US20140024598A1 - Methods and compositions for preserving retinal ganglion cells - Google Patents

Methods and compositions for preserving retinal ganglion cells Download PDF

Info

Publication number
US20140024598A1
US20140024598A1 US13/882,932 US201113882932A US2014024598A1 US 20140024598 A1 US20140024598 A1 US 20140024598A1 US 201113882932 A US201113882932 A US 201113882932A US 2014024598 A1 US2014024598 A1 US 2014024598A1
Authority
US
United States
Prior art keywords
necrostatin
inhibitor
eye
alkyl
caspase
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/882,932
Other languages
English (en)
Inventor
Demetrios Vavvas
Joan W. Miller
Maki Kayama
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Massachusetts Eye and Ear
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US13/882,932 priority Critical patent/US20140024598A1/en
Assigned to MASSACHUSETTS EYE AND EAR INFIRMARY reassignment MASSACHUSETTS EYE AND EAR INFIRMARY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KAYAMA, MAKI, MILLER, JOAN W., VAVVAS, DEMETRIOS G.
Publication of US20140024598A1 publication Critical patent/US20140024598A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: MASSACHUSETTS EYE AND EAR INFRIMARY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41661,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/005Enzyme inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics

Definitions

  • the field of the invention relates generally to methods and compositions for preserving the viability of retinal ganglion cells (RGCs), for example, in a subject affected with an ocular disorder wherein a symptom of the ocular disorder is loss of retinal ganglion cell viability. More particularly, the invention relates to the use of a necrosis inhibitor, e.g., a RIP kinase inhibitor, e.g., a necrostatin, either alone or in combination with an apoptosis inhibitor, e.g., a pan-caspase inhibitor, for preserving the viability of retinal ganglion cells for the treatment of the ocular disorder. The invention further relates to the use a necrosis inhibitor, either alone or in combination with an apoptosis inhibitor, for promoting axon regeneration with retinal ganglion cells.
  • a necrosis inhibitor e.g., a RIP kinase inhibitor, e.g., a necrostatin
  • the retina is a delicate neural tissue lining the back of the eye that converts light stimuli into electric signals for processing by the brain.
  • the optic nerve is a cable of retinal ganglion cells that carry the electric signals from the retina to the brain.
  • Diseases affecting the retina and optic nerve, including, for example, glaucoma, and optic nerve injury can lead to vision loss and blindness. Early detection and treatment are critical in correcting problems before vision is lost or in preventing further deterioration of vision.
  • Glaucoma is the second leading cause of blindness overall. Glaucoma is a progressive disease which leads to optic nerve damage and, ultimately, total loss of vision. The causes of this disease have been the subject of extensive studies for many years, but are still not fully understood.
  • the principal symptom of and/or risk factor for the disease is elevated intraocular pressure or ocular hypertension due to excess aqueous humor in the anterior chamber of the eye.
  • many of the drugs conventionally used to treat ocular hypertension have a variety of problems. For instance, miotics such as pilocarpine can cause blurring of vision and other visual side effects, which may lead either to decreased patient compliance or to termination of therapy.
  • therapies that control elevated intraocular pressure associated with glaucoma without the degree of undesirable side-effects attendant to most conventional therapies.
  • Optic nerve injury can be the result of glaucoma, trauma, toxicity, inflammation, ischemia, congenital diseases, or compression from tumors or aneurysms. To date, few effective treatments have been discovered to restore visual function and/or axon regeneration following optic nerve injury.
  • Apoptosis and necrosis represent two different mechanisms of cell death.
  • Apoptosis is a highly regulated process involving the caspase family of cysteine proteases, and characterized by cellular shrinkage, chromatin condensation, and DNA degradation.
  • necrosis is associated with cellular and organelle swelling and plasma membrane rupture with ensuing release of intracellular contents and secondary inflammation (Kroemer et al., (2009) C ELL D EATH D IFFER 16:3-11).
  • necrosis has been considered a passive, unregulated form of cell death; however, recent evidence indicates that some necrosis can be induced by regulated signal transduction pathways such as those mediated by receptor interacting protein (RIP) kinases, especially in conditions where caspases are inhibited or cannot be activated efficiently (Golstein P & Kroemer G (2007) T RENDS B IOCHEM . S CI . 32:37-43; Festjens et al. (2006) B IOCHIM . B IOPHYS . A CTA 1757:1371-1387).
  • RIP receptor interacting protein
  • DRs death domain receptors
  • caspase-8 or treated with pan-caspase inhibitor Z-VAD
  • stimulation of death domain receptors (DR) causes a RIP-1 kinase dependent programmed necrotic cell death instead of apoptosis (Holler et al. (2000) N AT . I MMUNOL . 1:489-495; Degterev et al. (2008) N AT . C HEM . B IOL. 4:313-321).
  • RECEPTOR Interacting Protein kinase 1 is a serine/threonine kinase that contains a death domain and forms a death signaling complex with the Fas-associated death domain and caspase-8 in response to death receptor (DR) stimulation (Festjens et al. (2007) C ELL D EATH D IFFER . 14:400-410).
  • RIP-1 During death domain receptor-induced apoptosis, RIP-1 is cleaved and inactivated by caspase-8, the process of which is prevented by caspase inhibition (Lin et al. (1999) G ENES . D EV . 13:2514-2526). It has been unclear how RIP-1 kinase mediates programmed necrosis, but recent studies revealed that the expression of RIP-3 and the RIP-1-RIP-3 binding through the RIP homotypic interaction motif is a prerequisite for RIP-1 kinase activation, leading to reactive oxygen species (ROS) production and necrotic cell death (He et al., (2009) C ELL 137:1100-1111; Cho et. al., (2009) CELL 137:1112-1123; Zhang et al., (2009) S CIENCE 325:332-336).
  • ROS reactive oxygen species
  • the invention is based, in part, on the discovery that a necrosis inhibitor, e.g., RIP kinase inhibitor, e.g., a necrostatin, e.g., necrostatin-1, can be used to reduce or prevent the loss of retinal ganglion cell viability, especially when the necrosis inhibitor is combined with an apoptotic inhibitor (e.g., a pan-caspase inhibitor, e.g., Z-VAD and/or IDN-6556).
  • an apoptotic inhibitor e.g., a pan-caspase inhibitor, e.g., Z-VAD and/or IDN-6556.
  • an apoptosis inhibitor i.e., a pan-caspase inhibitor
  • the studies described hereinbelow indicate that, in the presence of an apoptosis inhibitor (e.g., a pan-caspase inhibitor), retinal ganglion cells die by necrosis, including necroptosis (or programmed necrosis).
  • apoptosis inhibitor e.g., a pan-caspase inhibitor
  • necrosis or programmed necrosis
  • programmed necrosis is a critical mechanism for ocular disorders wherein a symptom of the disorder is the loss of retinal ganglion cell viability in the presence of a pan-caspase inhibitor.
  • it is possible to reduce the loss of visual function associated with an ocular disorder in particular while the ocular disorder is being treated, by reducing the loss of retinal ganglion cell viability.
  • a method preserving the visual function of an eye of a subject with an ocular condition, wherein a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of the eye with the condition.
  • the method comprises (a) administering to the eye of the subject an effective amount of a necrosis inhibitor and an effective amount of an apoptosis inhibitor thereby preserving the viability of the retinal ganglion cells disposed within the retina of the eye, and (b) then measuring the visual function (e.g., visual acuity) of the eye after the administration of the necrosis inhibitor and the apoptosis inhibitor.
  • the visual function (e.g., visual acuity) of the eye may be preserved or improved relative to the visual function of the eye prior to administration of the necrosis inhibitor and the apoptosis inhibitor.
  • the ocular condition may include, but is not limited to, glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • the ocular condition is glaucoma or optic nerve injury.
  • a symptom of the ocular condition may be the loss of retinal ganglion cells in the retina of the eye with the condition.
  • the ocular condition may be glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • the method comprises administering to the eye of the subject an effective amount of a necrosis inhibitor and an apoptosis inhibitor thereby preserving the viability of the retinal ganglion cells with the retina of the subject with the condition. After the administration of the necrosis inhibitor and the apoptosis inhibitor, the retinal ganglion cell is capable of supporting axonal regeneration.
  • a method of preserving visual function of an eye of a subject affected with an ocular condition such as glaucoma or optic nerve injury, wherein a symptom of the ocular condition is loss of retinal ganglion cell viability, e.g., glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • the method comprises reducing the production and/or activity of a RIP-1 kinase and/or RIP-3 kinase in the eye thereby preserving the viability of the retinal ganglion cells disposed with the retina of the eye.
  • the reduction in the production or activity of the RIP-1 kinase and/or the RIP-3 kinase can be achieved by administering an effective amount of RIP kinase (RIPK) inhibitor, e.g., a necrostatin.
  • RIPK RIP kinase
  • a method of preserving the visual function of an eye of a subject affected with an ocular condition wherein a symptom of the ocular condition is loss of retinal ganglion cell viability in the retina of the eye comprises (a) reducing the production or activity of a RIP-1 kinase and/or a RIP-3 kinase in the eye thereby to preserve the viability of the retinal ganglion cells disposed within the retina of the eye; and (b), after treatment, measuring visual function (e.g., visual acuity) of the eye.
  • the reduction in the production or activity of the RIP-1 kinase and/or the RIP-3 kinase can be achieved by administering an effective amount of RIPK inhibitor, e.g., a necrostatin.
  • RIPK inhibitor e.g., a necrostatin.
  • the provided herein is a method for promoting axonal regeneration in an eye of a subject with an ocular condition, wherein a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of an eye with the condition.
  • the method comprises administering to the eye of the subject with the condition an effective amount of a necrosis inhibitor and an effective amount of an apoptosis inhibitor thereby to promote the regeneration of a retinal ganglion cell axon within the retina of the eye.
  • the method may further comprise, after administration of the necrosis inhibitor and the apoptosis inhibitor, measuring visual function of the eye.
  • the ocular condition may include, but is not limited to, glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • a necrosis inhibitor e.g., a RIPK inhibitor, e.g., a necrostatin
  • an apoptosis inhibitor e.g., a pan-caspase inhibitor, e.g., Z-VAD or IDN-6556
  • the ocular condition may be glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • a necrosis inhibitor e.g., a RIPK inhibitor, e.g., a necrostatin
  • an apoptosis inhibitor e.g., a pan-caspase inhibitor, e.g., Z-VAD or IDN-6556
  • a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of the eye with the condition.
  • the ocular condition may be glaucoma, optic nerve injury, optic neuritis, optic neuropathies, central retinal artery occlusion, central retinal vein occlusion and diabetic retinopathy.
  • a necrosis inhibitor e.g., a RIPK inhibitor, e.g., a necrostatin
  • an apoptosis inhibitor e.g., a pan-caspase inhibitor, e.g., Z-VAD or IDN-6556
  • a necrosis inhibitor e.g., a RIPK inhibitor, e.g., a necrostatin
  • an apoptosis inhibitor e.g., a pan-caspase inhibitor, e.g., Z-VAD or IDN-6556
  • the necrosis inhibitor can be a RIP kinase inhibitor, for example, a necrostatin.
  • the necrostatin is necrostatin-1, necrostatin-2, necrostatin-3, necrostatin-4, necreostatin-5, necrostatin-7, or a combination thereof.
  • the necrostatin when a necrostatin is administered, the necrostatin is administered to provide a final concentration of necrostatin in the eye greater than about 5 ⁇ M.
  • the final concentration of necrostatin in the eye may range from about 5 ⁇ M to about 1000 ⁇ M, about 10 ⁇ M to about 1000 ⁇ M, about 100 ⁇ M to about 1000 ⁇ M, about 150 ⁇ M to about 1000 ⁇ M, from about 200 ⁇ M to about 800 ⁇ M or from about 200 ⁇ M to about 600 ⁇ M.
  • the final concentration of necrostatin in the eye is about 400 ⁇ M.
  • necrostatin when a necrostatin is administered, from about 0.05 mg to about 2 mg, 0.1 mg to about 1 mg, from about 0.2 mg to about 1 mg, or from about 0.2 mg to about 0.8 mg, of necrostatin can be administered locally to the eye of a mammal. In an exemplary embodiment, about 0.5 mg of necrostatin can be administered locally to the eye of a mammal.
  • the pan-caspase inhibitor when a pan-caspase inhibitor is administered, the pan-caspase inhibitor is administered to provide a final concentration of the pan-caspase inhibitor in eye greater than about 3 ⁇ M.
  • the final concentration of pan-caspase inhibitor in the eye may range from about 3 ⁇ M to about 500 ⁇ M, about 10 ⁇ M to about 500 ⁇ M, about 100 ⁇ M to about 500 ⁇ M, about 150 ⁇ M to about 500 ⁇ M, or from about 200 ⁇ M to about 400 ⁇ M.
  • the final concentration of the pan-caspase inhibitor in the eye is about 300 ⁇ M.
  • Exemplary pan-caspase inhibitors include zVAD, IDN-6556 or a combination thereof.
  • from about 0.05 mg to about 1.5 mg, from about 0.15 mg to about 1.5 mg, from about 0.2 mg to about 1 mg, from about 0.2 mg to about 0.8 mg, from about 0.4 mg to about 1 mg, or from about 0.5 mg to about 0.8 mg, of the pan-caspase inhibitor can be administered locally to the eye of a mammal.
  • about 0.7 mg of a pan-caspase inhibitor can be administered locally to the eye of a mammal.
  • the necrosis inhibitor e.g., a necrostatin, and/or the apoptosis inhibitor may be administered to the eye by intraocular, intravitreal, subretinal or trasscleral administration.
  • the necrosis inhibitor e.g., a necrostatin, and/or the apoptosis inhibitor may be solubilized in a viscoelastic carrier that is introduced into the eye.
  • the necrosis inhibitor, e.g., a necrostatin, and/or the apoptosis inhibitor may be administered systemically.
  • necrosis inhibitor e.g., a necrostatin
  • apoptosis inhibitor may be administered sequentially or simultaneously.
  • the necrosis inhibitor, e.g., a necrostatin, and the apoptosis inhibitor may be administered in the same or different carriers.
  • the necrostatin can be selected from one or more of the following necrostatins.
  • the necrostatin is a Nec-1 related compound of Formula I:
  • X is O or S
  • R 1 is hydrogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, or halogen
  • R 2 is hydrogen or C 1 -C 6 alkyl.
  • necrostatin can be a Nec-1 related compound of Formula I-A:
  • R 1 is H, alkyl, alkoxyl, or a halogen and R 2 is H or an alkyl.
  • necrostatin can be a Nec-1 related compound of Formula I-B:
  • necrostatin can be a Nec-1 related compound of Formula I-C:
  • necrostatin can be a Nec-1 related compound of Formula I-D:
  • necrostatin can be a Nec-1 related compound of Formula I-E:
  • R 1 is H, alkyl, alkoxyl, or a halogen (for example, F, Cl, Br or I) and R 2 is H or an alkyl.
  • necrostatin can be a Nec-1 related compound of Formula I-F:
  • necrostatin can be a Nec-1 related compound of Formula I-G:
  • necrostatin can be a Nec-2 related compound of Formula II:
  • X is —CH 2 —, —C(H)(R 14 )—, —C( ⁇ S)—, —C( ⁇ NH)—, or —C(O)—;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 each represent independently hydrogen, acyl, acetyl, alkyl, halogen, amino, C 1 -C 6 alkoxyl, nitro, —C(O)R 12 , —C(S)R 12 , —C(O)OR 12 , —C(O)NR 12 R 13 , —C(S)NR 12 R 13 , or —S(O 2 )R 12 ;
