humidified atmosphere enriched with 5% CO2. Cells were detached for passaging or plating for experiments using 0.25% Trypsin-EDTA (Thermo Fisher Scientific). For experiments, cells were plated on 96-well plates at a density of 15,000 cells/well and incubated for 72 h until completely confluent. The medium was changed to serum-free culture medium and cells were incubated with selected concentrations of PX-478 (MedChemExpress, Monmouth Junction, NJ, USA) for 48h. After 24h, hydroquinone (HQ, Merck KGaA, Darmstadt, Germany) was added and cells were incubated for the remaining 24h before sample collection and analysis of cell viability.
Cell Viability Assays
A 3-(4,5-dimethyldiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Merck KGaA) assay was used to assess cellular viability. Briefly, medium samples were removed from wells and replaced with MTT salt at a final concentration of 0.5 mg/ml in serum-free culture medium. Cells were incubated under absence of light for 90 min at 37°C, after which the MTT-containing medium was replaced with DMSO (Merck KGaA). DMSO dissolved the formed formazan crystals during an incubation step of 20 min at room temperature. The optical density of each well was then measured at a wavelength of 562 nm and results were calculated relative to untreated control, or hydroquinone-treated positive control, which was set to 100% viability.
The lactate dehydrogenase (FDH) assay (Cytotox 96® non-radioactive cytotoxicity assay, Promega, Madison, WI, USA) was used to determine cellular toxicity. The assay determined the amount of intracellular FDH that has leaked into the medium and thereby estimated the levels of membrane rupture and cell death. The assay was performed according to the manufacturer’s instructions and results were normalized to FDH levels in untreated control, which were set to be 1.
Statistical Analysis
All experiments were performed two to three times with comparable results. Data were combined and treatment groups were probed using pairwise comparison with the Mann- Whitney //-test. Differences between groups were considered statistically significant at p < 0.05. All statistical analyses were performed using GraphPad Prism 9.0.1 (GraphPad Software Inc., San Diego, CA, USA).
PROPHETIC EXAMPLE: Efficacy study of pharmaceutical composition containing an HIF inhibitor for treatment of an ischemic retinal disease A subject having an ischemic retinal disease or condition is selected for treatment.
The subject can be identified, for example, having, or being at risk of developing retinal atrophy. The subject can also be identified because he/she has retinal/subretinal neovascularization or macular edema and needs anti VEGF treatment that can be accompanied with retinal atrophy. Examples include dry atrophic AMD in which progression of atrophy has been established. Another example is neovascular AMD in which anti VEGF treatment is planned and retinal atrophy is expected or has started.
A pharmaceutical composition containing an HIF inhibitor is administered by injection or implantation into the vitreous cavity of the subject at a dose determined by a clinician to be therapeutically effective.
The duration of treatment is a period of time sufficient to treat one or more conditions selected from among retinal apoptosis, retinal atrophy and choroidal atrophy, or to otherwise improve the clinical condition of the subject. In some embodiments treatment can last over months to years. In some embodiments, intravitreal injections can be repeated monthly or every few months. Intravitreal injections can be administered, for example, daily, weekly, monthly, every few months, or the like. Eye drops can be applied 1-6 times a day, such as, for example, once per day, twice per day, three times per day, four times per day, five times per day, six times per day, or the like. Systemic administration can be administered, for example, 1-3 times per day (e.g., once per day, twice per day, or three times per day), weekly, monthly, every few months, or the like.
Improvement is determined by clinical signs or symptoms or by diagnostic tests. For example, assessments are performed to test for visual function (e.g., visual field, visual acuity, microperimetry, contrast sensitivity, color vision, combinations thereof, and the like). Imaging studies, including fundus photography and/or high speed spectral domain optical coherence tomography (SDOCT), are performed to determine reduction in atrophy.
Apoptosis and/or atrophy is assessed by a suitable morphological, functional, electric or metabolic method, or a combination thereof. For example, suitable methods include a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, annexin 5 staining, a combination thereof, and the like. Apoptotic cells are identified before administration to establish a baseline, and at a time after administration determined by a clinician to result in an a therapeutic improvement (e.g., 5 minutes, 30 minutes, 60 minutes, 120 minutes, 7 days, or 30 days, or any combination thereof). For example, fluorescently-labelled annexin 5 is intravenously administered to the subjects at a dose of 0.1-0.5 mg. Retinal imaging is performed to visualize fluorescent cells, which are quantified. Images are acquired with a confocal scanning laser ophthalmoscope (diode laser 786 nm excitation; photodetector with 800 nm barrier filter), after pupillary dilatation (1% tropicamide and 2.5% phenylephrine) .
Although the exemplary embodiments of the present disclosure have been described in detail with reference to the accompanying examples and drawings, the present disclosure is not limited thereto and can be embodied in many different forms without departing from the technical concept of the present disclosure. Therefore, the exemplary embodiments of the present disclosure are provided for illustrative purposes only and are not intended to limit the technical concept of the present disclosure. The protective scope of the present disclosure should be construed based on any appended claims and combinations thereof, and all the technical concepts in the equivalent scope thereof should be construed as falling within the scope of the present disclosure. As various changes could be made in the above methods and compositions without departing from the scope of the invention, it is intended that all matter contained in the above description shall be interpreted as illustrative and not in a limiting sense. Other embodiments within the scope of the claims herein will be apparent to one skilled in the art from consideration of the specification or practice of the exemplary embodiments disclosed herein. It is intended that the specification be considered exemplary only, with the scope and spirit of the described subject matter being indicated by the claims.

