WO2024091659A1 - Cyclodextrines modifiées et leurs utilisations ophtalmiques - Google Patents

Cyclodextrines modifiées et leurs utilisations ophtalmiques Download PDF

Info

Publication number
WO2024091659A1
WO2024091659A1 PCT/US2023/036113 US2023036113W WO2024091659A1 WO 2024091659 A1 WO2024091659 A1 WO 2024091659A1 US 2023036113 W US2023036113 W US 2023036113W WO 2024091659 A1 WO2024091659 A1 WO 2024091659A1
Authority
WO
WIPO (PCT)
Prior art keywords
ring
compound
nitrogen
sulfur
oxygen
Prior art date
Application number
PCT/US2023/036113
Other languages
English (en)
Inventor
Stephen Gitu MACHATHA
Charles Montgomery
Adam Brockman
Original Assignee
Aldeyra Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aldeyra Therapeutics, Inc. filed Critical Aldeyra Therapeutics, Inc.
Publication of WO2024091659A1 publication Critical patent/WO2024091659A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/724Cyclodextrins

Definitions

  • the present invention relates to modified cyclodextrin compounds useful as excipients and/or as compounds for trapping disease-causing aldehydes in aqueous formulations.
  • the present invention further relates to methods of use of such compounds for treating an ocular disease, disorder, or condition such as those described herein, as well as pharmaceutical compositions of such compounds.
  • A2E Age Related Macular Degeneration
  • Aldehydes are implicated in diverse pathological conditions such as dry eye, cataracts, keratoconus, Fuch’s endothelial dystrophy in the cornea, uveitis, allergic conjunctivitis, ocular cicatricial pemphigoid, conditions associated with photorefractive keratectomy (PRK) healing or other corneal healing, conditions associated with tear lipid degradation or lacrimal gland dysfunction, inflammatory ocular conditions such as ocular rosacea (with or without meibomian gland dysfunction), and non-ocular disorders or conditions such as skin cancer, psoriasis, contact dermatitis, atopic dermatitis, acne vulgaris, Sjogren-Larsson Syndrome, ischemic-reperfusion injury, inflammation, diabetes, neurodegeneration (e.g., Parkinson’s disease), scleroderma, amyotrophic lateral sclerosis, autoimmune disorders (e.g., lupus), cardiovascular disorders (e.g.
  • MDA, HNE and other toxic aldehydes are generated by a myriad of metabolic mechanisms involving: fatty alcohols, sphingolipids, glycolipids, phytol, fatty acids, arachadonic acid metabolism (Rizzo (2007)), polyamine metabolism (Wood et al. (2006)), lipid peroxidation, oxidative metabolism (Buddi et al. (2002), Zhou et al. (2005)), and glucose metabolism (Pozzi et al. (2009)).
  • Aldehydes can cross link with primary amino groups and other chemical moieties on proteins, phospholipids, carbohydrates, and DNA, leading in many cases to toxic consequences, such as mutagenesis and carcinogenesis (Marnett (2002)).
  • MDA is associated with diseased corneas, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy corneas (Buddi et al. (2002)).
  • skin disorders e.g., Sjogren-Larsson Syndrome
  • octadecanal and hexadecanal e.g., Sjogren-Larsson Syndrome
  • increased lipid peroxidation and resultant aldehyde generation are associated with the toxic effects of blister agents (Sciuto et al. (2004) and Pal et al. (2009)).
  • Dry Age-related macular degeneration currently affects over 1.8 million adults aged 40 years and older in the United States, with an additional 7.3 million at risk of developing AMD. Dry AMD is a leading cause of blindness with no FDA-approved therapy. Dry AMD is characterized by drusen that form under the retina and lead to retinal dysfunction. [0008] Accordingly, there remains a need for treating, preventing, and/or reducing a risk of a disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis.
  • the present invention provides a pharmaceutical composition comprising: a compound of formula I: or a pharmaceutically acceptable salt thereof, wherein each of A, R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is as defined herein; and a compound of formula II: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of formula X-I: X-I or a pharmaceutically acceptable salt thereof, wherein each of X, Y, W, R 1 , R 2 , R 3 , and R 5 is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’ described below, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula X-VI: X-VI or a pharmaceutically acceptable salt thereof, wherein each of R c , R d , R 7 , R 8 , R 9 , and R 10 is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-I: or a pharmaceutically acceptable salt thereof, wherein each of R d , R c , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-II: or a pharmaceutically acceptable salt thereof, wherein each of R d , R c , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: ’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-III: Y-III or a pharmaceutically acceptable salt thereof, wherein each of R b , R a , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-IV: or a pharmaceutically acceptable salt thereof, wherein each of R b , R a , R 1 , R 2 , R 3 , L 1 and Ring A is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides methods for treating an ocular disorder, such as age-related macular degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment with a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof, and a compound of formula II or a pharmaceutically acceptable salt thereof.
  • an ocular disorder such as age-related macular degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment
  • ALD age-related macular degeneration
  • RP retinitis pigmentosa
  • the present invention provides a compound of formula II: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula II, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: [0020] or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides methods for treating an ocular disorder, such as age-related macular degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment with a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, or a pharmaceutically acceptable salt thereof.
  • AMD age-related macular degeneration
  • RP retinitis pigmentosa
  • dark adaptation impairment with a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, or a pharmaceutically acceptable salt thereof.
  • FIG.1 is a graph showing the results of a rabbit vitreous PK profile of non-deuterated Compound A.
  • FIG.2 is a graph showing the results of a retinaldehyde binding assay of Compound A.
  • FIG.3A is a graph showing the results of a 4-HNE binding assay with Compound I- 1.
  • FIG. 3B is a graph showing the results of a 4-HNE binding assay with 3-Amino- ⁇ - cyclodextrin.
  • the present invention provides compounds, compositions, and methods for treatment, prevention, and/or reduction of a risk of diseases, disorders, or conditions in which aldehyde toxicity is implicated in the pathogenesis.
  • such compounds include those of the formulae described herein, or pharmaceutically acceptable salts thereof, wherein each variable is as defined herein and described in embodiments.
  • a disclosed compound contains an amino functionality and a carbinol functionality (such as a propan-2-ol group) that are believed to be capable of scavenging or trapping aldehydes by formation of an adduct.
  • a pharmaceutical composition comprises: a compound having a general formula I: or a pharmaceutically acceptable salt thereof, wherein each of A, R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is as defined herein; and a compound of general formula II: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of formula X-I: X-I or a pharmaceutically acceptable salt thereof, wherein each of X, Y, W, R 1 , R 2 , R 3 , and R 5 is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula X-VI: X-VI or a pharmaceutically acceptable salt thereof, wherein each of R c , R d , R 7 , R 8 , R 9 , and R 10 is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-I: or a pharmaceutically acceptable salt thereof, wherein each of R d , R c , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-II: Y-II or a pharmaceutically acceptable salt thereof, wherein each of R d , R c , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-III: Y-III or a pharmaceutically acceptable salt thereof, wherein each of R b , R a , R 1 , R 2 , R 3 , L 1 and Ring A are as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a compound of formula Y-IV: Y-IV or a pharmaceutically acceptable salt thereof, wherein each of R b , R a , R 1 , R 2 , R 3 , L 1 and Ring A is as defined herein; and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the administration of the compositions comprising a compound of general formula I and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula I in the eye compared to administration of a composition comprising a compound of general formula I that does not comprise the modified cyclodextrin.
  • the administration of the compositions comprising a compound of general formula X-I and and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula X-I in the eye compared to administration of a composition comprising a compound of general formula X-I that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • the administration of the compositions comprising a compound of general formula X-VI and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula X-VI in the eye compared to administration of a composition comprising a compound of general formula X-VI that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • the administration of the compositions comprising a compound of general formula Y-I and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula Y-I in the eye compared to administration of a composition comprising a compound of general formula Y-I that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • the administration of the compositions comprising a compound of general formula Y-II and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula Y-II in the eye compared to administration of a composition comprising a compound of general formula Y-II that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • the administration of the compositions comprising a compound of general formula Y-III and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula Y-III in the eye compared to administration of a composition comprising a compound of general formula Y-III that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • the administration of the compositions comprising a compound of general formula Y-IV and a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’, for example, a compound of formula II’, to an eye increases the residency time of the compound of general formula Y-IV in the eye compared to administration of a composition comprising a compound of general formula Y-IV that does not comprise a modified cyclodextrin, such as a compound of general formula II, II’, II’’, or II’’’’.
  • Compounds of General Formula I, X-I, X-VI, Y-I, Y-II, Y-III and Y-IV [0042] An exemplary compound of general formula I is Compound A.
  • Compound A is a small crystalline molecule that covalently and irreversibly binds proinflammatory reactive aldehyde species (RASP).
  • RASP proinflammatory reactive aldehyde species
  • Compound A is a deuterated benzoxazole derivative with a molecular weight of 273.3 g/mol.
  • Exemplary compounds of general formula X-I, X-VI, Y-I, Y-II, Y-III, and Y-IV are as described herein.
  • Compound A is a hydrophobic molecule (cLogP 2.24) with limited solubility. Cyclodextrins are excipients extensively used in drug delivery to increase the solubility and stability of hydrophobic drugs via the formation of water-soluble inclusion complexes. [0045] By sequestering reactive aldehydes, Compound A and other exemplary compounds of general formula X-I, X-VI, Y-I, Y-II, Y-III, and Y-IV may potentially represent a valuable treatment option for adults with the dry form of age-related macular degeneration (dry AMD) with evidence of dark adaptation impairment.
  • dry AMD age-related macular degeneration
  • Reactive aldehydes are toxic and react with cellular biomolecules, inducing inflammation and forming complex conjugates that contribute to the accumulation of drusen in the retina.
  • Compound A and other exemplary compounds described herein may protect against aldehyde toxicity through the targeting of all-trans- retinaldehyde, 4-HNE (4-hydroxynonenal), and other reactive aldehydes.
  • Retinol is converted into retinaldehyde by retinol dehydrogenase then retinaldehyde is converted into retinoic acid by retinaldehyde dehydrogenase, which is expressed in RPE (retinal pigment epithelium).
  • All-trans-retinaldehyde is a biologically chaperoned, reactive intermediate.
  • retinaldehyde can interact with amines, forming a dimerization of retinaldehyde. These dimers of retinaldehyde can be transformed into higher-order oligomers or smaller oxidative catabolites, or reactive vitamin A by-products.
  • Reffs et al., 2020.
  • Vitamin A by-products are not only implicated in the pathogenesis of dry AMD but also dark adaptation impairment.
  • Dark adaptation impairment is a defect in the ability of the visual system to adjust from photopic to scotopic conditions, and is one of the early symptoms of dry AMD.
  • Previous research demonstrated that the accumulation of Vitamin A by-products alone causes the worsening of dark adaptation.
  • dark adaptation impairment is positivity correlated with dry AMD severity (Flamendorf, et al., 2015; Nigalye, et al., 2022), and the measurement of dark adaptation has been identified to reveal abnormalities in subclinical dry AMD (Zhang, Robinson, Saad, and Washington, 2021).
  • Cyclodextrins are crystalline, non-hygroscopic, cyclic oligosaccharides with a hydrophilic outer surface and a less hydrophilic central cavity which is able to host hydrophobic compounds and form inclusion complexes (Loftsson 1996). These complexes are formed when a “guest” molecule, for example, a compound of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV, is partially or fully included inside a “host” molecule, for example, a cyclodextrin compound described herein, with no covalent bonding.
  • guest for example, a compound of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV
  • an Active Pharmaceutical Ingredient such as a compound capable of scavenging or trapping an aldehyde by formation of an adduct, e.g., Compound A or a compound of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV, with a a cyclodextrin compound described herein and forming an inclusion complex, and thus the half- life of the complex will be increased compared to API that is not formulated with a cyclodextrin compound described herein.
  • API Active Pharmaceutical Ingredient
  • Cyclodextrins are expected to have a longer half-life in the vitreous since they are very hydrophilic and are not expected to rapidly permeate across membranes. [0051]
  • the increased residency time in the vitreous may increase membrane permeability which is important for the drug to reach the Bruch’s membrane and retinal pigment epithelium (RPE), where excess retinaldehyde is thought to form toxic precipitates.
  • RPE retinal pigment epithelium
  • the rate of release of the API of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV from the core will be modified and potentially increase the ocular half-life of the drug in the retinal cavity, which should improve the activity of API of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV and potentially reduce the number of injections or other administrations to the eye required.