  • R 11 is hydrogen, acyl, acetyl, alkyl, or acylamino
  • R 12 and R 13 each represent independently hydrogen, an optionally substituted alkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted aralkyl, or an optionally substituted heteroaralkyl;
  • R 14 is acyl, acetyl, alkyl, halogen, amino, acylamino, nitro, —SR 11 , —N(R 11 ) 2 , or —OR 11 ;
  • the bond indicated by (a) can be a single or double bond
  • the bond indicated by (b) can be a single or double bond.
  • necrostatin can be a Nec-2 related compound of Formula IIA:
  • R 1 , R 2 , R 5 , R 6 , R 7 , and R 10 each represent independently hydrogen, alkyl, halogen, amino, or methoxyl;
  • R 3 , R 4 , R 8 , and R 9 are C 1 -C 6 alkoxyl.
  • necrostatin can be a Nec-3 related compound of Formula III:
  • Z is —CH 2 —, —CH 2 CH 2 —, —O—, —S—, —S(O)—, —S(O 2 )—, or —N(R 7 )—;
  • R 1 , R 3 , and R 5 each represent independently for each occurrence hydrogen, halogen, hydroxyl, amino, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkoxy-C 1 -C 6 alkyl, C 1 -C 6 alkanoyl, C 1 -C 6 alkylsulfinyl, C 1 -C 6 alkylsulfinyl-C 1 -C 6 alkyl, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonyl-C 1 -C 6 alkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl, or heteroaralkyl;
  • R 2 and R 4 are C 1 -C 6 alkoxy
  • R 6 is —C(O)R 8 , —C(S)R 8 , —C(O)NR 8 , —C(O)NR 8 R 9 , —C(S)NR 8 R 9 , —C(NH)R 8 , or —S(O 2 )R 8 ;
  • R 7 is alkyl, aralkyl, or heteroaralkyl
  • R 8 and R 9 each represent independently hydrogen, C 1 -C 6 alkyl, heteroalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
  • n independently for each occurrence 0, 1, or 2.
  • necrostatin can be a Nec-4 related compound of Formula IV:
  • R 2 and R 3 each represent independently for each occurrence hydrogen or methyl
  • R 4 represents independently for each occurrence halogen, hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 4 alkynyl;
  • R 5 is C 1 -C 4 alkyl
  • R 6 is hydrogen, halogen, or —CN
  • R 7 is hydrogen or C 1 -C 4 alkyl
  • R 8 is C 1 -C 6 alkyl, or R 8 taken together with R 9 , when present, forms a carbocyclic ring;
  • R 9 is hydrogen or C 1 -C 6 alkyl, or R 9 taken together with R 8 forms a carbocyclic ring;
  • R 10 is hydrogen or C 1 -C 6 alkyl
  • A is phenylene or a 5-6 membered heteroarylene
  • X is N or —C(R 9 )—
  • Y is N or —C(R 10 )—
  • Z is S or O
  • n and n each represent independently 1, 2, or 3.
  • the necrostatin can be a Nec-4
  • necrostatin can be a Nec-5 related compound of Formula V:
  • A is a saturated or unsaturated 5-6 membered carbocyclic ring
  • X is a bond or C 1 -C 4 alkylene
  • R 1 is C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • R 3 is —C 1 -C 6 alkylene-CN, —CN, C 1 -C 6 alkyl, or C 2 -C 6 alkenyl;
  • R 4 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl
  • R 5 represents independently for each occurrence C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • B is a 5-6 membered heterocyclic or carbocylic ring
  • n and p each represent independently 0, 1, or 2.
  • necrostatin can be a Nec-5 related compound of Formula V-A:
  • R 1 is C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, or —N(R 4 ) 2 ;
  • R 3 is —C 1 -C 6 alkylene-CN
  • R 4 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl
  • R 5 represents independently for each occurrence C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • B is a 5-6 membered heterocyclic or carbocylic ring
  • n and p each represent independently 0, 1, or 2.
  • necrostatin can be a Nec-7 related compound of Formula VII:
  • R 1 , R 2 , and R 3 each represent independently hydrogen or C 1 -C 4 alkyl
  • R 5 and R 6 each represent independently for each occurrence halogen, C 1 -C 6 alkyl, hydroxyl, C 1 -C 6 alkoxyl, —N(R 7 ) 2 , —NO 2 , —S—C 1 -C 6 alkyl, —S-aryl, —SO 2 —C 1 -C 6 alkyl, —SO 2 -aryl, —C(O)R 7 , —CO 2 R 7 , —C(O)N(R 7 ) 2 , heterocycloalkyl, aryl, or heteroaryl;
  • R 7 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl; or two occurrences of R 7 attached to the same nitrogen atom are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring;
  • A is a 5-6 membered heterocyclic ring
  • p 0, 1, or 2.
  • necrostatin can be a Nec-7 related compound of Formula VIII:
  • each X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is selected, independently, from N or CR X1 ;
  • each Y 1 , Y 2 , and Y 3 is selected, independently, from O, S, NR Y1 , or CR Y2 R Y3 ;
  • each Z 1 and Z 2 is selected, independently, from O, S, or NR Z1 ;
  • each R Y1 and R Z1 is selected, independently, from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, —C( ⁇ O)R 5A , —C( ⁇ O)OR 5A , or —C( ⁇ O)NR 5A R 6A ;
  • each R X1 , R Y2 , and R Y3 is selected, independently, from H, halogen, CN, NC, NO 2 , N 3 , OR 3 , SR 3 , NR 3 R 4 , —C( ⁇ O)R 5A , —C( ⁇ O)OR 5A , —C( ⁇ O)NR 5A R 6A , —S( ⁇ O)R 5A , —S( ⁇ O) 2 R 5A , —S( ⁇ O) 2 OR 5A , —S( ⁇ O) 2 NR 5A R 6A , optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • each R 1 , R 2 , R 5A , R 5B , R 6A , and R 6B is selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R 5A and R 6A , or R 5B and R 6B combine to form a heterocyclyl; and
  • each R 3 and R 4 is selected from H, optionally substituted C 1-6 alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, —C( ⁇ O)R 5B , —C( ⁇ S)R 5B , —C( ⁇ NR 6B )R 5B , —C( ⁇ O)OR 5B , —C( ⁇ O)NR 5B R 6B , —S( ⁇ O)R 5B , —S( ⁇ O)R 5B , —S( ⁇ O) 2 OR 5B , or —S( ⁇ O) 2 NR 5B R 6B .
  • R 1 is H
  • X 1 , X 2 , and X 4 are each CH
  • X 3 , X 5 , and X 6 are each N
  • Y 1 and Y 3 are each S
  • Y 2 is NH
  • Z 1 is NH
  • Z 2 is O
  • R 2 is not 4-fluorophenyl
  • necrostatin can be a Nec-4 related compound of Formula IX:
  • X 1 and X 2 are, independently, N or CR 4 ;
  • X 3 is selected from O, S, NR 5 , or —(CR 5 ) 2 ;
  • Y is selected from C(O) or CH 2 ;
  • Z is (CR 6 R 7 ) n ;
  • R 1 is selected from H, halogen, optionally substituted C 1-6 alkyl, or optionally substituted C 1-6 cycloalkyl, or optionally substituted aryl;
  • R 2 is selected from H or optionally substituted C 1-6 alkyl
  • R 3 is optionally substituted aryl
  • each R 4 is selected from H, halogen, carboxamido, nitro, cyano, optionally substituted C 1-6 alkyl, or optionally substituted aryl;
  • R 5 is selected from H, halogen, optionally substituted C 1-6 alkyl, or optionally substituted aryl;
  • each R 6 and R 7 is, independently, selected from H, optionally substituted C 1-6 alkyl, or aryl; and n is 0, 1, 2, or 3.
  • X 1 and X 2 are N
  • X 3 is S
  • Y is C(O)
  • Z is CH 2
  • R 2 is H
  • R 3 is 2-chloro-6-fluoro-phenyl
  • FIG. 1 provides a schematic diagram of retinal ganglion cell death.
  • FIGS. 2A-D provide graphs and a photograph showing TNF- ⁇ expression (A and B), RGC numbers per field (C), and IPL thickness (D) in mice that underwent ON injury and were injected with an anti-TNF- ⁇ neutralizing antibody.
  • FIGS. 2E-F provide graphs showing RIP1 and RIP3 expression in the retina of mice that underwent ON injury.
  • FIGS. 2G-I provide a photograph and graphs showing RIP1 and RIP3 protein levels in the retina of mice having undergone ON injury.
  • FIGS. 3A-G provide photographs and graphs showing TUNEL-positive cells (A and B), Brn3B-positive cells (C and D), IPL thickness (E), GCL and IPL thickness (F and G) in Z-VAD and/or Nec-1 treated mice that underwent ON injury at day one.
  • FIGS. 3H-J provide photographs and graphs showing caspase-8 (H), caspase-9 (I and K) and caspase-3 (J and K) expression in Z-VAD and/or Nec-1 treated mice that underwent ON injury one day after injury.
  • FIG. 3L provide a graph showing TNF- ⁇ expression in Z-VAD and/or Nec-1 treated mice that underwent ON injury one day after ON injury.
  • FIG. 3M-Q provide photographs and graphs showing TUNEL-positive cells (M and N), Brn3B-positive cells (0 and P), and IPL thickness (Q) in Z-VAD and/or Nec-1 treated mice that underwent ON injury at day one, day three, or day 7 after ON injury.
  • FIGS. 4A-B provide a photograph of PI staining (A) and graph showing quantification of PI-positive cells in ZVAD and/or Nec-1 treated mice that underwent ON injury.
  • FIGS. 4C-D provide (A) TEM photographs of RGCs one day after ON injury and (B) quantification of apoptotic and necrotic RGC death, where A denotes apoptosis and N denotes necrosis.
  • FIGS. 5A-G provide photographs and graphs showing TUNEL-positive cells (A and B), Brn3B-positive cells (C and D), IPL thickness (E), and PI-positive cells (F and G) in Z-VAD and/or Nec-1 treated RIP3 ⁇ / ⁇ mice that underwent ON injury.
  • FIGS. 7A-B provide a photograph and graph showing AIF translocation following Z-VAD and/or Nec-1 treatment in wildtype or RIP3 ⁇ / ⁇ mice.
  • FIG. 7C provides a graph showing ROS production in wildtype or RIP3 ⁇ / ⁇ mice.
  • FIG. 8A-B provide transmission electron microscope (TEM) photomicrographs depicting RGCs after ON injury.
  • FIG. 8B indicates formation of autophagosomes (black arrows) and autolysosome (black arrowheads) in necrotic cells with cellular swelling (left and middle panel) and swollen axons (right panel).
  • FIGS. 9A-C provide graphs showing AtgS (A), Atg7 (B), and Atg12 (C) expression after ON injury, as determined by quantitative real-time PCR analysis.
  • FIGS. 9D-E provide photographs showing LC3 protein levels and immunostaining at one day after ON injury.
  • FIGS. 10A-D provide photographs and a graph showing TUNEL-positive cells (A and B) and Brn3b-positive cells (C and D) in mice that were treated with 3-MA.
  • FIG. 10E-F provide graphs depicting IPL thickness (E) and carbonyl contents in mice that were treated with 3-MA (F).
  • FIG. 11 provides a graph showing RGC survival in mice that were treated with Z-VAD and/or Nec-1 following optic nerve crush injury.
  • FIGS. 12A-E provide photographs showing axon regeneration in mice treated with Z-VAD and/or Nec-1 following optic nerve crush injury.
  • FIGS. 12A-E show longitudinal sections of the optic nerve stained with an antibody against ⁇ III-tubulin, following optic nerve crush injury. The vertical arrows denote the locations of the injury sites, and the horizontal reference lines denote regions where axon regeneration were detected following treatment with Nec-1 and ZVAD ( FIGS. 12D-E versus FIGS. 12A-C ).
  • the invention relates to methods and composition for preserving the viability of retinal ganglion cells disposed within a retina of an eye of a subject with certain ocular condition, e.g., glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion, wherein the viability of the retinal ganglion cells are affected by the ocular condition.
  • ocular condition e.g., glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion
  • ocular condition e.g., glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion
  • the viability of the retinal ganglion cells are affected by the ocular condition.
  • apoptosis inhibitor e.g., a pan-caspase inhibitor.
  • FIG. 1 there are two pathways for cell death (apoptosis and necrosis), which appear to be mediated by RIP-1, a serine/threonine kinase.
  • RIP 1 forms a death inducing signaling complex with Fas-associated domain (FADD) and caspase-8, thereby activating caspase-8 and the downstream cascade leading to apoptosis.
  • FADD Fas-associated domain
  • RIP1 kinase when caspase pathway is blocked (for example, with a caspase inhibitor such as ZVAD), RIP1 kinase is activated in a RIP1-RIP3 complex and promotes RGC necrosis. Although autophagy is also activated during RGC death, it is mainly associated with necrotic RGC death. Thus, RIP kinases act as common intermediaries for various upstream death signals, and their blockade in addition to caspases is likely necessary for effective neuroprotection.
  • the methods and compositions described herein are directed to therapies that target both the necrotic and apoptotic pathways of programmed cell death.
  • the methods and compositions disclosed herein facilitate a combination therapy where a necrosis inhibitor, e.g., a necrostatin (e.g., necrostatin-1 or necrostatin-4), can be administered either alone or in combination (either sequentially or simultaneously) with an apoptosis inhibitor e.g., a pan-caspase inhibitor (e.g., Z-VAD or IDN-6556).
  • a necrosis inhibitor e.g., a necrostatin
  • an apoptosis inhibitor e.g., a pan-caspase inhibitor
  • the disclosed methods surprisingly use necrostatins at concentrations higher than those previously thought to be clinically tolerable.
  • necrostatin e.g., necrostatin-1 or necrostatin-4
  • a pan-caspase inhibitor e.g., Z-VAD or IDN-6556
  • cell death is understood to mean the death of a cell, e.g., by apoptosis or necrosis.
  • apoptosis is understood to mean caspase-dependent cell death, which is characterized by any of the following properties: cell shrinkage, nuclear condensation, DNA fragmentation or membrane blebbing.
  • apoptosis inhibitor is understood to mean any agent that, when administered to a mammal, reduces apoptotic cell death in retinal ganglion cells.
  • certain useful apoptosis inhibitors act by reducing or eliminating the activity of one or more members of the intrinsic or extrinsic or common apoptotic pathways.
  • an agent that either directly or indirectly affects the activity of one or more caspases e.g., a pan-caspase inhibitor
  • a caspase inhibitor can affect the activity of a caspase either directly by modulating a specific caspase in the apoptotic pathway or indirectly by modulating a downstream caspase present in the apoptotic pathway.
  • pan-caspase inhibitor is understood to mean a broad-spectrum caspase inhibitor that inhibits at least two, preferably at least three different caspases (e.g., caspase-1, caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, caspase-11, caspase-12, caspase-13, and/or caspase-14.
  • caspase-1, caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, caspase-11, caspase-12, caspase-13, and/or caspase-14 e.g., caspase-1, caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, caspase-11, caspase
  • Z-VAD also known as Benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone and carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone
  • IDN-6556 also known as “PF-3,491,390”
  • a “pan-caspase inhibitor” may also be a cocktail (e.g., a combination) of caspase inhibitors including two or more of specific caspase inhibitors (e.g., synthetic or endogenous caspase inhibitors).
  • necrosis is understood to mean caspase-independent cell death characterized by any of the following properties: cellular and/or organelle swelling, plasma membrane rupture, or discontinuity in plasma, nuclear and/or organelle membranes.
  • the terms “necroptosis” and “programmed necrosis” refer to a form of necrosis and is understood to mean one form of programmed or regulated necrosis, and in certain embodiments, necroptosis is mediated by the serine/threonine kinase activity of receptor interacting protein (RIP) kinases, for example, RIP-1 kinase and/or RIP-3 kinase.
  • RIP receptor interacting protein
  • necrosis inhibitor is understood to mean an agent, which, when administered to a mammal, reduces necrotic cell death in retinal ganglion cells.
  • necrosis inhibitors act by reducing or inhibiting necroptosis or programmed necrosis.
  • a necrosis inhibitor can be an agent that modulates the production and/or activity of one or more RIP kinases (e.g., RIP-1 kinase and/or RIP-3 kinase).
  • an inhibitor of RIP-1 kinase is understood to modulate the activity of RIP-1 kinase as well as downstream RIP kinases, e.g., RIP-3 kinase, in the necrosis cascade. Accordingly, a RIP-1 kinase inhibitor is also understood to modulate RIP-3 kinase activity.
  • necrostatin As used herein, the term “necrostatin” or “nec” is understood to mean an inhibitor of caspase-independent cell death or necroptosis.
  • exemplary necrostatins include necrostatin-1 (“Nec-1”), necrostatin-2 (“Nec-2”), necrostatin-3 (“Nec-3”), necrostatin-4 (“Nec-4”), necrostatin-5 (“Nec-5”) and necrostatin-7 (“Nec-7”).
  • the necrostatin is a Nec-1 related compound of Formula I:
  • X is O or S
  • R 1 is hydrogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, or halogen
  • R 2 is hydrogen or C 1 -C 6 alkyl.
  • X is O.