Claims

CLAIMS What is claimed is:
1. A method of treating, minimizing and/or inhibiting atrophy associated with retinal hypoxia, the method comprising administering a pharmaceutical composition comprising an HIF inhibitor to a subject having retinal hypoxia, wherein: the HIF inhibitor is administered in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia; and the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
2 The method of claim 1, wherein administration of the pharmaceutical composition containing an HIF inhibitor effects treatment of the atrophy associated with retinal hypoxia.
3. The method of claim 1 or claim 2, wherein treatment of the atrophy associated with retinal hypoxia comprises preventing, minimizing, slowing, alleviating and/or substantially inhibiting the atrophy.
4. The method of any one of claims 1-3, wherein treatment of the atrophy associated with retinal hypoxia comprises decreasing the severity, duration, or frequency of occurrence of the atrophy.
5. The method of any one of claims 1-4, further comprising, assessing the atrophy associated with retinal hypoxia.
6. The method of claim 5, wherein the assessing the atrophy associated with retinal hypoxia comprises a method selected from the group consisting of spectral-domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, and combinations thereof.
7. The method of claim 5 or claim 6, wherein the assessing the atrophy associated with retinal hypoxia occurs before administering the pharmaceutical composition comprising the HIF inhibitor.
8. The method of any one of claims 5-7, wherein the assessing the atrophy associated with the retinal hypoxia occurs after administering the pharmaceutical composition comprising the HIF inhibitor.
9. The method of any one of claims 5-8, wherein the atrophy associated with the retinal hypoxia is assessed before and after administering the HIF inhibitor.
10. The method of any one of claims 1-9, wherein the method comprises a reduction in retinal apoptosis associated with retinal hypoxia in the subject.
11. The method of claim 10, wherein the method comprises a reduction in the retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
12. The method of claim 10 or claim 11, wherein the method comprises a reduction in the retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 100%.
13. The method of any one of claims 1-12, comprising assessing retinal apoptosis by a method selected from among the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, and a combination thereof.
14. The method of claim 13, wherein the measuring phosphatidyl extemalization comprises annexin 5 staining.
15. The method of any one of claims 1-14, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of macular atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy, glaucomatous atrophy, ganglion cell atrophy, and combinations thereof.
16. The method of any one of claims 1-15, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), and dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor.
17. The method of any one of claims 1-16, wherein the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
18. The method of any one of claims 1-17, wherein the method comprises a reduction in a total area of the atrophy associated with retinal hypoxia.
19. The method of any one of claims 1-18, wherein the area of the atrophy associated with retinal hypoxia is reduced by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 100%.
20. The method of any one of claims 1-19, wherein the area of the atrophy associated with retinal hypoxia is assessed by a method selected from the group consisting of spectral- domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, biomicroscopy, multifocal electroretinography and combinations thereof.
21. The method of any one of claims 1-20, wherein the area of atrophy associated with retinal hypoxia is assessed by a method selected from among the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, and combinations thereof.
22. The method of any one of claims 1-21, wherein the method comprises a reduction in a severity grade of the atrophy associated with retinal hypoxia.
23. The method of any one of claims 1-22, wherein the method comprises an improvement in the Age-Related Eye Disease Study (AREDS) scale.
24. The method of any one of claims 1-23, wherein the subject is one who is being treated with an angiogenesis inhibitor.
25. The method of claim 24, wherein the angiogenesis inhibitor is a VEGF inhibitor and/or a VEGFR inhibitor.
26. The method of claim 24 or claim 25, wherein the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, and combinations thereof.
27. The method of claim 24 or claim 25, wherein the angiogenesis inhibitor is selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, and combinations thereof.
28. The method of any one of claims 1-27, wherein the retinal hypoxia is from a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof.
29. The method of any one of claims 1-28, wherein the retinal hypoxia is from retinal detachment.
30. The method of claim 29, wherein the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, serous retinal detachment, rhegmatogenous retinal detachment, tractional retinal detachment, proliferative vitreoretinopathy (PVR), and central serous chorioretinopathy .
31. The method of any one of claims 1-28, wherein the retinal hypoxia is from an ischemic retinal disease.
32. The method of claim 31, wherein administration of the pharmaceutical composition containing an HIF inhibitor effects treatment, minimizing and/or substantial inhibition of a symptom associated with the ischemic retinal disease.
33. The method of claim 32, wherein the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion and combinations thereof.
34. The method of any one of claims 32 and 33, wherein the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
35. The method of any one of claims 31-34, wherein the ischemic retinal disease is selected from the group consisting of age related macular degeneration, (dry atrophic AMD, geographic atrophy, diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy, retinal pigment epithelial detachment, central serous chorioretinopathy, and combinations thereof.
36. The method of any one of claims 31-35, wherein the ischemic retinal disease is diabetic macular edema.
37. The method of any one of claims 31-35, wherein the ischemic retinal disease is non-proliferative diabetic retinopathy.
38. The method of any one of claims 31-37, wherein the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, severe non-proliferative diabetic retinopathy, and traction retinal detachment in diabetic retinopathy.
39. The method of any one of claims 31-35, wherein the ischemic retinal disease is proliferative diabetic retinopathy.
40. The method of any one of claims 31-35, wherein the ischemic retinal disease is central retinal vein occlusion.
41. The method of any one of claims 31-35, wherein the ischemic retinal disease is branch retinal vein occlusion.
42. The method of any one of claims 31-35, wherein the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
43. The method of any one of claims 31-35, wherein the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
44. The method of any one of claims 1-43, wherein, administration of the pharmaceutical composition effects a decrease in expression of an HIF target gene or locus.
45. The method of claim 44, wherein the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI, TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG-VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, plasminogen-activator inhibitor- 1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2, a hexokinase activator, lactate dehydrogenase A/FDHA, a lactate dehydrogenase A/FDHA inhibitor, lactate dehydrogenase B/FDHB, iNOS, perilipin-2, PGK1, PKM2, cathepsin D, CCF2/JE/MCP-1, CTGF/CCN2, CXCR4, HGFR/c-MET, IF-6, IF-8/CXCF8, integrin alpha 5/CD49e, FOX-1/OFR1, FOXF1, lysyl oxidase homolog 2/FOXF2, MKP-1, MMP-1, MMP- 2, osteopontin/OPN, pref-l/DFKl/FAl, SNAI1, TCF-3/E2A, TRKB, TWIST-1, uPAR, ZEB1, KFF4, NANOG, OCT-3/4, OCT-4A, OCT-4B, and SOX2, Adrenomedullin/ADM, Cyclin Dl, Erythropoietin/EPO, IGF-II/IGF2, IGFBP-1, IGFBP-2, IGFBP-3, NOTCH 1, Survivin, TGF-a, keratin 14, keratin 18, keratin 19, vimentin, CXCR4, c-Met, autocrine motility factor (AMF/GPI), LDL receptor related protein 1 (LRPl), Transforming growth factor-a (TGF-a), Transforming growth factor^ (TGF- 3), Insulin-like growth factor 2 (IGF -2), IGF binding protein 1, 2 and 3 (IGF -BP), WAF1, Cyclin G2), Endothelin 1 (ET1), Adrenomedullin (ADM), Tyrosine hydroxylase, alB-adrenergic receptor, Inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), heme oxygenase- 1, atrial natriuretic peptide, insulin-like growth factor binding protein-1, NIP3, NIX, RTP801, Endoglin (ENG), Wilms' tumour suppressor, a-Fetoprotein, and Calcitonin-receptor like receptor), Erythropoetin (EPO), Leptin (LEP)), Glucose transporter 1 (GLUT1), Hexokinase 1 and 2 (HK1 and 2), 6-Phosphofructo-l -kinase L (PFKL), 6-Phosphofructo-2 -kinase, Glyceraldehyde-3-P dehydrogenase (GAPDH), Aldolase A (ALDA), Aldolase C (ALDC), Enolase 1 (ENOl), Phosphoglycerate kinase-1 (PGK1), Lactate dehydroxygenase A (LDHA), Pyruvate kinase M (PKM), Carbonic anhydrase 9 (CA9), Adenylate kinase 3, and Transglutaminase 2), Pro-collagen prolyl hydroxylase al, Collagen type V (al), Intestinal trefoil factor (TFF), Ecto-5 '-nucleotidase, Cathepsin D (CATHD), Fibronectin 1 (FN1), Matrix metalloproteinase 2 (MMP2)), DEC1, DEC2, ETS-1, CITED2/p35sq, and NUR77), Transferrin, Transferrin receptor, Ceruloplasmin, Multidrug resistance P-glycoprotein, halofuginone, a retrotransposon, retrotransposon VL30, and combinations thereof.