  • an API of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y- IV is formulated with a modified- ⁇ -cyclodextrin in a buffered aqueous solution.
  • Compounds of General Formula II [0054]
  • the pharmaceutical compositions disclosed herein comprise a modified cyclodextrin, such as those of general formula II, II’, II’’, or II’’’’.
  • the compounds and pharmaceutical compositions are in an aqueous solution buffered at an appropriate pH for intravitreal delivery, such as a pH of 7.0-7.8, for example, about 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, or 7.8.
  • the modified cyclodextrins of this disclosure are derived, for example, from native amino- ⁇ -cyclodextrin modified by covalently linking a hydrophilicity modifier, such as a semi- carbazide moiety, to its hydrophobic core.
  • a hydrophilicity modifier such as a semi- carbazide moiety
  • other cyclodextrins may be modified with a hydrophilicity modifier (“modifier” group described herein) to provide a disclosed cyclodextrin compound.
  • the present invention provides a compound of general formula II: II or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin; X is nitrogen, oxygen, or sulfur; R is H, OH, halogen, or C1-C12 alkyl; and Y is a modifier.
  • the present invention provides a compound of general formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin;
  • X is nitrogen, oxygen, sulfur, or a C 1-4 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -O-, -C(O), -C(O)O-, -OC(O)-, -C(S)-, -C(R X )2, -C(F)2, -N(R X )-, -C(O)N(R X )-, - R X NC(O)-, -OC(O)N(R X )-, -N(R X )C(O)N(R X )-, or -Cy-;
  • R is H, OH, halogen, -CN, or optionally substituted C1-C12 al
  • the present invention provides a compound of general formula II’’: II’’ or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin; X is a covalent bond, nitrogen, oxygen, or sulfur; or a C 1-4 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -O-, -C(O), -C(O)O-, -OC(O)-, -C(S)-, -C(R X )2, - C(F) 2 , -N(R X )-, -C(O)N(R X )-, -R X NC(O)-, -OC(O)N(R X )-, -N(R X )C(O)N(R X )-, or -Cy-; R is absent, H, -NO2, -OH,
  • the present invention provides a compound of general formula II’’’: or a pharmaceutically acceptable salt thereof, wherein: CD is a cyclodextrin; X is a covalent bond, nitrogen, oxygen, or sulfur; or a C1-4 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -O-, -C(O), -C(O)O-, -OC(O)-, -C(S)-, -C(R X )2, - C(F)2, -N(R X )-, -C(O)N(R X )-, -R X NC(O)-, -OC(O)N(R X )-, -N(R X )C(O)N(R X )-, or -Cy-; R is absent, H, -NO 2 , -CN, or optionally
  • the modifier is a functional moiety selected from the group consisting of hydrazide, thiohydrazide, vicinal diamine, aniline and vicinal amino alcohol
  • Y is substituted by R X , wherein R X is selected from -OH, - OMe, halogen, -CN, -C(O)Me, and -C(O)OMe.
  • the method of treating, preventing, or reducing a risk of a disease, disorder, condition, or cosmetic indication in which aldehyde toxicity is implicated in a subject in need thereof comprises administering topically or systemically to the subject a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II”, or II’” or a pharmaceutically acceptable salt thereof.
  • the disease, disorder, or condition is an ocular disorder. 2.
  • compositions and Compounds are useful for treating, preventing, and/or reducing a risk of a disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis.
  • the present invention provides a pharmaceutical composition comprising a compound of formula I: I or a pharmaceutically acceptable salt thereof, wherein each of A, R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is as defined herein; and a compound of general formula II: or a pharmaceutically acceptable salt thereof, wherein each of N, R, X and Y is as defined herein have been found to be useful in treating ocular diseases and condition.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of formula X-I: or a pharmaceutically acceptable salt thereof, wherein W is N or CR 4 ; X is S, NH, or O; Y is N or CR 6 ; provided that, if X is S or O, then Y is CR 6 ; each of R 1 , R 2 , R 3 , R 4 , R 5 , and R 6 is independently hydrogen, deuterium, halogen, -NH 2 , -CN, - OR’, -SR’, optionally substituted C1-6 aliphatic, or , R a is C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R a and R b , taken together with the carbon atom to which they are attached, form a 3-8 membered
  • the compound of formula X-I is a compound of formula X-II-a, X- II-b, X-II-c, X-II-d, X-II-e, X-II-f, X-II-g, X-II-h, X-II-i, or X-II-j: or a pharmaceutically acceptable salt thereof, wherein: each of W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R a , and R b is as defined above and described in embodiments herein, both singly and in combination.
  • the compound of formula X-I is a compound of formula X-III-a, X-III-b, X-III-c, X-III-d, X-III-e, X-III-f, X-III-g, X-III-h, X-III-i, or X-III-j: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R a , and R b is as defined above and described in embodiments herein, both singly and in combination.
  • the compound of formula X-I is a compound of formula X-IV-a, X-IV-b, X-IV-c, X-IV-d, X-IV-e, X-IV-f, X-IV-g, or X-IV-h: X-IV-a X-IV-b X-IV-c X-IV-d or a pharmaceutically acceptable salt thereof, wherein: each of W, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R a and R b is as defined above and described in embodiments herein, both singly and in combination.
  • the compound of formula X-I is a compound of formula X-V-a, X- V-b, X-V-c, X-V-d, X-V-e, X-V-f, X-V-g, X-V-h, X-V-i, or X-V-j: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R a , and R b is as defined above and described in embodiments herein, both singly and in combination.
  • the present invention provides a compound of formula X-VI: X-VI or a pharmaceutically acceptable salt thereof, each of R 7 , R 8 , R 9 , and R 10 is independently hydrogen, deuterium, halogen, -N(R)2, -CN, -OR’, -SR’, or optionally substituted C1-6 aliphatic;
  • R c is hydrogen or C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms;
  • R d is hydrogen or C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R c and R d , taken together with the carbon atom to which they are attached, form a 3-8 membered, saturated cycloalkyl or heterocyclyl ring containing 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; and each R’ is independently selected from hydrogen, deuterium, and an optionally substituted group selected from: C1-6 aliphatic optional
  • the present invention provides a compound of formula X-VII: X-VII or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 2 , R 3 , R 4 , R 5 , and R 6 is independently hydrogen, deuterium, halogen, -NH 2 , -CN, - OR, -SR’, -S(O)R’, -S(O)2R’, optionally substituted C1-6 aliphatic, or , provided that one of R 1 , R 2 , R 3 , R 4 , R 5 , and R 6 is –NH 2 and another one of R 1 , R 2 , R 3 , 4 5 6 R , R , and R is , and the -NH 2 and the are attached to adjacent carbon atoms or have a peri relationship; R 1' is hydrogen, deuterium, or C1-6 alkyl; R 6’ is hydrogen, deuterium, or C 1-6 alkyl; R a is C
  • the present invention provides a compound of formula X-VIII: X-VIII or a pharmaceutically acceptable salt thereof, wherein: each of R 2 , R 3 , R 4 , and R 5 is independently hydrogen, deuterium, halogen, -NH 2 , -CN, -OR’, - SR’, -S(O)R’, -S(O)2R’, optionally substituted C1-6 aliphatic, or , provided that one of R 2 , R 3 , R 4 , and R 5 is –NH 2 and another one of R 2 , R 3 , R 4 , and R 5 is , and the -NH 2 and the are attached to adjacent carbon atoms; R 1 and R 1' are each independently hydrogen, deuterium, or C 1-6 alkyl; R a is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C1-4 aliphatic optionally substituted with
  • the present invention provides a compound of formula X-IX: X-IX or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 2 , R 3 , and R 4 is independently hydrogen, deuterium, halogen, -NH2, -CN, -OR’, - SR’, -S(O)R’, -S(O) 2 R’, or optionally substituted C 1-6 aliphatic; R a is C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R a and R b , taken together with the carbon atom to which they are attached, form a 3-8 membered cycloalkyl or heterocyclyl ring containing 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; and each R’ is independently selected from hydrogen, deuterium, halogen,
  • the present invention provides a compound of formula X-X: X-X or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 2 , R 3 , R 4 , and R 5 is independently hydrogen, deuterium, halogen, -NH2, -CN, -OR’, -SR’, -S(O)R’, -S(O) 2 R’, optionally substituted C 1-6 aliphatic, or , provided that one of R 1 , R 2 , R 3 , R 4 , and R 5 is –NH2 and another one of R 1 , R 2 , R 3 , R 4 , 5 and R is , and the -NH 2 and the are attached to adjacent carbon atoms; R a is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms;
  • the present invention provides a compound of formula Y-I: Y-I or a pharmaceutically acceptable salt thereof, wherein each of R 1 , R 2 , and R 3 is independently hydrogen, deuterium, halogen, -N(R’)2, -CN, -OR’, -SR’, or optionally substituted C1-6 aliphatic; R c is hydrogen or C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; R d is hydrogen or C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R c and R d , taken together with the carbon atom to which they are attached, form a 3-8 membered, saturated cycloalkyl or heterocyclyl ring having 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; L 1 is -C(O)NR’-CR2- or -NR’C(O)-CR’2-, wherein either
  • the present invention provides a compound of formula Y-II: or a pharmaceutically acceptable salt thereof, wherein each of R 1 , R 2 , and R 3 is independently hydrogen, deuterium, halogen, -N(R’) 2 , -CN, -OR’, -SR’, or optionally substituted C 1-6 aliphatic; R c is hydrogen or C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; R d is hydrogen or C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R c and R d , taken together with the carbon atom to which they are attached, form a 3-8 membered, saturated cycloalkyl or heterocyclyl ring having 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; L 1 is -C(O)NR’-CR’ 2 - or -NR’C(O)-CR’ 2
  • the present invention provides a compound of formula Y-III: Y-III or a pharmaceutically acceptable salt thereof, wherein each of R 1 , R 2 , and R 3 is independently hydrogen, deuterium, halogen, -NH 2 , -CN, -OR’, -SR’, -S(O)R’, -S(O) 2 R’, or optionally substituted C1-6 aliphatic; R a is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R a and R b , taken together with the carbon atom to which they are attached, form a 3-8 membered cycloalkyl or heterocyclyl ring containing 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; L 1 is -C(O)NR’-CR’ 2 - or
  • the present invention provides a compound of formula Y-IV: or a pharmaceutically acceptable salt thereof, wherein each of R 1 , R 2 , and R 3 is independently hydrogen, deuterium, halogen, -NH 2 , -CN, -OR’, -SR’, -S(O)R’, -S(O) 2 R’, or optionally substituted C1-6 aliphatic; R a is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R a and R b , taken together with the carbon atom to which they are attached, form a 3-8 membered cycloalkyl or heterocyclyl ring containing 1-2 heteroatoms each independently selected from nitrogen, oxygen, and sulfur; L 1 is -C(O)NR’-CR’ 2 - or -NR’
  • the compound of formula Y-I is a compound depicted as follows: Y-I-a Y-I-b or a pharmaceutically acceptable salt thereof, wherein Ring A, R 4 , and n, and their constituent groups, are each as defined and described herein.
  • the compound of formula Y-II is a compound depicted as follows: Y-II-a Y-II-b or a pharmaceutically acceptable salt thereof, wherein Ring A, R 4 , and n, and their constituent groups are each as defined and described herein.
  • the compound of formula Y-III is a compound depicted as follows: Y-III-a Y-III-b or a pharmaceutically acceptable salt thereof, wherein Ring A, R 4 , and n, and their constituent groups are each as defined and described herein.
  • the compound of formula Y-IV is a compound depicted as follows: Y-IV-a Y-IV-b or a pharmaceutically acceptable salt thereof, wherein Ring A, R 4 , and n, and their constituent groups are each as defined and described herein.
  • the composition may contain the active compound of general formula I at a concentration of 0.01-20%, 0.02-15%, 0.04-10%, 0.06-5%, 0.08-1%, or 0.09-0.5% (weight/volume) with or without pH and/or osmotic adjustment to the solution. More particularly, the eye drop formulation may contain a compound of general formula I at a concentration of 0.09-0.5% (weight/volume), such as 0.1%, 0.25%, or 0.5%. [0086] The amount of a cyclodextrin compound described herein, such as one of general formula II, II’, II’’, or II’’’’ in the composition may range from about 10% to 50% weight/volume.
  • the concentration of the cyclodextrin compound is 20-40% weight/volume.
  • the amount of the cyclodextrin compound in the composition may range 5- 60%, 10-50%, 15-45%, 20-40%, 22-38%, 24-36%, 26-34% or 28-32% (weight/volume). More particularly, the composition may contain a cyclodextrin compound at a concentration of 25-35% (weight/volume), such as 30%.
  • the present invention provides a compound depicted in Table 1, or a pharmaceutically acceptable salt thereof. Table 1.
  • the present invention provides a compound or a pharmaceutically acceptable salt thereof according to general formula X-I or X-VI depicted as follows: [0089] In some embodiments, the present invention provides a compound or a pharmaceutically acceptable salt thereof according to general formula Y-I, Y-II, Y-III, or Y-IV depicted as follows: Y -I-1 Y-I-2 Y -I-3 Y-I-4 Y-I-57 [0090] In some embodiments, the present invention provides a compound or a pharmaceutically acceptable salt thereof depicted as follows: [0091] In certain embodiments, the present invention provides any compound described above and herein, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a compound of general formula II’: II’ or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin or a modified cyclodextrin; X is a covalent bond, nitrogen, oxygen, or sulfur; or a C 1-4 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -O-, -C(O), -C(O)O-, -OC(O)-, -C(S)-, -C(R X )2, -C(F)2, -N(R X )-, - C(O)N(R X )-, -R X NC(O)-, -OC(O)N(R X )-, -N(R)C(O)N(R X )-, or -Cy-; R is absent, H, -NO2,
  • the compound is a compound of formula III-A, R is H and Y .
  • X is N, R is H and R 11 is - NR 14 N(R 15 )2. In some embodiments of general formula IV-A, X is N, R is H and R 11 is - NHNH 2 . In some embodiments of general formula IV-A, X is N, R is H and R 11 is -NHNHOH . 1 1 In some embodiments of general formula IV-A, X is N, R is H and R is . [0099] In some embodiments of general formula IV-B, X is S, R is H, and R 11 is - NR 14 N(R 15 ) 2 .
  • the compound is a compound of general formula V-A, V-B, or V-C: V-A V-B V-C or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin; X is nitrogen, oxygen, or sulfur; R is absent, H, OH, halo, or C 1 -C 12 alkyl; each R 14 is independently H, -OH, halo, or C1-C6 alkyl; and each R 15 is independently H or C 1 -C 6 alkyl.
  • the compound is a compound of general formula VI-A, VI-B, or VI-C: VI-A VI-B VI-C or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin; X is nitrogen, oxygen, or sulfur; R is absent, H, OH, halo, or C1-C12 alkyl; each R 14 is independently H, -OH, halo, or C 1 -C 6 alkyl; and each R 15 is independently H or C 1 -C 6 alkyl.
  • CD is a cyclo