  • R 1 is hydrogen or halogen (such as chlorine).
  • R 2 is a methyl or ethyl.
  • R 1 is hydrogen or Cl, and R 2 is a methyl.
  • the necrostatin is a Nec-1 related compound of Formula I-A, shown below:
  • R 1 is H, alkyl, alkoxyl, or a halogen (for example, F, Cl, Br or I) and R 2 is H or an alkyl.
  • R 1 is H or Cl.
  • R 2 is a methyl or ethyl.
  • R 1 is H or Cl, and R 2 is a methyl.
  • the necrostatin is a Nec-1 related compound of Formula I-B, shown below:
  • the necrostatin is a Nec-1 related compound of Formula I-C, shown below:
  • the necrostatin is a Nec-1 related compound of Formula I-D, shown below:
  • the necrostatin is a Nec-1 related compound of Formula I-E, shown below:
  • R 1 is H, alkyl, alkoxyl, or a halogen (for example, F, Cl, Br or I) and R 2 is H or an alkyl.
  • R 1 is H or Cl.
  • R 2 is a methyl or ethyl.
  • R 1 is H or Cl, and R 2 is a methyl.
  • the necrostatin is a Nec-1 related compound of Formula I-F, shown below:
  • necrostatin is a Nec-1 related compound of Formula I-G, shown below:
  • Nec-1 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in Degterev et al., (2005) N AT C HEM B IOL 1:112-119; Degterev et al., (2008) N AT C HEM B IOL 4:313-321; and International Patent Application Publication No. WO 2007/075772, all of which are hereby incorporated by reference.
  • the necrostatin is a Nec-2 related compound of Formula II:
  • X is —CH 2 —, —C(H)(R 14 )—, —C( ⁇ S)—, —C( ⁇ NH)—, or —C(O)—;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 each represent independently hydrogen, acyl, acetyl, alkyl, halogen, amino, C 1 -C 6 alkoxyl, nitro, —C(O)R 12 , —C(S)R 12 , —C(O)OR 12 , —C(O)NR 12 R 13 , —C(S)NR 12 R 13 , or —S(O 2 )R 12 ;
  • R 11 is hydrogen, acyl, acetyl, alkyl, or acylamino
  • R 12 and R 13 each represent independently hydrogen, an optionally substituted alkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted aralkyl, or an optionally substituted heteroaralkyl;
  • R 14 is acyl, acetyl, alkyl, halogen, amino, acylamino, nitro, —SR 11 , —N(R 11 ) 2 , or —OR 11 ;
  • the bond indicated by (a) can be a single or double bond
  • the bond indicated by (b) can be a single or double bond.
  • X is —C(O)—.
  • R 1 , R 2 , R 5 , R 6 , R 7 , and R 10 each represent independently hydrogen, acyl, alkyl, halogen, or amino.
  • R 3 , R 4 , R 8 , and R 9 are C 1 -C 6 alkoxyl.
  • the bond indicated by (a) is a double bond; and the bond indicated by (b) is a double bond.
  • X is not —C(O)—, —CH 2 —, or —CH(OH)—.
  • the necrostatin is a Nec-2 related compound of Formula II-A:
  • R 1 , R 2 , R 5 , R 6 , R 7 , and R 10 each represent independently hydrogen, alkyl, halogen, amino, or methoxyl;
  • R 3 , R 4 , R 8 , and R 9 are C 1 -C 6 alkoxyl.
  • the Nec-2 related compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N
  • Nec-2 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in International Patent Application Publication No. WO 2007/075772, which is hereby incorporated by reference.
  • the necrostatin is a Nec-3 related compound of Formula III:
  • Z is —CH 2 —, —CH 2 CH 2 —, —O—, —S—, —S(O)—, —S(O 2 )—, or —N(R 7 )—;
  • R 1 , R 3 , and R 5 each represent independently for each occurrence hydrogen, halogen, hydroxyl, amino, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkoxy-C 1 -C 6 alkyl, C 1 -C 6 alkanoyl, C 1 -C 6 alkylsulfinyl, C 1 -C 6 alkylsulfinyl-C 1 -C 6 alkyl, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonyl-C 1 -C 6 alkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl, or heteroaralkyl;
  • R 2 and R 4 are C 1 -C 6 alkoxy
  • R 6 is —C(O)R 8 , —C(S)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —C(S)NR 8 R 9 , —C(NH)R 8 , or —S(O 2 )R 8 ;
  • R 7 is alkyl, aralkyl, or heteroaralkyl
  • R 8 and R 9 each represent independently hydrogen, C 1 -C 6 alkyl, heteroalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
  • n independently for each occurrence 0, 1, or 2.
  • Z is —CH 2 —.
  • R 1 , R 3 , and R 5 each represent independently for each occurrence hydrogen, halogen, hydroxyl, amino, or C 1 -C 6 alkyl.
  • R 2 and R 4 are methoxy.
  • R 6 is C(O)R 8
  • R 8 is C 1 -C 6 alkyl.
  • R 7 is alkyl.
  • R 8 and R 9 each represent independently hydrogen or C 1 -C 6 alkyl.
  • n is 0.
  • the Nec-3 related compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2
  • the Nec-3 related compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N
  • Nec-3 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in Degterev et al., (2008) N AT C HEM B IOL 4:313-321; and International Patent Application Publication No. WO 2007/075772, both of which is hereby incorporated by reference.
  • the necrostatin is a Nec-4 related compound of Formula IV:
  • R 2 and R 3 each represent independently for each occurrence hydrogen or methyl
  • R 4 represents independently for each occurrence halogen, hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 4 alkynyl;
  • R 5 is C 1 -C 4 alkyl
  • R 6 is hydrogen, halogen, or —CN
  • R 7 is hydrogen or C 1 -C 4 alkyl
  • R 8 is C 1 -C 6 alkyl, or R 8 taken together with R 9 , when present, forms a carbocyclic ring;
  • R 9 is hydrogen or C 1 -C 6 alkyl, or R 9 taken together with R 8 forms a carbocyclic ring;
  • R 10 is hydrogen or C 1 -C 6 alkyl
  • A is phenylene or a 5-6 membered heteroarylene
  • X is N or —C(R 9 )—
  • Y is N or —C(R 10 )—
  • Z is S or O
  • n and n each represent independently 1, 2, or 3.
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R 1 is
  • R 2 is hydrogen. In certain embodiments, R 3 is methyl. In certain other embodiments, R 3 is hydrogen. In certain embodiments, R 4 is halogen, such as fluorine or chlorine. In certain embodiments, R 4 is halogen. In certain embodiments, R 5 is methyl or ethyl. In certain embodiments, R 6 is —CN. In certain embodiments, A is phenylene. In certain embodiments, X is N. In certain embodiments, Y is N. In certain embodiments, Z is S. In certain embodiments, A is phenylene. In certain embodiments, R 1 is C 1 -C 6 alkyl, such as methyl. In certain embodiments, m is 1. In certain embodiments, n is 2.
  • the necrostatin is a Nec-4 related compound of Formula IV-A:
  • the necrostatin is a Nec-4 related compound of Formula IV-B:
  • Nec-4 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in Teng et al., (2007) B IOORG M ED C HEM L ETT , 17: 6836-6840; and Teng et al., (2008) B IOORG M ED C HEM L ETT , 18: 3219-3223, both of which are incorporated herein by reference.
  • the necrostatin is a Nec-5 related compound of Formula V:
  • A is a saturated or unsaturated 5-6 membered carbocyclic ring
  • X is a bond or C 1 -C 4 alkylene
  • R 1 is C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • R 3 is —C 1 -C 6 alkylene-CN, —CN, C 1 -C 6 alkyl, or C 2 -C 6 alkenyl;
  • R 4 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl
  • R 5 represents independently for each occurrence C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • B is a 5-6 membered heterocyclic or carbocylic ring
  • n and p each represent independently 0, 1, or 2.
  • X is a bond.
  • A is an unsaturated 6-membered carbocyclic ring.
  • R 1 is C 1 -C 6 alkyl, halogen, hydroxyl, or C 1 -C 6 alkoxyl.
  • R 2 is
  • R 3 is —C 1 -C 6 alkylene-CN, such as —CH 2 —CN.
  • R 4 represents independently for each occurrence hydrogen or C 1 -C 6 alkyl.
  • R 5 represents independently for each occurrence C 1 -C 6 alkyl, halogen, hydroxyl, or C 1 -C 6 alkoxyl.
  • B is a 5-6 membered heterocyclic ring.
  • n is O.
  • p is 0.
  • the necrostatin is a Nec-5 related compound of Formula V-A:
  • R 1 is C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, or —N(R 4 ) 2 ;
  • R 3 is —C 1 -C 6 alkylene-CN
  • R 4 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl
  • R 5 represents independently for each occurrence C 1 -C 6 alkyl, halogen, hydroxyl, C 1 -C 6 alkoxyl, —N(R 4 ) 2 , —C(O)R 4 , CO 2 R 4 , or C(O)N(R 4 ) 2 ;
  • B is a 5-6 membered heterocyclic or carbocylic ring
  • n and p each represent independently 0, 1, or 2.
  • the Nec-5 compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N
  • Nec-5 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in Degterev et al., (2008) N AT C HEM B IOL 4:313-321; and International Patent Application Publication No. WO 2008/045406, both of which is hereby incorporated by reference.
  • the necrostatin is a Nec-7 related compound of Formula VII:
  • R 1 , R 2 , and R 3 each represent independently hydrogen or C 1 -C 4 alkyl
  • R 5 and R 6 each represent independently for each occurrence halogen, C 1 -C 6 alkyl, hydroxyl, C 1 -C 6 alkoxyl, —N(R 7 ) 2 , —NO 2 , —S—C 1 -C 6 alkyl, —S-aryl, —SO 2 —C 1 -C 6 alkyl, —SO 2 -aryl, —C(O)R 7 , —CO 2 R 7 , —C(O)N(R 7 ) 2 , heterocycloalkyl, aryl, or heteroaryl;
  • R 7 represents independently for each occurrence hydrogen, C 1 -C 6 alkyl, aryl, or aralkyl; or two occurrences of R 7 attached to the same nitrogen atom are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring;
  • A is a 5-6 membered heterocyclic ring
  • p 0, 1, or 2.
  • R 1 is hydrogen. In certain embodiments, R 2 is hydrogen.
  • R 3 is hydrogen. In certain embodiments, R 4 is
  • R 5 is halogen, C 1 -C 6 alkyl, hydroxyl, C 1 -C 6 alkoxyl, or —N(R 7 ) 2 . In certain other embodiments, R 5 is halogen, such as fluorine or chlorine. In certain embodiments, p is O.
  • R 4 is
  • the Nec-7 related compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2
  • Nec-7 related compounds described above can be prepared based on synthetic procedures described in the literature, such as in Zheng et al., in B IOORG M ED C HEM L ETT , 2008, vol. 18, 4932-4935, which is incorporated herein by reference.
  • the necrostatin is a Nec-7 related compound of Formula VIII:
  • each X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is selected, independently, from N or CR X1 ;
  • each Y 1 , Y 2 , and Y 3 is selected, independently, from O, S, NR Y1 , or CR Y2 R Y3 ;
  • each Z 1 and Z 2 is selected, independently, from O, S, or NR Z1 ;
  • each R Y1 and R Z1 is selected, independently, from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, —C( ⁇ O)R 5A , —C( ⁇ O)OR 5A , or —C( ⁇ O)NR 5A R 6A ;
  • each R X1 , R Y2 , and R Y3 is selected, independently, from H, halogen, CN, NC, NO 2 , N 3 , OR 3 , SR 3 , NR 3 R 4 , —C( ⁇ O)R 5A , —C( ⁇ O)OR 5A , —C( ⁇ O)NR 5A R 6A , —S( ⁇ O)R 5A , —S( ⁇ O) 2 R 5A , —S( ⁇ O) 2 OR 5A , —S( ⁇ O) 2 NR 5A R 6A ; optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • each R 1 , R 2 , R 5A , R 5B , R 6A , and R 6B is selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; or R 5A and R 6A , or R 5B and R 6B combine to form a heterocyclyl; and
  • each R 3 and R 4 is selected from H, optionally substituted C 1-6 alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, —C( ⁇ O)R 5B , —C( ⁇ S)R 5B , —C( ⁇ NR 6B )R 5B , —C( ⁇ O)OR 5B , —C( ⁇ O)NR 5B R 6B , —S( ⁇ O)R 5B , —S( ⁇ O) 2 R 5B , —S( ⁇ O) 2 OR 5B , or —S( ⁇ O) 2 NR 5B R 6B .
  • R 1 is H
  • X 1 , X 2 , and X 4 are each CH
  • X 3 , X 5 , and X 6 are each N
  • Y 1 and Y 3 are each S
  • Y 2 is NH
  • Z 1 is NH
  • Z 2 is O
  • R 2 is not 4-fluorophenyl
  • the necrostatin is a Nec-7 related compound of Formula VIII-A:
  • X 1 , X 2 , X 4 , X 5 , R 1 , Y 2 , and R Z1 are as defined for Formula (VIII);
  • each R 2A , R 2B ; R 2C , R 2D , and R 2E is selected, independently, from H, halogen, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, CN, NC, NO 2 , N 3 , OR 7 , SR 7 , S( ⁇ O)R 12 , S( ⁇ O) 2 R 12 , S( ⁇ O)OR 12 , S( ⁇ O) 2 OR 12 , NR 7 R 8 , C( ⁇ O)R 12 , C( ⁇ O)OR 12 , C( ⁇ O)NR 12 R 13 , C( ⁇ S)R 12 , C( ⁇ S)OR 12 , C( ⁇ S)NR 12 R 13 , C( ⁇ NR 9 )R 12 , C( ⁇ NR 9 )OR 12
  • each R 7 , R 8 , and R 9 is selected, independently, from H, optionally substituted C 1-6 alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, S( ⁇ O)R 10 , S( ⁇ O) 2 R 10 , C( ⁇ O)R 10 , C( ⁇ O)OR 10 , C( ⁇ O)NR 10 R 11 , C( ⁇ S)R 10 , C( ⁇ S)OR 10 , C( ⁇ S)NR 10 R 11 , C( ⁇ NR 14 )R 10 , C(NR 14 )OR 10 , or C( ⁇ NR 14 )NR 10 R 11 , or R 7 and R 8 combine to form an optionally substituted heterocyclyl; and
  • each R 10 R 11 , R 12 , R 13 , and R 14 is selected, independently, from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or R 10 and R 11 or R 12 and R 13 combine to form an optionally substituted heterocyclyl.
  • each R 2A , R 2B , R 2C , R 2D , and R 2E is selected, independently, from H, halogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl.
  • the necrostatin is a Nec-7 related compound selected from:
  • Nec-7 related compounds described above can be prepared based on synthetic procedures described in the literature, such as International Patent Application Publication No. WO 2010/075290, which is hereby incorporated by reference.
  • the necrostatin is a Nec-4 related compound of Formula IX:
  • X 1 and X 2 are, independently, N or CR 4 ;
  • X 3 is selected from O, S, NR 5 , or —(CR 5 ) 2 ;
  • Y is selected from C(O) or CH 2 ;
  • Z is (CR 6 R 7 ) n ;
  • R 1 is selected from H, halogen, optionally substituted C 1-6 alkyl, or optionally substituted C 1-6 cycloalkyl, or optionally substituted aryl;
  • R 2 is selected from H or optionally substituted C 1-6 alkyl
  • R 3 is optionally substituted aryl
  • each R 4 is selected from H, halogen, carboxamido, nitro, cyano, optionally substituted C 1-6 alkyl, or optionally substituted aryl;
  • R 5 is selected from H, halogen, optionally substituted C 1-6 alkyl, or optionally substituted aryl;
  • each R 6 and R 7 is, independently, selected from H, optionally substituted C 1-6 alkyl, or aryl;
  • n 0, 1, 2, or 3.
  • X 1 and X 2 are N
  • X 3 is S
  • Y is C(O)
  • Z is CH 2
  • R 2 is H
  • R 3 is 2-chloro-6-fluoro-phenyl
  • the necrostatin is a Nec-4 related compound of Formula IX-A:
  • R 1 , R 2 , R 3 , R 6 and R 7 are as defined in Formula (IX).
  • the necrostatin is a Nec-4 related compound selected from:
  • Nec-4 related compounds described above can be prepared based on synthetic procedures described in the literature, such as U.S. Patent Application Publication No. 2009/0099242, which is hereby incorporated by reference.
  • alkyl is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has about 10 or fewer carbon atoms in its backbone (e.g., C 1 -C 10 for straight chain, C 3 -C 10 for branched chain), and alternatively, 5, 4, 3, 2 or 1 carbon atoms in its backbone.
  • cycloalkyls have from about 3 to about 10 carbon atoms in their ring structure, and alternatively about 5, 6 or 7 carbons in the ring structure.
  • exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, cyclopropyl, and cyclobutyl.
  • alkoxyl or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • An “ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as may be represented by one of —O-alkyl, —O-alkenyl, or —O-alkynyl.
  • alkylene refers to a diradical of an alkyl group. An exemplary alkylene group is —CH 2 CH 2 —.
  • aralkyl refers to an alkyl group substituted with an aryl group.
  • heteroarylkyl refers to an alkyl group substituted with a heteroaryl group.
  • alkenyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as a straight or branched group of 2-12, 2-10, or 2-6 carbon atoms, referred to herein as C 2 -C 12 alkenyl, C 2 -C 10 alkenyl, and C 2 -C 6 alkenyl, respectively.
  • alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2-ethylhexenyl, 2-propyl-2-butenyl, 4-(2-methyl-3-butene)-pentenyl, etc.
  • alkynyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond, such as a straight or branched group of 2-12, 2-8, or 2-6 carbon atoms, referred to herein as C 2 -C 12 alkynyl, C 2 -C 8 alkynyl, and C 2 -C 6 alkynyl, respectively.
  • alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-1-butynyl, 4-propyl-2-pentynyl, and 4-butyl-2-hexynyl, etc.
  • aryl is art-recognized and refers to a carbocyclic aromatic group.
  • aryl groups include phenyl, naphthyl, anthracenyl, and the like. Unless specified otherwise, the aromatic ring may be substituted at one or more ring positions with, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carboxylic acid, —C(O)alkyl, —CO 2 alkyl, carbonyl, carboxyl, alkylthio, sulfonyl, sulfonamido, sulfonamide, ketone, aldehyde, ester, heterocyclyl, heteroaryl, —CF 3 , —CN, or the like.
  • aryl also includes polycyclic ring systems having two or more carbocyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, and/or aryls.