46. The method of any one of claims 1-45, wherein expression of a vascular endothelial growth factor (VEGF) is reduced in an eye of the subject.
47. The method of any one of claims 1-46, wherein activity of a vascular endothelial growth factor (VEGF) is reduced in an eye of the subject.
48. The method of any one of claims 1-47, wherein expression of a vascular endothelial growth factor receptor (VEGFR) is reduced in an eye of the subject.
49. The method of any one of claims 1-48, wherein activity of a vascular endothelial growth factor receptor (VEGFR) is reduced in an eye of the subject.
50. The method of any one of claims 1-49, wherein the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v.
51. The method of any one of claims 1-50, wherein the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to about 0.1%, about 0.1% to about 0.5%, about 0.5% to about 1%, about l%to about 1.5%, about 1.5% to about 2%, about 2% to about 2.5%, about 3% to about 4%, about 4% to about 5%, about 5% to about 6%, about 6% to about 7%, or about 7% to about 8%.
52. The method of any one of claims 1-51, wherein the HIF inhibitor is administered at a dose of or about 0.001 mg, about 0.002 mg, about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg, about 4.5 mg, about 5 mg, about 10 mg or more.
53. The method of any one of claims 1-52, wherein the HIF inhibitor is administered at a dose of or about 0.001 mg to about 0.01 mg, about 0.01 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
54. The method of any one of claims 1-53, wherein administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the retina of the subject.
55. The method of any one of claims 1-54, wherein administering the pharmaceutical composition comprises delivery of the HIF inhibitor to the choroid, or to the suprachoroidal space, of the subject.
56. The method of any one of claims 1-55, wherein the HIF inhibitor is selected from among the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, and a combination thereof.
57. The method of any one of claims 1-56, wherein the HIF inhibitor is an HIF-1 inhibitor.
58. The method of any one of claims 1-57, wherein the HIF inhibitor is an HIF-la inhibitor.
59. The method of any one of claims 1-58, wherein the HIF inhibitor inhibits HIF by one or more pathways selected from the group consisting of inhibiting HIF-Ia mRNA expression, inhibiting HIF-la translation, and inhibiting deubiquitination.
60. The method of any one of claims 1-59, wherein the HIF inhibitor is an HIF-2 inhibitor.
61. The method of any one of claims 1-60, wherein the HIF inhibitor is an HIF-1 inhibitor and an HIF-2 inhibitor.
62. The method of any one of claims 1-61, wherein the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3- [[(lS,2S,3R)-2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4-yl]oxy]-5- fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro- lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)- camptothecin (NSC-639174), a cardenolide, EZN-2208 (PEG-SN38), SN38 (7-Ethyl-10- hydroxy-camptothecin), a Ca2+ channel blocker, NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH-benzimidazol-2-yl)propyl]methylamino]ethyl]-6-fluoro-l,2,3,4- tetrahydro-l-(l-methylethyl)-2-naphthalenyl ester, dihydrochloride, PX-478 (,S'-2-amino-3- [4'-N,N,-bis(chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride), an inhibitor of the PI3K/Akt/TOR pathway, an inhibitor of the MAPK pathway, resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, sorafenib, LY294002, wortmannin, nelfmavir, aHSP90 inhibitor, a glyceollin, IDF-11774 (2-(4-((3r,5r,7r)-adamantan-l-yl)phenoxy)-l-(4- methylpiperazin-l-yl)ethan-l-one), a histone deacetylase (HDAC) inhibitor, panobinostat (LBH589, (E)-N-hydroxy-3-[4-[[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop- 2-enamide), the indole-3-ethylsulfamoylphenylacrylamide compound MPT0G157, a diazepinquinazolin-amine derivate, BIX01294 (N-(l-benzylpiperidin-4-yl)-6,7-dimethoxy-2- (4-methyl-l,4-diazepan-l-yl)quinazolin-4-amine), a benzopyranyl 1,2,3-triazole, 4-(4- methoxyphenyl)- 1 -((2-methyl-6-nitro-2H-chromen-2-yl)methyl)- 1H- 1 ,2,3-triazole, Kresoxim-methyl, an analog of Kresoxim -methyl, a nanoparticle or nanoparticle conjugate, camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer, CRLX-101, PT2399, 0X3 (N-(3-Chloro-5-fluorophenyl)-4-nitrobenzo[c][l,2,5]oxadiazol-5- amine), acriflavine (ACF), a CBP inhibitor, a p300 inhibitor, CG13250, CCS1477 ((,Sj- 1 - (3,4-Difluorophenyl)-6-(5-(3,5-dimethylisoxazol-4-yl)-l-((lr,4S)-4-methoxycyclohexyl)-lH- benzo[d]imidazol-2-yl)piperidin-2-one), bortezomib ([(lR)-3-methyl-l-[[(2S)-3-phenyl-2- (pyrazine-2-carbonylamino)propanoyl] amino] butyl] boronic acid), chetomin, Erotinib, Gefitinib, Genistein, apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, pleurotin, aminoflavone, belinostat, CG1350, chidamide, cyclo-CLLFVY, digoxin, EZN-2968, glyceollins, IDF-1174, MPTOG1S7, NNC55-0396, romidepsin (Istodax/FK228), siRNA, tetrathiomolybdate, vorinostat (suberanilohydroxamic acid), and combinations thereof.
63. The method of any one of claims 1-62, wherein the HIF inhibitor is PX-478 (,S'-2- amino-3-[4'-N,N,-bis(chloroethyl)amino]phenyl propionic acid N-oxide dihydrochloride).
64. The method of any one of claims 1-62, wherein the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK-6482, 3-[[(lS,2S,3R)-2,3- difluoro- 1 -hydroxy-7 -methylsulfonyl-2,3 -dihydro- lH-inden-4-yl]oxy] -5 -fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)- 5 -fluorobenzonitrile), and combinations thereof.
65. The method of any one of claims 1-64, wherein the administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition.
66. The method of any one of claims 1-65, wherein administering the pharmaceutical composition comprises administration into the vitreous cavity of the eye.
67. The method of any one of claims 1-66, wherein the administering the pharmaceutical composition comprises injecting or implanting the pharmaceutical composition into the vitreous cavity of an eye of the subject.
68. The method of any one of claims 1-67, wherein the administering the pharmaceutical composition comprises injecting the pharmaceutical composition.
69. The method of any one of claims 1-68, wherein administering the pharmaceutical composition comprises intravitreal injection.
70. The method of any one of claims 1-67, wherein the administering the pharmaceutical composition comprises implanting the pharmaceutical composition.
71. The method of any one of claims 1-67 and 70, wherein administering the pharmaceutical composition comprises implanting the pharmaceutical composition into the vitreous cavity.
72. The method of any one of claims 1-64, wherein administering the pharmaceutical composition comprises administration selected from the group consisting of intravitreal injection, intravitreal implant, administering an eye drop, suprachoroidal injection, oral administration, parenteral injection, and combinations thereof.
73. The method of any one of claims 1-64, wherein administering the pharmaceutical composition comprises topical administration of an eye drop.
74. The method of claim 73, wherein administering the pharmaceutical composition comprises delivery to the retina.
75. The method of any one of claims 1-64, wherein administering the pharmaceutical composition comprises administration to the suprachoroidal space.
76. The method of any one of claims 1-75, wherein the administering the pharmaceutical composition comprises repeated administration of the pharmaceutical composition.
77. The method of any one of claims 1-76, wherein the administering the pharmaceutical composition comprises administration of the pharmaceutical composition hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly or every few months.