  • the compound is a compound of general formula XII-A, XII- B, or XII-C: or a pharmaceutically acceptable salt thereof, wherein CD is a cyclodextrin; R is H, -OH, halo, or C1-C12 alkyl; R 17 is H, -OH, -NH2, -OR 18 , or R 18 ; R 18 is C 1 -C 6 alkyl or C 1 -C 6 alkenyl, wherein the C 1 -C 6 alkyl and the C 1 -C 6 alkenyl each has 0 to 5 substituents selected from halo, -OH, -SH, -OR, -SR, -NR 2 , and wherein the C 1 -C 6 alkenyl has 1, 2, or 3 double bonds.
  • the cyclodextrin portion of formula II may be any cyclodextrin known in the art, either naturally occurring or modified.
  • Cyclodextrins are a family of cyclic oligosaccharides, consisting of a macrocyclic ring of glucose subunits joined by ⁇ -1,4 glycosidic bonds. Cyclodextrins are composed of 5 or more ⁇ -D-glucopyranoside units linked 1->4, as in amylose (a fragment of starch). Cyclodextrins may have as many as 150-membered cyclic oligosaccharides are also known.
  • the cyclodextrin portion of formula II, II’, II”, II’ may be any cyclodextrin known in the art, either naturally occurring or modified or a compound as described herein.
  • An alternate way to depict the structure of a cyclodextrin shows the glucose subunits in chair conformation:
  • a cyclodextrin having six glucose subunits is an alpha cyclodextrin ( ⁇ -cyclodextrin); a cyclodextrin having seven glucose subunits is a beta cyclodextrin ( ⁇ -cyclodextrin); and cyclodextrin having eight glucose subunits is a gamma cyclodextrin ( ⁇ -cyclodextrin).
  • Cyclodextrins are generally regarded as safe by the United States FDA. Cyclodextrins have toroidal shapes, with the larger and the smaller openings of the toroid exposing to the solvent secondary and primary hydroxyl groups respectively, as shown in this ⁇ - cyclodextrin:
  • the cyclodextrin of formula II may be any modified cyclodextrin known in the art. Common modifications include chemical modification of native hydroxyl groups in the cyclodextrin. O-methylation and acylation are common modifications, with tunability of the degree of functionalization can be controlled.
  • Cyclodextrins react with propylene oxide to give hydroxypropylated derivatives.
  • the primary -OH moieties may be tosylated.
  • derivation of a cyclodextrin by reaction with a primary hydroxyl group results in modification at the larger opening of the toroid shape, while derivatization by reaction with a secondary hydroxyl group results in modification at the smaller opening of the toroid shape.
  • Suitable cyclodextrins for additional modification according to the modifiers of this invention include trimethyl- ⁇ -cyclodextrin, 2-hydroxyethyl- ⁇ -cyclodextrin, 2- hydroxypropyl- ⁇ -cyclodextrin, 3-hydroxypropyl- ⁇ -cyclodextrin, and ⁇ -cyclodextrin sulfobutylether sodium salt (or potassium salt) or an oligomeric or a polymeric carrier such as ⁇ - cyclodextrin sulfobutylether sodium salt.
  • the cyclodextrin of a compound of general formulas II to XI is ⁇ -cyclodextrin.
  • the cyclodextrin of a compound of general formulas II to XI is ⁇ -cyclodextrin. In some embodiments, the cyclodextrin of a compound of general formulas II to XI is ⁇ -cyclodextrin. [00115] In some embodiments, the cyclodextrin of a compound of general formulas II to XI is an amino- ⁇ -cyclodextrin. In some embodiments, the cyclodextrin of a compound of general formulas II to XI is an amino- ⁇ -cyclodextrin.
  • the cyclodextrin of a compound of general formulas II to XI is an amino- ⁇ -cyclodextrin.
  • amino-cyclodextrins may be further modified to semi- carbazido cyclodextrins, e.g., semi-carbazido- ⁇ -cyclodextrin: semi-carbazido- ⁇ -cyclodextrin
  • the cyclodextrin of a compound of general formulas II to XI is a semi-carbazido- ⁇ -cyclodextrin.
  • the cyclodextrin of a compound of general formulas II to XI is a semi-carbazido- ⁇ -cyclodextrin. In some embodiments, the cyclodextrin of a compound of general formulas II to XI is a semi-carbazido- ⁇ -cyclodextrin.
  • the modified cyclodextrin compounds of general formula II, II’ II” or II’ have been designed to tune the physicochemical properties of the API of general formula I, X-I, X-VI, Y-I to Y-IV: cyclodextrin compound of general formula II, II’ II” or II’” complex, resulting in enhanced overall solubility and stability. Incorporation of a polar, electron-rich “warhead” into the hydrophobic core of native amino- ⁇ -cyclodextrin achieves this goal by increasing the hydrophilicity of the overall system through introduction of new, polar, and in some cases, heteroatomic, functionality. [00119] In some embodiments, the compound of general formula II, II’ II” or II’” is:
  • the present invention provides methods for treating an ocular disease, disorder, or condition, such as age-related macular degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment with a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I to Y-IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II” or II”’ or a pharmaceutically acceptable salt thereof.
  • AMD age-related macular degeneration
  • RP retinitis pigmentosa
  • the method of treating an ocular disease or condition whose etiology involves accumulation of A2E and/or lipofuscin in a subject comprises administering to the subject an effective amount of a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I to Y-IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II” or II”’ or a pharmaceutically acceptable salt thereof, and thereby reducing the level of A2E accumulation relative to the level of A2E accumulation in said subject prior to administration of the compound or composition or pharmaceutically acceptable salt thereof.
  • the method of treating, preventing, or reducing a risk of a disease, disorder, condition, or cosmetic indication in which aldehyde toxicity is implicated in a subject in need thereof comprises administering topically or systemically to the subject a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I to Y- IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II” or II”’ or a pharmaceutically acceptable salt thereof.
  • the disease, disorder, or condition is an ocular disorder.
  • “ocular disorder” includes any disease, disorder, or condition affecting any structure of the eye.
  • the ocular disorder is selected from the group consisting of dry eye disease, age-related macular degeneration (AMD), retinitis pigmentosa (RP), dark, adaptation impairment, cataracts, keratoconus, bullous and other keratopathy, Fuch’s endothelial dystrophy, allergic conjunctivitis, ocular cicatricial pemphigoid, a condition associated with PRK healing and other corneal healing, a condition associated with tear lipid degradation or lacrimal gland dysfunction, uveitis, scleritis, Stargardt’s disease, ocular Stevens Johnson Syndrome, and ocular rosacea.
  • AMD age-related macular degeneration
  • RP retinitis pigmentosa
  • dark adaptation impairment
  • cataracts keratoconus
  • bullous and other keratopathy Fuch’s endothelial dystrophy
  • allergic conjunctivitis ocular cicatricial pemphigoid
  • the ocular disorder is dry eye syndrome.
  • the ocular disorder is Age-related macular degeneration (AMD).
  • the ocular disorder is dry age-related macular degeneration (AMD).
  • the ocular disorder is dry age-related macular degeneration (AMD) with evidence of dark adaptation impairment.
  • the ocular disorder is retinitis pigmentosa (RP).
  • the ocular disorder is diabetic macular edema (DME), atopic keratoconjunctivitis (AKC), vernal keratoconjunctivitis (VKC), age-related macular degeneration (AMD), dry eye disease (DED), allergic conjunctivitis (AC), dry eye disease with allergic conjunctivitis, noninfectious anterior uveitis, posterior uveitis, pan-uveitis, post-surgical ocular pain and inflammation.
  • the ocular disorder is selected from the group consisting of uveitis, scleritis, ocular Stevens Johnson Syndrome, and ocular rosacea.
  • the ocular disorder is ocular rosacea or uveitis.
  • the ocular disorder is selected from the group consisting of keratoconus, cataracts, bullous and other keratopathy, Fuchs’ endothelial dystrophy, ocular cicatricial pemphigoid, and allergic conjunctivitis.
  • the present invention provides a method of reducing levels of one or more toxic aldehydes in a subject, comprises administering to the subject a a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I to Y- IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II” or II”’ or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition comprising a compound of general formula I, X-I, X-VI, Y-I to Y- IV or a pharmaceutically acceptable salt thereof, and a compound of general formula II, II’, II” or II”’ or a pharmaceutically acceptable salt thereof.
  • Any of the compounds of general formula II, II’, II” or II”’ or formulas III to XI may be used in the absence of a compound of general formula I, X-I, X-VI, Y-I to Y-IV for any ocular indication or disorder.
  • CD is a cyclodextrin
  • X is nitrogen, oxygen, or sulfur
  • R is absent, H, OH, halo, or C 1 -C 12 alkyl
  • Y is a modifier.
  • the compound is selected from any one of formulas III-XI.
  • the cyclodextrin is selected from a modified ⁇ -cyclodextrin, a modified ⁇ -cyclodextrin, or a modified ⁇ cyclodextrin.
  • the cyclodextrin is a modified ⁇ -cyclodextrin.
  • the compound of general formula II is: degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment comprising administering to a subject in need thereof, a compound of general formula II, II’, II” or II”’ or of any one of formulas III to XII, or a pharmaceutically acceptable salt thereof. 3.
  • aliphatic or “aliphatic group,” as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle,” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • “cycloaliphatic” (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C 3 -C 6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • bridged bicyclic refers to any bicyclic ring system, i.e. carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge.
  • a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen).
  • a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups.
  • any substitutable nitrogen of a bridged bicyclic group is optionally substituted.
  • exemplary bridged bicyclics include: [00146]
  • the term “lower alkyl” refers to a C1-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
  • the term “lower haloalkyl” refers to a C 1-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • unsaturated as used herein, means that a moiety has one or more units of unsaturation.
  • alkylene refers to a bivalent alkyl group.
  • An “alkylene chain” is a polymethylene group, i.e., –(CH2)n–, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3.
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent.
  • Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent.
  • Suitable substituents include those described below for a substituted aliphatic group.
  • halogen means F, Cl, Br, or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • aryl may be used interchangeably with the term “aryl ring.”
  • aryl refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non–aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and “heteroar—,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ⁇ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and “heteroar—”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H–quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3–b]–1,4–oxazin– 3(4H)–one.
  • heteroaryl group may be mono– or bicyclic.
  • heteroaryl may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5– to 7–membered monocyclic or 7–10–membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4– dihydro–2H–pyrrolyl), NH (as in pyrrolidinyl), or + NR (as in N–substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H–indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl.
  • a heterocyclyl group may be mono– or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
  • partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • compounds of the invention may contain “optionally substituted” moieties.
  • substituted means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; –(CH 2 ) 0–4 R ; o –(CH 2 ) 0–4 OR ; o -O(CH 2 ) 0-4 R o , –O– (CH 2 ) 0–4 C(O)OR°; –(CH 2 ) 0–4 CH(OR ) o 2 ; –(CH 2 ) 0–4 SR ; o –(CH 2 ) 0–4 Ph, which may be substituted with R°; –(CH 2 ) 0–4 O(CH 2 ) 0–1 Ph which may be substituted with R°; –
  • Suitable monovalent substituents on R o are independently halogen, —(CH2)0–2R ⁇ , –(haloR ⁇ ), –(CH2)0–2OH, –(CH2)0–2OR ⁇ , –(CH2)0–2CH(OR ⁇ )2; -O(haloR ⁇ ), –CN, –N 3 , –(CH 2 ) 0–2 C(O)R ⁇ , –(CH 2 ) 0–2 C(O)OH, –(CH 2 ) 0–2 C(O)OR ⁇ , –(CH 2 ) 0–2 SR ⁇ , –(CH2)0–2SH, –(CH2)0–2NH2, –(CH2)0–2NHR ⁇ , –(CH2)0–2NR ⁇ 2, –NO2,
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: –O(CR * 2 ) 2–3 O–, wherein each independent occurrence of R * is selected from hydrogen, C1–6 aliphatic which may be substituted as defined below, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, –R ⁇ , -(haloR ⁇ ), -OH, –OR ⁇ , –O(haloR ⁇ ), –CN, –C(O)OH, –C(O)OR ⁇ , –NH2, –NHR ⁇ , –NR ⁇ 2, or –NO2, wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include –R ⁇ , –C(O)C(O)R ⁇ , –C(O)CH2C(O)R ⁇ , -S(O)2R ⁇ , -S(O) 2 NR ⁇ 2 , –C(S)NR ⁇ 2 , –C(NH)NR ⁇ 2 , or –N(R ⁇ )S(O) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, C 1–6 aliphatic which may be substituted as defined below, unsubstituted –OPh, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form an unsubstit
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, –R ⁇ , -(haloR ⁇ ), –OH, –OR ⁇ , –O(haloR ⁇ ), –CN, –C(O)OH, –C(O)OR ⁇ , –NH2, –NHR ⁇ , –NR ⁇ 2, or -NO2, wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1–4 aliphatic, –CH 2 Ph, –O(CH 2 ) 0–1 Ph, or a 5–6– membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1–19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, besylate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2–hydroxy–ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, mesylate, 2–naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pec
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1–4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. [00169] The “retina” is a region of the central nervous system with approximately 150 million neurons.
  • retinal pigment epithelium The retina initiates the first stage of visual processing by transducing visual stimuli in specialized neurons called “photoreceptors”. Their synaptic outputs are processed by elaborate neural networks in the retina and are then transmitted to the brain.
  • the retina has evolved two specialized classes of photoreceptors to operate under a wide range of light conditions. “Rod” photoreceptors transduce visual images under low light conditions and mediate achromatic vision. “Cone” photoreceptors transduce visual images in dim to bright light conditions and mediate both color vision and high acuity vision.
  • Every photoreceptor is compartmentalized into two regions called the “outer” and “inner” segment.
  • the inner segment is the neuronal cell body containing the cell nucleus. The inner segment survives for a lifetime in the absence of retinal disease.
  • the outer segment is the region where the light sensitive visual pigment molecules are concentrated in a dense array of stacked membrane structures. Part of the outer segment is routinely shed and regrown in a diurnal process called outer segment renewal. Shed outer segments are ingested and metabolized by RPE cells.
  • the “macula” is the central region of the retina which contains the fovea where visual images are processed by long slender cones in high spatial detail (“visual acuity”).
  • “Macular degeneration” is a form of retinal neurodegeneration which attacks the macula and destroys high acuity vision in the center of the visual field.