  • the aromatic group is not substituted, i.e., it is unsubstituted.
  • phenylene refers to a multivalent radical (e.g., a divalent or trivalent radical) of benzene.
  • a divalent valent radical of benzene is illustrated by the formula
  • heterocyclyl or “heterocyclic group” are art-recognized and refer to saturated, partially unsaturated, or aromatic 3- to 10-membered ring structures, alternatively 3-to 7-membered rings, whose ring structures include one to four heteroatoms, such as nitrogen, oxygen, and sulfur. Heterocycles may also be mono-, bi-, or other multi-cyclic ring systems. A heterocycle may be fused to one or more aryl, partially unsaturated, or saturated rings.
  • Heterocyclyl groups include, for example, biotinyl, chromenyl, dihydrofuryl, dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, homopiperidinyl, imidazolidinyl, isoquinolyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, oxolanyl, oxazolidinyl, phenoxanthenyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrazolinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolidin-2-onyl, pyrrolinyl, tetrahydrofuryl, tetrahydroisoquinolyl, tetrahydropyranyl, tetrahydroquinolyl, thiazolidinyl, th
  • the heterocyclic ring is optionally substituted at one or more positions with substituents such as alkanoyl, alkoxy, alkyl, alkenyl, alkynyl, amido, amidino, amino, aryl, arylalkyl, azido, carbamate, carbonate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, imino, ketone, nitro, phosphate, phosphonato, phosphinato, sulfate, sulfide, sulfonamido, sulfonyl and thiocarbonyl.
  • the heterocyclyl group is not substituted, i.e., it is unsubstituted.
  • heteroaryl is art-recognized and refers to aromatic groups that include at least one ring heteroatom. In certain instances, a heteroaryl group contains 1, 2, 3, or 4 ring heteroatoms. Representative examples of heteroaryl groups include pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrazolyl, pyridinyl, pyrazinyl, pyridazinyl and pyrimidinyl, and the like.
  • the heteroaryl ring may be substituted at one or more ring positions with, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carboxylic acid, —C(O)alkyl, —CO 2 alkyl, carbonyl, carboxyl, alkylthio, sulfonyl, sulfonamido, sulfonamide, ketone, aldehyde, ester, heterocyclyl, aryl, —CF 3 , —CN, or the like.
  • heteroaryl also includes polycyclic ring systems having two or more rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, and/or aryls.
  • heteroarylene refers to a multi-valent (e.g., di-valent or trivalent) aromatic group that comprises at least one ring heteroatom.
  • An exemplary “heteroarylene” is pyridinylene, which is a multi-valent radical of pyridine.
  • a divalent radical of pyridine is illustrated by the formula
  • ortho, meta and para are art-recognized and refer to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively.
  • 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formula:
  • R 50 and R 51 each independently represent hydrogen, alkyl, alkenyl, or —(CH 2 ) m —R 61 ; or R 50 and R 51 , taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; wherein R 61 is aryl, cycloalkyl, cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8.
  • R 50 and R 51 each independently represent hydrogen or alkyl.
  • amide or “amido” as used herein refers to a radical of the form —R a C(O)N(R b )—, —R a C(O)N(R b )R c —, —C(O)NR b R c , or —C(O)NH 2 , wherein R a , R b and R c are each independently selected from alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, and nitro.
  • the amide can be attached to another group through the carbon, the nitrogen, R b , R c or R a .
  • the amide also may be cyclic, for example R b and R c , R a and R b , or R a and R c may be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or a 5- to 6-membered ring.
  • the term “carboxamido” refers to the structure —C(O)NR b R c .
  • sulfonamide or “sulfonamido” as used herein refers to a radical having the structure —N(R r )—S(O) 2 —R s — or —S(O) 2 —N(R r )R s , where R r , and R s can be, for example, hydrogen, alkyl, aryl, cycloalkyl, and heterocyclyl.
  • Exemplary sulfonamides include alkylsulfonamides (e.g., where R s is alkyl), arylsulfonamides (e.g., where R s is aryl), cycloalkyl sulfonamides (e.g., where R s is cycloalkyl), and heterocyclyl sulfonamides (e.g., where R s is heterocyclyl), etc.
  • sulfonyl refers to a radical having the structure R u SO 2 —, where R u can be alkyl, aryl, cycloalkyl, and heterocyclyl, e.g., alkylsulfonyl.
  • alkylsulfonyl refers to an alkyl group attached to a sulfonyl group.
  • the term “optionally substituted” as used herein means that the specified group may be substituted at one, two or more positions with, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carboxylic acid, —C(O)alkyl, —CO 2 alkyl, carbonyl, carboxyl, alkylthio, sulfonyl, sulfonamido, sulfonamide, ketone, aldehyde, ester, heterocyclyl, heteroaryl, —CF 3 , —CN, or the like.
  • the term “therapeutically effective amount” is understood to mean the amount of an active ingredient, for example, a necrostatin (e.g., necrostatin-1 or necrostatin-4) and/or a pan-caspase inhibitor (e.g., Z-VAD or IDN-6556) that is sufficient to reduce, minimize or eliminate the death of retinal ganglion cells associated with certain ocular conditions described herein.
  • the compounds of the invention are administered in amounts effective at, e.g., reducing the death of retinal ganglion cells, increasing efficacy compared to monotherapy with either drug alone, preserving or improving vision, preserving or improving visual function, preventing vision loss, and/or promoting axon regeneration. It is understood that preserving vision or visual function, includes stabilizing vision or visual function and/or slowing the decline of vision or visual function prior to treatment.
  • “pharmaceutically acceptable” or “pharmacologically acceptable” mean molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or to a human, as appropriate.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • a method of preserving the visual function of an eye of a subject with ocular conditions wherein a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of the eye with the conditions.
  • the method comprises (a) administering to the eye of the subject an effective amount of a necrosis inhibitor (e.g., a necrostatin, e.g., necrostatin-1 or necrostatin-4) and an effective amount of an apoptosis inhibitor thereby preserving the viability of the retinal ganglion cells disposed within the retina of the eye, and (b) measuring the visual function (e.g., visual acuity) of the eye after the administration of the necrosis inhibitor and the apoptosis inhibitor.
  • a necrosis inhibitor e.g., a necrostatin, e.g., necrostatin-1 or necrostatin-4
  • an apoptosis inhibitor e.g., apoptosis inhibitor
  • the visual function of the eye may be preserved or improved relative to the visual function of the eye prior to administration of the necrosis inhibitor and the apoptosis inhibitor. Further, after the administration of the necrosis inhibitor and the apoptosis inhibitor, the preserved retinal ganglion cell is capable of supporting axonal regeneration.
  • a method of preserving the visual function of an eye of a subject with ocular condition wherein a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of the eye with the condition.
  • the method comprises reducing the production and/or activity of a RIP-1 kinase and/or a RIP-3 kinase in the eye to preserve the viability of the retinal ganglion cells disposed within the eye.
  • the reduction in the production and/or activity of the RIP-1 kinase and/or RIP-3 kinase may be achieved by administering an effective amount of a necrosis inhibitor (e.g., RIPK inhibitor, e.g., a necrostatin).
  • a necrosis inhibitor e.g., RIPK inhibitor, e.g., a necrostatin
  • the reduction in the production and/or activity of the RIP-1 kinase and/or RIP-3 kinase may be direct (e.g., the necrosis inhibitor modulates the production and/or activity the RIP-1 kinase and/or RIP-3 kinase directly) or indirect (e.g., the necrosis inhibitor acts upstream of the RIP-1 kinase and/or RIP-3 kinase but the administration of which indirectly modulates the production and/or activity of the RIP-1 kinase and/or RIP-3 kinase).
  • direct e.g., the necrosis inhibitor modulates the production and/or activity the RIP-1 kinase and/or RIP-3 kinase directly
  • indirect e.g., the necrosis inhibitor acts upstream of the RIP-1 kinase and/or RIP-3 kinase but the administration of which indirectly modulates the production and/or activity of the RIP-1 kinase and/
  • Visual function of the eye may be measured before and/or after the administration of the necrosis inhibitor that directly or indirectly reduces the production and/or activity of a RIP-1 kinase and/or RIP-3 kinase. After administration of the necrosis inhibitor the visual function of the eye may be preserved or improved relative to the visual function of the eye prior to administration of the necrosis inhibitor.
  • the ocular condition wherein a symptom of the condition is the loss of retinal ganglion cell viability in the retina of the eye, includes but is not limited to glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion.
  • optic neuropathies such as ischemic optic neuropathy (e.g., arteritic or non-arteritic anterior ischemic neuropathy and posterior ischemic optic neuropathy), compressive optic neuropathy, infiltrative optic neuropathy, traumatic optic neuropathy, mitochondrial optic neuropathy (e.g., Leber's optic neuropathy), nutritional optic neuropathy, toxic optic neuropathy, and hereditary optic neuropathy (e.g., Leber's optic neuropathy, Dominant Optic Atrophy, Behr's syndrome).
  • ischemic optic neuropathy e.g., arteritic or non-arteritic anterior ischemic neuropathy and posterior ischemic optic neuropathy
  • compressive optic neuropathy e.g., infiltrative optic neuropathy
  • traumatic optic neuropathy e.g., mitochondrial optic neuropathy (e.g., Leber's optic neuropathy), nutritional optic neuropathy, toxic optic neuropathy, and hereditary optic neuropathy (e.g., Leber's optic neuropathy, Dominant Op
  • the method comprises administering to the eye of the subject an effective amount of a necrostatin and an effective amount of an apoptosis inhibitor thereby preserving the viability of the retinal ganglion cells disposed within the retina of the eye and the visual function of the eye.
  • a method of preserving the viability of retinal ganglion cells disposed within a retina of a mammalian eye affected by, for example, glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion comprises administering a necrosis inhibitor and/or an apoptosis inhibitor to the eye in which a region of the retina has been affected in amounts sufficient to preserve the viability of retinal ganglion cells disposed within the region of the affected retina.
  • the preserved retinal ganglion cell is capable of supporting axonal regeneration.
  • a method for promoting axon regeneration in a eye of a subject with an ocular condition wherein a symptom of the ocular condition is the loss of retinal ganglion cell viability in the retina of the eye with the condition.
  • the method comprises administering to the eye of the subject an effective amount of a necrostatin and an effective amount of an apoptosis inhibitor thereby promoting axon regeneration of the retinal ganglion cell within the retina of the eye.
  • the methods and compositions described herein can be used to preserve the viability and/or promote axon regeneration of retinal ganglion cells during treatment of the underlying conditions including, but not limited to, glaucoma, optic nerve injury, optic neuritis, optic neuropathies, diabetic retinopathy, central retinal artery occlusion, and central retinal vein occulusion.
  • the necrostatin can be administered to give a final concentration of greater than about 5 ⁇ M, for example, in the range of about 5 ⁇ M to about 1000 ⁇ M. In certain embodiments, the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 10 ⁇ M. In another embodiment, the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 50 ⁇ M. In another embodiment, the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 100 ⁇ M.
  • the necrostatin may be administered in an amount sufficient to give a final concentration of necrostatin in the eye in the range from about 5 ⁇ M to about 1000 ⁇ M, 10 ⁇ M to about 1000 ⁇ M, 50 ⁇ M to about 1000 ⁇ M, 80 ⁇ M to about 1000 ⁇ M, about 100 ⁇ M to about 1000 ⁇ M, about 150 ⁇ M to about 1000 ⁇ M, from about 200 ⁇ M to about 800 ⁇ M, or from about 200 ⁇ M to about 600 ⁇ M.
  • the necrostatin is administered in an amount sufficient to give a final concentration of necrostatin in the eye of about 400 ⁇ M.
  • the apoptosis inhibitor for example, the pan-caspase inhibitor, can be administered in an amount sufficient to give a final concentration of the inhibitor in the eye of greater than about 3 ⁇ M, for example, in the range of about 3 ⁇ M to about 500 ⁇ M.
  • the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 3 ⁇ M.
  • the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 30 ⁇ M.
  • the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 50 ⁇ M.
  • the necrostatin can be administered in an amount sufficient to give a final concentration of necrostatin in the eye of greater than about 100 ⁇ M.
  • the apoptosis inhibitor can be administered in an amount sufficient to give a final concentration of the inhibitor in the eye in the range from about 3 ⁇ M to about 500 ⁇ M, from about 80 ⁇ M to about 500 ⁇ M, 100 ⁇ M to about 500 ⁇ M, 125 ⁇ M to about 500 ⁇ M, 150 ⁇ M to about 500 ⁇ M or from about 200 ⁇ M to about 400 ⁇ M.
  • apoptosis inhibitor e.g., the pan-caspase inhibitor
  • the volume of the eye in a given subject is known (for example, typical human eye contains 4 to 6 mL of fluid (humor)
  • the dosage of the necrostatin and/or the pan-caspase inhibitor is calculated to give the therapeutically effective concentrations noted above.
  • the dosage of the necrostatin and/or the pan-caspase inhibitor can be administered to achieve the concentrations noted above.
  • from about 0.025 mg to about 4 mg, from about 0.035 mg to about 2 mg, from about 0.05 mg to about 2 mg, from about 0.1 mg to about 2 mg, from about 0.2 mg to about 1 mg, or from about 0.2 mg to about 0.8 mg of the necrosis inhibitor (e.g., a necrostatin) can be administered locally to the eye of a mammal. In one embodiment, 0.5 mg of necrostatin is administered locally to the eye of a mammal.
  • from about 0.05 mg to about 2 mg, from about 0.2 mg to about 2 mg, from about 0.05 mg to about 1.5 mg, from about 0.15 mg to about 1.5 mg, from about 0.4 mg to about 1 mg, or from about 0.5 mg to about 0.8 mg of an apoptosis inhibitor (e.g., a pan-caspase inhibitor, e.g., Z-VAD) can be administered locally to the eye of a mammal.
  • a pan-caspase inhibitor e.g., Z-VAD
  • about 0.7 mg of a pan-caspase inhibitor, e.g., Z-VAD is administered locally to the eye of a mammal.
  • the invention relates to the use of a necrosis inhibitor, either alone or in combination with an apoptosis inhibitor, for preserving the viability and/or promoting the axon regeneration of retinal ganglion cells for the treatment of the ocular disorder.
  • the invention also relates to the use of an apoptois inhibitor, either alone or in combination with a necrosis inhibitor, for preserving the viability and/or promoting the axon regeneration of retinal ganglion cells for the treatment of the ocular disorder.
  • one or more of a necrosis inhibitor, one or more of an apoptosis inhibitor, or one or more of a necrosis inhibitor and one or more of an apoptosis inhibitor can be administered to the eye in which a region of the retina has been affected in amounts sufficient to preserve the viability and/or promote axon regeneration of retinal ganglion cells disposed within the region of the affected retina.
  • the necrosis inhibitor is a necrostatin, for example, necrostatin-1, a necrostatin-2, a necrostatin-4, a necrostatin-5, and a necrostatin-7.
  • a necrostatin for example, necrostatin-1, a necrostatin-2, a necrostatin-4, a necrostatin-5, and a necrostatin-7.