78. The method of any one of claims 1-77, further comprising administering a second therapeutic agent or treatment to the subject for treatment of an ischemic retinal disease.
79. The method of claim 78, wherein the pharmaceutical composition is administered before, after or with the second therapeutic agent or treatment.
80. The method of claim 78 or claim 79, wherein the second therapeutic agent is an angiogenesis inhibitor.
81. The method of any one of claims 78-80, wherein the second therapeutic agent is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, and combinations thereof.
82. The method of any one of claims 78-81, wherein the second therapeutic agent is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, and combinations thereof.
83. The method of any one of claims 78-81, wherein the second therapeutic agent is selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, and combinations thereof.
84. The method of any one of claims 78-80, wherein the second therapeutic agent or treatment is selected from among the group consisting of a corticosteroid, dexamethasone, triamcinolone, and combinations thereof.
85. The method of any one of claims 78-84, wherein the second therapeutic agent is formulated in a second pharmaceutical composition.
86. The method of any one of claims 78 and 79, wherein the second therapeutic treatment is selected from the group consisting of laser photocoagulation, macular laser photocoagulation, panretinal photocoagulation (scatter photocoagulation), laser photocoagulation for retinal tears, oxygen therapy, hyperbaric oxygen therapy, carotid surgery, and combinations thereof.
87. The method of any one of claims 78-86, wherein the pharmaceutical composition and the second therapeutic agent are administered as a single composition or as two compositions.
88. A pharmaceutical composition, comprising an HIF inhibitor in an amount effective to treat, minimize and/or inhibit atrophy associated with retinal hypoxia, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, choroidal atrophy, and combinations thereof.
89. The pharmaceutical composition of claim 88, wherein the amount of the HIF inhibitor is effective to treat, minimize and/or inhibit retinal apoptosis associated with retinal hypoxia in the subject.
90. The pharmaceutical composition of claim 89, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
91. The pharmaceutical composition of any one of claims 89-90, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 100%.
92. The pharmaceutical composition of any one of claims 89-91, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the retinal apoptosis as determined by a method selected from the group consisting of retinal photography, OCT imaging, retinal fluorescein angiography, electroretinopathy (ERG) techniques, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, measuring phosphatidyl extemalization, and a combination thereof.
93. The pharmaceutical composition of claim 92, wherein the measuring phosphatidyl extemalization comprises annexin 5 staining.
94. The pharmaceutical composition of any one of claims 88-93, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of retinal atrophy, macular atrophy, and choroidal atrophy.
95. The pharmaceutical composition of claim 94, wherein the retinal atrophy is selected from the group consisting of macular atrophy, iris atrophy, ciliary body atrophy, optic nerve atrophy, glaucomatous atrophy, ganglion cell atrophy, and combinations thereof.
96. The pharmaceutical composition of any one of claims 88-95, wherein the atrophy associated with retinal hypoxia is selected from the group consisting of dry retinal atrophy in AMD (geographic atrophy), dry AMD (early dry stage), dry AMD (intermediate dry stage), dry (nonexudative) AMD (advanced atrophic without subfoveal involvement), dry (nonexudative) AMD (advanced atrophic with subfoveal involvement), macular atrophy in macular ischemia in diabetic retinopathy, macular ischemia and atrophy in retinal vein occlusion, retinal atrophy (thinning) in retinal detachment, and retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor.
97. The pharmaceutical composition of any one of claims 88-96, wherein the atrophy associated with retinal hypoxia is retinal or macular atrophy associated with administration of a VEGF or VEGFR inhibitor for treatment of a disease or condition selected from the group consisting of neovascular AMD, diabetic macular edema, and proliferative diabetic retinopathy.
98. The pharmaceutical composition of any one of claims 88-97, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the total area of the atrophy associated with retinal hypoxia.
99. The pharmaceutical composition of any one of claims 88-98, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of the atrophy associated with retinal hypoxia by about 1% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, about 50% to about 60%, about 60% to about 70%, about 70% to about 80%, about 80% to about 90%, or about 100%.
100. The pharmaceutical composition of any one of claims 88-99, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce the area of the atrophy associated with retinal hypoxia by about 1% or more, about 2% or more, about 3% or more, about 4% or more, about 5% or more, about 6% or more, about 7% or more, about 8% or more, about 9% or more, about 10% or more, about 12% or more, about 15% or more, about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 100%.
101. The pharmaceutical composition of any one of claims 88-100, wherein the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the total area of atrophy as assessed by a method selected from the group consisting of spectral- domain optical coherence tomography (OCT), near-infrared reflectance, fundus photography, visual acuity testing, microperimetry, visual field testing, biomicroscopy, and combinations thereof.
102. The pharmaceutical composition of any one of claims 88-101, wherein the pharmaceutical composition comprises the HIF inhibitor in an amount effective to reduce the area of atrophy associated with retinal hypoxia as assessed by a method selected from the group consisting of retinal photography, retinal fluorescein angiography, electroretinopathy (ERG) techniques, visual acuity testing, near-infrared reflectance, fundus photography, biomicroscopy, multifocal retinal electroretinopathy (ERG), retinal perimetry (visual field analysis), microperimetry, retinal oximetry, and combinations thereof.
103. The pharmaceutical composition of any one of claims 88-102, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to reduce and/or substantially maintain a severity grade of the atrophy associated with retinal hypoxia.
104. The pharmaceutical composition of any one of claims 88-103, wherein the pharmaceutical composition comprises an amount of the HIF inhibitor effective to improve the Age-Related Eye Disease Study (AREDS) scale.
105. The pharmaceutical composition of any one of claims 88-104, wherein the amount of the HIF inhibitor is effective to treat, minimize and/or inhibit the atrophy associated with retinal hypoxia in a subject being treated with an angiogenesis inhibitor.
106. The pharmaceutical composition of claim 105, wherein the angiogenesis inhibitor is a VEGF inhibitor and/or a VEGFR inhibitor.
107. The pharmaceutical composition of claim 105 or claim 106, wherein the angiogenesis inhibitor is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, and combinations thereof.
108. The pharmaceutical composition of claim 105 or claim 106, wherein the angiogenesis inhibitor is selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, and combinations thereof.
109. The pharmaceutical composition of any one of claims 88-108, wherein the retinal hypoxia is associated with a disease or condition selected from the group consisting of retinal ischemia, retinal detachment, proliferative vitreoretinopathy, and combinations thereof.
110. The pharmaceutical composition of any one of claims 88-109, wherein the retinal hypoxia is associated with retinal detachment.
111. The pharmaceutical composition of claim 110, wherein the retinal detachment is selected from the group consisting of grade A proliferative vitreoretinopathy, grade B proliferative vitreoretinopathy, grade C P proliferative vitreoretinopathy, grade C A proliferative vitreoretinopathy, serous retinal detachment, rhegmatogenous retinal detachment, tractional retinal detachment, proliferative vitreoretinopathy (PVR), and central serous chorioretinopathy.
112. The pharmaceutical composition of any one of claims 88-109, wherein the retinal hypoxia is associated with an ischemic retinal disease.