  • Age-Related Macular Degeneration begins in a “dry form” characterized by residual lysosomal granules called lipofuscin in RPE cells, and by extracellular deposits called “drusen”. Drusen contain cellular waste products excreted by RPE cells. “Lipofuscin” and drusen can be detected clinically by ophthalmologists and quantified using fluorescence techniques. They can be the first clinical signs of macular degeneration. [00172] Lipfuscin contains aggregations of A2E.
  • Lipofuscin accumulates in RPE cells and poisons them by multiple known mechanisms. As RPE cells become poisoned, their biochemical activities decline and photoreceptors begin to degenerate. Extracellular drusen may further compromise RPE cells by interfering with their supply of vascular nutrients. Drusen also trigger inflammatory processes, which lead to choroidal neovascular invasions of the macula in one patient in ten who progresses to wet form AMD. Both the dry form and wet form progress to blindness. [00173] “ERG” is an acronym for electroretinogram, which is the measurement of the electric field potential emitted by retinal neurons during their response to an experimentally defined light stimulus.
  • ERG is a non-invasive measurement which can be performed on either living subjects (human or animal) or a hemisected eye in solution that has been removed surgically from a living animal.
  • RAL retinaldehyde.
  • RAL-trap means a therapeutic compound that binds free RAL and thereby prevents the RAL from Schiff base condensation with membrane phosphatidylethanolamine (PE).
  • Free RAL is defined as RAL that is not bound to a visual cycle protein.
  • trans-RAL and “all-trans-RAL” are used interchangeably and mean all trans-retinaldehyde.
  • A2E is a reaction by-product of a complex biochemical pathway called the “visual cycle” which operates collaboratively in both RPE cells and photoreceptor outer segments.
  • the visual cycle recycles a photoreactive aldehyde chromophore called “retinaldehyde” which is derived from vitamin A and is essential for vision.
  • the visual cycle has four principal steps: 1) it converts vitamin A in the RPE into an aldehyde chromophore with one photoreactive strained double bond (11-cis-RAL); 2) it transports 11-cis-RAL to the retina where it binds to a specialized photoreceptor protein called opsin; 3) light photoisomerizes bound 11-cis-RAL to trans-RAL, which initiates the release of bound RAL from the opsin binding site; and 4) it converts trans-RAL (an aldehyde) to vitamin A (an alcohol) and transports vitamin A back to the RPE where the cycle begins again.
  • trans-RAL an aldehyde
  • the aldehyde group of RAL helps bind the molecule to opsin by forming a reversible chemical bond to an amino acid sidechain in the opsin binding site. While the aldehyde group on RAL is essential for anchoring the molecule to the opsin binding site, it is otherwise hazardous because of its propensity to form Schiff bases with other biological amines.
  • the first three reactions take place in photoreceptor outer segments and produce an intermediary product called A2PE. Once formed, A2PE partitions into the lipid phase and accumulates in photoreceptor outer segment membranes.
  • RAL-traps such as certain compounds disclosed herein. RAL-traps lower the amount of A2E formed, for example by forming a covalent bond with RAL that has escaped sequestering. RAL that has reacted with a RAL-trap compound is thereby unavailable to react with phosphatidylethanolamine.
  • parenteral administration and “administered parenterally” are art- recognized terms, and include modes of administration other than enteral and topical administration, such as injections, and include, without limitation, intravenous, intramuscular, intrapleural, intravascular, intrapericardial, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra- articular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • “about” or “approximately” in reference to a numerical value means that the stated numerical value may vary by up to 10% of the stated value.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. 4. Uses of Compounds and Pharmaceutically Acceptable Compositions Thereof [00181] Certain compounds described herein are found to be useful in scavenging toxic aldehydes, such as MDA and HNE.
  • compounds described herein undergo a Schiff base condensation with MDA, HNE, or other toxic aldehydes, and form a complex with the aldehydes in an energetically favorable reaction, thus reducing or eliminating aldehydes available for reaction with a protein, lipid, carbohydrate, or DNA.
  • compounds described herein can react with aldehydes to form a compound having a cyclic structure that contains the aldehydes, thus trapping the aldehydes and preventing the aldehydes from being released back into the cellular milieu.
  • the present invention provides a method for reducing levels of one or more toxic aldehydes in a subject, comprising administering to a subject in need thereof a disclosed compound or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as described herein.
  • the present invention provides a method for reducing levels of one or more toxic aldehydes in a biological sample, comprising contacting the biological sample with a disclosed compound or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as described herein. In some embodiments, the method is carried out in vitro.
  • the toxic aldehyde is selected from formaldehyde, acetaldehyde, acrolein, glyoxal, methylglyoxal, hexadecanal, octadecanal, hexadecenal, succinic semi-aldehyde, malondialdehyde, 4-hydroxynonenal, 4-hydroxy-2E-hexenal, 4-hydroxy-2E,6Z- dodecadienal, retinaldehyde, leukotriene B4 aldehyde, and octadecenal.
  • the toxic aldehyde is formaldehyde.
  • the toxic aldehyde is acetaldehyde. In some embodiments, the toxic aldehyde is acrolein. In some embodiments, the toxic aldehyde is glyoxal. In some embodiments, the toxic aldehyde is methylglyoxal. In some embodiments, the toxic aldehyde is hexadecanal. In some embodiments, the toxic aldehyde is octadecanal. In some embodiments, the toxic aldehyde is hexadecenal. In some embodiments, the toxic aldehyde is succinic semi-aldehyde (SSA).
  • SSA succinic semi-aldehyde
  • the toxic aldehyde is malondialdehyde (MDA). In some embodiments, the toxic aldehyde is 4-hydroxynonenal. In some embodiments, the toxic aldehyde is retinaldehyde. In some embodiments, the toxic aldehyde is 4-hydroxy-2E-hexenal. In some embodiments, the toxic aldehyde is 4-hydroxy-2E,6Z-dodecadienal. In some embodiments, the aldehyde is leukotriene B4 aldehyde. In some embodiments, the aldehyde is octadecenal. [00186] In some embodiments, the compound reduces systemic inflammation in the patient.
  • MDA malondialdehyde
  • the toxic aldehyde is 4-hydroxynonenal. In some embodiments, the toxic aldehyde is retinaldehyde. In some embodiments, the toxic aldehyde is 4-hydroxy-2E-hexenal. In some embodiments,
  • the compound reduces plasma levels of a biomarker selected from IL-1 ⁇ , IL-6, IL-10, and tumor necrosis factor alpha. In some embodiments, the compound reduces plasma levels of a biomarker selected from a RASP. In some embodiments, the RASP is malondialdehyde (MDA) and/or 4-hydroxynonenal (4-HNE). [00188] In some embodiments, the method further comprises a reduction in the level of a reactive aldehyde species (RASP) in the patient’s blood, such as malondialdehyde (MDA) or 4- hydroxynonenal (HNE).
  • RASP reactive aldehyde species
  • the level of RASP is reduced by at least 30%, at least 40%, or at least 50%. In some embodiments, the level of RASP is reduced by about 30% to 75%. In some embodiments, the level of RASP is reduced by about 20% to about 60%, or about 20% to about 50%, or about 20% to about 30%. [00190] In another aspect, the present invention provides a method for treating a disease, disorder, or condition described herein, comprising administering to a subject in need thereof a disclosed compound or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as described herein.
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment is administered after one or more symptoms have developed.
  • treatment is administered in the absence of symptoms.
  • treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors).
  • treatment is also continued after symptoms have resolved, for example to prevent, delay or lessen the severity of their recurrence.
  • the invention relates to compounds described herein for the treatment, prevention, and/or reduction of a risk of diseases, disorders, or conditions in which aldehyde toxicity is implicated in the pathogenesis.
  • diseases, disorders, or conditions in which aldehyde toxicity is implicated include an ocular disease, disorder, or condition, including, but not limited to, a corneal disease (e.g., dry eye syndrome, cataracts, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy), other ocular disorders or conditions (e.g., allergic conjunctivitis, ocular cicatricial pemphigoid, conditions associated with PRK healing and other corneal healing, and conditions associated with tear lipid degradation or lacrimal gland dysfunction), and other ocular conditions associated with high aldehyde levels as a result of inflammation (e.g., uveitis, scleritis, ocular Stevens Johnson Syndrome,
  • a corneal disease e
  • the ocular disease, disorder, or condition is dry eye syndrome, ocular rosacea, or uveitis.
  • the invention relates to the treatment, prevention, and/or reduction of a risk of an ocular disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis, comprising administering to a subject in need thereof a compound described herein.
  • the ocular disease, disorder, or condition includes, but is not limited to, a corneal disease (e.g., dry eye syndrome, cataracts, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy in the cornea), other ocular disorders or conditions (e.g., allergic conjunctivitis, ocular cicatricial pemphigoid, conditions associated with PRK healing and other corneal healing, and conditions associated with tear lipid degradation or lacrimal gland dysfunction), and other ocular conditions where inflammation leads to high aldehyde levels (e.g., uveitis, scleritis, ocular Stevens Johnson Syndrome, ocular rosacea (with or without meibomian gland dysfunction)).
  • a corneal disease e.g., dry eye syndrome, cataracts, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy in the cornea
  • the ocular disease, disorder, or condition is macular degeneration. In some embodiments, the ocular disease, disorder, or condition is AMD or Stargardt’s disease.
  • the amount or concentration of MDA or HNE is increased in the ocular tissues or cells.
  • the amount or concentration of aldehydes e.g., MDA or HNE
  • the amount or concentration of aldehydes is increased for at least 1.1 fold, 1.2 fold, 1.3 fold, 1.4 fold, 1.5 fold, 2 fold, 2.5 fold, 5 fold, 10 fold as compared to that in normal ocular tissues or cells.
  • Compounds described herein decrease aldehyde (e.g., MDA and/or HNE) concentration in a concentration-dependent manner.
  • the amount or concentration of aldehydes can be measured by methods or techniques known in the art, such as those described in Tukozkan et al., Furat Tip Dergisi 11: 88- 92 (2006).
  • the ocular disease, disorder, or condition is dry eye syndrome.
  • the ocular disease, disorder, or condition is a condition associated with PRK healing and other corneal healing.
  • the invention is directed to advancing PRK healing or other corneal healing, comprising administering to a subject in need thereof a compound described herein.
  • the ocular disease, disorder, or condition is an ocular condition associated with high aldehyde levels as a result of inflammation (e.g., uveitis, scleritis, ocular Stevens Johnson Syndrome, and ocular rosacea (with or without meibomian gland dysfunction).
  • the ocular disease, disorder, or condition is keratoconus, cataracts, bullous and other keratopathy, Fuchs’ endothelial dystrophy, ocular cicatricial pemphigoid, or allergic conjunctivitis.
  • the compound described herein may be administered topically or systemically, as described herein below.
  • the present invention is also directed to the use of a compound described herein in the manufacture of a medicament for the treatment, prevention, and/or reduction of a risk of a disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis.
  • this aspect of the invention is directed to the use of a compound described herein in the manufacture of a medicament for the treatment, prevention, and/or reduction of a risk of (1) an ocular disease, disorder, or condition, including, but not limited to, a corneal disease (e.g., dry eye syndrome, cataracts, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy), other ocular disorders or conditions (e.g., allergic conjunctivitis, ocular cicatricial pemphigoid, conditions associated with PRK healing and other corneal healing, and conditions associated with tear lipid degradation or lacrimal gland dysfunction), and other ocular conditions associated with high aldehyde levels as a result of inflammation (e.g., uveitis, scleritis, ocular Stevens Johnson Syndrome, and ocular rosacea (with or without meibomian gland dysfunction)), (2) a skin disorder or condition or a cosmetic indication.
  • the present invention is also directed to the use of a compound described herein in treating, preventing, and/or reducing a risk of a disease, disorder, or condition in which aldehyde toxicity is implicated in the pathogenesis. More specifically, this aspect of the invention is directed to the use of a compound described herein in treating, preventing, and/or reducing a risk of an ocular disease, disorder, or condition, including, but not limited to, a corneal disease (e.g., dry eye syndrome, cataracts, keratoconus, bullous and other keratopathy, and Fuch’s endothelial dystrophy), other ocular disorders or conditions (e.g., allergic conjunctivitis, ocular cicatricial pemphigoid, conditions associated with PRK healing and other corneal healing, and conditions associated with tear lipid degradation or lacrimal gland dysfunction), and other ocular conditions associated with high aldehyde levels as a result of inflammation (e.g., uve
  • the atopic dermatitis treated is in the acute phase. In some embodiments, acute atopic dermatitis presents with a vesicular, weeping, crusting eruption. [00199] In some embodiments, the atopic dermatitis treated is in the subacute phase. In some embodiments, subacute atopic dermatitis presents with dry, scaly, erythematous papules and plaques. [00200] In some embodiments, the DME treated is clinically significant macular edema (CSME).
  • CSME macular edema
  • CSME is defined as DME meeting at least one of the criteria presented as follows: (a) thickening of the retina at or within 500 ⁇ m of the center of the macula; (b) hard exudates at or within 500 ⁇ m of the center of the macula, if associated with thickening of adjacent retina (not counting residual hard exudates remaining after disappearance of retinal thickening); and (c) any zone(s) of retinal thickening 1 disc area or larger, any part of which is within 1 disc diameter of the center of the macula.
  • the DME treated is center-involved DME.
  • the central macula is generally the thickest portion of the retina and is an inversion of the normal morphology.
  • the DME treated is non-center-involved DME. Non-central DME lack involvement of the center of the macula.