  • apoptosis inhibitors e.g., IDN-6556
  • one or more of the necrostatins shown by Formua I, I-A, I-B, I-C, I-D, I-E, I-F, I-G, II, II-A, III, IV, IV-A, IV-B, V, V-A, VII, VIII, VIII-A, IX, or IX-A can be administered with one or more of the apoptosis inhibitors (e.g., IDN-6556 or IDN-6734) listed below.
  • the apoptosis inhibitors e.g., IDN-6556 or IDN-6734
  • the necrosis inhibitor reduces the production and/or activity of a RIP-1 kinase and/or a RIP-3 kinase.
  • RIP kinase inhibitors e.g., RIP-1 kinase and/or RIP-3 kinase inhibitors
  • RNAs including small inhibitory RNAs (siRNAs) and short hairpin RNAs (shRNAs). Methods for designing and synthesizing siRNAs and shRNAs are well known in the art.
  • Exemplary RIP-1 kinase inhibitors include, for example, a pSIREN-RIP-1 shRNA construct which targets RIP-1 kinase as disclosed in Kaiser et al., (2008) J OURNAL OF I MMUNOLOGY 181:6427-6434.
  • Exemplary RIP-3 kinase inhibitors include, for example, sc-61482-SH and sc-135170 available from Santa Cruz Biotechnology.
  • RIP kinase inhibitors may include inhibitor of apoptosis proteins (IAPB), active fragments thereof, and nucleic acids encoding the same. It is well established that IAPB inhibit RIP-1 kinase by functioning as a E3 ligase for RIP-1 kinase (see, for example, Van Wegakker et al., (2010)).
  • IAPB inhibitor of apoptosis proteins
  • the one or more apoptosis inhibitors may include a pan-caspase inhibitor.
  • the pan-caspase inhibitor can be Z-VAD (i.e., Z-Val-Ala-Asp(OMe)-CH 2 F*), IDN-6556 available from Conatus Pharmaceuticals (i.e., (3- ⁇ 2-[(2-tert-butyl-phenylaminooxalyl)-amino]-propionylamino ⁇ -4-oxo-5-(2,3,5,6-tetrafluoro-phenoxy)-pentanoic acid) (3- ⁇ 2-[(2-tert-butyl-phenylaminooxalyl)-amino]-propionylamino ⁇ -4-oxo-5-(2,3,5,6-tetrafluoro-phenoxy)-pentanoic acid), IDN-6734 available from Conatus Pharmaceuticals, VX-799 available from Vertex Pharmaceuticals, MX1013 and MX20
  • the pan-caspase inhibitor can be a cocktail of caspase inhibitors including two or more specific caspase inhibitors (e.g., synthetic caspase inhibitors) such as a caspase 1 inhibitor, a caspase 2 inhibitor, a caspase 3 inhibitor, a caspase 4 inhibitor, a caspase 5 inhibitor, a caspase 6 inhibitor, a caspase 7 inhibitor, a caspase 8 inhibitor, and a caspase 9 inhibitor. It is contemplated that one or more of the pan-caspase inhibitors may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • necrostatins e.g., necrostain-1 and/or necrostatin-4
  • Exemplary synthetic caspase 1 inhibitors include, for example, Ac-N-Me-Tyr-Val-Ala-Asp-aldehyde (SEQ ID NO: 7), Ac-Trp-Glu-His-Asp-aldehyde (SEQ ID NO: 8), Ac-Tyr-N-Me-Val-Ala-N-Me-Asp-aldehyde (SEQ ID NO: 9), Ac-Tyr-Val-Ala-Asp-Aldehyde (SEQ ID NO: 10), Ac-Tyr-Val-Ala-Asp-chloromethylketone (SEQ ID NO: 11), Ac-Tyr-Val-Ala-Asp-2,6-dimethylbenzoyloxymethylketone (SEQ ID NO: 12), Ac-Tyr-Val-Ala-Asp(OtBu)-aldehyde-dimethyl acetol (SEQ ID NO: 13), Ac-Tyr
  • exemplary caspase 1 inhibitors include, for example, Z-Val-Ala-Asp-fluoromethylketone, biotin-X-Val-Ala-Asp-fluoromethylketone, Ac-Val-Ala-Asp-aldehyde, Boc-Asp-fluoromethylketone, Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Pro-Tyr-Val-Ala-Asp-aldehyde (SEQ ID NO: 1), biotin-Tyr-Val-Ala-Asp-fluoroacyloxymethylketone (SEQ ID NO: 20), Ac-Tyr-Val-Ala-Asp-acyloxymethylketone (SEQ ID NO: 21), Z-Asp-CH 2 -DCB, and Z-Tyr-Val-Ala-Asp-fluoromethylketone (SEQ ID
  • Exemplary synthetic caspase 2 inhibitors include, for example, Ac-Val-Asp-Val-Ala-Asp-aldehyde (SEQ ID NO: 23), which can be obtained from Bachem Bioscience Inc., PA, and Z-Val-Asp-Val-Ala-Asp-fluoromethylketone (SEQ ID NO: 24), which can be obtained from Calbiochem, Calif.
  • SEQ ID NO: 23 Ac-Val-Asp-Val-Ala-Asp-aldehyde
  • SEQ ID NO: 24 Z-Val-Asp-Val-Ala-Asp-fluoromethylketone
  • Exemplary synthetic caspase 3 precursor protease inhibitors include, for example, Ac-Glu-Ser-Met-Asp-aldehyde (pseudo acid) (SEQ ID NO: 25) and Ac-Ile-Glu-Thr-Asp-aldehyde (pseudo acid) (SEQ ID NO: 26) which can be obtained from Bachem Bioscience Inc., PA.
  • Exemplary synthetic caspase 3 inhibitors include, for example, Ac-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 27), Ac-Asp-Met-Gln-Asp-aldehyde (SEQ ID NO: 28), biotinyl-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 29), Z-Asp-Glu-Val-Asp-chloromethylketone (SEQ ID NO: 30), Z-Asp(OMe)-Glu(OMe)-Val-DL-Asp(OMe)-fluoromethylketone (SEQ ID NO: 31), and Z-Val-Ala-DL-Asp(OMe)-fluoromethylketone which can be obtained from Bachem Bioscience Inc., PA.
  • caspase 3 inhibitors include, for example, Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 2), biotin-X-Asp-Glu-Val-Asp-fluoromethylketone (SEQ ID NO: 32), Ac-Asp-Glu-Val-Asp-chloromethylketone (SEQ ID NO: 33), all of which are available from Calbiochem.
  • SEQ ID NO: 2 Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Leu-Ala-Pro-Asp-Glu-Val-Asp-aldehyde
  • SEQ ID NO: 32 biotin-X-Asp-Glu-Val-Asp-fluoromethylketone
  • Another exemplary caspase 3 inhibitor includes, the caspase 3 inhibitor N-benzyloxycarbonal-Asp(OMe)-Glu(OMe)-Val-Asp(Ome)-fluoromethyketone (z-Asp-Glu-Val-Asp-fmk) (SEQ ID NO: 34), which is available from Enzyme Systems Products.
  • Additional exemplary caspase 3 inhibitors include M-826 and M-791 available from Merck-Frosst, Immunocasp-3, Ad-G/iCasp3, and PEF-F8-CP3.
  • Exemplary synthetic caspase 4 inhibitors include, for example, Ac-Leu-Glu-Val-Asp-aldehyde (SEQ ID NO: 35) and Z-Tyr-Val-Ala-DL-Asp-fluoromethylketone (SEQ ID NO: 36), which can be obtained from Bachem Bioscience Inc., PA, and Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Leu-Glu-Val-Pro-aldehyde (SEQ ID NO: 3), which can be obtained from Calbiochem, Calif.
  • Exemplary synthetic caspase 5 inhibitors include, for example, Z-Trp-His-Glu-Asp-fluoromethylketone (SEQ ID NO: 37), which can be obtained from Calbiochem, Calif., and Ac-Trp-Glu-His-Asp-aldehyde (SEQ ID NO: 38) and Z-Trp-Glu(O-Me)-His-Asp(O-Me) fluoromethylketone (SEQ ID NO: 39), which can be obtained from Sigma Aldrich, Germany.
  • Exemplary synthetic caspase 6 inhibitors include, for example, Ac-Val-Glu-Ile-Asp-aldehyde (SEQ ID NO: 40), Z-Val-Glu-Ile-Asp-fluoromethylketone (SEQ ID NO: 41), and Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Val-Glu-Ile-Asp-aldehyde (SEQ ID NO: 4), which can be obtained from Calbiochem.
  • Another exemplary caspase 6 inhibitor includes Immunocasp-6.
  • Exemplary synthetic caspase 7 inhibitors include, for example, Z-Asp(OMe)-Gln-Met-Asp(OMe) fluoromethylketone (SEQ ID NO: 42), Ac-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 43), Biotin-Asp-Glu-Val-Asp-fluoromethylketone (SEQ ID NO: 44), Z-Asp-Glu-Val-Asp-fluoromethylketone (SEQ ID NO: 45), Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 2), which can be obtained from Sigma Aldrich, Germany.
  • Exemplary synthetic caspase 8 inhibitors include, for example, Ac-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 46), Ac-Ile-Glu-Pro-Asp-aldehyde (SEQ ID NO: 47), Ac-Ile-Glu-Thr-Asp-aldehyde (SEQ ID NO: 48), Ac-Trp-Glu-His-Asp-aldehyde (SEQ ID NO: 49) and Boc-Ala-Glu-Val-Asp-aldehyde (SEQ ID NO: 50) which can be obtained from Bachem Bioscience Inc., PA.
  • exemplary caspase 8 inhibitors include, for example, Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Ile-Glu-Thr-Asp-aldehyde (SEQ ID NO: 5) and Z-Ile-Glu-Thr-Asp-fluoromethylketone (SEQ ID NO: 51), which can be obtained from Calbiochem, Calif.
  • Exemplary synthetic caspase 9 inhibitors include, for example, Ac-Asp-Glu-Val-Asp-aldehyde (SEQ ID NO: 52), Ac-Leu-Glu-His-Asp-aldehyde (SEQ ID NO: 53), and Ac-Leu-Glu-His-Asp-chloromethylketone (SEQ ID NO: 54) which can be obtained from Bachem Bioscience Inc., PA.
  • exemplary caspase 9 inhibitors include, for example, Z-Leu-Glu-His-Asp-fluoromethylketone (SEQ ID NO: 55) and Ac-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Leu-Glu-His-Asp-aldehyde (SEQ ID NO:6), which can be obtained from Calbiochem, Calif.
  • Another exemplary caspase 9 inhibitor includes FKBP12/caspase-9 fusion protein.
  • the pan-caspase inhibitor may also be an endogenous caspase inhibitor or a combination of an endogenous caspase inhibitor with one or more synthetic caspase inhibitors.
  • one useful class of endogenous caspase inhibitor includes proteins known as inhibitors of apoptosis proteins (IAPB) (Deveraux et al., (1998) EMBO J. 17(8): 2215-2223) including bioactive fragments and analogs thereof.
  • IAP X-linked inhibitor of apoptosis protein
  • Another exemplary IAP includes survivin (see, U.S. Pat. No.
  • the one or more apoptosis inhibitors may target the inhibitor of apoptosis proteins (IAPB) and second mitochondria-derived activator of caspases (SMACs).
  • IAPB apoptosis proteins
  • SMACs second mitochondria-derived activator of caspases
  • Exemplary apoptosis inhibitors that target IAPB and SMACs include, for example, BIR3 antagonists available from Idun Pharmaceuticals, capped tripeptide XIAP antagonists from Abbot Laboratories, TWX024, polyphenylurea derivatives, SMAC-mimetic compounds, embelin, XIAP antisense and RNAi constructs, AEG35156/GEM®640 available from Aegera Therapeutics, HIV-Tat- and polyarginine conjugated SMAC peptides, and nonpeptide small-molecule SMAC mimetics. It is contemplated that one or more of the apoptosis inhibitors which target IAPB and SMACs may be used in
  • the one or more apoptosis inhibitors may target the TNF-related apoptosis-inducing ligand (TRAIL) receptors.
  • TRAIL TNF-related apoptosis-inducing ligand
  • Exemplary apoptosis inhibitors that target the TRAIL receptors include, for example, HGS-ETR1, HGS-ETR2, and HGS-TR2J available from Human Genome Sciences, and PRO1762 available from Amgen. It is contemplated that one or more of the apoptosis inhibitors which target the TRAIL receptors may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • necrostatins e.g., necrostain-1 and/or necrostatin-4.
  • the one or more apoptosis inhibitors may target CD95/Fas.
  • exemplary apoptosis inhibitors that target CD95/FAS include, for example, CD95-Fc available from ApoGenix GmbH. It is contemplated that one or more of the apoptosis inhibitors which target CD95/Fas may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • the one or more apoptosis inhibitors may be an anti-FasL factors.
  • anti-FasL factors include, for example, anti-FasL neutralizing antibody (available, for example, from Pharmingen, San Diego, Calif.); peptides and nucleic acids (for example, anti-FasL aptamers) that bind FasL to prevent or reduce its binding to its cognate receptor; certain antibodies and antigen binding fragments thereof and peptides that bind preferentially to the Fas receptor; antisense nucleotides and double stranded RNA for RNAi that ultimately reduce or eliminate the production of either FasL or the Fas receptor; soluble Fas; soluble FasL; decoy receptor-3 (DcR3) and analogues thereof; matrix metalloproteinases (MMPs); vasoactive intestinal peptide (VIP); pituitary adenylate cyclase-activating polypeptide (PACAP); forskolin
  • the one or more apoptosis inhibitors may target the tumor necrosis factor (TNF).
  • TNF tumor necrosis factor
  • exemplary apoptosis inhibitors that target TNF include, for example, recombinant TNF- ⁇ , adalimumab available from Abbott, infliximab available from Centocor Ortho Biotech Inc., etanercept from Amgen, CDP571 available from Celltech, and ISIS 104838 (a 2′-O-methoxyethyl antisense construct against TNF-alpha) available from ISIS Pharmaceuticals. It is contemplated that one or more of the apoptosis inhibitors which target TNF may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • necrostatins e.g., necrostain-1 and/or necrostatin-4.
  • the one or more apoptosis inhibitors may target survivin.
  • exemplary apoptosis inhibitors that target survivin include, for example, LY2181308 available from ISIS Pharmaceuticals and Ad-survivin T34A. It is contemplated that one or more of the apoptosis inhibitors which target survivin may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • the one or more apoptosis inhibitors may target the Bcl-2 proteins.
  • Exemplary apoptosis inhibitors that target the Bcl-2 proteins include, for example, Bcl-2 blockers available from Idun Pharmaceuticals and Abbot Laboratories, Gx01 series of compounds available from Gemin X Pharmaceuticals, Bcl-2 small-molecule antagonist, Tetrocarcin-A derivatives available from Kyowa Hakko Kogyo Co., Chelerythrine, antimycin A derivatives, HA14-1, synthetic compound binding to the BH3 of Bcl-2, Genasense available from Sanofi-Aventis, ISIS 22783 available from ISIS Pharmaceuticals, bispecific Bcl-2/Bcl-XL antisense, BH3 peptides from Bax, Bak, Bid or Bad, SAHBs, and BH3Is. It is contemplated that one or more of the apoptosis inhibitors which target the Bcl-2 proteins may be used in combination with one
  • the one or more apoptosis inhibitors may target p53.
  • exemplary apoptosis inhibitors that target p53 include, for example, INGN201 available from Invitrogen Therapeutics, SCH58500 available from Schering-Plough, ONYX-015 available from Onyx Pharmaceuticals, C-terminal p53 peptides, CDB3, Amifostine, CP31398 available from Pfizer, Prima-1, HPF E6-binding peptide aptamers, Nutlins available from Roche, Chalcones, Small peptide compounds, and Pifithrin- ⁇ . It is contemplated that one or more of the apoptosis inhibitors which target p53 may be used in combination with one or more necrostatins (e.g., necrostain-1 and/or necrostatin-4).
  • necrostatins e.g., necrostain-1 and/or necrostatin-4
  • necrostatins may be used in combination with a pan-caspase inhibitor.
  • necrostain-1 and/or necrostatin-4 may be used in combination with Z-VAD available from R&D Systems (Cat. No. FMK001) and Promega (Cat. No. G7231).
  • necrostain-1 and/or necrostatin-4 may be used in combination with IDN-6556 available from Conatus Pharmaceuticals.
  • necrostain-1 and/or necrostatin-4 may be used in combination with IDN-6734 available from Conatus Pharmaceuticals.
  • necrostatins may be used in combination with a TNF inhibitor.
  • necrostain-1 and/or necrostatin-4 may be used in combination with adalimumab available from Abbot Laboratories.
  • necrostain-1 and/or necrostatin-4 may be used in combination with etanercept available from Amgen, Inc.
  • necrostain-1 and/or necrostatin-4 may be used in combination with infiximab available from Centocor Ortho Biotech, Inc.
  • necrostatins may be used in combination with a p53 agonist.
  • necrostain-1 and/or necrostatin-4 may be used in combination with INGN 201 available from Invitrogen Therapeutics.
  • necrostain-1 and/or necrostatin-4 may be used in combination with nutlins, for example, nutlin-3 available from Cayman Chemical (Cat. No. 10004372).
  • necrostain-1 and/or necrostatin-4 may be used in combination with CP31398 available from Tocris Bioscience (Cat. No. 3023).
  • necrostatins may be used in combination with an anti-FasL factor.
  • necrostain-1 and/or necrostatin-4 may be used in combination with anti-FasL neutralizing antibody available from Pharmingen (San Diego, Calif.).
  • apoptotic inhibitor can modulate both the apoptotic and necrotic pathways
  • the necrosis inhibitor can modulate both the necrotic and apoptotic pathways.
  • a RIP-1 inhibitor may inhibit both necrotic and apoptotic cell death thus preserving the viability of the retinal ganglion cells in the retina of the eye of a subject with an ocular condition as disclosed herein.
  • the methods and compositions of the invention can preserve the visual function of an eye of a subject with an ocular condition.
  • Assessment of axonal regeneration may also be monitored through visual function tests as disclosed herein.
  • Visual function can be measured using one or more of a variety of methods well-known in the art.
  • visual function can be assessed by measuring visual acuity.
  • Visual acuity can be assessed, for example, by using conventional “eye charts” in which visual acuity is evaluated by the ability to discern letters of a certain size, with five letters of a given size present on each line (see, e.g., the “ETDRS” eye chart described in the Murphy, R.