113. The pharmaceutical composition of claim 112, wherein the amount of the HIF inhibitor is effective to treat, minimize and/or substantially inhibit a symptom associated with the ischemic retinal disease.
114. The pharmaceutical composition of claim 113, wherein the symptom associated with the ischemic retinal disease is selected from the group consisting of retinal detachment, glaucoma, optic nerve damage, vision impairment, blindness, macular edema, macular ischemia, angiogenesis, retinal neovascularization, choroidal neovascularization, iris neovascularization, vision loss, vitreous hemorrhage, subretinal haemorrhage, retinal hemorrhages, retinal venous congestion or occlusion and combinations thereof.
115. The pharmaceutical composition of any one of claims 113-114, wherein the symptom associated with the ischemic retinal disease comprises macular edema and/or angiogenesis.
116. The pharmaceutical composition of any one of claims 112-115, wherein the ischemic retinal disease is selected from the group consisting of dry atrophic age related macular degeneration (dry atrophic AMD; geographic atrophy), diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity (ROP), sickle cell retinopathy, retinal pigment epithelial detachment, central serous chorioretinopathy, and combinations thereof.
117. The method of any one of claims 112-116, wherein the ischemic retinal disease is diabetic macular edema.
118. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is non-proliferative diabetic retinopathy.
119. The pharmaceutical composition of any one of claims 112-118, wherein the ischemic retinal disease is selected from the group consisting of mild non-proliferative diabetic retinopathy, moderate non-proliferative diabetic retinopathy, and severe non proliferative diabetic retinopathy.
120. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is proliferative diabetic retinopathy.
121. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is central retinal vein occlusion.
122. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is branch retinal vein occlusion.
123. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is selected from the group consisting of stage I retinopathy of prematurity, stage II retinopathy of prematurity, stage III retinopathy of prematurity, stage IV retinopathy of prematurity and stage V retinopathy of prematurity.
124. The pharmaceutical composition of any one of claims 112-116, wherein the ischemic retinal disease is selected from the group consisting of stage I sickle cell retinopathy, stage II sickle cell retinopathy, stage III sickle cell retinopathy, stage IV sickle cell retinopathy, and stage V sickle cell retinopathy.
125. The pharmaceutical composition of any one of claims 88-124, wherein the HIF inhibitor is present in an amount effective to decrease expression of an HIF target gene or locus.
126. The pharmaceutical composition of claim 125, wherein the HIF target gene or locus is selected from the group consisting of angiopoietin-1, angiopoietin-2, angiopoietin-4, angiopoietin-like protein 4/ANGPTL4, CXCL12/SDF-1, FGF-3, PDGF, P1GF, TGF-bI,
TGF- b3, VEGF, endothelial gland derived vascular endothelial growth factor (EG-VEGF), VEGFRl/Flt-1, VEGFR2/KDR/Flk-1, plasminogen-activator inhibitor-1 (PAI1), urokinase plasminogen activator receptor (UPAR)), GAPDH, glutl, glut3, hexokinase 1, hexokinase 1/2, hexokinase 2, a hexokinase activator, lactate dehydrogenase A/LDHA, a lactate dehydrogenase A/LDHA inhibitor, lactate dehydrogenase B/LDHB, iNOS, perilipin-2,
PGK1, PKM2, cathepsin D, CCL2/JE/MCP-1, CTGF/CCN2, CXCR4, HGFR/c-MET, IL-6, IL-8/CXCL8, integrin alpha 5/CD49e, LOX-1/OLR1, LOXL1, lysyl oxidase homolog 2/LOXL2, MKP-1, MMP-1, MMP-2, osteopontin/OPN, pref-l/DLKl/FAl, SNAI1, TCF- 3/E2A, TRKB, TWIST- 1, uPAR, ZEB1, KLF4, NANOG, OCT-3/4, OCT-4A, OCT-4B, and SOX2, Adrenomedullin/ADM, Cyclin Dl, Erythropoietin/EPO, IGF-II/IGF2, IGFBP-1, IGFBP-2, IGFBP-3, NOTCH1, Survivin, TGF-a, keratin 14, keratin 18, keratin 19, vimentin, CXCR4, c-Met, autocrine motility factor (AMF/GPI), LDL receptor related protein 1 (LRPl), Transforming growth factor-a (TGF-a), Transforming growth factor^3 (TGF^3), Insulin- like growth factor 2 (IGF-2), IGF binding protein 1, 2 and 3 (IGF-BP), WAF1, Cyclin G2), Endothelin 1 (ET1), Adrenomedullin (ADM), Tyrosine hydroxylase, alB-adrenergic receptor, Inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), heme oxygenase- 1, atrial natriuretic peptide, insulin-like growth factor binding protein- 1, NIP3, NIX, RTP801, Endoglin (ENG), Wilms' tumour suppressor, a-Fetoprotein, and Calcitonin-receptor like receptor), Erythropoetin (EPO), Leptin (LEP)), Glucose transporter 1 (GLUT1), Hexokinase 1 and 2 (HK1 and 2), 6-Phosphofructo-l -kinase L (PFKL), 6- Phosphofructo-2 -kinase, Glyceraldehyde-3-P dehydrogenase (GAPDH), Aldolase A (ALDA), Aldolase C (ALDC), Enolase 1 (ENOl), Phosphoglycerate kinase- 1 (PGK1), Lactate dehydroxygenase A (LDHA), Pyruvate kinase M (PKM), Carbonic anhydrase 9 (CA9), Adenylate kinase 3, and Transglutaminase 2), Pro-collagen prolyl hydroxylase al, Collagen type V (al), Intestinal trefoil factor (TFF), Ecto-5 '-nucleotidase, Cathepsin D (CATHD), Fibronectin 1 (FN1), Matrix metalloproteinase 2 (MMP2)), DEC1, DEC2, ETS-1, CITED2/p35sq, and NUR77), Transferrin, Transferrin receptor, Ceruloplasmin, Multidrug resistance P-gly coprotein, halofuginone, a retrotransposon, retrotransposon VL30, and combinations thereof.
127. The pharmaceutical composition of any one of claims 88-126, wherein the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor (VEGF) in an eye of the subject.
128. The pharmaceutical composition of any one of claims 88-127, wherein the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor (VEGF) in an eye of the subject.
129. The pharmaceutical composition of any one of claims 88-128, wherein the HIF inhibitor is present in an amount effective to decrease expression of a vascular endothelial growth factor receptor (VEGFR) in an eye of the subject.
130. The pharmaceutical composition of any one of claims 88-129, wherein the HIF inhibitor is present in an amount effective to decrease activity of a vascular endothelial growth factor receptor (VEGFR) in an eye of the subject.
131. The pharmaceutical composition of any one of claims 88-130, wherein the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001%, about 0.002%, about 0.003%, about 0.004%, about 0.005%, about 0.006%, about 0.007%, about 0.008%, about 0.009%, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, or about 8% w/w or w/v.
132. The pharmaceutical composition of any one of claims 88-131, wherein the HIF inhibitor is present in the pharmaceutical composition at a concentration of or about 0.001% to about 0.01%, about 0.01% to about 0.1%, about 0.1% to about 0.5%, about 0.5% to about 1%, about l% to about 1.5%, about 1.5% to about 2%, about 2% to about 2.5%, about 3% to about 4%, about 4% to about 5%, about 5% to about 6%, about 6% to about 7%, or about 7% to about 8%.
133. The pharmaceutical composition of any one of claims 88-132, wherein the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg, about 0.002 mg, about about 0.003 mg, about 0.004 mg, about 0.005 mg, about 0.006 mg, about 0.007 mg, about 0.008 mg, about 0.009 mg, about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.15 mg, about 0.2 mg, about 0.25 mg, about 0.3 mg, about 0.35 mg, about 0.4 mg, about 0.45 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, about 4 mg, about 4.5 mg, about 5 mg, about 10 mg or more.
134. The pharmaceutical composition of any one of claims 88-133, wherein the HIF inhibitor is formulated for administration at a dose of or about 0.001 mg to about 0.01 mg, about 0.01 mg to about 1 mg, about 1 mg to about 2 mg, about 2 mg to about 3 mg, about 3 mg to about 4 mg, about 4 mg to about 5 mg, or about 5 mg to about 10 mg.
135. The pharmaceutical composition of any one of claims 88-134, wherein the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the retina of the subject.
136. The pharmaceutical composition of any one of claims 88-135, wherein the pharmaceutical composition is formulated for delivery of the HIF inhibitor to the choroid of the subject.
137. The pharmaceutical composition of any one of claims 88-136, wherein the HIF inhibitor is selected from among the group consisting of an inhibitor of HIF mRNA transcription, an inhibitor of HIF protein expression, an inhibitor of HIF protein stabilization, an inhibitor of HIF-a/b dimerization, an inhibitor of HIF transcription complex formation, an inhibitor of HIF binding to DNA, an inhibitor of transcription of HIF target genes, an inhibitor of the HIF/von Hippel-Lindau pathway, an activator of prolyl-4-hydroxylase, a CBP inhibitor, a p300 inhibitor, a receptor tyrosine kinase inhibitor, an EGFR tyrosine kinase inhibitor, and a combination thereof.