  • the DME treated is focal DME. Focal edema often occurs associated with a cluster of microaneurysms, sometimes surrounded by an incomplete ring of hard exudates. It may be associated with less macular thickening, better visual acuity, and less severe retinopathy severity.
  • the DME treated is diffuse type DME.
  • Diffuse macular edema occurs from dilated retinal capillaries in the retina and involve a larger area of retinal thickening.
  • the DME treated is accompanied by retinal detachment or severe non-clearing vitreous hemorrhage.
  • the patient treated has undergone focal laser photocoagulation therapy.
  • the patient treated has undergone grid laser photocoagulation therapy.
  • a method of the disclosure is directed to treatment of Stargardt’s disease.
  • a method of treating or reducing the risk of Stargardt’s disease comprises administering to a patient in need thereof an effective amount of a compound disclosed herein.
  • Stargardt’s disease is inherited form of macular dystrophy characterized by bilateral vision loss, including dyschromatopsia and central scotomata, with characteristic macular atrophy and yellow-white flecks at the level of the retinal pigment epithelium (RPE) at the posterior pole.
  • Stargardt’s disease may also be referred to as Stargardt macular dystrophy, juvenile macular degeneration, or fundus flavimaculatus.
  • Onset of Stargardt’s disease occurs most commonly in childhood, with the next peak being early adulthood, and least frequently in later adulthood. Better prognosis is generally associated with a later onset.
  • the Stargardt’s disease treated is childhood-onset Stargardt’s disease.
  • the Stargardt’s disease treated is adult-onset or late onset Stargardt’s disease.
  • the severity of Stargardt’s disease can be classified based on electrophysiological assessment (see, e.g., Tanna et al., British Journal of Ophthalmology 2017; 101:25-30).
  • the Stargardt’s disease treated is classified in Group 1.
  • Stargardt’s disease in Group 1 display a severe pattern electroretinogram (ERG) abnormality (macular dysfunction) with normal full-field ERGs.
  • ERG electroretinogram
  • the Stargardt’s disease treated is classified in Group 2.
  • Stargardt’s disease in Group 2 display the characteristics of Group 1 and have in addition generalised loss of cone function. Patients in Group 2 have intermediate variable prognosis.
  • the Stargardt’s disease treated is classified in Group 3.
  • Stargardt’s disease in Group 3 display an additional generalised loss of both cone and rod function. Patients in Group 3 show the worst prognosis.
  • the patient treated is identified as having a mutation in retina- specific ATP-binding cassette transporter (ABCA4) gene resulting in reduction or defect in ABCA4 function. Mutations in ABCA4 are the most common forms of inherited Stargardt’s disease.
  • the patient treated is identified as having mutation in ABCA4 gene that are associated with childhood-onset Stargardt’s disease.
  • Exemplary mutations associated with childhood-onset Stargardt’s disease include, among others, 634C>T, 768G>T, 1317G>A, 1531C>T, 1557C>A, 5308T>G, 6088C>T, or 6449G>A.
  • the patient treated is identified as having mutation in ABCA4 gene that are associated with adult-onset or late onset Stargardt’s disease.
  • Exemplary mutations associated with adult-onset or late onset Stargardt’s disease include, among others, 769-784C>T, 2486C>T, 5603A>T, or 5882G>A.
  • the compound or pharmaceutically acceptable salt thereof described herein can be administered systemically to treat the indications described herein.
  • the pharmaceutical composition is a liquid.
  • the disease, disorder, or condition for treatment with the compounds of the disclosure is an ocular inflammatory disorder.
  • the ocular inflammatory disorder is diabetic macular edema (DME), atopic keratoconjunctivitis (AKC), vernal keratoconjunctivitis (VKC), age-related macular degeneration (AMD), retinitis pigmentosa (RP), dry eye disease (DED), allergic conjunctivitis (AC), dry eye disease with allergic conjunctivitis, noninfectious anterior uveitis, posterior uveitis, pan-uveitis, post-surgical ocular pain and inflammation.
  • DME diabetic macular edema
  • ATC age-related macular degeneration
  • RP retinitis pigmentosa
  • DED dry eye disease
  • AC allergic conjunctivitis
  • dry eye disease with allergic conjunctivitis noninfectious anterior uveitis, posterior uveitis, pan-uveitis, post-surgical ocular pain and inflammation.
  • the disease, disorder, or condition is one of
  • the disease, disorder, or condition is one of those described in WO 2023/278816, which is hereby incorporated by reference.
  • the compound or pharmaceutically acceptable salt thereof is administered once, twice, thrice, or four times per day. In some embodiments, the compound or pharmaceutically acceptable salt thereof is administered twice per day.
  • the dose of the compound or pharmaceutically acceptable salt thereof is administered systemically.
  • the dose of the compound or pharmaceutically acceptable salt thereof is administered orally.
  • the pharmaceutical composition is a liquid. In some embodiments, the pharmaceutical composition is administered as a liquid via nasogastric tube.
  • a disclosed compound may be administered to a subject in order to treat or prevent macular degeneration and other forms of retinal disease whose etiology involves the accumulation of A2E and/or lipofuscin. Other diseases, disorders, or conditions characterized by the accumulation of A2E may be similarly treated.
  • a compound is administered to a subject that reduces the formation of A2E.
  • the compound may compete with PE for reaction with trans- RAL, thereby reducing the amount of A2E formed.
  • a compound is administered to a subject that prevents the accumulation of A2E.
  • the compound competes so successfully with PE for reaction with trans-RAL, no A2E is formed.
  • compositions are administered topically or systemically at one or more times per month, week or day. Dosages may be selected to avoid side effects, if any, on visual performance in dark adaptation. Treatment is continued for a period of at least one, three, six, or twelve or more months. Patients may be tested at one, three, six, or twelve months or longer intervals to assess safety and efficacy. Efficacy is measured by examination of visual performance and retinal health as described above. [00229] In one embodiment, a subject is diagnosed as having symptoms of macular degeneration, and then a disclosed compound is administered.
  • a subject may be identified as being at risk for developing macular degeneration (risk factors include a history of smoking, age, female sex, and family history), and then a disclosed compound is administered.
  • risk factors include a history of smoking, age, female sex, and family history
  • a disclosed compound is administered.
  • a subject may have dry AMD in both eyes, and then a disclosed compound is administered.
  • a subject may have wet AMD in one eye but dry AMD in the other eye, and then a disclosed compound is administered.
  • a subject may be diagnosed as having Stargardt’s disease and then a disclosed compound is administered.
  • a subject is diagnosed as having symptoms of other forms of retinal disease whose etiology involves the accumulation of A2E and/or lipofuscin, and then the compound is administered.
  • a subject may be identified as being at risk for developing other forms of retinal disease whose etiology involves the accumulation of A2E and/or lipofuscin, and then the disclosed compound is administered.
  • a compound is administered prophylactically.
  • a subject has been diagnosed as having the disease before retinal damage is apparent. For example, a subject is found to carry a gene mutation for ABCA4 and is diagnosed as being at risk for Stargardt’s disease before any ophthalmologic signs are manifest, or a subject is found to have early macular changes indicative of macular degeneration before the subject is aware of any effect on vision.
  • a human subject may know that he or she is in need of the macular generation treatment or prevention.
  • a subject may be monitored for the extent of macular degeneration.
  • a subject may be monitored in a variety of ways, such as by eye examination, dilated eye examination, fundoscopic examination, visual acuity test, and/or biopsy. Monitoring can be performed at a variety of times. For example, a subject may be monitored after a compound is administered. The monitoring can occur, for example, one day, one week, two weeks, one month, two months, six months, one year, two years, five years, or any other time period after the first administration of a compound. A subject can be repeatedly monitored. In some embodiments, the dose of a compound may be altered in response to monitoring.
  • the disclosed methods may be combined with other methods for treating or preventing macular degeneration or other forms of retinal disease whose etiology involves the accumulation of A2E and/or lipofuscin, such as photodynamic therapy.
  • a patient may be treated with more than one therapy for one or more diseases or disorders.
  • a patient may have one eye afflicted with dry form AMD, which is treated with a compound of the invention, and the other eye afflicted with wet form AMD which is treated with, e.g., photodynamic therapy.
  • a compound for treating or preventing macular degeneration or other forms of retinal disease whose etiology involves the accumulation of A2E and/or lipofuscin may be administered chronically.
  • the compound may be administered daily, more than once daily, twice a week, three times a week, weekly, biweekly, monthly, bimonthly, semiannually, annually, and/or biannually. 5.
  • the present invention provides a combination pharmaceutical composition comprising: a compound of formula I: or a pharmaceutically acceptable salt thereof, wherein each of A, R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is as defined herein; and a compound of formula II: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a combination pharmaceutical composition comprising an aldehyde trap; and a compound of formula II: or a pharmaceutically acceptable salt thereof, wherein each of CD, R, X and Y is as defined herein.
  • the present invention provides a combination pharmaceutical composition
  • a combination pharmaceutical composition comprising: a compound of formula I: or a pharmaceutically acceptable salt thereof, wherein each of A, R 1 , R 2 , R 3 , R 4 , R 5 , and R 8 is as defined herein; and an aldehyde trap.
  • An “aldehyde trap” is any compound that can react to form a covalent bond with an aldehyde moiety. In some cases, the aldehydes are by-products of a disorder or a disease process.
  • the compounds described herein are compounds that have aldehyde trapping activity, and have been described for use in treating disorders and diseases associated with the effects of toxic aldehydes.
  • the compound for use in a combination therapy with a compound of formula I and/or a compound of formula II is a compound of formula III: III or a pharmaceutically acceptable salt thereof, wherein: Q, T and V are independently S, N, O, or -C-R; each of R 1 , R 6 , R 7 , and R 8 is independently H, D, halogen, -NH2, -CN, -OR, -SR, optionally substituted C1-6 aliphatic, or , wherein one of R 1 , R 6 , R 7 , and R 8 is –NH2 and other one of R 1 , R 6 , R 7 , and R 8 is ; R a is C 1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; and R b is C1-4 aliphatic optionally substituted with 1, 2, or 3 deuterium or halogen atoms; or R a and R b , taken together with the
  • the –NH 2 on one of R 1 , R 6 , R 7 , and R 8 and the carbinol on the other of R 1 , R 6 , R 7 , and R 8 are on adjacent carbon atoms of the phenyl moiety.
  • one of Q, T and V is N, and other of Q, T and V is O.
  • Q is O
  • V is N
  • T is C-R.
  • Q is N
  • T is O and V is C-R.
  • the compound is a compound of formula III-a or III-b: III-a III-b or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C1-6 aliphatic, or , wherein one of R 1 , R 6 , R 7 , and R 8 is Q, T, V, R, R a and R b are as defined in formula III.
  • the compound is a compound of formula III-c, III-d or III-e: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , and R 7 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C1-6 aliphatic; and Q, T, V, R, R a and R b are as defined in formula III.
  • the compound is a compound of formula III-f, III-g, III-h or III-i: III-f III-g III-h III-i or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C1-6 aliphatic or , wherein one of R 1 , R 6 , R 7 , and R 8 is R, R a and R b are as defined in formula III.
  • the compound is a compound of formula III-j, III-k, III-l or III-m: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C 1-6 aliphatic; and R, R a and R b are as defined in formula III.
  • the compound is a compound of formula III-n: III-n or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 is independently H, D, halogen, -NH 2 , -CN, -OR, -SR, optionally substituted C1-6 aliphatic, or rein one of R 1 , R 6 , R 7 , and R 8 is –NH2 and other one of R 1 , R 6 , R 7 , and R 8 R, R a , and R b are as defined in formula III.
  • the compound is a compound of formula III-o, III-p, III-q or III-r: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C1-6 aliphatic, or , wherein one of R 1 , R 6 , R 7 , and R 8 is R, R a , and R b are as defined in formula III.
  • the compound is a compound of formula III-s, III-t, III-u, III-v, III-w, or III-x: or a pharmaceutically acceptable salt thereof, wherein: each of R 1 , R 6 , R 7 , and R 8 when present is independently H, D, halogen, -CN, -OR, -SR, optionally substituted C1-6 aliphatic; and R, R a and R b are as defined in formula III.
  • the compound for use in a combination therapy with a compound of formula I and/or a compound of formula II is a compound selected from the following: [00249] In some embodiments, the compound for use in combination therapy with a compound of formula I and/or a compound of formula II is a compound selected from the following:
  • the compound for use in a combination therapy with a compound of formula I, X-I, X-VI, Y-I, Y-II, Y-III, or Y-IV and/or a compound of formula II is a compound selected from: [00251]
  • the present invention provides methods for treating an ocular disorder, such as age-related macular degeneration (AMD), retinitis pigmentosa (RP), and dark adaptation impairment with a combination pharmaceutical composition comprising a compound of formula I, X-I, X-VI, Y-I to Y-IV or a pharmaceutically acceptable salt thereof, and a compound of formula II or a modified cyclodextrin compound described herein or a pharmaceutically acceptable salt thereof.
  • AMD age-related macular degeneration
  • RP retinitis pigmentosa
  • dark adaptation impairment with a combination pharmaceutical composition comprising a compound of formula I, X-I, X-VI, Y-I to Y-IV or a pharmaceutically
  • a compound of formula II may be used in other combination therapies, with or without a compound of formula I, X-I, X-VI, Y-I to Y-IV present.
  • the compound of formula II or a modified cyclodextrin compound described herein is used, with or without an aldehyde trapping compound, in combination with a co-administered agent in the form of eye drops, a sustained release delivery system to the eye, a cream, a paste, or an ointment containing an anti-inflammatory agent, antihistamine, antibiotic, or other agent.
  • a method of treating wet AMD further comprises co- administering a known treatment for AMD, such as a VEGF inhibitor or photodynamic therapy.