  • Evaluation of visual acuity may also be achieved by measuring reading speed and reading time. Visual acuity may be measured to evaluate whether administration of a necrosis inhibitor and/or an apoptosis inhibitor to the affected eye preserves or permits improvement of visual acuity (e.g., to 20/40 vision or to 20/20 vision).
  • Visual function may also be measured by determining whether there is an increase in the thickness of the Nerve Fiber layer (NFL) (e.g., NFL thickness is 15% thicker than, 35% thicker than, 50% thicker than, 60% thicker than, 70% thicker than, or 80% thicker than a macula without the treatment as measured by optical coherence tomography (OCT); an improvement of the ganglion cell layer or bipolar cell layer or photoreceptor cell layer or its subdivisions as seen in the OCT; an improvement of visual field (e.g., by at least 10% in the mean standard deviation on the Humphrey Visual Field Test; an improvement of an electroretinograph (ERG), a measurement of the electrical response of the retina to light stimulation, (e.g., to increase ERG amplitude by at least 15%); and/or preservation or improvement of multifocal ERG, which evaluates the response of the retina to multifocal stimulation and allows characterization of the function of a limited area of the retina.
  • NNL Nerve Fiber layer
  • OCT optical co
  • Visual function may also be measured by electrooculography (EOG), which is a technique for measuring the resting potential of the retina.
  • EOG is particularly useful for the assessment of RPE function.
  • EOG may be used to evaluate whether administration of a necrosis inhibitor and/or an apoptosis inhibitor to the retina of the affected eye preserves or permits improvement in, for example, the Arden ratio (e.g., an increase in Arden ratio of at least 10%).
  • Visual function may also be assessed through fundus autofluorescence (AF) imaging, which is a clinical tool that allows evaluation of the interaction between photoreceptor cells and the RPE.
  • AF fundus autofluorescence
  • Fundus AF imaging may be used to evaluate whether administration of a necrosis inhibitor and/or an apoptosis inhibitor to the retina of the affected eye slows disease progression.
  • Visual function may also be assessed by evaluation of contrast sensitivity, which a measurement of the ability to discern between luminances of different levels in a static image.
  • contrast sensitivity a measurement of the ability to discern between luminances of different levels in a static image.
  • An evaluation of contrast sensitivity may be used to assess whether administration of a necrosis inhibitor and/or an apoptosis inhibitor to the retina of the affected eye preserves or permits improvement in the resolving power of the eye.
  • Visual function may also be assessed by microperimetry, which monitors retinal visual function against retinal thickness or structure and the condition of the subject's fixation over time. Microperimetry may be used to assess whether administration of a necrosis inhibitor and/or an apoptosis inhibitor to the retina of the affected eye preserves or permits improvement in retinal sensitivity and fixation.
  • the necrosis inhibitor e.g., a necrostatin
  • the apoptosis inhibitor e.g., a pan-caspase inhibitor such as Z-VAD or IDN-6556
  • OCT optical coherence
  • the necrosis inhibitor e.g., a necrostatin
  • the apoptosis inhibitor e.g., a pan-caspase inhibitor such as Z-VAD or IDN-6556
  • Both agents may be administered on the day of diagnosis and/or the same day that the retina has gone through other treatments and/or in the post-operative period.
  • the agents may then be administered every three days, every five days, or every seven days until the mammal, e.g., a human, has improved vision (e.g., visual acuity has improved to 20/40 vision or to 20/20 vision), the thickness of the NFL or macula has increased (e.g., macula thickness is 15% thicker than, 35% thicker than, 50% thicker than, 60% thicker than, 70% thicker than, or 80% thicker than without treatment as measured by OCT); the appearance of the ganglion cell layer or bipolar cell layer or photoreceptor cell layer and RPE as detected by OCT; the visual field has improved by at least 10% in the mean standard deviation as determined by Humphrey Visual Field testing; and/or the mammal's retina shows an increased response to light stimulation (e.g., at least a 15% increase in amplitude as determined by electroretinography).
  • vision e.g., visual acuity has improved to 20/40 vision or to 20/20 vision
  • the necrosis inhibitor and the apoptosis inhibitor can be administered by the same route or by different routes.
  • the necrosis inhibitor and/or the apoptosis inhibitor may be administered locally to the eye, for example, by intravitreal, intraocular, intraorbital, subconjuctival, subretinal or transscleral routes.
  • the necrosis inhibitor and/or the apoptosis inhibitor may be administered locally to the eye by intravitreal injection. It is contemplated that local modes of administration may reduce or eliminate the incidence of potential side effects (e.g., systemic toxicity) that may occur during systemic administration.
  • necrosis inhibitor and/or the apoptosis inhibitor may be administered systemically, e.g., by oral or parenteral routes.
  • Parenteral routes include, for example, intravenous, intrarterial, intramuscular, intradermal, subcutaneous, intranasal and intraperitoneal routes.
  • the necrosis inhibitor and the apoptosis inhibitor may be administered to a subject simultaneously or sequentially. It will be appreciated that when administered simultaneously, the necrosis inhibitor and the apoptosis inhibitor may be in the same pharmaceutically acceptable carrier (e.g., solubilized in the same viscoelastic carrier that is introduced into the eye) or the two drugs may be dissolved or dispersed in separate pharmaceutical carriers, which are administered at the same time. Alternatively, the drugs may be provided in separate dosage forms and administered sequentially.
  • the necrostatin may be administered before the pan-caspase inhibitor. In other examples, the pan-caspase inhibitor may be administered before the necrostatin.
  • a single active agent may inhibit both necrosis and apoptosis.
  • Administration may be provided as a periodic bolus (for example, intravitreally or intravenously) or as continuous infusion from an internal reservoir (for example, from an implant disposed at an intra- or extra-ocular location (see, U.S. Pat. Nos. 5,443,505 and 5,766,242)) or from an external reservoir (for example, from an intravenous bag, or a contact lens slow release formulation system).
  • an internal reservoir for example, from an implant disposed at an intra- or extra-ocular location (see, U.S. Pat. Nos. 5,443,505 and 5,766,242)
  • an external reservoir for example, from an intravenous bag, or a contact lens slow release formulation system.
  • the necrosis inhibitor and/or the apoptosis inhibitor may be administered locally, for example, by continuous release from a sustained release drug delivery device immobilized to an inner wall of the eye or via targeted transscleral controlled release into the choroid (see, for example, PCT/US00/00207, PCT/US02/14279, Ambati et al., (2000) I NVEST . O PHTHALMOL . V IS . S CI. 41:1181-1185, and Ambati et al., (2000) I NVEST . O PHTHALMOL . V IS . S CI. 41:1186-1191).
  • a variety of devices suitable for administering the disclosed necrosis and/or apoptosis inhibitors locally to the inside of the eye are known in the art. See, for example, U.S. Pat. Nos. 6,251,090, 6,299,895, 6,416,777, 6,413,540, and 6,375,972, and PCT/US00/28187.
  • the necrosis inhibitor and/or the apoptosis inhibitor may be solubilized in a carrier, for example, a viscoelastic carrier, that is introduced locally into the eye.
  • a carrier for example, a viscoelastic carrier
  • One or both inhibitors also may be administered in a pharmaceutically acceptable carrier or vehicle so that administration does not otherwise adversely affect the recipient's electrolyte and/or volume balance.
  • the carrier may comprise, for example, physiologic saline or other buffer system.
  • the necrostatin, the pan-caspase inhibitor, or both the necrostatin and the pan-caspase inhibitor may be solubilized in PBS or another aqueous buffer by sonication.
  • one or both drugs may be solubilized using conventional solvent or solubilization systems, for example, dimethyl sulfoxide (DMSO), dimethoxyethane (DME), dimethylformamide (DMF), cyclodextran, micelles, liposomes, liposomal agents, and other solvents known in the art to aid in the solubilization and administration of hydrophobic agents.
  • DMSO dimethyl sulfoxide
  • DME dimethoxyethane
  • DMF dimethylformamide
  • cyclodextran cyclodextran
  • micelles micelles
  • liposomes liposomal agents
  • liposomal agents and other solvents known in the art to aid in the solubilization and administration of hydrophobic agents.
  • the necrosis inhibitor and/or the apoptosis inhibitor may be solubilized in a liposome or microsphere.
  • Methods for delivery of a drug or combination of drugs in liposomes and/or microspheres are well-known in the art.
  • necrosis inhibitor and/or the apoptosis inhibitor may be formulated so as to permit release of one or both inhibitors over a prolonged period of time.
  • a release system can include a matrix of a biodegradable material or a material, which releases the incorporated active agents.
  • the active agents can be homogeneously or heterogeneously distributed within a release system.
  • release systems may be useful in the practice of the invention, however, the choice of the appropriate system will depend upon the rate of release required by a particular drug regime. Both non-degradable and degradable release systems can be used.
  • Suitable release systems include polymers and polymeric matrices, non-polymeric matrices, or inorganic and organic excipients and diluents such as, but not limited to, calcium carbonate and sugar (for example, trehalose). Release systems may be natural or synthetic. However, under certain circumstances, synthetic release systems are preferred because generally they are more reliable, more reproducible and produce more defined release profiles.
  • the release system material can be selected so that inhibitors having different molecular weights are released by diffusion through or degradation of the material.
  • Representative synthetic, biodegradable polymers include, for example: polyamides such as poly(amino acids) and poly(peptides); polyesters such as poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), and poly(caprolactone); poly(anhydrides); polyorthoesters; polycarbonates; and chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), copolymers and mixtures thereof.
  • polyamides such as poly(amino acids) and poly(peptides)
  • polyesters such as poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), and poly(caprolactone)
  • poly(anhydrides) polyorthoesters
  • polycarbonates and chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylation
  • Representative synthetic, non-degradable polymers include, for example: polyethers such as poly(ethylene oxide), poly(ethylene glycol), and poly(tetramethylene oxide); vinyl polymers-polyacrylates and polymethacrylates such as methyl, ethyl, other alkyl, hydroxyethyl methacrylate, acrylic and methacrylic acids, and others such as poly(vinyl alcohol), poly(vinyl pyrolidone), and poly(vinyl acetate); poly(urethanes); cellulose and its derivatives such as alkyl, hydroxyalkyl, ethers, esters, nitrocellulose, and various cellulose acetates; polysiloxanes; and any chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), copolymers and mixtures thereof.
  • polyethers such as poly(ethylene oxide), poly(ethylene glycol), and poly(
  • the microspheres are composed of a polymer of lactic acid and glycolic acid, which are structured to form hollow spheres. These spheres can be approximately 15-30 ⁇ m in diameter and can be loaded with a variety of compounds varying in size from simple molecules to high molecular weight proteins such as antibodies. The biocompatibility of these microspheres is well established (see, Sintzel et al., (1996)), and microspheres have been used to deliver a wide variety of pharmacological agents in numerous biological systems.
  • poly(lactide-co-glycolide) microspheres are hydrolyzed by the surrounding tissues, which cause the release of the contents of the microspheres (Zhu et al., (2000)).
  • the in vivo half-life of a microsphere can be adjusted depending on the specific needs of the system.
  • compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present invention also consist essentially of, or consist of, the recited components, and that the processes of the present invention also consist essentially of, or consist of, the recited processing steps. Further, it should be understood that the order of steps or order for performing certain actions are immaterial so long as the invention remains operable. Moreover, two or more steps or actions may be conducted simultaneously.
  • RIP-3 ⁇ / ⁇ mice were generated as described previously and backcrossed to C57BL/6 mice (Newton et al., (2004) M OL C ELL B IOL 24:1464-1469). Except as noted otherwise, the animals were anesthetized with ketamine hydrochloride (30 mg/kg; Ketalar, Parke-Davis, Morris Plains, N.J.) and xylazine hydrochloride (5 mg/kg; Rompun, Harver-Lockhart, Morris Plains, N.J.) before all experimental manipulations.
  • NMDA NMDA (2 ⁇ l of a 0.1M stock; Sigma-Aldrich) was injected intravitreously in combination with ZVAD, Nec-1, or ZVAD plus Nec-1 and mice were separated into the aforementioned 4 groups.
  • mice received an intravitreal injection of 3-methyladenine (3-MA), in DMSO (33.3 mM; Sigma-Aldrich, St. Louis, Mo.), a goat anti-mouse TNF- ⁇ blocking antibody, or the appropriate control goat antibody (R & D Systems, Minneapolis, Minn.).
  • 3-methyladenine 3-methyladenine
  • DMSO 33.3 mM
  • Sigma-Aldrich St. Louis, Mo.
  • a goat anti-mouse TNF- ⁇ blocking antibody or the appropriate control goat antibody (R & D Systems, Minneapolis, Minn.).
  • Intravitreal injections were performed as follows. Briefly, the tip of a 33 gauge needle (Hamilton, Reno, Nev.) was carefully inserted through the sclera into the intravitreal space to reduce intraocular pressure. Then, the needle was extracted, loaded with compounds and tangentially reinserted through the sclera into the intravitreal space, inducing a self-sealing wound tunnel. After injection, the absence of choroidal bleeding was confirmed. At specified times after injury, mice were sacrificed with an overdose of sodium pentobarbital, and eyes were enucleated.
  • RNA extraction, cDNA synthesis and PCR amplification have been performed as previously reported (Kayama et al., (2010) O PHTHALMIC R ES 43:79-91).
  • a real-time PCR assay was performed with Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, Calif.). The primers are shown below in Table 1.
  • TUNEL and quantification of TUNEL (+) cells were performed as previously described (Nakazawa et al., (2007) P ROC N ATL A CAD S CI USA 104:2425-2430) by using the ApopTag Fluorescein In Situ Apoptosis Detection Kit (S7110; Chemicon International, Temecula, Calif.).
  • Glaucoma is a group of ocular disorders, characterized by optic nerve injury. In most cases of glaucoma, the optic nerve injury is caused by elevated intraocular pressure. Furthermore, higher intraocular pressures are generally associated with greater nerve damage.
  • mouse models of optic nerve (ON) injury was used to assess the role of RIP-mediated programmed necrosis and apoptosis in this ocular disorder. Given that ON injury is a hallmark of glaucoma, this model also provided an assessment of RIP-mediated programmed necrosis in RGC loss in glaucoma.
  • TNF- ⁇ c retinal ganglion cell
  • TNF- ⁇ c is involved in RGC cell death in mice subjected to optic nerve (ON) injury (induced by physical injury to the optic nerve).
  • ON optic nerve
  • TNF- ⁇ level in the retina was measured using quantitative real-time RT-PCR.
  • FIG. 2A As depicted in FIG. 2A , at one day after ON injury, TNF- ⁇ mRNA levels increased almost 10-to 13-fold relative to non-injured mice.
  • IPL inner plexiform layer
  • mice treated with the control antibody showed significant reduction both in RGCs and IPL thickness ( FIGS. 2B-2D ), whereas, mice treated with TNF- ⁇ neutralizing antibody showed negligible loss of RGCs ( FIGS. 2B and 2C ) and minimal change in IPL thickness ( FIGS. 2B and 2D ). These data indicate that TNF- ⁇ plays a critical role in RGC death after ON injury.
  • TNF- ⁇ c has been shown to be a potent inducer of programmed necrosis as well as apoptosis (Degterev et al., (2005) N AT C HEM B IOS 1:112-119, Balkwill, (2009)).
  • the kinases RIP3 and RIP1 are key signaling molecules in cellular apoptotic and necrotic pathways and are regulated by TNF- ⁇ (He et al., (2009) C ELL 137:1100-1111, Vandenabeele et al., (2010) S CI S IGNAL 3:re4)).
  • TNF- ⁇ He et al., (2009) C ELL 137:1100-1111, Vandenabeele et al., (2010) S CI S IGNAL 3:re4
  • RIP3 and RIP1 increased significantly up to 9- and 5-fold, respectively, compared to non-injured mice ( FIGS. 2E and 2F ).
  • RIP3 and RIP1 protein levels were also assessed using Western Blot analysis. After ON injury, expressions of RIP3 and RIP1 were found to be approximately 3-fold up-regulated compared with non-injured retina ( FIGS. 2G-21 ). These results suggest that RIP kinases may contribute to ON injury-induced RGC death.
  • Nec-1 which is a potent and selective inhibitor of programmed necrosis targeting RIP1 kinase activity (Degterev et al., (2008)), was employed to investigate the effect of RIP kinase inhibition in RGC death after ON injury.
  • Mice received an intravitreal injection of Z-VAD (300 uM) and/or Nec-1 (400 uM). The dose of these compounds was selected based on studies that established that their half-life inside the eye is around 6 hours.
  • Administration of Nec-1 and/or Z-VAD did not affect increase of TNF- ⁇ levels after ON injury ( FIG. 3L ).
  • FIGS. 3A and 3B By day one after ON injury, there was a dramatic decrease in apoptotic RGCs in mice that received a combination of Z-VAD and Nec-1 ( FIGS. 3A and 3B ). Injection of Nec-1 alone, did not affect the number of TUNEL positive cells ( FIGS. 3A and 3B ), which declined significantly at day three and day seven after ON injury ( FIGS. 3M and 3N ).
  • IPL Inner plexiform layer
  • GCL ganglion cell layer
  • IPL thickness location of RGC cell bodies and axons
  • SD-OCT Spectral Domain Optical Coherence Tomography
  • Mice were positioned on a custom cassette, which allowed three-dimensional free rotation and alignment of the mouse eye for imaging. Hydration with normal saline was used to preserve corneal clarity. A volume analysis centered on the ON head was performed, using 100 horizontal, raster, and consecutive B-scan lines, each one composed by 1200 A-scans.
  • the thickness of GCL+IPL was assessed at 500 ⁇ m, 400 ⁇ m, 300 ⁇ m distance from the ON head (nasally and temporally) as well as at 200 ⁇ m, 400 ⁇ m above and below the ON head.
  • Co-administration of Z-VAD and Nec-1 remarkably reversed the decrease in IPL thickness ( FIG. 3Q ) and GCL with IPL thickness that was seen in the vehicle group ( FIGS. 3E-3G ).
  • these data suggest that inhibition of RIP kinases together with a broad-spectrum caspase inhibitor are required for effective neuroprotection after ON injury.
  • TEM transmission electron microscopy
  • RGC death was categorized into apoptosis, necrosis and unclassified end-stage of death, as previously described (Trichonas et al., (2010) P ROC N ATL A CAD S CI USA). Consistent with the PI study, at one day after ON injury, both apoptotic and necrotic RGC death was observed in the vehicle-treated retina (apoptotic cells: 13.4 ⁇ 5.8%, necrotic cells: 16.9 ⁇ 4.2%, unclassified: 2.2 ⁇ 2.4%; FIG. 3 , a and b ). Nec-1 treatment slightly decreased necrotic RGC death (% apoptotic cells: 13.0 ⁇ 8.4%, necrotic cells: 10.6 ⁇ 2.6%, unclassified: 1.0 ⁇ 1.8%; FIGS.
  • RIP3 deficient mice were used (He et al., (2009) C ELL 137:1100-1111, Zhang et al., (2009) S CIENCE 325:332-336)), because RIP1 ⁇ / ⁇ mice die postnatally at day 1-3 (Kelliher et al., (1998) I MMUNITY 8:297-303)).
  • RIP3 ⁇ / ⁇ mice exhibited significantly less TUNEL positive cells 1 day compared to wildtype controls ( FIGS. 5A and 5B ).
  • the number of viable RGCs were assessed by Brn3b immunohistochemistry and measurement of IPL thickness.
  • RIP3 ⁇ / ⁇ mice demonstrated increased number of Brn3b positive cells compared to wildtype mice, (vehicle, white box) which was further enhanced with Z-VAD administration ( FIGS. 5C and 5D ).
  • RIP3 deficiency combined with Z-VAD administration also resulted in preservation of IPL thickness compared to RIP3 ⁇ / ⁇ mice that received only vehicle solution ( FIGS. 5C and 5E ).
  • the number of Brn3b positive cells and IPL thickness did not alter after Nec-1 coadministration ( FIGS. 5C-5E ).
  • PI staining shows the number of cells with disrupted plasma membrane.
  • RIP3 deficient mice exhibited significantly less PI positive cells compared with Z-VAD treated WT mice ( FIGS. 5F and 5G ). This result suggests that RIP3 kinase plays an essential role in ON injury-induced programmed necrosis, especially in the presence of caspase inhibitor.
  • TNF- ⁇ mRNA levels increased at 1 day after NMDA injury over 10-fold compared with non-injured mice ( FIG. 6A ).
  • Quantitative real-time PCR and Western blot analyses revealed that expressions of RIP3 and RIP1 after NMDA injury were up-regulated compared with those in non-injured retina ( FIGS. 6B-6D ).
  • Treatment with ZVAD or Nec-1 alone decreased the number of TUNEL positive cells and prevented the reduction of Brn3b positive cells, and these protective effects were further enhanced by co-administration of ZVAD plus Nec-1 ( FIGS. 6E-6H ).
  • Rip3 deficiency also decreased RGC loss after NMDA injury ( FIGS. 6E-6H ).
  • Apoptosis inducing factor is a mitochondrial flavoprotein which is involved in initiating caspase-independent apoptosis. After loss of mitochondrial membrane potential, AIF translocates into the nucleus, induces DNA fragmentation and peripheral chromatin condensation (Susin et al., (1999), Susin et al., (2000), Candé et al., (2004)). To analyze whether mitochondrial release of AIF is associated with RGC death after ON injury, immunostaining for AIF was performed together with TUNEL assay. One day after ON injury, there was a prominent nuclear translocation of AIF, which was not affected by pan-caspase inhibition ( FIGS. 7A and 7B ).
  • Apoptosis is characterized by activation of caspases, DNA fragmentation, and membrane blebbing (Kroemer et al., (2009) C ELL D EATH D IFFER 16:3-11, Y 1 et al., (2009)), whereas necrosis is characterized by swelling of the endoplasmic reticulum, mitochondria, and cytoplasm, with subsequent rupture of the plasma membrane and lysis of the cells (Kroemer et al., (2009) C ELL D EATH D IFFER 16:3-11, Festjens et al., (2006)).
  • RGCs that were unable to be classified were defined as end-stage cell death/unclassified. Under normal conditions, RGCs had a well-defined continuous plasma membrane, and a non-uniform distribution of organelles in the cytoplasm, with maximum concentration in the perinuclear region. RGCs contained tubular sacs of rough endoplasmic reticulum (rER) surrounded by large numbers of ribosomes (Nissl bodies). Mitochondria were identified as round or oval double-membrane structures with characteristic cristae.
  • rER rough endoplasmic reticulum
  • Nissl bodies ribosomes
  • the cytoplasm contained elements of Golgi apparatus (GA), free ribosomes and microtubules sectioned at various angles.
  • FIG. 8A homogeneously dispersed karyoplasm
  • FIG. 8A homogeneously dispersed karyoplasm
  • FIG. 8A homogeneously dispersed karyoplasm
  • FIG. 8A homogeneously dispersed karyoplasm
  • FIG. 8A homogeneously dispersed karyoplasm
  • Autophagic cell death is recognized by the formation of autophagosomes, double-membrane autophagic vacuoles that eventually fuse with lysosomes to form autolysosomes (Levine et al., (2004)).
  • the Atg6-Vps34 complex was suggested to be critical for autophagosome-vesicle nucleation (Levine et al., (2005)).
  • Elongation of the autophagosomal membrane formed by the Atg6-Vps34 complex is assisted by two Atg12 and Atg8 ubiquitin-like conjugation systems (Ichimura et al., (2000)).
  • Atg12 activated by Atg7, covalently attaches to Atg5, forming the irreversible conjugate.
  • FIGS. 9A-9C To investigate whether autophagy influences RGC death after ON injury, expression of several genes critical for autophagosome formation including Atg5, 7 and 12 was measured by quantitative RT-PCR. These transcripts were found to be up-regulated 2.0- to 2.5-fold at one day after ON injury ( FIGS. 9A-9C ). ZVAD administration further increased Atg12 expression compared with vehicle treatment ( FIG. 9C ). In contrast, administration of ZVAD plus Nec-1, or Rip3 deficiency suppressed expression of Atg5, 7 and 12 transcripts ( FIGS. 9A-9C ).
  • LC3-II an isoform associated with autophagy activity (Kabeya et al, 2000)
  • protein level of LC3-II increased more than vehicle following Z-VAD administration ( FIG. 9D ).
  • the optic nerve cannot regenerate if injured, leaving victims of traumatic nerve injury or degenerative diseases such as glaucoma with life-long visual losses.
  • This situation can be, at least, partially reversed by enhancing the intrinsic growth state of retinal ganglion cells (RGCs).
  • RGCs retinal ganglion cells
  • the efficacy of necrosis inhibitor and a pan-caspase inhibitor in promoting RGC survival and axon regeneration is investigated using a mouse optic nerve crush model.
  • a Necrosis Inhibitor in Combination with a Caspase Inhibitor Promotes RGC Survival in a Optic Nerve Crush Model
  • mice were subjected to optic nerve crush surgery. Specifically, animals were anesthetized with an intraperitoneal injection of ketamine (60-80 mg/kg: Phoenix Pharmaceutical, St. Joseph, Mo.) and xylazine (10-15 mg/kg: Bayer, Shawnee Mission, Kans.). Animals were positioned in a stereotaxic apparatus and a 1-1.5 cm incision was made in the skin above the right orbit. Under microscopic illumination, the lachrymal glands and extraocular muscles were resected to expose 3-4 mm of the optic nerve.
  • ketamine 60-80 mg/kg: Phoenix Pharmaceutical, St. Joseph, Mo.
  • xylazine 10-15 mg/kg: Bayer, Shawnee Mission, Kans.
  • the epineurium was slit open along the long axis, and the nerve was crushed 2 mm behind the eye with angled jeweler's forceps (Dumont #5) for 10 seconds, avoiding injury to the ophthalmic artery. Nerve injury was verified by the appearance of a clearing at the crush site, while the vascular integrity of the retina was evaluated by fundoscopic examination. Cases in which the vascular integrity of the retina was in question were excluded from the study.
  • mice were divided into four groups for treatment: vehicle group, ZVAD group (300 ⁇ M; given at day 0, day 3 and day 7 after injury), Nec-1 group (4 mM; given at day 0, day 3 and day 7 after injury), and ZVAD plus Nec-1 group (300 ⁇ M and 4 mM, respectively; given either once or at day 0, day 3 and day 7 after injury).
  • ZVAD group 300 ⁇ M; given at day 0, day 3 and day 7 after injury
  • Nec-1 group 4 mM; given at day 0, day 3 and day 7 after injury
  • ZVAD plus Nec-1 group 300 ⁇ M and 4 mM, respectively; given either once or at day 0, day 3 and day 7 after injury.
  • Zymosan 12.5 ⁇ g/ ⁇ l
  • yeast cell wall preparation known to stimulate axonal regeneration.
  • a combination of ZVAD and Nec-1 significantly prevented RGC death and promoted RGC survival following optic nerve crush injury when compared to treatment with Zymosan alone (p ⁇ 0.05).
  • the effect of the ZVAD and Nec-1 combination treatment on RGC survival was even more pronounced when the treatment was given at day 0, day 3 and day 7 after injury when compared to a single treatment at day 0 (p ⁇ 0.05).
  • a Necrosis Inhibitor in Combination with a Caspase Inhibitor Promotes Axon Regeneration
  • mice were subjected to optic nerve crush surgery as previously described. Subsequently, injured mice were divided into five groups of treatment: vehicle group, ZVAD group (300 ⁇ M; given at day 0, day 3 and day 7 after injury), Nec-1 group (4 mM; given at day 0, day 3 and day 7 after injury), ZVAD plus Nec-1 group (300 ⁇ M and 4 mM, respectively; given once at day 0), and ZVAD plus Nec-1 group (300 ⁇ M and 4 mM, respectively; given at day 0, day 3 and day 7 after injury).
  • ZVAD group 300 ⁇ M; given at day 0, day 3 and day 7 after injury
  • Nec-1 group 4 mM; given at day 0, day 3 and day 7 after injury
  • ZVAD plus Nec-1 group 300 ⁇ M and 4 mM, respectively; given once at day 0
  • ZVAD plus Nec-1 group 300 ⁇ M and 4 mM, respectively; given at day 0, day 3 and day 7 after injury.
  • Axon regeneration was assessed by obtaining longitudinal sections of the optic nerve and counting the number of axons at pre-specified distances from the injury site. Specifically, mice were sacrificed at 14 days after optic nerve injury and were perfused with saline and 4% paraformaldehyde (PFA). Optic nerves and eyes were dissected and postfixed in PFA. Nerves were impregnated with 10% and then 30% sucrose, embedded in OCT Tissue Tek Medium (Sakura Finetek), frozen, cut in the longitudinal plane at 14 ⁇ m, and mounted on coated slides. Regenerating axons were visualized by staining with a sheep antibody to 13111-tubulin, followed by staining with a fluorescently labeled secondary antibody.
  • PFA paraformaldehyde
  • Axons were counted manually in at least eight longitudinal sections per case at pre-specified distances from the injury site. The number of regenerating axons at various distances are determined as described previously (Leon et al., (2000) J N EUROSCI 20:4615-4626). To determine the number of surviving cells, staining with an anti-Brn3a antibody was used.
  • FIGS. 12A-12E show longitudinal sections of the optic nerve following optic nerve crush injury.
  • the sections are stained with an antibody against ⁇ III-tubulin, which marks axon fibers.
  • an arrow indicates the sites of optic nerve injury, and staining beyond the injury site starting from left to right indicates axon regeneration (e.g., axons regenerate from the site of injury into the nerve).
  • axon regeneration e.g., axons regenerate from the site of injury into the nerve.
  • No significant axon regeneration was seen in mice treated with vehicle control, as demonstrated by the lack of axon staining ( FIG. 12A ).
  • Treatment with Nec-1 or ZVAD alone had minimal effects on axon regeneration ( FIGS. 12B and 12C ).
  • ZVAD plus Nec-1 combination treatment significantly enhanced axon outgrowth as demonstrated by the increase in axon staining ( FIGS. 12D and 12E ; see the regions denoted by the horizontal reference lines under each figure).
  • FIGS. 12D and 12E the effect of the ZVAD and Nec-1 combination treatment on axon regeneration was more pronounced when the treatment was given at day 0, day 3 and day 7 after injury when compared to a single treatment at day 0.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Ophthalmology & Optometry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US13/882,932 2010-11-01 2011-10-21 Methods and compositions for preserving retinal ganglion cells Abandoned US20140024598A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/882,932 US20140024598A1 (en) 2010-11-01 2011-10-21 Methods and compositions for preserving retinal ganglion cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US40905510P 2010-11-01 2010-11-01
US41486210P 2010-11-17 2010-11-17
US201161472144P 2011-04-05 2011-04-05
US13/882,932 US20140024598A1 (en) 2010-11-01 2011-10-21 Methods and compositions for preserving retinal ganglion cells
PCT/US2011/057327 WO2012061045A2 (fr) 2010-11-01 2011-10-21 Méthodes et compositions permettant de préserver les cellules ganglionnaires de la rétine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/057327 A-371-Of-International WO2012061045A2 (fr) 2010-11-01 2011-10-21 Méthodes et compositions permettant de préserver les cellules ganglionnaires de la rétine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/093,480 Continuation US9993517B2 (en) 2010-11-01 2016-04-07 Methods and compositions for preserving retinal ganglion cells

Publications (1)

Publication Number Publication Date
US20140024598A1 true US20140024598A1 (en) 2014-01-23

Family

ID=46025008

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/882,932 Abandoned US20140024598A1 (en) 2010-11-01 2011-10-21 Methods and compositions for preserving retinal ganglion cells
US15/093,480 Active US9993517B2 (en) 2010-11-01 2016-04-07 Methods and compositions for preserving retinal ganglion cells
US15/956,133 Active US10617735B2 (en) 2010-11-01 2018-04-18 Methods and compositions for preserving retinal ganglion cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/093,480 Active US9993517B2 (en) 2010-11-01 2016-04-07 Methods and compositions for preserving retinal ganglion cells
US15/956,133 Active US10617735B2 (en) 2010-11-01 2018-04-18 Methods and compositions for preserving retinal ganglion cells

Country Status (2)

Country Link
US (3) US20140024598A1 (fr)
WO (1) WO2012061045A2 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492432B2 (en) 2010-04-23 2016-11-15 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving photoreceptor and retinal pigment epithelial cells
US9556152B2 (en) 2013-02-15 2017-01-31 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US9993517B2 (en) 2010-11-01 2018-06-12 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving retinal ganglion cells
US10022419B2 (en) 2011-10-21 2018-07-17 Massachusetts Eye And Ear Infirmary Methods for treating spinal cord injury
CN110177577A (zh) * 2016-07-05 2019-08-27 马萨诸塞大学 Sfasl的aav2介导的基因递送作为青光眼的神经保护疗法
US20220017944A1 (en) * 2011-09-26 2022-01-20 Qiagen Gmbh Stabilisation and isolation of extracellular nucleic acids
US11786694B2 (en) 2019-05-24 2023-10-17 NeuroLight, Inc. Device, method, and app for facilitating sleep

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9725452B2 (en) 2013-03-15 2017-08-08 Presidents And Fellows Of Harvard College Substituted indoles and pyrroles as RIP kinase inhibitors
US10143703B2 (en) 2014-01-02 2018-12-04 Massachusetts Eye And Ear Infirmary Treating ocular neovascularization
EP3229807A4 (fr) 2014-12-11 2018-10-17 President and Fellows of Harvard College Inhibiteurs de nécrose cellulaire et procédés associés
CN107779437B (zh) * 2016-08-26 2022-01-21 中国科学院脑科学与智能技术卓越创新中心 自噬诱导剂作为微管稳定药物促进神经再生的用途
US20210330735A1 (en) * 2018-07-16 2021-10-28 Stealth Biotherapeutics Corp Compositions and methods for the treatment of traumatic optic neuropathy
JP2022504764A (ja) * 2018-10-11 2022-01-13 ザ ユナイテッド ステイツ オブ アメリカ アズ リプリゼンテッド バイ ザ セクレタリー、デパートメント オブ ヘルス アンド ヒューマン サービシーズ 細胞培養のための組成物および方法
CN113134076A (zh) * 2020-01-16 2021-07-20 上海科技大学 一种利用转录因子再生具备功能的视网膜神经节细胞的方法

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5443505A (en) 1993-11-15 1995-08-22 Oculex Pharmaceuticals, Inc. Biocompatible ocular implants
US5725493A (en) 1994-12-12 1998-03-10 Avery; Robert Logan Intravitreal medicine delivery
US6299895B1 (en) 1997-03-24 2001-10-09 Neurotech S.A. Device and method for treating ophthalmic diseases
ATE414149T1 (de) 1996-11-20 2008-11-15 Univ Yale Survivin, ein protein das zelluläre apoptosis hemmt, und dessen modulation
EP1154691A4 (fr) 1999-01-05 2004-07-07 Massachusetts Eye & Ear Infirm Administration ciblee de medicament a liberation regulee vers la retine et la choroide a travers la sclerotique
WO2001028493A2 (fr) 1999-10-15 2001-04-26 President And Fellows Of Harvard College Inhibiteurs a petite molecule de la necrose
US6416777B1 (en) 1999-10-21 2002-07-09 Alcon Universal Ltd. Ophthalmic drug delivery device
DE60016271T2 (de) 1999-10-21 2005-12-01 Alcon Inc. Medikamentenzuführvorrichtung
WO2001039792A2 (fr) * 1999-12-03 2001-06-07 Alcon Universal Ltd. Utilisation d'inhibiteurs de caspase 9 pour traiter une pathologie oculaire nerveuse
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
WO2002063959A1 (fr) 2001-02-09 2002-08-22 The Schepens Eye Research Institute Procedes et compositions pour stimuler la regeneration axonale et prevenir la degenerescence des cellules neuronales
EP1387671A1 (fr) 2001-05-03 2004-02-11 MASSACHUSETTS EYE & EAR INFIRMARY Dispositif d'administration de medicament implantable et utilisation associee
AU2003209297A1 (en) 2002-01-18 2003-09-02 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving the viability of photoreceptor cells
US20050130877A1 (en) 2003-03-14 2005-06-16 Children's Medical Center Corporation Axon regeneration with PKC inhibitors
EP3081214A3 (fr) 2003-08-29 2016-11-16 The Brigham And Women's Hospital, Inc. Inhibiteurs de nécrose cellulaire
US8912144B2 (en) 2003-12-16 2014-12-16 Children's Medical Center Corporation Method for treating stroke via administration of NEP1-40 and inosine
US20070049565A1 (en) 2005-08-24 2007-03-01 Neurotech Pharmaceuticals Co., Ltd. Combination of cell necrosis inhibitor and lithium for treating neuronal death or neurological dysfunction
US20070298129A1 (en) * 2005-08-24 2007-12-27 Neurotech Pharmaceuticals Co., Ltd. Compounds and compositions for treating neuronal death or neurological dysfunction
EP1968583A4 (fr) 2005-12-20 2010-09-15 Harvard College Composes, essais et methodes de traitement
SG163525A1 (en) 2005-12-23 2010-08-30 Partnership & Corp Technology Synthetic peptides for use as inhibitors of neurotransmitter secretion and as inducers of muscle relaxation
CA2666060C (fr) 2006-10-10 2015-02-03 President And Fellows Of Harvard College Composes de thieno-[2,3-d]-pyrimidin-4-one et procedes d'utilisation de ceux-ci
EP2192838A4 (fr) 2007-08-15 2011-07-27 Harvard College Inhibiteurs hétérocycliques de la nécroptose
CN102186350A (zh) 2008-08-19 2011-09-14 诺普神经科学股份有限公司 使用(r)-普拉克索的组合物与方法
WO2010075290A1 (fr) 2008-12-22 2010-07-01 President And Fellows Of Harvard College Inhibiteurs hétérocycliques insaturés de la nécroptose
US7622106B1 (en) 2009-03-06 2009-11-24 Board Of Regents, The University Of Texas System Necrosis assay
JP5723294B2 (ja) 2009-12-09 2015-05-27 北海道公立大学法人 札幌医科大学 スーパーオキシド製造方法、スーパーオキシド消去能評価方法、スーパーオキシド製造装置およびスーパーオキシド消去能評価装置
US20130137642A1 (en) 2010-04-23 2013-05-30 Demetrios Vavvas Methods and compositions for preserving photoreceptor and retinal pigment epithelial cells
WO2012061045A2 (fr) 2010-11-01 2012-05-10 Massachusetts Eye And Ear Infirmary Méthodes et compositions permettant de préserver les cellules ganglionnaires de la rétine
EP2773341A2 (fr) 2011-10-21 2014-09-10 Massachusetts Eye & Ear Infirmary Compositions comprenant des inhibiteurs de la necrose, tels que des necrostatines, seuls ou en combinaison, pour la promotion de la régénération axonale et de la fonction nerveuse, ainsi pour traiter les maladies du snc

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492432B2 (en) 2010-04-23 2016-11-15 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving photoreceptor and retinal pigment epithelial cells
US10639345B2 (en) 2010-04-23 2020-05-05 Massachusetts Eye And Ear Infirmary Compositions for preserving photoreceptor cells, retinal pigment epithelial cells, or visual function
US10149884B2 (en) 2010-04-23 2018-12-11 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving photoreceptor and retinal pigment epithelial cells
US9993517B2 (en) 2010-11-01 2018-06-12 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving retinal ganglion cells
US10617735B2 (en) 2010-11-01 2020-04-14 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving retinal ganglion cells
US20220017944A1 (en) * 2011-09-26 2022-01-20 Qiagen Gmbh Stabilisation and isolation of extracellular nucleic acids
US10022419B2 (en) 2011-10-21 2018-07-17 Massachusetts Eye And Ear Infirmary Methods for treating spinal cord injury
US10799552B2 (en) 2011-10-21 2020-10-13 Massachusetts Eye And Ear Infirmary Methods for treating diabetic neurotherapy
US9624202B2 (en) 2013-02-15 2017-04-18 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10292987B2 (en) 2013-02-15 2019-05-21 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US9556152B2 (en) 2013-02-15 2017-01-31 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10933070B2 (en) 2013-02-15 2021-03-02 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10940154B2 (en) 2013-02-15 2021-03-09 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
CN110177577A (zh) * 2016-07-05 2019-08-27 马萨诸塞大学 Sfasl的aav2介导的基因递送作为青光眼的神经保护疗法
US11786694B2 (en) 2019-05-24 2023-10-17 NeuroLight, Inc. Device, method, and app for facilitating sleep

Also Published As

Publication number Publication date
WO2012061045A2 (fr) 2012-05-10
US10617735B2 (en) 2020-04-14
US20160367619A1 (en) 2016-12-22
US20190099467A1 (en) 2019-04-04
US9993517B2 (en) 2018-06-12
WO2012061045A3 (fr) 2013-07-11

Similar Documents

Publication Publication Date Title
US10617735B2 (en) Methods and compositions for preserving retinal ganglion cells
US10639345B2 (en) Compositions for preserving photoreceptor cells, retinal pigment epithelial cells, or visual function
US10799552B2 (en) Methods for treating diabetic neurotherapy
AU2016276170B2 (en) Compositions and methods for treating pterygium
JP2019518730A (ja) 眼の炎症性障害および疾患の組合せ処置
US20180369172A1 (en) Methods and compositions for promoting wound healing with decreased scar formation after glaucoma filtration surgery

Legal Events

Date Code Title Description
AS Assignment

Owner name: MASSACHUSETTS EYE AND EAR INFIRMARY, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAVVAS, DEMETRIOS G.;MILLER, JOAN W.;KAYAMA, MAKI;SIGNING DATES FROM 20111213 TO 20130205;REEL/FRAME:030510/0923

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:MASSACHUSETTS EYE AND EAR INFRIMARY;REEL/FRAME:052582/0294

Effective date: 20200505