138. The pharmaceutical composition of any one of claims 88-137, wherein the HIF inhibitor is an HIF-1 inhibitor.
139. The pharmaceutical composition of any one of claims 88-138, wherein the HIF inhibitor is an HIF- la inhibitor.
140. The pharmaceutical composition of any one of claims 88-139, wherein the HIF inhibitor inhibits HIF by one or more pathways selected from the group consisting of inhibiting HIF-la mRNA expression, inhibiting HIF-la translation, and inhibiting deubiquitination.
141. The pharmaceutical composition of any one of claims 88-140, wherein the HIF inhibitor is an HIF -2 inhibitor.
142. The pharmaceutical composition of any one of claims 88-141, wherein the HIF inhibitor is an HIF-1 inhibitor and an HIF -2 inhibitor.
143. The pharmaceutical composition of any one of claims 88-142, wherein the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan (NSC-609699), belzutifan (MK-6482, 3-[[(lS,2S,3R)-2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro- lH-inden-4-yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7- (methylsulfonyl)-2,3-dihydro-lH-inden-4-yl)oxy)-5-fluorobenzonitrile), a topoisomerase inhibitor, camptothecin or a camptothecin analog, camptothecin 20-ester(S) (NSC-606985), 9-glycineamido-20(S)-camptothecin (NSC-639174), a cardenolide, EZN-2208 (PEG-SN38), SN38 (7-Ethyl- 10-hydroxy-camptothecin), a Ca2+ channel blocker, NNC 55-0396 (cyclopropanecarboxylic acid, (lS,2S)-2-[2-[[3-(lH-benzimidazol-2- yl)propyl]methylamino] ethyl] -6-fluoro- 1 ,2,3 ,4-tetrahydro- 1 -( 1 -methylethyl)-2-naphthalenyl ester, dihydrochloride, PX-478 (S'-2-ami no-3- |4'-N.N.-bis(chloroethyl)ami no | phenyl propionic acid N-oxide dihydrochloride), an inhibitor of the PI3K/Akt/TOR pathway, an inhibitor of the MAPK pathway, resveratrol, everolimus, rapamycin, silibinin, temsirolimus, PD98059, sorafenib, LY294002, wortmannin, nelfmavir, aHSP90 inhibitor, a glyceollin, IDF- 11774 (2-(4-((3r,5r,7r)-adamantan- 1 -yl)phenoxy)- 1 -(4-methylpiperazin- 1 -yl)ethan- 1 - one), a histone deacetylase (HD AC) inhibitor, panobinostat (LBH589, (E)-N-hydroxy-3-[4- [[2-(2-methyl-lH-indol-3-yl)ethylamino]methyl]phenyl]prop-2-enamide), the indole-3- ethylsulfamoylphenylacrylamide compound MPT0G157, a diazepinquinazolin-amine derivate, BIX01294 (N-( 1 -benzylpiperidin-4-yl)-6,7-dimethoxy-2-(4-methyl- 1 ,4-diazepan- 1 - yl)quinazolin-4-amine), a benzopyranyl 1,2,3-triazole, 4-(4-methoxyphenyl)-l-((2-methyl-6- nitro-2H-chromen-2-yl)methyl)-lH-l, 2, 3-triazole, Kresoxim-methyl, an analog of Kresoxim- methyl, a nanoparticle or nanoparticle conjugate, camptothecin (CPT) conjugated to a linear, cyclodextrin-polyethylene glycol co-polymer, CRLX-101, PT2399, 0X3 (N-(3-Chloro-5- fluorophenyl)-4-nitrobenzo[c][l,2,5]oxadiazol-5-amine), acriflavine (ACF), a CBP inhibitor, ap300 inhibitor, CG13250, CCS 1477 ((<S)-l-(3,4-Difluorophenyl)-6-(5-(3,5- dimethylisoxazol-4-yl)- 1 -(( lr,4S)-4-methoxycyclohexyl)- lH-benzo[d]imidazol-2- yl)piperidin-2-one), bortezomib ([(1 R)-3 -methyl- 1 - [[(2S)-3 -phenyl-2-(pyrazine-2- carbonylamino)propanoyl]amino]butyl]boronic acid), chetomin, Erotinib, Gefitinib, Genistein, apigenin, deguelin, geldanamycin, FK228, SAHA, Trichostatin A, flavopiridol, cisplatin, doxorubicin, echinomycin, a pyrrole-imidazole polyamide, 2-methoxyestradiol (2ME2), curcumin, antimycin Al, chetomin, ECyd, YC-1, pleurotin, aminoflavone, belinostat, CG1350, chidamide, cyclo-CLLFVY, digoxin, EZN-2968, glyceollins, IDF-1174, MPTOG1S7, NNC55-0396, romidepsin (Istodax/FK228), siRNA, tetrathiomolybdate, vorinostat (suberanilohydroxamic acid), and combinations thereof.
144. The pharmaceutical composition of any one of claims 88-143, wherein the HIF inhibitor is PX-478 (.S'-2-am i no-3 -| 4'-N.N.-bis(chlorocthyl)ami no | phenyl propionic acid N- oxide dihydrochloride).
145. The pharmaceutical composition of any one of claims 88-143, wherein the HIF inhibitor is selected from the group consisting of doxorubicin, topotecan, belzutifan (MK- 6482; 3-[[(lS,2S,3R)-2,3-difluoro-l-hydroxy-7-methylsulfonyl-2,3-dihydro-lH-inden-4- yl]oxy]-5-fluorobenzonitrile), PT2385 ([S]-3((2,2-difluoro-l-hydroxy-7-(methylsulfonyl)- 2,3-dihydro-lH-inden-4-yl)oxy)-5-fluorobenzonitrile), and combinations thereof.
146. The pharmaceutical composition of any one of claims 88-145, wherein the pharmaceutical composition is formulated for administration by injection and/or implantation.
147. The pharmaceutical composition of any one of claims 88-146, wherein the pharmaceutical composition is formulated for administration into the vitreous cavity of the eye.
148. The pharmaceutical composition of any one of claims 88-147, wherein the pharmaceutical composition is formulated for administration by injection and/or implantation into the vitreous cavity of an eye of the subject.
149. The pharmaceutical composition of any one of claims 88-148, wherein the pharmaceutical composition is formulated for administration by injection.
150. The pharmaceutical composition of any one of claims 88-149, wherein the pharmaceutical composition is formulated for administration by intravitreal injection.
151. The pharmaceutical composition of any one of claims 88-150, wherein the pharmaceutical composition is formulated for administration by implantation.
152. The pharmaceutical composition of any one of claims 88-148 and 151, wherein the pharmaceutical composition is formulated for administration by implantation into the vitreous cavity.
153. The pharmaceutical composition of any one of claims 88-145, wherein the pharmaceutical composition is formulated for administration selected from the group consisting of intravitreal injection, intravitreal implant, eye drop, suprachoroidal injection, oral administration, parenteral injection, and combinations thereof.
154. The pharmaceutical composition of any one of claims 88-145, wherein the pharmaceutical composition is formulated for topical administration as an eye drop.
155. The pharmaceutical composition of any one of claims 88-154, wherein the pharmaceutical composition is formulated for delivery to the retina and/or to the choroid.
156. The pharmaceutical composition of any one of claims 88-145, wherein the pharmaceutical composition is formulated for administration to the suprachoroidal space.
157. The pharmaceutical composition of any one of claims 88-156, wherein the pharmaceutical composition is formulated for repeated administration.
158. The pharmaceutical composition of any one of claims 88-157, wherein the pharmaceutical composition is formulated for administration selected from the group consisting of hourly, every several hours, three times daily, twice daily, once daily, every other day, every third day, every week, every other week, every third week, monthly and every few months.
159. A combination, comprising: the pharmaceutical composition of any one of claims 88-158; and a second pharmaceutical composition comprising a second therapeutic agent for treatment of an ischemic retinal disease.
160. The combination of claim 159, wherein the pharmaceutical composition containing an HIF inhibitor is for administration before, after or with the second pharmaceutical composition.
161. The combination of claim 159 or claim 160, wherein the second therapeutic agent is an angiogenesis inhibitor.
162. The combination of any one of claims 159-161, wherein the second therapeutic agent is selected from the group consisting of a VEGF inhibitor, a VEGFR inhibitor, and combinations thereof.
163. The combination of any one of claims 159-162, wherein the second therapeutic agent is selected from the group consisting of an anti-VEGF antibody, ranibizumab, bevacizumab, aflibercept, pegaptanib, and combinations thereof.
164. The combination of any one of claims 159-162, wherein the second therapeutic agent is selected from the group consisting of an anti-VEGFR antibody, cediranib, Cabozantinib, pazopanib, lenvatinib, sunitinib, axitinib, sorafenib, regorafenib, ponatinib, vandetanib, ramucirumab, sunitinib, dovitenib, and combinations thereof.
165. The combination of any one of claims 159-161, wherein the second therapeutic agent is selected from among the group consisting of dexamethasone, triamcinolone, a corticosteroid, and combinations thereof.
166. The combination of any one of claims 159-165, wherein the pharmaceutical composition containing an HIF inhibitor and the second pharmaceutical composition are formulated for administration as a single composition or as two compositions.
PCT/IB2022/054013 2021-04-29 2022-04-29 Hypoxia inducible factor (hif) inhibitors for treatment of atrophy associated with retinal hypoxia WO2022229932A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163181611P 2021-04-29 2021-04-29
US63/181,611 2021-04-29