  • VEGF inhibitors include brolucizumab (Beovu®), aflibercept (Eylea®), ranibizumab (Lucentis®), pegaptanib sodium (Macugen®), faricimab-svoa (VABYSMO®), ranibizumab injection for intravitreal use via ocular implant (SUSVIMO TM ), and Avastin® (bevacizumab).
  • the co-administered treatment is RGX-314.
  • a method of treating retinitis pigmentosa further comprises co-administering a known treatment for RP, such as UV-reducing glasses or voretigene neparvovec-ryzl (Luxturna®).
  • a known treatment for RP such as UV-reducing glasses or voretigene neparvovec-ryzl (Luxturna®).
  • the compound of formula II or a modified cyclodextrin compound described herein above is administered in combination with an anti-inflammatory agent.
  • Anti-inflammatory agents useful for treating ocular inflammation in combination with the compound of formula II or a modified cyclodextrin compound described herein include, among others, a steroid, non-steroidal anti-inflammatory compound, anti-metabolite, immunosuppressive antibiotic, alkylating agent, and an anti- inflammatory cytokine antibody.
  • the anti-inflammatory agent is a steroid, particularly a steroid with immunomodulatory, anti-inflammatory properties.
  • the anti- inflammatory is a glucocorticosteroid, which is widely used as effective treatments to control inflammatory and autoimmune diseases.
  • the glucocorticosteroid is a corticosteroid or a prodrug thereof.
  • the corticosteroid is selected from cortisol, cortisone, prednisone, prednisolone, methylprednisone, triamcinolone, betamethasone, dexamethasone and a prodrug thereof.
  • the corticosteroid prodrug is prednisolone acetate.
  • the compound of formula II or a modified cyclodextrin compound described herein is used in combination with a non-steroidal anti-inflammatory compound.
  • cyclooxygenase COX
  • COX-1 COX-1
  • COX-2 cyclooxygenase-2
  • Different classes of non-steroidal anti-inflammatory compounds includes salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid, anthranilic acid derivatives, and selective cox inhibitors.
  • non-steroidal anti-inflammatory compounds useful in the combination include, among others, acetylsalicylic acid, diflunisal, salsalate, ibuprofen, dexibuprofen, naioxen, fenoprofen, ketoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxoprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, aceclofenac, nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, phenylbutazone, mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, and celecoxib.
  • the non-steroidal anti-inflammatory compound can be selected from diclofenac, flurbiprofen, ketoprofen, bromfenac, and nepafenac.
  • the anti-inflammatory agent for use in combination with the compound of formula II or a modified cyclodextrin compound described herein is an anti- metabolite.
  • the anti-metabolite is a cytotoxic purine analog, for example, azathioprine and mercaptopurine.
  • the anti-metabolite is an anti-folate compound.
  • Exemplary anti-folate compound is methotrexate.
  • the anti- metabolite is an inosine monophosphate dehydrogenase inhibitor (IMPDH), such as mycophenolic acid and its prodrug mycophenolate mofetil.
  • IMPDH inosine monophosphate dehydrogenase inhibitor
  • the anti- metabolite for use in the combination therapy is a dihydroorotate dehydrogenase inhibitor, such as leflunomide.
  • the anti-inflammatory agent for use in combination with the compound of formula II or a modified cyclodextrin compound described herein is an alkylating agent.
  • alkylating agents include, among others, cyclophosphamide and chlorambucil.
  • the anti-inflammatory agent for use in combination with the compound of formula II or a modified cyclodextrin compound described herein is an immunomodulatory antibiotic.
  • Immunosuppressive antibiotics useful for the treatments herein include those that bind cyclophilin and/or calcineurin and inhibit T-lymphocyte activity.
  • the immunomodulatory antibiotic is selected from cyclosporine, tacrolimus, rapamycin, and derivatives thereof.
  • the anti-inflammatory agent for use in the combination therapy is an anti-inflammatory antibody, such as an antibody targeting an inflammatory cytokine, such as TNF, IL-1, IL-4, IL-5, or IL-17, or targeting an immune system cell active in the inflammatory response, such as B cells expressing CD20.
  • the anti- inflammatory biologic agent is selected from etanercept, infliximab, adalimumab, daclizumab, rituximab, toclizumab, certolizumab pegol, and golimumab.
  • the compounds of general formula II or a modified cyclodextrin compound described herein may be used with a therapeutic agent for treating inflammatory diseases or disorders of the eye.
  • the composition for treating eye inflammation comprises a cyclodextrin and a therapeutic agent selected from an anti-histamine, alpha-adrenergic agonist, non-steroidal anti-inflammatory drug (NSAID), mast cell inhibitor or stabilizer, corticosteroid, adhesion molecule inhibitor, T-cell activation inhibitor and combinations thereof.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and an anti-histamine.
  • the anti-histamine is selected from the group consisting of ketotifen, phenylephrine, tetrahydrozaline, pheniramine, alcaftadine, azelastine, emedastine, epinastine, and bepotastine.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and an alpha-adrenergic agonist.
  • the alpha-adrenergic agonist is selected from the group consisting of oxymetazoline, brimonidine, cirazoline, apraclonidine hydrochloride, and naphazoline.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and a non-steroidal anti-inflammatory drug.
  • the non-steroidal anti-inflammatory drug is selected from the group consisting of diclofenac, flurbiprofen, ketorolac, bromfenac, indomethacin and nepafenac.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and a mast cell inhibitor or stabilizer.
  • the mast cell inhibitor or stabilizer is selected from the group consisting of cromolyn, nedocromil, pemirolast, nedocromil sodium, tranilast, lodoxamide (e.g., lodoxamide tromethane), and lodoxamine (e.g., lodoxamine ethyl).
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and an anti-inflammatory steroid.
  • the anti-inflammatory steroid is a corticosteroid.
  • the steroid is selected from the group consisting of lotoprednol, rimexolone, difluprednate, fluorometholone, prednisone and prednisolone.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and an adhesion molecule inhibitor, in particular an integrin antagonist.
  • the integrin agonist is lifitegrast (lymphocyte function-associated antigen 1 – LFA-1 antagonist).
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and a T-cell activation inhibitor.
  • the T-cell activation inhibitor is selected from the group consisting of cyclosporine, tacrolimus, pimecrolimus, and rapamycin.
  • the composition comprises a compound of general formula II or a modified cyclodextrin compound described herein and two or more of therapeutic agents selected from anti-histamine, alpha-adrenergic agonist, non-steroidal anti-inflammatory drug (NSAID), mast cell inhibitor or stabilizer, corticosteroid, adhesion molecule inhibitor, and T-cell activation inhibitor.
  • the compound of general formula II or a modified cyclodextrin compound described herein is present in excess of the therapeutic agent.
  • the composition further comprises one or more ophthalmic pharmaceutically acceptable additive or excipient.
  • the one or more pharmaceutically acceptable additive or excipient is selected from a tonicity agent, solubilizing agent, stabilizing agent, preservative, buffering agent, wetting agent, viscosity enhancing agent, lubricating agent, chelating agent, and antioxidant.
  • the composition comprising a compound of general formula II or a modified cyclodextrin compound described herein and a therapeutic agent is used for treating an eye disease or disorder having an inflammatory response, or in some embodiments, for treating an inflammatory eye disease or disorder.
  • the compositions herein are used for treating allergic conjunctivitis, scleritis, blepharitis, keratitis, retinitis, cyclitis, iridocyclitis choroiditis, dry-eye syndrome (keratoconjunctivitis sicca), ulceris, meibomian gland dysfunction, sarcoidosis, or uveitis.
  • the compositions are used in treating inflammation of the eye associated with injury or trauma to the eye.
  • the compositions are used for treating inflammation associated with cataract surgery, laser surgery, keratotomy, keratoplasty, corneal surgery, accidental (non-iatrogrenic) trauma or vitrectomy.
  • the compositions herein are used for treating ocular inflammation associated with eye infections.
  • the compositions are used for treating inflammation associated with fungal (e.g., keratomycosis, fungal keratitis, etc.), bacterial, protozoal or viral infections, including conjunctivitis, keratitis, cyclitis, iridocyclitis and blepharitis associated with eye infections.
  • the compositions herein are used for treating irritation or inflammation associated with exposure of the eye to chemical irritants. These include irritants such as smoke, volatile organic compounds, cosmetics), dust, and use of computer monitors.
  • the ocular inflammatory disorder treatable with the combination of the compound of formula II or a modified cyclodextrin compound described herein and the anti-inflammatory agent include allergic conjunctivitis, including vernal keratoconjunctivitis and atopic keratoconjunctivitis; dry eye syndrome and meibomian gland dysfunction; cataracts; keratoconus; bullous and other keratopathy; Fuch's endothelial dystrophy; ocular cicatricial pemphigoid; conditions associated with photoreactive keratotomy (PRK) healing and other corneal healing; conditions associated with tear lipid degradation or lacrimal gland dysfunction; uveitis, including anterior uveitis, intermediate uveitis, posterior uveitis, panuveitis, non- infectious uveitis, and infectious uveitis; keratitis; scleritis; ulceris; cyclitis; ocular graft
  • the combination therapy is used to treat uveitis.
  • the uveitis treated with the combination is anterior uveitis, which includes the disorders of iridocyclitis and ulceris and involves the anterior chamber and iris, with the latter indication also affecting the ciliary body.
  • the vast majority of uveitis is anterior uveitis.
  • the uveitis treatable with the combination is intermediate uveitis, also referred to as pars planitis, which involves inflammation of cells in the vitreous cavity or deposition of inflammatory material on the pars plana.
  • the uveitis treatable with the combination is posterior uveitis, which involves inflammation of the uveal tract in the retina and the choroid.
  • the uveitis treatable with the combination is panuveitis (i.e., diffuse uveitis), which is inflammation of the uveal tract of both the anterior segment (e.g., iris and ciliary body) and the posterior segment (e.g., retina and choroid).
  • the combination of the compound of formula II and the anti- inflammatory agent is used to treat macular degeneration.
  • a method of treating macular degeneration comprises administering to a subject in need thereof a therapeutically effective amount a compound of formula II or a modified cyclodextrin compound described herein, particularly compound I-1, for example, and an anti-inflammatory agent, as described herein.
  • a compound of formula II or a modified cyclodextrin compound described herein particularly compound I-1, for example
  • an anti-inflammatory agent as described herein.
  • the form of macular degeneration which can be treated is selected from age-related macular degeneration; Stargardt disease, an inherited form of juvenile macular degeneration; wet macular degeneration; dry macular degeneration; and geographic atrophy (GA) secondary to dry age related macular degeneration.
  • the compounds and compositions, according to the method of the present invention are administered using any amount and any route of administration effective for treating or lessening the severity of a disease, disorder, or condition provided above. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • Unit dosage form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention are administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents,
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a compound of the present invention In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • additional substances other than inert diluents e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin.
  • the rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of the invention can also be administered topically, such as directly to the eye, e.g., as an eye-drop or ophthalmic ointment.
  • Eye drops typically comprise an effective amount of at least one compound of the invention and a carrier capable of being safely applied to an eye.
  • the eye drops are in the form of an isotonic solution, and the pH of the solution is adjusted so that there is no irritation of the eye.
  • the epithelial barrier interferes with penetration of molecules into the eye.
  • penetration enhancers work by loosening the tight junctions of the most superior epithelial cells (Burstein, 1985, Trans Ophthalmol Soc U K 104(Pt 4): 402-9; Ashton et al., 1991, J Pharmacol Exp Ther 259(2): 719-24; Green et al., 1971, Am J Ophthalmol 72(5): 897-905).
  • Topical administration may be in the form of a cream, suspension, emulsion, ointment, drops, oil, lotion, patch, tape, inhalant, spray, or controlled release topical formulations including gels, films, patches, and adhesives.
  • Intra-ocular administration may take the form of subconjunctival, subtenon’s capsule, retrobulbar or intravitreal injections, depots or implants.
  • Compounds administered by these routes may be in solution or suspension form.
  • Administration of compounds by depot injection may contain pharmaceutically acceptable carriers or excipients; these may be natural or synthetic and may be biodegradable or non-biodegradable and facilitate drug release in a controlled manner.
  • Implants used for controlled release of compound may be composed of natural or synthetic, biodegradable or non-biodegradable materials.
  • the carrier is acceptable in that it is compatible with the other components of the composition and is not injurious to the patient.
  • Formulations for topical administration to the skin can include, for example, ointments, creams, gels and pastes comprising the primary amine compound in a pharmaceutical acceptable carrier.
  • the formulation of the primary amine compound for topical use includes the preparation of oleaginous or water-soluble ointment bases, as is well known to those in the art.
  • these formulations may include vegetable oils, animal fats, and, for example, semisolid hydrocarbons obtained from petroleum.
  • Particular components used may include white ointment, yellow ointment, cetyl esters wax, oleic acid, olive oil, paraffin, petrolatum, white petrolatum, spermaceti, starch glycerite, white wax, yellow wax, lanolin, anhydrous lanolin and glyceryl monostearate.
  • Various water-soluble ointment bases may also be used, including glycol ethers and derivatives, polyethylene glycols, polyoxyl 40 stearate and polysorbates.
  • the formulations for topical administration may contain the compound used in the present application at a concentration in the range of 0.001-10%, 0.05-10%, 0.1-10%, 0.2-10%, 0.5-10%, 1-10%, 2-10%, 3-10%, 4-10%, 5-10%, or 7-10% (weight/volume), or in the range of 0.001-2.0%, 0.001-1.5%, or 0.001-1.0%, (weight/volume), or in the range of 0.05-2.0%, 0.05- 1.5%, or 0.05-1.0%, (weight/volume), or in the range of 0.1-5.0%, 0.1-2.0%, 0.1-1.5%, or 0.1- 1.0% (weight/volume), or in the range of 0.5-5.0%, 0.5-2.0%, 0.5-1.5%, or 0.5-1.0% (weight/volume), or in the range of 1-5.0%, 1-2.0%, or 1-1.5% (weight/volume).
  • the formulations for topical administration may also contain the compound used in the present application at a concentration in the range of 0.001-2.5%, 0.01-2.5%, 0.05-2.0%, 0.1-2.0%, 0.2- 2.0%, 0.5-2.0%, or 1-2.0% (weight/weight), or in the range of 0.001-2.0%, 0.001-1.5%, 0.001- 1.0%, or 0.001-5% (weight/weight).
  • the composition may contain the active compound at a concentration of 0.01-20%, 0.02-15%, 0.04-10%, 0.06-5%, 0.08-1%, or 0.09-0.5% (weight/volume) with or without pH and/or osmotic adjustment to the solution.
  • the eye drop formulation may contain a compound described herein at a concentration of 0.09-0.5% (weight/volume), such as 0.1%, 0.25%, or 0.5%.
  • the compounds described herein may be administered orally and as such the pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum starches, agar, alginic acid or its sodium salt, or effervescent mixtures, croscarmellose or its sodium salt, and the like.
  • Diluents include, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • Parenteral formulations comprising a compound described herein can be prepared in aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • the formulations may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutic agents.
  • compositions are prepared according to conventional methods, and may contain about 0.1 to 75%, preferably about 1 to 50%, of a compound described herein.
  • the present invention is directed to a composition, as described herein, comprising a prodrug of a compound of formula I, X-I, X-VI, Y-I, Y-II, Y-III or Y-IV or II or a modified cyclodextrin compound as described herein, or a pharmaceutically acceptable salt thereof.
  • prodrug means a compound that is convertible in vivo by metabolic means (e.g.
  • prodrugs are known in the art such as those discussed in, for example, Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard- Larsen, et al., (ed).
  • Impairments in Dark Adaptation are Associated with Age-Related Macular Degeneration Severity and Reticular Pseudodrusen. Ophthalmology, 122(10), 2053–2062. 3. Loftsson T, Brewster ME. Pharmaceutical applications of cyclodextrins.1. Drug solubilization and stabilization. J Pharm Sci.1996 Oct;85(10):1017-25. doi: 10.1021/js950534b. PMID: 8897265. 4.
  • reaction mixture was cooled to room temperature and water (1.65 L) was added. After stirring for 10-15 min, ethyl acetate (550 mL) was added, and the reaction mass was filtered through celite and washed with ethyl acetate (110 mL). The organic layer was separated, and the aqueous layer was extracted with ethyl acetate (2.0 vol). The combined organic layers were washed with water (2.0 vol) and then distilled under vacuum below 50 °C to obtain the crude.
  • Step 2 Synthesis of Semi-carbazido- ⁇ -cyclodextrin I-1: [00314] To a stirred solution of Boc-Semi-carbazido- ⁇ -cyclodextrin 7 (500 mg, 1.0 eq) in DCM (5.0 mL) was added trifluoro acetic acid (2.5 mL) at room temperature under nitrogen atmosphere.
  • Step 2 Synthesis of Semi-thiocarbazido- ⁇ -cyclodextrin I-2: [00316] To a stirred solution of Boc-Semi-carbazido- ⁇ -cyclodextrin 7 (23 mg, 1.0 eq) in DCM (5.0 mL) was added trifluoro acetic acid (2.5 mL) at room temperature under nitrogen atmosphere. The reaction mixture was stirred for 3 h at room temperature. After starting material was consumed (as shown by TLC), volatiles were removed under reduced pressure to obtain crude. The crude material was co-distilled with water followed by toluene.
  • Step 1 Synthesis of Boc-Serinol- ⁇ -cyclodextrin 8: [00317] To a stirred solution of carbonyldiimidazole 6 (5.0 eq) in DMF (5.0 mL) was added Boc-serinol (5.0 eq) at room temperature under nitrogen atmosphere. After stirring for 15-20 min, amino- ⁇ -cyclodextrin (50.0 mg, 1.0 eq) was added and stirred the reaction mixture for 12 h at room temperature. After starting material was consumed (as shown by TLC), acetone (2.0 mL) was added to the reaction mass and the precipitated solid was filtered.
  • Step 2 Synthesis of Serinol- ⁇ -cyclodextrin (I-3) [00318] To a stirred solution of Boc-Serinol- ⁇ -cyclodextrin 8 (30 mg, 1.0 eq) in DCM (5.0 mL) was added trifluoro acetic acid (2.5 mL) at room temperature under nitrogen atmosphere. The reaction mixture was stirred for 3 h at room temperature. After starting material was consumed (as shown by TLC), volatiles were removed under reduced pressure to obtain crude.
  • Example X.1 Synthesis of Compounds X-I-1 to X-I-37 Synthesis of 2-(6-Aminobenzo[d] [1,3] dioxol-5-yl propan-2-ol (X-I-29): Synthesis of X-I-29: [00324] To a stirred solution of 1-(6-aminobenzo[d] [1,3] dioxol-5-yl) ethanone 1 (0.5 g, 1.0 eq) in THF (15 mL) was added methyl magnesium bromide (4.0 eq, 2M solution) at 0°C under nitrogen. The reaction was stirred for 2 h at 0°C.
  • 4-HNE 4-hydroxynonenal
  • Example X.4 Evaluation of Prophylactic Anti-Inflammatory Activity of Test Compounds in Acute LPS-Induced Sepsis in C57BL/6 Mice Summary
  • LPS lipopolysaccharide
  • This study models acute sepsis, which is a systemic inflammatory syndrome initiated by Gram-negative and Gram-positive bacteria and fungi which infect the lungs, abdomen, bloodstream, and renal or genitourinary tracts.
  • Sepsis is characterized by an early systemic inflammatory response phase featuring symptoms, such as tachycardia, fever, hyperventilation, and activation of the complement and coagulation cascades.
  • symptoms such as tachycardia, fever, hyperventilation, and activation of the complement and coagulation cascades.
  • a compensatory anti-inflammatory response phase follows, characterized by neuroendocrine-mediated immunosuppression. Since these processes are the result of interaction between cells of inflammation and organs, investigation of the treatment of this syndrome requires the use of intact animal models. The use of ten mice per group allows useful statistical modelling of the results.
  • mice female, 18-22 gram, C57BL/6 were purchased from ENVIGO. The mice were housed in 20 cages of 5 mice per cage with 1 cage of 3 mice as extra. The cages had filter tops and autoclaved bedding, and the animals were placed in quarantine with daily inspection.
  • Table X.2A Table X.2A:
  • mice Female, 18-22 gram, C57BL/6 were purchased from ENVIGO. The mice were housed in 37 cages of 5 mice per cage. The cages had filter tops and autoclaved bedding, and the animals were placed in quarantine with daily inspection.
  • mice were injected IP with 1.5 mg/kg MPS (Sigma).
  • MPS 1.5 mg/kg
  • all mice were anesthetized and exsanguinated into pre-chilled EDTA- treated tubes.
  • the blood was processed to plasma which was stored in labeled 0.5 polypropylene snap-cap tubes (0.5 mL Eppendorf Safe-Lock Tubes (Fisher Scientific) at -80 °C.
  • Preparation of vehicle 1.0 gram of methyl cellulose (Sigma) was dissolved in 200 mL of water (USP purified) to make a solution of 0.5% methyl cellulose in water.
  • LPS solution 6 mg of LPS (from Escherichia Coli 055:B5, Cat# L2880, Sigma Lot# 057m4013) was dissolved in 20 ml of saline to give a LPS solution of 0.3 mg/ml. All mice were injected with 5ml/kg (1.5 mg/kg) at the scheduled time by intraperitoneal injection.
  • Cytokine Panel Summary [00339] A 32-plex cytokine panel was obtained from Eve Technologies. Each cytokine was assessed based against a 7 point curve range in duplicate. The average of the two replicate analyses was used for statistics. Unpaired t-tests were performed using ExcelTM contrasting vehicle against each treatment group.
  • Example Y.1 Synthesis of Compounds Y-I-1 to Y-I-169 Key int-1 synthesis: Synthesis of methyl 3-amino-4-((trimethylsilyl)ethynyl)benzoate 2
  • reaction mixture was slowly heated to 60 °C and stirred for 16 h in a sealed tube. After consumption of starting material (by TLC), the reaction mass was cooled to room temperature and diluted with ethyl acetate. The organic layer was washed with water (3 x 150 mL). The ethyl acetate layer was separated, dried over Na2SO4 and evaporated under reduced pressure to obtain crude compound which was purified by column chromatography eluted with (ethyl acetate: Hexane, 5:95), pure fraction was distilled to give 2 (15.5 g, 71.7%) as thick syrup.
  • Example Y.2 Experimental Conditions Used for Initial Trapping Experiments
  • 4-HNE 4- hydroxynonenal
  • 4-HNE 4-hydroxynonenal
  • ethanol 10 mg/mL solution
  • Example Y.4 Evaluation of Prophylactic Anti-Inflammatory Activity of Test Compounds in Acute LPS-Induced Sepsis in C57BL/6 Mice Summary
  • LPS lipopolysaccharide
  • This study models acute sepsis, which is a systemic inflammatory syndrome initiated by Gram-negative and Gram-positive bacteria and fungi which infect the lungs, abdomen, bloodstream, and renal or genitourinary tracts.
  • Sepsis is characterized by an early systemic inflammatory response phase featuring symptoms, such as tachycardia, fever, hyperventilation, and activation of the complement and coagulation cascades.
  • symptoms such as tachycardia, fever, hyperventilation, and activation of the complement and coagulation cascades.
  • a compensatory anti-inflammatory response phase follows, characterized by neuroendocrine-mediated immunosuppression. Since these processes are the result of interaction between cells of inflammation and organs, investigation of the treatment of this syndrome requires the use of intact animal models. The use of ten mice per group allows useful statistical modelling of the results.
  • mice female, 18-22 gram, C57BL/6 may be purchased from ENVIGO.
  • the mice housed in 20 cages of 5 mice per cage with 1 cage of 3 mice as extra.
  • the cages have filter tops and autoclaved bedding, and the animals are placed in quarantine with daily inspection.
  • 10 mice are included in each group.
  • the mice are dosed by oral gavage (PO) at 10 ml/kg.
  • PO oral gavage
  • 185 mice female, 18-22 gram, C57BL/6 may be purchased from ENVIGO.
  • the mice housed in 37 cages of 5 mice per cage.
  • the cages having filter tops and autoclaved bedding, and the animals placed in quarantine with daily inspection. 10 mice may be included in each group.
  • mice are to be dosed by oral gavage (PO) at 10 ml/kg.
  • PO oral gavage
  • 0.5 polypropylene snap-cap tubes 0.5 mL Eppendorf Safe-Lock Tubes (Fisher Scientific) at -80 °C.
  • ALDH2 is a NAD(p)+-dependent mitochondrial matrix enzyme.
  • Major substrates (high affinity) of ALDH2 are short-chain aliphatic lipid aldehydes (e.g., 4-HNE, acetaldehyde, malondialdehyde and acrolein).
  • ALDH2 dysfunction has been associated with neurodegeneration, apoptotic cell death, pain and the process of aging (Chen Y, He Y, DeVivo MJ. Changing demographics and injury profile of new traumatic spinal cord injuries in the United States, 1972-2014. Arch Phys Med Rehabil. 2016b;97:1610–1619).
  • ALDH2 KO mice had a significantly high 4-HNE load, increased cytokine levels, and persistent behavioral deficits after closed head traumatic brain injury, supporting the anti-inflammatory and antioxidant role of ALDH2 activity in neurotrauma (Knopp RC, Lee SH, Hollas M, Nepomuceno E, Gonzalez D, Tam K, Aamir D, Wang Y, Pierce E, BenAissa M, Thatcher GRJ. Interaction of oxidative stress and neurotrauma in ALDH2(-/-) mice causes significant and persistent behavioral and pro- inflammatory effects in a tractable model of mild traumatic brain injury. Redox Biol. 2020;32:101486).
  • ALDH2 is an established druggable target, and it can be selectively and robustly activated by Alda-1 and inhibited by daidzin (Chen et al., 2014). See Khan, M. et al., Neural Regen Res.2022 Jan; 17(1): 185–193 for an assay procedure. [00428] Compounds Y-I-36, Y-I-55, and Y-I-68 showed an increased in ALDH activity (agonism). EC50s: Y-I-36: 0.29 ⁇ M; Y-I-55: 0.56 ⁇ M; Y-I-68: 0.83 ⁇ M.
  • Example 3 Rabbit Vitreous PK Profile
  • Ocular PK was conducted with a non-deuterated analog of Compound A.
  • a single injection of 500 micrograms of the non-deuterated version was administered to New Zealand White rabbits.
  • the vitreous humor was collected at various time points and analyzed by LCMS.
  • the half-life of non-deuterated Compound A was determined to be approximately 24 hours as shown in FIG.1. Fit parameters for the data in FIG.1 are shown below.
  • Parameter Unit Value Example 4 - Retinaldehyde Binding Profile of Compound A
  • Compound A is designed to trap reactive aldehydes species (RASP) in diverse ocular regions and inhibit RASP-mediated toxicity.
  • RASP reactive aldehydes species
  • Compound A is a small, membrane-permeable molecule that should readily reach the Bruch's membrane and retinal pigment epithelium (RPE), where excess retinaldehyde is thought to form toxic precipitates.
  • RPE retinal pigment epithelium
  • All-trans-retinaldehyde is a highly reactive intermediate, that is toxic retina cells and contributes to retinal and macular degenerations (Dreffs, et al., 2020). Retinol is converted into retinaldehyde by retinol dehydrogenase, which is expressed in RPE cells. Retinaldehyde is converted into retinoic acid by the enzyme retinaldehyde dehydrogenase (RALDH), which is expressed in RPE, and retina.
  • RALDH retinaldehyde dehydrogenase
  • modulating retinaldehyde and inhibiting retinoic acid may mitigate further vision loss (Telias, et al., 2022).
  • Compound A is designed to prevent retinal toxicity by sequestering aldehydes before they can form reactive precipitates in the eye.
  • An in vitro study was executed demonstrating the irreversible covalent binding of retinaldehyde by Compound A.
  • Compound A (2mM) was added to a buffered solution (pH ⁇ 7.4) of retinaldehyde (1mM). The loss of free Compound A and increase in Compound A -CD complex formation was monitored at various time points to determine completeness of the reaction as shown in FIG.2.
  • Example 5 In Vitro ADMET Assays
  • the following in-vitro ADMET assessments have been performed and reported below in Table 2.
  • Table 2 Additional Planned ADMET Studies
  • Metabolite ID 2 Time Points with Hepatocytes (Human, Beagle Dog, Rat, NZW Rabbit)
  • Plasma Protein Binding (rat, rabbit, dog, human)
  • Example 6 Clinical Study [00347] Protocol Synopsis:
  • Example 9 In vitro Binding Data for Compounds I-1, I-2, I-3, and I-4 and Amino-CD Compound I-1/4-HNE Binding Data
  • Fig 3A shows Compound I-1 binding with 4-HNE (4-hydroxynonenol) as a model aldehyde.
  • Fig 3B shows Compound 6 of Example 2.1 binding with 4-HNE (4-hydroxynonenol) as a model aldehyde.
  • Aldehyde traps, including Compound A have increased residency time in the eye when delivered in a composition having a modified cyclodextrin of this disclosure.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Ophthalmology & Optometry (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne le traitement, la prévention et/ou la réduction d'un risque d'une maladie, d'un trouble ou d'une affection oculaire où la toxicité de l'aldéhyde est impliquée dans la pathogenèse, grâce à l'utilisation de formulations décrites.
PCT/US2023/036113 2022-10-28 2023-10-27 Cyclodextrines modifiées et leurs utilisations ophtalmiques WO2024091659A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263420303P 2022-10-28 2022-10-28
US63/420,303 2022-10-28