Publications (1)

Publication Number Publication Date
WO2022229932A1 true WO2022229932A1 (en) 2022-11-03

Family

ID=81648071

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/054013 WO2022229932A1 (en) 2021-04-29 2022-04-29 Hypoxia inducible factor (hif) inhibitors for treatment of atrophy associated with retinal hypoxia

Country Status (1)

Country Link
WO (1) WO2022229932A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020179747A1 (en) * 2019-03-01 2020-09-10 学校法人慶應義塾 Hif inhibitory composition

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020179747A1 (en) * 2019-03-01 2020-09-10 学校法人慶應義塾 Hif inhibitory composition

Non-Patent Citations (95)

* Cited by examiner, † Cited by third party
Title
ALBADARI ET AL., EXPERT OPIN DRUG DISCOV, vol. 14, no. 7, July 2019 (2019-07-01), pages 667 - 682
ALQAWI ET AL., PROSTATE CANCER PROSTATIC DIS., vol. 9, no. 2, 2006, pages 126 - 35
AMMAR ET AL., CURR. OPIN. OPHTHALMOL., vol. 31, no. 3, 2020, pages 215 - 221
ARCH OPHTHALMOL, vol. 123, no. 11, 2005, pages 1484 - 1498
ARJAMAA ET AL., AGEING RES REV, vol. 8, no. 4, October 2009 (2009-10-01), pages 349 - 58
ARJAMAA ET AL., AGEING RES REV., vol. 8, no. 4, October 2009 (2009-10-01), pages 349 - 58
BAE ET AL., ONCOL. REP., vol. 15, no. 6, 2006, pages 1557 - 62
BAN ET AL., CELL DEATH DIS, vol. 8, no. 6, 2017, pages e2843
BEFANI ET AL., J. MOL. MED., vol. 90, 2012, pages 45 - 54
BHATTARAI ET AL., INT. J. MOL. SCI., vol. 21, no. 6, 2020, pages 2066
BUCHLER ET AL., CANCER, vol. 100, no. 1, 2004, pages 201 - 10
CAS , no. 357400-13-6
CHO ET AL., NATURE, vol. 539, 2016, pages 107 - 111
COURTNEY ET AL., J. CLIN. ONCOL., vol. 36, no. 9, 2018, pages 867 - 874
DENKO ET AL., NAT REV CANCER, vol. 8, 2008, pages 705 - 713
DUYNDAM, BIOCHEM. PHARMACOL., vol. 74, no. 2, 2007, pages 191 - 201
EVANS ET AL., JAMA OPHTHALMOL, vol. 138, no. 10, 2020, pages 1043 - 1051
FANDREY ET AL., CARDIOVASC. RES., vol. 71, 2006, pages 642 - 651
FANG ET AL., CARCINOGENESIS, vol. 28, no. 4, 2007, pages 858 - 64
FREDE ET AL., METHODS ENZYMOL., vol. 435, 2007, pages 405 - 419
GIRMENS ET AL., INTRACTABLE RARE DIS., vol. 1, no. 3, 2012, pages 103 - 114
GRUNWALD ET AL., CLINICAL TRIAL OPHTHALMOLOGY, vol. 124, no. 1, 2017, pages 97 - 104
HIDESHIMA ET AL., BLOOD J. AM. SOC. HEMATOL., vol. 101, 2003, pages 1530 - 1534
HUANG ET AL., ONCOTARGET, vol. 6, 2015, pages 18590
IMAYOSHI ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 484, 2017, pages 262 - 268
INOUE ET AL., BR. J. OPHTHALMOL., vol. 91, 2007, pages 1720 - 1721
JIANG ET AL., CELL GROWTH DIFFER., vol. 12, no. 7, 2001, pages 363 - 9
KAARNIRANTA ET AL., FRONT BIOSCI, vol. 2, no. 4, 1 June 2010 (2010-06-01), pages 1374 - 84
KAMIYAMA ET AL., J. CANCER RES. CLIN. ONCOL., vol. 131, 2005, pages 205 - 213
KANDASAMY ET AL., ASIA PAC. J. OPHTHALMOL, vol. 6, no. 6, 2017, pages 508 - 513
KAUPPINEN ET AL., CELL MOL LIFE SCI, vol. 73, no. 9, May 2016 (2016-05-01), pages 1765 - 86
KIM ET AL., DRUGS AGING, vol. 38, no. 1, 2021, pages 17 - 27
KIM ET AL., J. MOL. MED., vol. 93, 2015, pages 499 - 509
KNUROWSKI ET AL., BLOOD, vol. 134, 2019, pages 1266
KOH ET AL., MOL CANCER THER., vol. 7, no. 1, 2008, pages 90 - 100
KOH ET AL., MOL. CANCER THER., vol. 7, 2008, pages 90 - 100
KONG ET AL., CANCER RES., vol. 65, no. 19, 2005, pages 9047 - 55
KOO, NAT. COMMUN., vol. 9, 2018, pages 1855
KOVACS ET AL., MOL. CELL, vol. 18, 2005, pages 601 - 607
KUNG ET AL., CANCER CELL, vol. 6, no. 1, 2004, pages 33 - 43
KURIHARA ET AL., J. CLIN. INVESTIG., vol. 122, 2012, pages 4213 - 4217
LEE ET AL., BIOCHEM. PHARMACOL., vol. 80, 2010, pages 982 - 989
LEE ET AL., BIOORG. MED. CHEM. LETT., vol. 27, 2017, pages 3026 - 3029
LEE ET AL., J. CELL. PHYSIOL., vol. 230, 2015, pages 853 - 862
LEE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 106, 2009, pages 17910 - 17915
LEE ET AL., PROC. NATL. ACAD. SCI. USA., vol. 106, 2009, pages 2353 - 8
LI ET AL., EXPERT. OPIN INVESTIG. DRUGS, vol. 26, no. 10, 2017, pages 1103 - 1114
MABJEESH ET AL., CANCER CELL, vol. 3, no. 4, 2003, pages 363 - 75
MAEDA ET AL., BIOL PHARM BULL, vol. 29, no. 7, 2006, pages 1344 - 8
MASOUD ET AL., ACTA PHARM SIN B, vol. 5, no. 5, September 2015 (2015-09-01), pages 378 - 89
MASOUD, ACTA PHARM SIN B, vol. 5, no. 5, 2015, pages 378 - 89
MAXWELL ET AL., NATURE, vol. 399, no. 6733, 1999, pages 271 - 5
MAXWELL ET AL., NATURE, vol. 399, no. 6733, 20 May 1999 (1999-05-20), pages 271 - 5
MIE ET AL., BIOCHEM BIOPHYS RES COMMUN., vol. 300, no. 1, 2003, pages 241 - 6
NEWCOMB ET AL., NEURO. ONCOL., vol. 7, no. 3, 2005, pages 225 - 35
OH ET AL., MOL. CELLS, vol. 38, 2015, pages 528
OLENYUK, PROC NATL ACAD SCI USA, vol. 101, no. 48, 2004, pages 16768 - 73
PAN XIAOYAN ET AL: "Effects and Mechanism of Action of PX-478 in Oxygen-Induced Retinopathy in Mice", OPHTHALMIC RESEARCH, vol. 63, no. 2, 17 January 2020 (2020-01-17), CH, pages 182 - 193, XP055935730, ISSN: 0030-3747, Retrieved from the Internet <URL:https://www.karger.com/Article/Pdf/504023> DOI: 10.1159/000504023 *
PARVEEN ET AL., TAIWAN J OPHTHALMOL., vol. 8, no. 4, 2018, pages 205 - 215
PATEL ET AL., WORLD J. PHARMACOL., vol. 2, no. 2, 2013, pages 47 - 64
PAWLUS ET AL., CELL SIGNAL, vol. 25, no. 9, September 2013 (2013-09-01), pages 1895 - 903
PHAM ET AL., CLIN. CANCER RES., vol. 21, 2015, pages 808 - 818
PONS ET AL., PLOS ONE., vol. 6, no. 2, 2011, pages e16722
PORE ET AL., CANCER RES., vol. 66, no. 18, 2006, pages 9252 - 9
PORE ET AL., CANCER RES., vol. 69, no. 4, 2009, pages 1624 - 204
PRABHAKAR ET AL., PHYSIOL REV, vol. 92, no. 3, July 2012 (2012-07-01), pages 967 - 1003
RAPISARDA ET AL., CANCER RES, vol. 62, 2002, pages 4316 - 4324
RAPISARDA ET AL., CANCER RES, vol. 64, 2004, pages 6845 - 6848
ROFAGHA ET AL., OPHTHALMOLOGY, vol. 120, 2013, pages 2292 - 2299
SCHEUERMANN ET AL., NAT. CHEM. BIOL., vol. 9, 2013, pages 271
SCHLINGEMANN, GRAEFES ARCH. CLIN. EXP. OPHTHALMOL., vol. 242, 2004, pages 91 - 101
SCHOFIELD ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 338, 2005, pages 617 - 626
SEMENZA ET AL., BIOCHEM PHARMACOL, vol. 64, no. 5-6, September 2002 (2002-09-01), pages 993 - 8
SEMENZA ET AL., BIOCHEM PHARMACOL., vol. 64, no. 5-6, September 2002 (2002-09-01), pages 993 - 8
SHANKAR ET AL., MOL. CANCER THER., vol. 8, no. 6, 2009, pages 1596 - 605
SHIMADA ET AL., GRAEFES ARCH CLIN EXP OPHTHALMOL., vol. 245, no. 2, 2007, pages 295 - 300
SHIN ET AL., BLOOD, vol. 111, no. 6, 2008, pages 3131 - 6
SHINOJIMA ET AL., J. CLIN. MED., vol. 10, no. 23, 2021, pages 5496
SHINOJIMA ET AL., J. CLIN. MED., vol. 10, no. 23, 24 November 2021 (2021-11-24), pages 5496
SHIRAI ET AL., CANCERS (BASEL, vol. 13, no. 11, 2021, pages 2813
SOBOLEWSKA ET AL., CLIN OPHTHALMOL, vol. 15, 2021, pages 4317 - 4326
STEFANSSON ET AL., PROG RETIN EYE RES, vol. 30, no. 1, January 2011 (2011-01-01), pages 72 - 80
STEFANSSON ET AL., PROG. RETIN. EYE RES., vol. 30, no. 1, 2011, pages 72 - 80
STEFANSSON ET AL., PROG. RETIN. EYE RES., vol. 70, 2019, pages 1 - 22
THOMAS ET AL., MED CLIN NORTH AM, vol. 105, no. 3, May 2021 (2021-05-01), pages 473 - 491
THOMAS ET AL., MED. CLIN. NORTH AM, vol. 105, no. 3, May 2021 (2021-05-01), pages 473 - 491
TOLONEN ET AL., CELL. MOL. LIFE SCI., vol. 77, 2020, pages 3627 - 3642
WALLACE ET AL., CANCER RES., vol. 76, 2016, pages 5491 - 5500
WEINHOUSE ET AL., SCIENCE, vol. 124, 1956, pages 267 - 272
WELSH ET AL., MOL. CANCER. THER., vol. 2, no. 3, 2003, pages 235 - 43
WONG ET AL., LANCET GLOB HEALTH, vol. 2, no. 2, February 2014 (2014-02-01), pages el06 - 16
YANG ET AL., INVEST OPHTHALMOL VIS SCI., vol. 61, no. 10, 2020, pages 35
YANG, J. EXP. CLIN. CANCER RES., vol. 25, no. 4, 2006, pages 593 - 9
YASUI ET AL., BR. J. CANCER, vol. 99, no. 9, 2008, pages 1442 - 52
ZHAO ET AL., PANCREAS, vol. 34, no. 2, 2007, pages 242 - 7