Publications (1)

Publication Number Publication Date
WO2024091659A1 true WO2024091659A1 (fr) 2024-05-02

Family

ID=90831796

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/036113 WO2024091659A1 (fr) 2022-10-28 2023-10-27 Cyclodextrines modifiées et leurs utilisations ophtalmiques

Country Status (1)

Country Link
WO (1) WO2024091659A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006045071A (ja) * 2004-07-30 2006-02-16 Daikin Ind Ltd 包接体及びアルデヒド類又はケトン類の捕捉剤
US20120028944A1 (en) * 2005-07-22 2012-02-02 Oculis Ehf Cyclodextrin nanotechnology for ophthalmic drug delivery
US20170266220A1 (en) * 2016-02-28 2017-09-21 Aldeyra Therapeutics, Inc. Treatment of allergic eye conditions with cyclodextrins
WO2023278816A1 (fr) * 2021-07-02 2023-01-05 Aldeyra Therapeutics, Inc. Composés hétérocycliques de piégeage d'aldéhyde et leurs utilisations

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006045071A (ja) * 2004-07-30 2006-02-16 Daikin Ind Ltd 包接体及びアルデヒド類又はケトン類の捕捉剤
US20120028944A1 (en) * 2005-07-22 2012-02-02 Oculis Ehf Cyclodextrin nanotechnology for ophthalmic drug delivery
US20170266220A1 (en) * 2016-02-28 2017-09-21 Aldeyra Therapeutics, Inc. Treatment of allergic eye conditions with cyclodextrins
WO2023278816A1 (fr) * 2021-07-02 2023-01-05 Aldeyra Therapeutics, Inc. Composés hétérocycliques de piégeage d'aldéhyde et leurs utilisations

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PUBCHEM SUBSTANCE 1 February 2023 (2023-02-01), ANONYMOUS: "SID 475569291", XP093168695, Database accession no. 475569291 *
DATABASE PUBCHEM SUBSTANCE 13 August 2020 (2020-08-13), ANONYMOUS: "SID 426898190", XP093168693, Database accession no. 426898190 *

Similar Documents

Publication Publication Date Title
JP7332186B2 (ja) アルデヒドコンジュゲートおよびその使用
CA3032609A1 (fr) Composes de piegeage d'aldehydes et leurs procedes d'utilisation
JP6602802B2 (ja) Ssaoの置換3−ハロアリルアミン阻害剤およびその使用
US20180050989A1 (en) Aldehyde trapping compounds and uses thereof
JP7311162B2 (ja) 炎症性障害の処置
EP1888548B1 (fr) Dérivé de quinoline pour le traitement de maladies rétiniennes
AU2017253937A1 (en) Compounds and compositions for treating conditions associated with NLRP activity
JP5815552B2 (ja) 眼疾患を治療する化合物および方法
JP2018523700A (ja) 重水素化化合物およびその使用
AU2003243282B2 (en) Amelioration of the development of cataracts and other opthalmic diseases
AU2019351122A1 (en) Inhibitors of VAP-1
CA2546053A1 (fr) Amelioration de la cataracte, de la degenerescence maculaire et autres maladies ophtalmiques
US20050131025A1 (en) Amelioration of cataracts, macular degeneration and other ophthalmic diseases
WO2024091659A1 (fr) Cyclodextrines modifiées et leurs utilisations ophtalmiques
CA3220889A1 (fr) Composes heterocycliques de piegeage d'aldehyde et leurs utilisations
KR20210015892A (ko) 통증 치료를 위한 화합물, 이를 포함하는 조성물 및 이를 사용하는 방법
WO2022267930A1 (fr) Composé tricyclique, son procédé de préparation et son utilisation médicale
ZA200604402B (en) Amelioration of cataracts, macular degeneration and other ophthalmic diseases