Similar Documents

Publication Publication Date Title
AU2009240470B2 (en) Inhibition of neovascularization by cerium oxide nanoparticles
Stewart Corticosteroid use for diabetic macular edema: old fad or new trend?
Algvere et al. A prospective study on intravitreal bevacizumab (Avastin®) for neovascular age‐related macular degeneration of different durations
JP7278257B2 (en) Treatment of eye conditions such as macular degeneration, glaucoma, and diabetic retinopathy with pharmaceutical agents that eliminate senescent cells
Sivaprasad et al. Intravitreal steroids in the management of macular oedema
Adams et al. Glaucoma-next generation therapeutics: impossible to possible
Chen et al. Subconjunctival injection of bevacizumab (avastin) on corneal neovascularization in different rabbit models of corneal angiogenesis
WO2010125416A1 (en) Drug delivery to the anterior and posterior segments of the eye
Hou et al. Photodynamic therapy leads to time-dependent regression of pathologic corneal (lymph) angiogenesis and promotes high-risk corneal allograft survival
JP2013049701A (en) Pharmaceutical for preventing or treating disorder accompanied by ocular angiogenesis and/or elevated ocular vascular permeability
TW200410699A (en) Use of ANECORTAVE acetate for the protection of visual acuity in patients with age related macular degeneration
Marangoz et al. Comparison of the neuroprotective effects of brimonidine tartrate and melatonin on retinal ganglion cells
WO2022229932A1 (en) Hypoxia inducible factor (hif) inhibitors for treatment of atrophy associated with retinal hypoxia
US11478465B2 (en) Compounds for treatment of eye diseases associated with excessive vascularisation
Veiga Reis et al. Combined intravitreal dexamethasone and bevacizumab injection for the treatment of persistent diabetic macular edema (DexaBe study): a phase I clinical study
Jain et al. Neuroprotection of the Optic Nerve and the Retina
Rocha-de-Lossada et al. Ocular surface toxicity of depatuxizumab mafoditin (ABT-414)
RU2815482C2 (en) Treatment of eye diseases such as macular degeneration, glaucoma and diabetic retinopathy with medications that eliminate aging cells
Williams et al. Drug Induced Maculopathies
Rocha-de-Lossada et al. Toxicidade do depatuxizumabe mafodotina (ABT-414) na superfície ocular: relatos de casos
Elsayed Comparisons of Ranibizumab Injection and Dexamethasone Implant in Macular Oedema Secondary to Central Retinal Vein Occlusion
EA045318B1 (en) COMPOUNDS FOR THE TREATMENT OF EYE DISEASES ASSOCIATED WITH EXCESSIVE VASCULARIZATION
Golan et al. Anti VEGF Agents for 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: 22723199

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE