US20130273065A1 - Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues - Google Patents

Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues Download PDF

Info

Publication number
US20130273065A1
US20130273065A1 US13/795,327 US201313795327A US2013273065A1 US 20130273065 A1 US20130273065 A1 US 20130273065A1 US 201313795327 A US201313795327 A US 201313795327A US 2013273065 A1 US2013273065 A1 US 2013273065A1
Authority
US
United States
Prior art keywords
composition
ocular
inflammatory
seq
cytokine
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
US13/795,327
Inventor
Reza Dana
Mohammad Dastjerdi
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.)
Schepens Eye Research Institute Inc
Original Assignee
Schepens Eye Research Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Schepens Eye Research Institute Inc filed Critical Schepens Eye Research Institute Inc
Priority to US13/795,327 priority Critical patent/US20130273065A1/en
Assigned to THE SCHEPENS EYE RESEARCH INSTITUTE INC. reassignment THE SCHEPENS EYE RESEARCH INSTITUTE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DASTJERDI, MOHAMMAD, DANA, REZA
Publication of US20130273065A1 publication Critical patent/US20130273065A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/16Ginkgophyta, e.g. Ginkgoaceae (Ginkgo family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/25Araliaceae (Ginseng family), e.g. ivy, aralia, schefflera or tetrapanax
    • A61K36/258Panax (ginseng)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/45Ericaceae or Vacciniaceae (Heath or Blueberry family), e.g. blueberry, cranberry or bilberry
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/53Lamiaceae or Labiatae (Mint family), e.g. thyme, rosemary or lavender
    • A61K36/537Salvia (sage)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/82Theaceae (Tea family), e.g. camellia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/87Vitaceae or Ampelidaceae (Vine or Grape family), e.g. wine grapes, muscadine or peppervine
    • 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
    • A61K38/063Glutathione
    • 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
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1816Erythropoietin [EPO]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2006IL-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K9/0051Ocular inserts, ocular implants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the composition comprises a polynucleotide, a polypeptide, an antibody, or a small molecule that binds or modifies the function of IL-1a, IL-1b, IL-1R1, IL-1R2, IL-1Ra3, IL-1RAP, or IRAK1.
  • the composition comprises morpholino antisense oligonucleotides, microRNAs (miRNAs), short hairpin RNA (shRNA), or short interfering RNA (siRNA) to silence gene expression.
  • Vectors are constructed by recombinant DNA technology methods standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the short polynucleotide can be placed under the control of any promoter known in the art to act in mammalian, preferably human cells. Promoters are inducible or constitutive.
  • subjects are identified by measuring their intraocular pressure and determining if the measured intraocular pressure is elevated above normal levels.
  • normal level is meant to describe value within an acceptable range of values that one of ordinary skill in the art and/or a medical professional would expect a healthy subject of similar physical characteristics and medical history to have.
  • IOP normal intraocular pressure
  • Intraocular pressure is maintained by the liquid aqueous humor, which is produced by the ciliary body of the eye.
  • Aqueous humor normally does not go into the posterior segment of the eye; it is kept out of this area by the lens and the Zonule of Zinn Instead, it stays only in the anterior segment, which is divided into the anterior and posterior chambers. While the anterior and posterior chambers are very similarly named to the anterior and posterior segments, they are not synonymous. The anterior and posterior chambers are both parts of the anterior segment.
  • Intraocular pressure may become elevated due to anatomical problems, inflammation of the eye, genetic factors, as a side-effect from medication, or during exercise. Intraocular pressure usually increases with age and is genetically influenced.
  • Anakinra/Kineret® is encoded by the following mRNA sequence (NCBI Accession No. M55646 and SEQ ID NO: 15):
  • IL-1RAP transcript variant 2
  • NBI Accession No. NM — 134470 and SEQ ID NO: 27 amino acid sequence
  • the invention comprises a contact lens and a composition that inhibits an activity of an inflammatory interleukin-1 cytokine.
  • the composition is incorporated into or coated onto said lens.
  • the composition is chemically bound or physically entrapped by the contact lens polymer.
  • a color additive is chemically bound or physically entrapped by the polymer composition that is released at the same rate as the therapeutic drug composition, such that changes in the intensity of the color additive indicate changes in the amount or dose of therapeutic drug composition remaining bound or entrapped within the polymer.
  • an ultraviolet (UV) absorber is chemically bound or physically entrapped within the contact lens polymer.
  • the contact lens is either hydrophobic or hydrophilic.

Abstract

The present invention comprises a composition with means to inhibit the function of the inflammatory cytokine IL-1 and methods for using this composition to treat inflammatory disease of ocular and adnexal tissues by topical administration. The present invention also discloses devices for delivering this composition to target tissues.

Description

    RELATED APPLICATIONS
  • This application claims priority to U.S. application Ser. No. 12/298,380, filed Aug. 15, 2008, which claims priority to International Application No. PCT/US08/09776, filed Apr. 28, 2010, which claims priority to U.S. Provisional Application Nos. 60/965,135, filed Aug. 16, 2007 and 61/130,687, filed Jun. 2, 2008, the contents of which are each herein incorporated by reference in their entirety.
  • FIELD OF THE INVENTION
  • This invention relates generally to the field of ophthalmology.
  • BACKGROUND OF THE INVENTION
  • Inflammation of the ocular and adnexal tissues can occur by a variety of mechanisms and is associated, either primarily or secondarily, with a large number of disease conditions. Current treatments for inflammation of these tissues involve the systemic administration of antibiotics, steroids, and immune-system inhibitors. The difficulty of using these systemic drugs becomes apparent through damaging long-term side effects in the case of steroids, long-term drug resistance in the case of antibiotics, or insufficient long-term persistence at the target site in the case of signaling inhibitors. Moreover, the systemic inhibition of signaling within the immune system can have deleterious outcomes for individuals already afflicted with disease, whose susceptibility to additional complications is increased as a result of the systemic use of these treatments.
  • SUMMARY OF THE INVENTION
  • The present invention overcomes these obstacles by administering a topical composition comprising one or more antagonists of IL-1 function, or a combination of IL-1 and other inflammatory antagonists, to locally decrease or prevent inflammation of the ocular and adnexal tissues.
  • A method for inhibiting or reducing the severity of an ocular inflammatory disorder is carried out by locally administering to an ocular or adnexal tissue of a subject a composition that inhibits an activity of an inflammatory interleukin-1 cytokine such as binding of an inflammatory IL-1 cytokine to an IL-1 receptor. The subject is identified as suffering from a ocular inflammatory disorder by detecting a sign or symptom selected from the group consisting of epithelial overexpression of an inflammatory cytokine, vascular hyperplasia or thickening of lid margin, neovascularization of lid margin or corneal periphery, increase of leukocytes at an ocular or adnexal tissue, or overexpression of a matrix metalloprotease at an ocular or adnexal tissue. The method of therapy inhibits or reduces the severity of at least one of these signs or symptoms. For example, the inflammatory disorder is blepharitis. The method comprises administration of a compound that inhibits binding of an inflammatory IL-1 cytokine to an IL-1 receptor. Optionally, the composition also contains an antibiotic compound. The composition does not comprise tetracycline alone, e.g. in the absence of a functional antagonist that specifically targets IL-1. For example, the composition comprises an antibiotic composition administered in combination with a functional antagonist specifically targeting IL-1.
  • The composition that inhibits binding of an inflammatory IL-1 cytokine to an IL-1 receptor comprises the amino acid sequence of SEQ ID NO: 16. For example, the composition is present in a concentration range of 0.1-10%, with preferred ranges between 1-5%, or 2-2.5% (mg/ml). Exemplary liquid formulations for eye drops contain 2-2.5% (mg/ml) of the composition. Preferred formulations are in the form of a solid, a paste, an ointment, a gel, a liquid, an aerosol, a mist, a polymer, a film, an emulsion, or a suspension. The formulations are administered topically, e.g., the composition is delivered to an ocular or adnexal tissue to directly contact that tissue. The method does not involve systemic administration or substantial dissemination of the composition to non-ocular or non-adnexal tissue. For example, subcutaneous injection of Kineret (see SEQ ID NO: 15 and 16) at 1-2 mg/kg results in an estimated peak blood serum concentration of about 1200-1500 ng/ml 7 hours post-injection. Topical administration of Kineret, as disclosed herein, temporally and spatially restricts absorption of the drug to a much greater degree than subcutaneous injection. The systemic dissemination of a topically administered Kineret composition contributes significantly less drug to the blood serum concentration than a subcutaneous injection.
  • Optionally, the composition further contains a compound selected from the group consisting of a physiological acceptable salt, poloxamer analogs with carbopol, carbopol/hydroxypropyl methyl cellulose (HPMC), carbopol-methyl cellulose, carboxymethylcellulose (CMC), hyaluronic acid, cyclodextrin, and petroleum.
  • The invention comprises a composition that inhibits an activity of an inflammatory interleukin-1 cytokine, the composition being in the form of a solid, a paste, an ointment, a gel, a liquid, an aerosol, a mist, a polymer, a film, an emulsion, or a suspension. The composition is present at a concentration of 0.1-10% (mg/ml). An exemplary composition includes a polypeptide comprising the amino acid sequence of SEQ ID NO: 16.
  • A method for inhibiting or reducing the severity of an ocular inflammatory disorder is also carried out by locally administering to an ocular or adnexal tissue of a subject a composition comprising a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule that inhibits the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding an inflammatory interleukin-1 cytokine (IL-1a, SEQ ID NO: 1 and 2, or IL-1b, SEQ ID NO: 3 and 4), an IL-1 receptor (type 1, SEQ ID NO: 17 and 18, or type 2, SEQ ID NO: 17-21), an IL-1R binding protein (IL-1RAP, SEQ ID NO: 24-27), or an IL-1R downstream signaling effector (IRAK1, SEQ ID NO: 28-33).
  • Alternatively, the composition inhibits or enhances the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding the IL-1 receptor, type 2 (IL-1R2). IL-1R2 binds IL-1 and can inhibit the function of IL-1R1. Thus, in one embodiment, enhancement of IL-1R2 function provides another mechanism by which IL-1R1 activity is inhibited. In this same embodiment, inhibition of an antagonist of IL-1R2, specifically, IL-1Ra3, inhibits IL-1R1 function. Thus, the composition alone, or in combination with an enhancer of IL-1R2, inhibits the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding IL-1Ra3, SEQ ID NO: 22 or 23. Alternatively, in an embodiment wherein IL-1R2 receptor function augments the activity of IL-1R1, the composition contains one or more regions of a polynucleotide or polypeptide encoding IL-1Ra3 to augment IL-1R2 inhibition. Furthermore, the composition of this embodiment comprises the whole polynucleotide or polypeptide encoding IL-1Ra3.
  • The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding an accessory protein of an IL-1 Receptor. For example, this IL-1 receptor accessory protein is IL-1RAP, which directly binds IL-1 and IL-1R1, and is defined by the polynucleotide sequence of SEQ ID NO: 24 or 26 and the polypeptide sequence of SEQ ID NO: 25 or 27. IL-1RAP belongs to a signaling complex that is required for signal transduction from IL-1R1. Thus, inhibition of IL-1RAP antagonizes IL-1R1 function.
  • In another embodiment, the composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding an associated kinase to an IL-1 receptor. For example, IL-1 receptor-associated kinase is IRAK1. IRAK1 is a downstream signaling effector that leads to transcriptional events associated with escalating inflammatory responses and is defined by the polynucleotide sequence of SEQ ID NO: 28, 30, or 32 and the polypeptide sequence of SEQ ID NO: 29, 31, or 33. Upon IL-1 receptor binding by IL-1, IRAK1 is recruited to the receptor complex, becomes hyperphosphorylated, and participates in the formation of a new protein complex consisting of hyperphosphorylated IRAK1 and TRAF6. The formation of this IRAK1/TRAF6 complex is a prerequisite for tumor necrosis factor (TNF) associated factor 6 (TRAF6)-mediated activation of nuclear factor-KB (NF-κB) and subsequent induction of an inflammatory response. Thus, the inhibition of IRAK1 expression and/or function provides an additional mechanism for inhibiting an IL-1-mediated immune response.
  • The composition comprises a polynucleotide, a polypeptide, an antibody, or a small molecule that binds or modifies the function of IL-1a, IL-1b, IL-1R1, IL-1R2, IL-1Ra3, IL-1RAP, or IRAK1. Moreover the composition comprises morpholino antisense oligonucleotides, microRNAs (miRNAs), short hairpin RNA (shRNA), or short interfering RNA (siRNA) to silence gene expression. Exemplary compounds to be adapted for topical administration include, but are not limited to, anakinra/Kineret® (recombinant human IL-1Ra, rhIL-1Ra, and SEQ ID NO: 15 and 16), IL-1R antisense oligomers (U.S. Patent No. 2005033694), IL-1Ra-like nucleic acid molecule (Amgen, U.S. Patent No. 2001041792), and polynucleotide encoding a soluble IL-1R accessory molecule (Human Genome Sciences, U.S. Pat. No. 6,974,682).
  • The composition comprises microRNA molecules adapted for topical administration to ocular or adnexal tissues in order to silence gene expression. Exemplary miRNAs that bind to human IL-1α include, but are not limited to, miR-30c (SEQ ID NO: 34), miR-30b (SEQ ID NO: 35), miR-30a-5p (SEQ ID NO: 36), and miR-24 (SEQ ID NO: 37). Exemplary miRNAs (and corresponding sequences) that bind to human IL-1R1 include, but are not limited to, miR-135b (SEQ ID NO: 38), miR-326 (SEQ ID NO: 39), miR-184 (SEQ ID NO: 40), miR-214 (SEQ ID NO: 41), miR-203 (SEQ ID NO: 42), miR-331 (SEQ ID NO: 43), and miR-205 (SEQ ID NO: 44).
  • Exemplary polypeptides to be adapted for topical administration to ocular or adnexal tissues include, but are not limited to, anakinra/Kineret® (recombinant human IL-1Ra, rhIL-1Ra, and SEQ ID NO: 15 and 16), AF12198 (binds human IL-1R1, Ac-FEWTPGWYQJYALPL-NH2 where J represents the unnatural amino acid, 2-azetidine-1-carboxylic acid, SEQ ID NO: 45), IL-1R and IL-1RAP peptide antagonists (U.S. Patent No. 20060094663), IL-1R accessory molecule polypeptides (U.S. Patent No. 20050171337), IL-1Ra peptides (U.S. Patent No. 2005105830), and IL-1Ra-related peptides (Amgen, U.S. Patent No. 2001042304).
  • Exemplary antibodies to be adapted for topical administration to ocular or adnexal tissues include, but are not limited to, IL-1 TRAP (inline fusion double chain protein of IL1R-gp130 with hIgGFc, Regeneron, U.S. Issued U.S. Pat. No. 6,927,044), anti-IL-1α (U.S. Patent No. 20030026806), anti-IL-1β (U.S. Patent No. 20030026806 and Yamasaki et al. Stroke. 1995; 26:676-681), and humanized monoclonal anti-IL-1R (Amgen, U.S. Patent No. 2004022718 and Roche, U.S. Patent No. 2005023872).
  • Small molecules are organic or inorganic. Exemplary organic small molecules include, but are not limited to, aliphatic hydrocarbons, alcohols, aldehydes, ketones, organic acids, esters, mono- and disaccharides, aromatic hydrocarbons, amino acids, and lipids. Exemplary inorganic small molecules comprise trace minerals, ions, free radicals, and metabolites. Alternatively, small molecule inhibitors can be synthetically engineered to consist of a fragment, or small portion, or a longer amino acid chain to fill a binding pocket of an enzyme. Typically small molecules are less than one kilodalton. An exemplary small molecule to be adapted for topical administration to ocular or adnexal tissues is ZnPP (IL-1 blocker zinc protoporphyrin, naturally-occurring metabolite, Yamasaki et al. Stroke. 1995; 26:676-681).
  • The composition does or, alternatively, does not comprise one or more antibiotic compositions to be used in combination with an antagonist of IL-1 function. The antibiotic and IL-1 antagonist compositions are administered simultaneously or sequentially. Exemplary antibiotic compositions used for combination-therapy with antagonists of IL1-mediated inflammation include but are not limited to, amikacin, gentamicin, kanamycin, neomycin, netilmicin, streptomycin, tobramycin, teicoplanin, vancomycin, azithromycin, clarithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, amoxicillin, ampicillin, azlocillin, carbenicillin, clozacillin, dicloxacillin, flucozacillin, mezlocillin, nafcillin, penicillin, piperacillin, ticarcillin, bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, moxifloxacin, norfloxacin, oflazacin, trovafloxacin, mafenide, sulfacetamide, sulfamethizole, sulfasalazine, sulfisoxazole, trimethoprim, cotrimoxazole, demeclocycline, soxycycline, minocycline, oxytetracycline, or tetracycline.
  • The composition comprises an antagonist of an IL-1 cytokine or an IL-1 receptor, administered simultaneously or sequentially with a second immunosuppressive composition. The immunosuppressive compound comprises cyclosporin A or analogs thereof a concentration of 0.05-4.0% (mg/ml). Alternatively, or in addition, the immunosuppressive composition comprises a glucocorticoid, a cytostatic agent, an alkylating agent (nitrogen mustards/cyclophosphamide, nitrosoureas, platinum compounds), an antimetabolic agent (methotrexate, any folic acid analog, azathioprine, mercaptopurine, any purine analog, any pyrimidine analog, any inhibitor of protein synthesis), a cytotoxic antibiotic (dactinomycin, an anthracycline, mitomycin C, bleomycin, mithramycin), a polyclonal antibody (Atgam®, Thympglobuline®, any antibody against the antilymphocyte or antithymocyte antigens), a monoclonal antibody (OKT3®, any antibody against the T-cell receptor, any antibody against IL-2, basiliximab/Simulect®, declizumab/Zenapax®), Tacrolimus/Prograf™/FK506, Sirolimus/Rapamune™/Rapamycin, interferon beta, interferon gamma, an opioid, a TNFα binding protein, mycophenolate, or FTY720.
  • The composition comprises a polynucleotide, a polypeptide, an antibody, or a small molecule that binds or modifies the function of IL-1α, IL-1b, IL-1Ra, IL-1R1, IL-1R2, Il-1Ra3, IL-1RAP, or IRAK1, administered topically with a pharmaceutically appropriate carrier. Delivery methods for polynucleotide compositions include, but are not limited to, liposomes, receptor-mediated delivery systems, naked DNA, and engineered viral vectors such as herpes viruses, retroviruses, adenoviruses and adeno-associated viruses, among others. Polynucleotide compositions are administered topically with a pharmaceutically acceptable liquid carrier, e.g., a liquid carrier, which is aqueous or partly aqueous. Alternatively, polynucleotide sequences within the composition are associated with a liposome (e.g., a cationic or anionic liposome).
  • A number of methods have been developed for delivering short DNA or RNA sequences into cells; e.g., polynucleotide molecules can be contacted directly onto the tissue site, or modified polynucleotide molecules, designed to specifically target desired cell types (e.g., sequences linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface).
  • A preferred approach uses a recombinant DNA construct in which the short polynucleotide sequence is placed under the control of a strong polymerase III or polymerase II promoter. The use of such a construct will result in the transcription of sufficient amounts of polynucleotide that will form complementary base pairs with the endogenous transcripts of nucleic acids of the invention and thereby prevent translation of endogenous mRNA transcripts. The invention encompasses the construction of a short polynucleotide using the complementary strand as a template. For example, a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an interfering RNA or precursor to a double stranded RNA molecule. Alternatively, the template for the short polynucleotide transcript is placed under the transcriptional control of a cell-type specific promoter or other regulatory element. Thus, diffusion or absorption of a topically administered composition beyond the intended ocular or adnexal target tissue does not cause deleterious or systemic side effects. The vector remains episomal or becomes chromosomally integrated, as long as it can be transcribed to produce the desired polynucleotide.
  • Vectors are constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells. Expression of the sequence encoding the short polynucleotide can be placed under the control of any promoter known in the art to act in mammalian, preferably human cells. Promoters are inducible or constitutive. Exemplary promoters include, but are not limited to: the SV40 early promoter region (Bernoist et al., Nature 290:304, 1981); the promoter contained in the 3′ long terminal repeat of Rous sarcoma virus (Yamamoto et al., Cell, 22:787-797, 1988); the herpes thymidine kinase promoter (Wagner et al., Proc. Natl. Acad. Sci. USA, 78:1441, 1981); or the regulatory sequences of the metallothionein gene (Brinster et al., Nature, 296:39, 1988).
  • Polypeptide compositions are associated with liposomes alone or in combination with receptor-mediated delivery systems, to enable transport across the plasma membrane. Polypeptide compositions are soluble or membrane-bound. An exemplary receptor-mediated delivery system involves fusion of a low-density or very-low-density lipoprotein containing particle or vesicle to the low-density lipoprotein (LDL) receptor (LDLR) as observed with Hepatitis C Virus (HCV) infection and HCV-mediated drug delivery methods.
  • Compositions comprise one or more extracellular or intracellular antibodies, also called intrabodies, raised against one or more of the following: IL-1a, IL-1b, IL-1Ra, IL-1R1, IL-1R2, Il-1Ra3, IL-1RAP, or IRAK1. Extracellular antibodies are topically administered with a pharmacologically appropriate aqueous or non-aqueous carrier. Sequences encoding intracellular antibodies are subcloned into a viral or mammalian expression vector, packed in a lipophilic device to facilitate transport across the plasma membrane, and topically administered to ocular or adnexal tissue with a pharmacologically appropriate aqueous or non-aqueous carrier. Once inside the plasma membrane, host cell machinery transcribes, translates, and processes the intrabody code to generate an intracellular function-blocking antibody targeted against IL-1a, IL-1b, IL-1Ra, IL-1R1, IL-1R2,1′-1Ra3, IL-1RAP, or IRAK1. In the case of secreted molecules, intracellular antibodies prevent post-translational modification or secretion of the target protein. In the case of membrane-bound molecules, intracellular antibodies prevent intracellular signaling events upon receptor engagement by IL-1 cytokines.
  • The composition comprises an antagonist of IL-1 and/or IL-1R function in combination with other inhibitory elements. Antagonists of IL-1 and/or IL-1R and other inhibitory elements are administered simultaneously or sequentially. In one embodiment, the composition comprises an antagonist of IL-1 and/or IL-1R function and an antagonist of tumor necrosis factor alpha (TNFα). Exemplary functional blockers of TNFα include, but are not limited to, recombinant and/or soluble TNFα receptors, monoclonal antibodies, and small molecule antagonists and/or inverse agonists. One or more commercially-available TNF-α blocking agents are reformulated for topical administration in this embodiment. Exemplary commercial TNF-α blocking agents used for reformulation include, but are not limited to, etanerept/Embrel, infliximab/Remicade, and adalimumab/Humira. Alternatively, the composition comprises an antagonist of IL-1 and/or IL-1R function and antagonist(s) of one or more interleukin cytokines. Exemplary cytokines include, but are not limited to, IL-2, IL-4, IL-5, IL-6, IL-8, IL-12, IL-17, IL-18, and IL-23. In another embodiment, the composition comprises an antagonist of IL-1 and/or IL-1R function and antagonist(s) of one or more member(s) of the vascular epithelial growth factor (VEGF) family composed of growth factors and receptors (VEGFR). Exemplary members include, but are not limited to, VEGF-A, VEGF-C, VEGFR-2, and VEGFR-3. In another embodiment, the composition comprises an antagonist of IL-1 and/or IL-1R function and an antagonist of interferon-gamma. In another embodiment, the composition comprises an antagonist of IL-1 and/or IL-1R function and antagonist(s) of one or more chemokines and their receptors. Exemplary chemokines and receptors comprised by the composition of this embodiment include, but are not limited to, chemokine (C-C motif) receptor 1 (CCR1), chemokine (C-C motif) receptor 2 (CCR2), chemokine (C-C motif) receptor 5 (CCR5), chemokine (C-C motif) receptor 7 (CCR7), and chemokine (C-X-C motif) receptor 3 (CXCR3).
  • In embodiments wherein the composition comprises an antagonist of IL-1 and/or IL-1R function and antagonist(s) of one or more inflammatory species, the respective doses of the IL-1 antagonist to the other inflammatory antagonist(s) is a ratio between 1:10 and 10:1 (mass/weight). Alternatively, the ratio is 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, or 9:1.
  • The invention also comprises a contact lens device consisting of a composition that inhibits an activity of an inflammatory interleukin-1 cytokine and a pharmaceutically compatible polymer. This composition also comprises a combination of antagonists of IL-1 or IL-1R function as well as antagonists of other inflammatory agents. For example, the composition is incorporated into or coated onto said lens. The composition is either chemically bound or physically entrapped by the contact lens polymer. The contact lens is either hydrophobic or hydrophilic.
  • The invention comprises a drug-delivery device consisting of a composition that inhibits an activity of an inflammatory interleukin-1 cytokine and a pharmaceutically compatible polymer. This composition also comprises a combination of antagonists of IL-1 or IL-1R function as well as antagonists of other inflammatory agents. For example, the composition is incorporated into or coated onto said polymer. The composition is either chemically bound or physically entrapped by the polymer. The polymer is either hydrophobic or hydrophilic. The polymer device comprises multiple physical arrangements. Exemplary physical forms of the polymer device include, but are not limited to, a film, a scaffold, a chamber, a sphere, a microsphere, a stent, or other structure. The polymer device has internal and external surfaces. The device has one or more internal chambers. These chambers contain one or more compositions. The device contains polymers of one or more chemically-differentiable monomers. The subunits or monomers of the device polymerize in vitro or in vivo.
  • Exemplary mucoadhesive polyanionic natural or semi-synthetic polymers from which the device is formed include, but are not limited to, polygalacturonic acid, hyaluronic acid, carboxymethylamylose, carboxymethylchitin, chondroitin sulfate, heparin sulfate, and mesoglycan. In one embodiment, the device comprises a biocompatible polymer matrix that may optionally be biodegradable in whole or in part. A hydrogel is one example of a suitable polymer matrix material. Examples of materials which can form hydrogels include polylactic acid, polyglycolic acid, PLGA polymers, alginates and alginate derivatives, gelatin, collagen, agarose, natural and synthetic polysaccharides, polyamino acids such as polypeptides particularly poly(lysine), polyesters such as polyhydroxybutyrate and poly-.epsilon.-caprolactone, polyanhydrides; polyphosphazines, poly(vinyl alcohols), poly(alkylene oxides) particularly poly(ethylene oxides), poly(allylamines) (PAM), poly(acrylates), modified styrene polymers such as poly(4-aminomethylstyrene), pluronic polyols, polyoxamers, poly(uronic acids), poly(vinylpyrrolidone) and copolymers of the above, including graft copolymers. In another embodiment, the scaffolds may be fabricated from a variety of synthetic polymers and naturally-occurring polymers such as, but not limited to, collagen, fibrin, hyaluronic acid, agarose, and laminin-rich gels.
  • One preferred material for the hydrogel is alginate or modified alginate material. Alginate molecules are comprised of (1-4)-linked β-D-mannuronic acid (M units) and a L-guluronic acid (G units) monomers which vary in proportion and sequential distribution along the polymer chain. Alginate polysaccharides are polyelectrolyte systems which have a strong affinity for divalent cations (e.g. Ca+2, Mg+2, Ba+2) and form stable hydrogels when exposed to these molecules. See Martinsen A., et al., Biotech. & Bioeng., 33 (1989) 79-89.
  • An embodiment of the invention utilizes an alginate or other polysaccharide of a lower molecular weight, preferably of size which, after dissolution, is at the renal threshold for clearance by humans. Polymeric devices are located topically or subcutaneously, though very superficially, wherein either a composition chemically bound or physically entrapped by the polymeric device or the device itself, degrades and must be cleared from the body. For a biodegradable polymeric device, it is preferred that the alginate or polysaccharide is reduced to a molecular weight of 1000 to 80,000 daltons, more preferably 1000 to 60,000 daltons, particularly preferably 1000 to 50,000 daltons. It is also useful to use an alginate material of high guluronate content since the guluronate units, as opposed to the mannuronate units, provide sites for ionic crosslinking through divalent cations to gel the polymer.
  • Internal and external surfaces optionally contain pores. Pores are either created prior to administration into a subject or result from the inclusion of pore-forming agents within the device that perforate surfaces upon administration to a subject. Exemplary pore forming agents include, but are not limited to, water soluble compounds such as inorganic salts and sugars. Pore forming agents are added as particulates and comprise between one and thirty percent (weight/weight of polymer). Pore size is sufficient for diffusion of proteins but not large enough cell migration into or out of the device.
  • The device is administered topically, subconjuntively, or in the episcleral space, subcutaneously, or intraductally. Specifically, the device is placed on or just below the surface if an ocular or adnexal tissue. Alternatively, the device is placed inside a tear duct or gland. The composition incorporated into or onto the polymer is released or diffuses from the device.
  • The invention comprises a method of contacting a composition, with means to inhibit IL-1R activity, to an ocular or adnexal tissue surface of a subject who presents symptoms or associated conditions of posterior blepharitis. Unlike all other treatments for this condition, this method comprises a composition that is topically administered to the subject and affects local disease mechanisms without the side effects present with systemically administered treatments. The composition is not only effective immediately, but also safe for long-term administration.
  • The invention comprises a method of treating non-infectious eye disease, wherein a composition containing an inhibitor of IL-1 or IL-1R function alleviates, prevents, or attenuates a symptom, cause, or mechanism of said disease by contacting said composition to the ocular surface of an affected subject. Exemplary disease mechanisms comprise inflammation, hyperplasia, neovascularization, leukocyte recruitment, or cytokine production within superficial eye structures and those structures juxtaposed to the ocular surface. For instance, non-infectious eye disease is caused by obstruction, thickening, inflammation, neovascularization, or atrophy of the meibomian glands. Alternatively, non-infectious eye disease is caused by damage to structures other than the meibomian glands. Exemplary alternative causes include, but are not limited to, oily tear film, papillary hypertrophy of the tarsal conjunctiva, corneal punctate epitheliopathy, vernal keratoconjunctivitis, atopic keratocojunctivitis, chemical burn, trachoma, pterygium, pemphigoid, corneal angiogenesis (growth of new blood vessels), psoriasis, iethyosis, erythema mutiforme, anhydrotic ectodermal dysplasia, systemic retinoid therapy, or exposure to polychlorinated byphenols. Finally, non-infectious eye disease can also result from a deficient tear lipid layer, an increase in tear evaporation, or the occurrence of an evaporative dry eye. The methods treat non-infectious eye disease that is or is not coincident with the presentation of dermatoses comprising acne rosacea, seborrhoeic dermatitis, or atopic dermatitis. Furthermore, methods are applicable to treat a primary or secondary non-infectious eye disease that is coincident with chalazia, pannus, phlyctenules, recurrent conjunctivitis, ocular surface damage, ocular rosacea, corneal ulceration, corneal perforation or secondary complications of ocular infection.
  • The methods described herein are not intended to treat eye disease that results from an immune response that consequently arises following the transplantation of foreign tissue onto or into an ocular or adnexal tissue. For instance, inflammatory responses occur following host rejection of corneal transplant or as a consequence of infection following surgical procedures. The instant application is not intended to treat eye conditions that result directly from tissue rejection or infectious disease. However, immune responses to foreign tissues or to infectious agents can lead to protean secondary complications, such as growth of blood or lymph vessels, or overexpression of molecules that cause tissue injury; in such cases the methods described in the present application are useful as a treatment for reducing inflammation associated with such secondary complications. Furthermore, the methods and devises comprising the instant application are not intended to treat conditions arising from autoimmune responses, e g immune responses raised against healthy host tissues.
  • In a preferred embodiment, the non-infectious eye disease comprises blepharitis and/or posterior blepharitis that is or is not coincident with inflammation of the posterior lid margin, inflammation of the ocular surface, burning, irritation, or patient discomfort. Alternatively, the present invention is intended to treat blepharitis that is coincident with meibomian gland dysfunction.
  • The methods alleviate symptoms of non-infectious eye disease. Exemplary symptoms include, but are not limited to, dryness, discomfort, burning, itching, irritation, inflammation, skin abnormalities surrounding the eye, photophobia, blurred vision, and contact lens intolerance.
  • The present invention comprises a composition with variable physical and chemical forms; however, the composition is topically administered and contacts an ocular or adnexal tissue directly. The composition is administered as a solid, a paste, an ointment, a gel, a liquid, an aerosol, a mist, a polymer, a film, an emulsion, or a suspension. Furthermore, the composition is incorporated into or coated onto a contact lens, from which one or more molecules diffuse away from the lens or are released in a temporally-controlled manner. In this embodiment, the contact lens composition either remains on the ocular surface, e.g. if the lens is required for vision correction, or the contact lens dissolves as a function of time simultaneously releasing the composition into closely juxtaposed tissues.
  • In one preferred embodiment, the present invention comprises a composition with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or receptor binding of IL-1α, IL-1β, or a combination of both cytokines. In one embodiment, the composition comprises a polynucleotide capable of binding to a region of the IL-1α mRNA transcript, defined by SEQ ID NO: 1. In another embodiment, the composition comprises a polynucleotide capable of binding to a region of the IL-1β mRNA transcript, defined by SEQ ID NO: 3.
  • In another embodiment, the composition is capable of increasing the abundance of the naturally-occurring IL-1 Receptor antagonist (IL-1Ra). The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule that binds to a region of the IL-1Ra gene, mRNA transcript defined by SEQ ID NO: 5, 7, 9, 11, or 13, a polypeptide isoform of IL-1Ra defined by SEQ ID NO: 6, 8, 10, 12, or 14, or a recombinant IL-1Ra protein defined by SEQ ID NO: 16. Alternatively, the composition contains mRNA transcripts or polypeptides encoding a region or the entirety of the IL-1Ra gene.
  • The composition comprises an antagonist or inverse agonist of a receptor for IL-1α or IL-1β, specifically, IL-1R1. In this embodiment an antagonist is defined as a binding partner, or ligand, of an IL-1R that inhibits the function of an agonist, IL-1, or inverse agonist by blocking its binding to the receptor. An inverse agonist is defined as a molecule which binds to the same IL-1R binding-site as an agonist, for instance, IL-1, but exerts the opposite pharmacological effect. The composition contains a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule that binds to a region of the IL-1R1 defined by the polynucleotide and polypeptide sequences SEQ ID NO: 17-21. In an alternative embodiment, the composition comprises a molecule with means to inhibit IL-1R transcription, transcript stability, translation, modification, localization, secretion, ligand binding, or association with an accessory protein of an IL-1R (IL-1RAP). IL-1RAP is defined by the polynucleotide sequence of SEQ ID NO: 24 or 26 and the amino acid sequence of SEQ ID NO: 25 or 27.
  • The composition comprises a molecule with means to inhibit IL-1α- or IL-β-mediated modulation of matrix metalloproteinase (MMP) overexpression, which degrades collagen matrices within tissues and retards wound healing.
  • In another preferred embodiment, the composition comprises a human recombinant IL-1R antagonist either in pure form, or as a component of a mixture. The human recombinant IL-1R antagonist is combined with balanced saline, carboxymethylcellulose (CMC), or hyaluronic acid (HA), or other vehicles prior to the composition contacting the ocular or lid surface. Within these mixtures, the human recombinant IL-1R antagonist comprises at least 0.1%, 2.0%, 2.5%, 5%, or at most 10% of the total volume administered. Preferred aqueous formulations contain 2-2.5% of the purified antagonist. Purified is defined as the antagonist in the absence of unrelated polynucleotides, polypeptides, cellular organelles, or lipids. Purified is defines a degree of sterility that is safe for administration to a human subject, e.g. lacking infectious or toxic agents.
  • Affected subjects to which this invention is administered are identified by a variety of methods. The following examinations are used individually or combinatorially to assess the presence or absence of non-infectious eye disease (for explanation of examinations, see Examples). Subjects are identified by a break-up time of less than 10 seconds following a standard Tear Film Break-up Time (TFBT) test. Subjects are identified by a significant corneal fluorescein staining of tear film and/or conjunctival lissamine green or rose bengal staining. Subjects are identified by a result of 10 mm or less for the Schrimer test with or without anesthesia. Subjects are identified by significantly viscous tear excreta and/or a decrease in the number of meibomian glands or meibomian gland orifices capable of excreting tears. Subjects are identified by shades of red discoloration of the lid margin, palpebral conjunctiva, and/or bulbar conjunctiva indicating increasing extreme vascular injection (erythema). Subjects are also classified as presenting increasingly severe non-infectious eye disease with the increasing abundance and severity of the above factors.
  • The compositions and methods provided herein are used modify intraocular pressure. In one aspect of the invention, the compositions and methods provided herein are used to decrease intraocular pressure Inhibitors or antagonists of IL-1 cytokines and/or IL-1 receptors are used alone or in combination with other compounds to modify intraocular pressure. In a preferred embodiment of the invention, inhibitors or antagonists of IL-1 cytokines and/or IL-1 receptors are used alone or in combination with other compounds to decrease intraocular pressure. Alternatively, or in addition, inhibitors or antagonists of IL-1 cytokines and/or IL-1 receptors are used alone or in combination with other compounds to treat ocular hypertension.
  • Exemplary compounds to be used in combination with IL-1 cytokines and/or IL-1 receptors include, but are not limited to, prostaglandin analogs (such as latanoprost (Xalatan), bimatoprost (Lumigan) and travoprost (Travatan) which increase uveoscleral or trabecular outflow of aqueous humor); topical beta-adrenergic receptor antagonists (such as timolol, levobunolol (Betagan), and betaxolol, which decrease aqueous humor production by the ciliary body); Alpha2-adrenergic agonists (such as brimonidine (Alphagan), which work by a dual mechanism, decreasing aqueous production and increasing uveo-scleral outflow); less-selective sympathomimetics (such as epinephrine and dipivefrin (Propine), which increase outflow of aqueous humor through trabecular meshwork and possibly through uveoscleral outflow pathway, probably by a beta2-agonist action); miotic agents (parasympathomimetics) (such as pilocarpine, which work by contraction of the ciliary muscle, tightening the trabecular meshwork and allowing increased outflow of the aqueous humour); carbonic anhydrase inhibitors (such as dorzolamide (Trusopt), brinzolamide (Azopt), acetazolamide (Diamox), which lower secretion of aqueous humor by inhibiting carbonic anhydrase in the ciliary body); physostigmine which is also used to treat glaucoma and delayed gastric emptying; fish oil; omega 3 fatty acids, bilberries, vitamin E, cannabinoids, carnitine, coenzyme Q10, curcurmin, Salvia miltiorrhiza, dark chocolate, erythropoietin, folic acid, Ginkgo biloba, Ginseng, L-glutathione, grape seed extract, green tea, magnesium, melatonin, methylcobalamin, N-acetyl-L cysteine, pycnogenols, resveratrol, quercetin, and fludrocortisone.
  • The invention also provides compositions and methods for treating individuals or subjects with ocular hypertension including administrating to these subjects a composition comprising an IL-1 or IL-1R inhibitor or antagonist. In one aspect of the invention, the IL-1 or IL-1R inhibitor or antagonist is administered topically to the ocular surface as a liquid. In another aspect of the invention, the IL-1 or IL-1R inhibitor or antagonist is administered intraocularly by injection.
  • The invention also provides compositions and methods for preventing glaucoma in individuals or subjects with ocular hypertension including administrating to these subjects a composition comprising an IL-1 or IL-1R inhibitor or antagonist. In one aspect of the invention, the IL-1 or IL-1R inhibitor or antagonist is administered topically to the ocular surface as a liquid. In another aspect of the invention, the IL-1 or IL-1R inhibitor or antagonist is administered intraocularly by injection.
  • The invention provides a method for reducing intraocular pressure, including identifying a subject suffering from or at risk of a condition associated with above-normal intra-ocular pressure and locally administering to the subject a composition that inhibits an activity of an inflammatory interleukin-1 cytokine. In one preferred aspect of the invention, the condition is glaucoma.
  • In one aspect of the invention, subjects are identified by measuring their intraocular pressure and determining if the measured intraocular pressure is elevated above normal levels. As used herein, the term “normal level” is meant to describe value within an acceptable range of values that one of ordinary skill in the art and/or a medical professional would expect a healthy subject of similar physical characteristics and medical history to have. For example, normal intraocular pressure (IOP) is defined as IOP in the range of 10 mmHg to 21 mmHg.
  • In another aspect of the invention, subjects are identified as those individuals who are at risk for developing elevated intraocular pressure based upon non-limiting factors such as medical history (for instance, diabetes), side effects of medications, lifestyle and/or diet, medical intervention (such as surgery to the eye), trauma/injury, hormone changes, and aging. Compositions of the invention are administered to these subjects for preventative means.
  • All polynucleotides and polypeptides of the invention are purified and/or isolated. As used herein, an “isolated” or “purified” nucleic acid molecule, polynucleotide, polypeptide, or protein, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized.
  • Publications, U.S. patents and applications, Genbank/NCBI accession numbers, and all other references cited herein, are hereby incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is the Ocular Surface Disease Index (OSDI) 12-item questionnaire.
  • FIG. 2 the Oxford Schema for grading corneal and conjunctival staining.
  • FIG. 3 is a graph representing the effect of IL-1a on intraocular pressure (IOP).
  • FIG. 4 is a schematic representation of signaling pathways that are transduced from the IL-1R and the downstream effectors involved in carrying these intracellular signals (drawing reproduced from BioCarta website).
  • FIG. 5 is a questionnaire given to subjects of a study to diagnose Meibomian Gland Dysfunction (Posterior Blepharitis).
  • FIG. 6 is a flow diagram showing the sequence of clinical tests performed on study subjects during the screening process described in Example 8.
  • DETAILED DESCRIPTION Posterior Blepharitis
  • Posterior blepharitis is a common chronic eyelid condition that is described as generalized inflammation of the posterior lid margin and associated with inflammation of the ocular surface and with symptoms of burning, irritation, and discomfort. Posterior blepharitis is associated with various disorders of the meibomian glands, known collectively as meibomian gland dysfunction (MGD). It is associated either with obstruction and inflammation of the meibomian glands or, less commonly, atrophy of the meibomian glands (Foulks, G. et al. 2003. Ocul Surf. 107-26; Bron, A. J. et al. 2004. Ocul Surf. 2:149-65).
  • Clinically, MGD often presents with inspissated meibomian glands, oily tear film, as well as inflammation and vascularization of the meibomian gland orifices. Papillary hypertrophy of the tarsal conjunctiva and corneal punctate epitheliopathy are often present, and there are prominent associations with dermatoses, such as acne rosacea, seborrhoeic dermatitis, and atopic dermatitis. Other causes and associations of MGD are: vernal keratoconjunctivitis, atopic keratoconjunctivitis, chemical burns, trachoma, pemphigoid, psoriasis, iethyosis, erythema multiforme, anhydrotic ectodermal dysplasia, and systemic retinoid therapy. Additionally, exposure to polychlorinated biphenyls, through ingestion of contaminated cooking oils, causes a chronic form of MGD with gross and extensive acneiform skin changes (Fu, Y. A. 1984. Am J Ind Med. 5:127-32).
  • MGD of sufficient extent and degree is associated with a deficient tear lipid layer, an increase in tear evaporation, and the occurrence of an evaporative dry eye. In fact MGD is considered to be the most common cause of evaporative dry eye (Foulks, G. et al. 2003. Ocul Surf. 107-26). Individuals with MGD often complain of significant discomfort, including burning, itching, irritation, and photophobia. They may also have other associated symptoms of dry eye and may be plagued by blurred vision, gradual contact lens intolerance. Furthermore, these patients may become functionally handicapped by the negative impact of dry eye on their crucial daily activities such as working, reading, using computer, and driving (Goto, E. et al. 2002. Am J Opthalmol. 133:181-186; Miljanovic, B. et al. 2007. Am J Opthalmol. 143: 409-15).
  • In addition to dry eye, other sequelae of MGD that confront the ophthalmologist include chalazia, pannus, phlyctenules, and recurrent conjunctivitis which increase risk of ocular surface damage, ocular infection, corneal ulceration, and perforation.
  • Despite the high incidence of posterior blepharitis, there is currently no consistently effective treatment for this condition and it still remains a therapeutic challenge. Posterior blepharitis has traditionally been managed with eyelid hygiene, topical antibiotics (erythromycin or bacitracin ointments), oral tetracyclines (tetracycline, doxycycline, or minocycline) and corticosteroids which are often time consuming, frustrating, and frequently ineffective or variably effective.
  • The current treatments for posterior blepharitis are limited, in part, by their inability to target the underlying pathophysiologic processes. There is ample evidence that posterior blepharitis is the result of an underlying cytokine and inflammatory-mediated process affecting both the meibomian glands and the ocular surface (Kocak-Altintas, A. G. et al. 2003. Eur J Opthalmol. 13:351-359; McCulley, J. P. et al. 2000. Cornea. 19:650-658). In an experimental blepharoconjunctivitis, it has been shown that T-cells are the prime orchestrator driving the infiltration of other inflammatory cells into the conjunctiva, as well as the upregulation of chemokines (Fukushima, A. et al. 2003. Invest. Opthalmol. Vis. Sci. 44:4366-4374).
  • In ocular rosacea which belongs to the large pathologic group of blepharitis and MGD, inflammation of the ocular surface was clearly demonstrated with an increase of inflammatory mediators in tears such as interleukin (IL)-1α (Barton, K. et al. 1997. Opthalmology. 104:1868-1874). Flow cytometry and impression cytology of conjunctival epithelium in ocular rosacea has demonstrated a significantly higher level of intercellular adhesion molecule-1 (ICAM-1) which is known to play a significant role in inflammation associated with dry eye disease. This expression of ICAM-1 could be due to the liberation of one or more pro-inflammatory cytokines such as IL-1β or interferon-γ (Pisella, P. J. et al. 2000. Opthalmology. 107:1841-9).
  • Importance of inflammation in the pathogenesis of posterior blepharitis is underscored by reports that the signs and symptoms of MGD markedly improve with anti-inflammatory therapies such as topical steroids (Marsh, P. et al. 1999. Opthalmology. 106:811-816).
  • Systemically administered tetracycline antibiotics have long been recognized as effective therapies for ocular surface inflammatory diseases. The semisynthetic tetracycline, doxycycline, has been reported to successfully treat the common dry eye condition acne rosacea. One of the mechanisms of action of doxycycline in MGD and dry eye may be the downregulation of the IL-1-mediated inflammatory cascade in the corneal epithelium (Solomon, A. et al. 2000. Invent. Opthalmol. Vis. Sci. 41:2544-57).
  • In addition, findings from several studies indicate that MGD is also associated with aqueous deficiency dry eye patients, especially those with Sjogren syndrome (SS) (McCulley, J. P. et al. 1977. A, J. Opthalmol. 84:788-93; Shimazaki, J. et al. 1998. Opthalmology. 105: 1485-1488). In these studies, at least 35% of patients with aqueous tear deficiency had MGD. Classically, SS affects the exocrine glands, not the sebaceous glands such as the meibomian glands. Underlying inflammation as well as desiccation and keratinization of ocular surface epithelia occurs in chronic dry eye disease and plays a role in pathogenesis of MGD.
  • Consistent with the concept that inflammation is a key feature in the pathophysiology of dry eye syndrome is the finding that both aqueous tear deficiency and meibomian gland disease are effectively treated with the corticosteroid methylprednisolone (Marsh, P. et al. 1999. Opthalmology. 107:967-74). Unfortunately the long-term use of topical corticosteroids is limited by potential sight-threatening side effects, such as glaucoma and cataracts. Therefore, there is a clinical need for nontoxic steroid-sparing anti-inflammatory therapies that target the underlying inflammatory environment of the ocular surface in this common condition.
  • Based on the concept that inflammation is a key component of the pathogenesis of MGD and dry eye, therapies targeting the underlying inflammatory environment of the ocular surface represent a major improvement in the management of these conditions and will have a major clinical impact. Given the facts that IL-1-mediated inflammatory activities play a critical role on the ocular surface in patients with MGD and dry eye, therefore, local blockade of IL-1 activity by IL-1Ra, is used to reduce one or more symptoms of dry eye syndrome.
  • Ocular and Adnexal Tissues:
  • Ocular tissues or compartments that contact the compositions comprised by the present invention include, but are not limited to, the cornea, aqueous humor, iris, and sclera. The term “adnexal” is defined in general terms as the appendages of an organ. In the present invention, adnexal defines a number of tissues or surfaces that are in immediate contact with the ocular surface but are not, by definition, comprised by the ocular surface. Exemplary adnexal tissues include, but are not limited to, the eyelids, lacrimal glands, and extraocular muscles. Topical administration of the presently invented compositions contact the following tissues and structures within the eyelid: skin, subcutaneous tissue, orbicularis oculi, orbital septum, tarsal plates, palpebral conjuntiva, and meibomian glands. The adnexal tissues comprise all subdivisions of the lacrimal glands, including the orbital and palpebral portions, as well as all tissues contacted by these glands. Extraocular muscles belonging to this category of adnexal tissues include, but are not limited to, the superior and inferior rectus, lateral and medial rectus, and superior and inferior oblique muscles. Compositions comprised by the present invention are applied topically and contact these tissues either alone, or in combination with ocular tissues.
  • Intraocular Pressure
  • Intraocular pressure is maintained by the liquid aqueous humor, which is produced by the ciliary body of the eye. Aqueous humor normally does not go into the posterior segment of the eye; it is kept out of this area by the lens and the Zonule of Zinn Instead, it stays only in the anterior segment, which is divided into the anterior and posterior chambers. While the anterior and posterior chambers are very similarly named to the anterior and posterior segments, they are not synonymous. The anterior and posterior chambers are both parts of the anterior segment.
  • When the ciliary bodies produce the aqueous humor, it first flows into the posterior chamber (bounded by the lens and the iris). It then flows through the pupil of the iris into the anterior chamber (bounded by the iris and the cornea). From here, it flows through a structure known as the trabecular meshwork to enter the normal body circulation. Thus, the two main mechanisms of ocular hypertension are an increased production of aqueous humor, or a decreased outflow of aqueous humor.
  • Ocular hypertension (OHT) is intraocular pressure higher than normal in the absence of optic nerve damage or visual field loss. Current consensus in ophthalmology defines normal intraocular pressure (IOP) as that between 10 mmHg and 21 mmHg. Intraocular pressure is measured with a tonometer. Elevated IOP is the most important risk factor for glaucoma, so those with ocular hypertension are frequently considered to have a greater chance of developing the condition. Intraocular pressure can increase when a patient lies down. There is evidence that some glaucoma patients (e.g., normal tension glaucoma patients) with normal IOP while sitting or standing may have intraocular pressure that is elevated enough to cause problems when they are lying down.
  • Differences in pressure between the two eyes is often clinically significant, and potentially associated with certain types of glaucoma, as well as iritis or retinal detachment.
  • Because of the effect of corneal thickness and rigidity on measured value of intraocular pressure, some forms of refractive surgery (such as photorefractive keratectomy) can cause traditional intraocular pressure measurements to appear normal when in fact the pressure may be abnormally high.
  • Intraocular pressure may become elevated due to anatomical problems, inflammation of the eye, genetic factors, as a side-effect from medication, or during exercise. Intraocular pressure usually increases with age and is genetically influenced.
  • Hypotony, or ocular hypotony, is typically defined as intraocular pressure equal to or less than 5 mmHg. Such low intraocular pressure could indicate fluid leakage and deflation of the eyeball.
  • Glaucoma
  • Glaucoma is a leading cause of irreversible blindness. A primary risk factor for glaucoma is elevated intraocular pressure (IOP), which can contribute to significant optic nerve damage and vision loss. Elevated IOP due to reduction in aqueous outflow facility is a major causal risk factor. The main aqueous outflow pathway of the eye consists of a series of endothelial-cell-lined channels in the angle of the anterior chamber comprising the trabecular meshwork (TM), Schlemm's canal, the collector channels and the episcleral venous system.
  • Glaucoma is a group of diseases of the optic nerve involving loss of retinal ganglion cells in a characteristic pattern of optic neuropathy. Although raised intraocular pressure is a significant risk factor for developing glaucoma, there is no set threshold for intraocular pressure that causes glaucoma. Untreated glaucoma leads to permanent damage of the optic nerve and resultant visual field loss, which can progress to blindness. Loss of visual field often occurs gradually over a long time and may only be recognized when it is already quite advanced. Once lost, this damaged visual field can never be recovered. Worldwide, it is the second leading cause of blindness. Glaucoma affects one in two hundred people aged fifty and younger, and one in ten over the age of eighty.
  • Ocular hypertension is the largest risk factor in most glaucomas. Though, in some populations only 50% of patients with primary open angle glaucoma have elevated ocular pressure. Diabetics and those of African descent are three times more likely to develop primary open angle glaucoma. Higher age, thinner corneal thickness, and myopia are also risk factors for primary open angle glaucoma. People with a family history of glaucoma have about a six percent chance of developing glaucoma. Asians are prone to develop angle-closure glaucoma, and Inuit have a twenty to forty times higher risk than Caucasians of developing primary angle closure glaucoma. Women are three times more likely than men to develop acute angle-closure glaucoma due to their shallower anterior chambers. Use of steroids can also cause glaucoma.
  • Primary open angle glaucoma (POAG) has been found to be associated with mutations in genes at several loci. Normal tension glaucoma, which comprises one third of POAG, is associated with genetic mutations.
  • There is increasing evidence of ocular blood flow to be involved in the pathogenesis of glaucoma. Current data indicate that fluctuations in blood flow are more harmful in glaucomatous optic neuropathy than steady reductions. Unstable blood pressure and dips are linked to optic nerve head damage and correlate with visual field deterioration.
  • A number of studies also suggest that there is a correlation, not necessarily causal, between glaucoma and systemic hypertension (i.e. high blood pressure). In normal tension glaucoma, nocturnal hypotension may play a significant role. On the other hand there is no clear evidence that vitamin deficiencies cause glaucoma in humans, nor that oral vitamin supplementation is useful in glaucoma treatment.
  • Various rare congenital/genetic eye malformations are associated with glaucoma. Occasionally, failure of the normal third trimester gestational atrophy of the hyaloid canal and the tunica vasculosa lentis is associated with other anomalies. Angle closure induced ocular hypertension and glaucomatous optic neuropathy may also occur with these anomalies. These rare developmental causes of glaucoma are modeled in mice.
  • Only a few glaucomas are associated with visible cellular inflammation in the ocular compartments; however, Interleukin-1 (IL-1) is consistently up-regulated by glaucomatous TM cells and acts as a key factor in accelerated TM cell injury. This condition is mediated by both IL-1 effect on the TM cells functioning as well as their expression of adhesion factors that can affect outflow facility. The net effect is that IL-1 can compromises the function of the trabecular meshwork, causing reduced outflow facility in the open angle glaucoma. Interleukin-1 receptor antagonist (IL-1Ra) is a naturally occurring IL-1 isoform that with high-affinity binds to the IL-1 receptors and blocks its effects. Therefore, topical application of IL-1Ra, such as human recombinant forms (anakinra/Kineret) suppresses IL-1-mediated cell injury in TM, enhances outflow facility, and reduces IOP in glaucomatous eyes.
  • This mechanism of action is in contrast with the other views that IL-1 itself increases matrix metalloproteinase expression in the TM and increases outflow facility. Therefore, others believed that inhibition of action of IL-1 by an IL-1 neutralizing antibody or with IL-1 receptor antagonist causes reduced outflow facility in trabecular meshwork.
  • Interleukin-1 (IL-1):
  • The IL-1 family is a group of cytokines that function as major mediators of inflammation and immune response (Dinarello, C. A. 1996. Blood. 15:2095-2147). This family is composed of three forms: two proinflammatory forms, IL-1α and IL-1β, each having a precursor form, and an anti-inflammatory form, IL-1 receptor antagonist (IL-1Ra). The proinflammatory cytokine IL-1 plays an important role in inflammation and immunity by increasing chemokine production, adhesion factors, macrophage infiltration and activity, and lymphocyte proliferation. IL-1 has been implicated in the pathogenesis of human inflammatory diseases, such as rheumatoid arthritis, septic shock, and periodontitis (Jiang, Y. et al. 2000. Arthritis Rheum. 43:1001-1009; Okusawa, S. et al. 1988. J Clin Invest. 81: 1162-1172; McDevitt, M. J. et al. 2000. J. Periodontol. 71:156-163).
  • The IL-1 cytokines play an important role in the regulation of inflammation and wound healing in the corneal and ocular surface diseases (Fabre, E. J. et al. 1991. Curr Eye Res. 10:585-592; Rosenbaum, J. T. et al. 1995. Invest Opthalmol Vis Sci. 36: 2151-2155). Both IL-1α and IL-1β have been found to modulate matrix metalloproteinase (MMP) expression by corneal stromal fibroblasts (Fini, M. E. et al. 1990. Invest Opthalmol Vis Sci. 31:1779-1788). Increased levels of IL-1α and IL-1β have been shown in patients with Sjögren syndrome and ocular rosacea (Pflugfelder, S. C. et al. 1999. Curr Eye Res. 19:201-211; Solomon, A. et al. 2001. Invest Opthalmol Vis Sci. 42:2283-2292). Levels of proinflammatory forms of IL-1 are directly correlated with the intensity of corneal fluorescein staining and are inversely correlated with conjunctival goblet cell density. In both types of dry eye syndrome, evaporative and aqueous deficiency, as tear clearance from the ocular surface decreases, the concentrations of both isoforms of the proinflammatory cytokine IL-1 increase in the tear fluid (Solomon, A. et al. 2001. Invest Opthalmol Vis Sci. 42:2283-2292). The results of a study on the effects of doxycycline on the expression patterns of the IL-1 gene family in the human limbal epithelium in response to a strong inflammatory stimulus, have demonstrated an inhibitory effect of doxycycline on the expression of the inflammatory cytokine IL-1β, with a concomitant upregulation of the anti-inflammatory IL-1Ra (Solomon, A. et al. 2000. Invest Opthalmol Vis Sci. 41:2544-57). These results imply that some of the clinically proven benefits of tetracycline compounds (tetracycline, doxycycline, and minocycline) in treating the ocular surface disease of MGD and dry eye may be mediated through their regulatory effects on the synthesis, processing, or release of IL-1.
  • IL-1 induces ocular surface disease, e.g., the chronic subclinical ocular surface inflammation of MGD and dry eye. The compositions and methods described herein inhibit the activity of human IL-1α and/or IL-1β, as defined by the ability to induce signal transduction or initiate/activate a downstream signaling cascade from an IL-1 receptor. Compositions that contain an inhibitor of human IL-1α or IL-1β function antagonize the activity of an IL-1 receptor. The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding human IL-1α or IL-1β. Moreover, the inhibitory polynucleotide or polypeptide composition binds to one or more region(s) of IL-1α or IL-1β comprised by SEQ ID NO: 1 and SEQ ID NO: 2 (IL-1α) or SEQ ID NO: 3 and SEQ ID NO: 4 (IL-1β). The inhibitory polynucleotide or polypeptide composition binds to one or more fragments of IL-1α or IL-1β comprised by SEQ ID NO: 1 and SEQ ID NO: 2 (IL-1α) or SEQ ID NO: 3 and SEQ ID NO: 4 (IL-1β).
  • A fragment, in the case of these sequences and all others provided herein, is defined as a part of the whole that is less than the whole. Moreover, a fragment ranges in size from a single nucleotide or amino acid within a polynucleotide or polypeptide sequence to one fewer nucleotide or amino acid than the entire polynucleotide or polypeptide sequence. Finally, a fragment is defined as any portion of a complete polynucleotide or polypeptide sequence that is intermediate between the extremes defined above.
  • Human IL-1α is encoded by the following mRNA sequence (NCBI Accession No. NM000575 and SEQ ID NO: 1): (For all mRNA transcripts incorporated into the present application, the initiator methionine, encoded by the codon “atg,” is bolded and capitalized to delineate the start of the coding region.)
  • accaggcaacaccattgaaggctcatatgtaaaaatccatgccttcctttctcccaatctccattcccaa
    acttagccactggcttctggctgaggccttacgcatacctcccggggcttgcacacaccttcttctacag
    aagacacaccttgggcatatcctacagaagaccaggcttctctctggtccttggtagagggctactttac
    tgtaacagggccagggtggagagttctctcctgaagctccatcccctctataggaaatgtgttgacaata
    ttcagaagagtaagaggatcaagacttctttgtgctcaaataccactgttctcttctctaccctgcccta
    accaggagcttgtcaccccaaactctgaggtgatttatgccttaatcaagcaaacttccctcttcagaaa
    agatggctcattttccctcaaaagttgccaggagctgccaagtattctgccaattcaccctggagcacaa
    tcaacaaattcagccagaacacaactacagctactattagaactattattattaataaattcctctccaa
    atctagccccttgacttcggatttcacgatttctcccttcctcctagaaacttgataagtttcccgcgct
    tccctttttctaagactacatgtttgtcatcttataaagcaaaggggtgaataaatgaaccaaatcaata
    acttctggaatatctgcaaacaacaataatatcagctatgccatctttcactattttagccagtatcgag
    ttgaatgaacatagaaaaatacaaaactgaattcttccctgtaaattccccgttttgacgacgcacttgt
    agccacgtagccacgcctacttaagacaattacaaaaggcgaagaagactgactcaggcttaagctgcca
    gccagagagggagtcatttcattggcgtttgagtcagcaaagaagtcaagATGgccaaagttccagacat
    gtttgaagacctgaagaactgttacagtgaaaatgaagaagacagttcctccattgatcatctgtctctg
    aatcagaaatccttctatcatgtaagctatggcccactccatgaaggctgcatggatcaatctgtgtctc
    tgagtatctctgaaacctctaaaacatccaagcttaccttcaaggagagcatggtggtagtagcaaccaa
    cgggaaggttctgaagaagagacggttgagtttaagccaatccatcactgatgatgacctggaggccatc
    gccaatgactcagaggaagaaatcatcaagcctaggtcagcaccttttagcttcctgagcaatgtgaaat
    acaactttatgaggatcatcaaatacgaattcatcctgaatgacgccctcaatcaaagtataattcgagc
    caatgatcagtacctcacggctgctgcattacataatctggatgaagcagtgaaatttgacatgggtgct
    tataagtcatcaaaggatgatgctaaaattaccgtgattctaagaatctcaaaaactcaattgtatgtga
    ctgcccaagatgaagaccaaccagtgctgctgaaggagatgcctgagatacccaaaaccatcacaggtag
    tgagaccaacctcctcttcttctgggaaactcacggcactaagaactatttcacatcagttgcccatcca
    aacttgtttattgccacaaagcaagactactgggtgtgcttggcaggggggccaccctctatcactgact
    ttcagatactggaaaaccaggcgtaggtctggagtctcacttgtctcacttgtgcagtgttgacagttca
    tatgtaccatgtacatgaagaagctaaatcctttactgttagtcatttgctgagcatgtactgagccttg
    taattctaaatgaatgtttacactctttgtaagagtggaaccaacactaacatataatgttgttatttaa
    agaacaccctatattttgcatagtaccaatcattttaattattattcttcataacaattttaggaggacc
    agagctactgactatggctaccaaaaagactctacccatattacagatgggcaaattaaggcataagaaa
    actaagaaatatgcacaatagcagttgaaacaagaagccacagacctaggatttcatgatttcatttcaa
    ctgtttgccttctacttttaagttgctgatgaactcttaatcaaatagcataagtttctgggacctcagt
    tttatcattttcaaaatggagggaataatacctaagccttcctgccgcaacagttttttatgctaatcag
    ggaggtcattttggtaaaatacttcttgaagccgagcctcaagatgaaggcaaagcacgaaatgttattt
    tttaattattatttatatatgtatttataaatatatttaagataattataatatactatatttatgggaa
    ccccttcatcctctgagtgtgaccaggcatcctccacaatagcagacagtgttttctgggataagtaagt
    ttgatttcattaatacagggcattttggtccaagttgtgcttatcccatagccaggaaactctgcattct
    agtacttgggagacctgtaatcatataataaatgtacattaattaccttgagccagtaattggtccgatc
    tttgactcttttgccattaaacttacctgggcattcttgtttcaattccacctgcaatcaagtcctacaa
    gctaaaattagatgaactcaactttgacaaccatgagaccactgttatcaaaactttcttttctggaatg
    taatcaatgtttcttctaggttctaaaaattgtgatcagaccataatgttacattattatcaacaatagt
    gattgatagagtgttatcagtcataactaaataaagcttgcaacaaaattctctgacaaaaaaaaaaaaa
    aaa.
  • Human IL-1α is encoded by the following amino acid sequence (NCBI Accession No. NM000575 and SEQ ID NO: 2):
  • MAKVPDMFEDLKNCYSENEEDSSSIDHLSLNQKSFYHVSYGPLHDSEEEIIKPRSAPFSFL
    SNVKYNFMRIIKYEFILNDALNQSIIRANDQYLTAAALHNLDEAVKFDMGAYKSSKDDA
    KITVILRISKTQLYVTAQDEDQPVLLKEMPEIPKTITGSETNLLFFWETHGTKNYFTSVAH
    PNLFIATKQDYWVCLAGGPPSITDFQILENQA.
  • Human IL-1β is encoded by the following mRNA sequence (NCBI Accession No. NM000576 and SEQ ID NO: 3):
  • accaaacctcttcgaggcacaaggcacaacaggctgctctgggattctcttcagccaatcttcattgctc
    aagtgtctgaagcagccATGgcagaagtacctgagctcgccagtgaaatgatggcttattacagtggcaa
    tgaggatgacttgttctttgaagctgatggccctaaacagatgaagtgctccttccaggacctggacctc
    tgccctctggatggcggcatccagctacgaatctccgaccaccactacagcaagggcttcaggcaggccg
    cgtcagttgttgtggccatggacaagctgaggaagatgctggttccctgcccacagaccttccaggagaa
    tgacctgagcaccttctttcccttcatctttgaagaagaacctatcttcttcgacacatgggataacgag
    gcttatgtgcacgatgcacctgtacgatcactgaactgcacgctccgggactcacagcaaaaaagcttgg
    tgatgtctggtccatatgaactgaaagctctccacctccagggacaggatatggagcaacaagtggtgtt
    ctccatgtcctttgtacaaggagaagaaagtaatgacaaaatacctgtggccttgggcctcaaggaaaag
    aatctgtacctgtcctgcgtgttgaaagatgataagcccactctacagctggagagtgtagatcccaaaa
    attacccaaagaagaagatggaaaagcgatttgtcttcaacaagatagaaatcaataacaagctggaatt
    tgagtctgcccagttccccaactggtacatcagcacctctcaagcagaaaacatgcccgtcttcctggga
    gggaccaaaggcggccaggatataactgacttcaccatgcaatttgtgtcttcctaaagagagctgtacc
    cagagagtcctgtgctgaatgtggactcaatccctagggctggcagaaagggaacagaaaggtttttgag
    tacggctatagcctggactttcctgttgtctacaccaatgcccaactgcctgccttagggtagtgctaag
    aggatctcctgtccatcagccaggacagtcagctctctcctttcagggccaatccccagcccttttgttg
    agccaggcctctctcacctctcctactcacttaaagcccgcctgacagaaaccacggccacatttggttc
    taagaaaccctctgtcattcgctcccacattctgatgagcaaccgcttccctatttatttatttatttgt
    ttgtttgttttattcattggtctaatttattcaaagggggcaagaagtagcagtgtctgtaaaagagcct
    agtttttaatagctatggaatcaattcaatttggactggtgtgctctctttaaatcaagtcctttaatta
    agactgaaaatatataagctcagattatttaaatgggaatatttataaatgagcaaatatcatactgttc
    aatggttctgaaataaacttcactgaag.
  • Human IL-1β is encoded by the following amino acid sequence (NCBI Accession No. NM000576 and SEQ ID NO: 4):
  • MAEVPELASEMMAYYSGNEDDLFFEADGPKQMKCSFQDLDLCPLDGGIQLRISDHHYS
    KGFRQAASVVVAMDKLRKMLVPCPQTFQENDLSTFFPFIFEEEPIFFDTWDNEAYVHDA
    PVRSLNCTLRDSQQKSLVMSGPYELKALHLQGQDMEQQVVFSMSFVQGEESNDKIPVA
    LGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEKRFVFNKIEINNKLEFESAQFP
    NWYISTSQAENMPVFLGGTKGGQDITDFTMQFVSS.
  • Interleukin-1 Receptor (Type 1) Antagonist (IL-1Ra):
  • IL-1Ra is an endogenous receptor antagonist which is primarily produced by activated monocytes and tissue macrophages, inhibits the activities of the proinflammatory forms of IL-1 by competitively binding to IL-1 receptor. (Gabay, C. et al. 1997. 159: 5905-5913). IL-1Ra is an inducible gene that is typically upregulated in inflammatory conditions, such as rheumatoid arthritis (Arend, W. P. 1993. Adv Immunol. 54: 167-223).
  • Normal tear fluid has been found to contain high concentrations of IL-1Ra in concentrations 25,000 and 40,000 times greater than both proinflammatory forms of IL-1 (Solomon, A. et al. 2001. Invest Opthalmol Vis Sci. 42: 2283-2292). The high concentration of IL-1Ra in the tear fluid may be a natural homeostatic mechanism for preventing inappropriate activation of IL-1-mediated inflammatory events on the ocular surface (Dinarello, C. A. 1996. Blood. 15:2095-2147).
  • An increased concentration of IL-1Ra has been detected in the tear fluid of patients with dry eye and in the conjunctival epithelium of patients with dry eye syndrome (Solomon, A. et al. 2001. Invest Opthalmol Vis Sci. 42: 2283-2292). Despite this increased level of expression, the ratio of IL-1Ra to proinflammatory forms IL-1 dry-eye was significantly lower than in normal subjects. Reports of placebo-controlled clinical trials in which IL-1Ra was administered to patients with rheumatoid arthritis have noted that increasing the ratio of IL-1Ra to the proinflammatory forms significantly improves clinical symptoms (Dinarello, C. A. 2000. N Engl J Med. 343:732-734).
  • In the present invention, compositions comprise one or more regions of IL- 1Ra transcripts 1, 2, 3, or 4, intacellular IL-1Ra (icIL-1Ra), or their corresponding polypeptide isoforms. Alternatively, compositions comprise the entirety of IL- 1Ra transcripts 1, 2, 3, or 4, intacellular IL-1Ra (icIL-1Ra), or their corresponding polypeptide isoforms. Compositions comprising any form of human IL-1Ra, or fragments thereof, inhibit the function of IL-1R1. These polynucleotides and polypeptides are defined by the following sequences.
  • Human IL-1Ra, transcript 1, is encoded by the following mRNA sequence (NCBI Accession No. NM173842 and SEQ ID NO: 5):
  • atttctttataaaccacaactctgggcccgcaatggcagtccactgccttgctgcagtcacagaATGgaa
    atctgcagaggcctccgcagtcacctaatcactctcctcctcttcctgttccattcagagacgatctgcc
    gaccctctgggagaaaatccagcaagatgcaagccttcagaatctgggatgttaaccagaagaccttcta
    tctgaggaacaaccaactagttgctggatacttgcaaggaccaaatgtcaatttagaagaaaagatagat
    gtggtacccattgagcctcatgctctgttcttgggaatccatggagggaagatgtgcctgtcctgtgtca
    agtctggtgatgagaccagactccagctggaggcagttaacatcactgacctgagcgagaacagaaagca
    ggacaagcgcttcgccttcatccgctcagacagcggccccaccaccagttttgagtctgccgcctgcccc
    ggttggttcctctgcacagcgatggaagctgaccagcccgtcagcctcaccaatatgcctgacgaaggcg
    tcatggtcaccaaattctacttccaggaggacgagtagtactgcccaggcctgcctgttcccattcttgc
    atggcaaggactgcagggactgccagtccccctgccccagggctcccggctatgggggcactgaggacca
    gccattgaggggtggaccctcagaaggcgtcacaagaacctggtcacaggactctgcctcctcttcaact
    gaccagcctccatgctgcctccagaatggtctttctaatgtgtgaatcagagcacagcagcccctgcaca
    aagcccttccatgtcgcctctgcattcaggatcaaaccccgaccacctgcccaacctgctctcctcttgc
    cactgcctcttcctccctcattccaccttcccatgccctggatccatcaggccacttgatgacccccaac
    caagtggctcccacaccctgttttacaaaaaagaaaagaccagtccatgagggaggtttttaagggtttg
    tggaaaatgaaaattaggatttcatgatttttttttttcagtccccgtgaaggagagcccttcatttgga
    gattatgttctttcggggagaggctgaggacttaaaatattcctgcatttgtgaaatgatggtgaaagta
    agtggtagcttttcccttctttttcttctttttttgtgatgtcccaacttgtaaaaattaaaagttatgg
    tactatgttagccccataattttttttttccttttaaaacacttccataatctggactcctctgtccagg
    cactgctgcccagcctccaagctccatctccactccagattttttacagctgcctgcagtactttacctc
    ctatcagaagtttctcagctcccaaggctctgagcaaatgtggctcctgggggttctttcttcctctgct
    gaaggaataaattgctccttgacattgtagagcttctggcacttggagacttgtatgaaagatggctgtg
    cctctgcctgtctcccccaccgggctgggagctctgcagagcaggaaacatgactcgtatatgtctcagg
    tccctgcagggccaagcacctagcctcgctcttggcaggtactcagcgaatgaatgctgtatatgttggg
    tgcaaagttccctacttcctgtgacttcagctctgttttacaataaaatcttgaaaatgcctaaaaaaaa
    aaaaaaaaaa.
  • Human IL-1Ra, transcript 1, is encoded by the following amino acid sequence (NCBI Accession No. NM173842 and SEQ ID NO: 6):
  • MEICRGLRSHLITLLLFLFHSETICRPSGRKSSKMQAFRIWDVNQKTFYLRNNQLVAGYL
    QGPNVNLEEKIDVVPIEPHALFLGIHGGKMCLSCVKSGDETRLQLEAVNITDLSENRKQD
    KRFAFIRSDSGPTTSFESAACPGWFLCTAMEADQPVSLTNMPDEGVMVTKFYFQEDE.
  • Human IL-1Ra, transcript 2, is encoded by the following mRNA sequence (NCBI Accession No. NM173841 and SEQ ID NO: 7):
  • gggcagctccaccctgggagggactgtggcccaggtactgcccgggtgctactttatgggcagcagctca
    gttgagttagagtctggaagacctcagaagacctcctgtcctatgaggccctccccATGgctttagctga
    cttgtatgaagaaggaggtggaggaggaggagaaggtgaagacaatgctgactcaaaggagacgatctgc
    cgaccctctgggagaaaatccagcaagatgcaagccttcagaatctgggatgttaaccagaagaccttct
    atctgaggaacaaccaactagttgctggatacttgcaaggaccaaatgtcaatttagaagaaaagataga
    tgtggtacccattgagcctcatgctctgttcttgggaatccatggagggaagatgtgcctgtcctgtgtc
    aagtctggtgatgagaccagactccagctggaggcagttaacatcactgacctgagcgagaacagaaagc
    aggacaagcgcttcgccttcatccgctcagacagcggccccaccaccagttttgagtctgccgcctgccc
    cggttggttcctctgcacagcgatggaagctgaccagcccgtcagcctcaccaatatgcctgacgaaggc
    gtcatggtcaccaaattctacttccaggaggacgagtagtactgcccaggcctgcctgttcccattcttg
    catggcaaggactgcagggactgccagtccccctgccccagggctcccggctatgggggcactgaggacc
    agccattgaggggtggaccctcagaaggcgtcacaagaacctggtcacaggactctgcctcctcttcaac
    tgaccagcctccatgctgcctccagaatggtctttctaatgtgtgaatcagagcacagcagcccctgcac
    aaagcccttccatgtcgcctctgcattcaggatcaaaccccgaccacctgcccaacctgctctcctcttg
    ccactgcctcttcctccctcattccaccttcccatgccctggatccatcaggccacttgatgacccccaa
    ccaagtggctcccacaccctgttttacaaaaaagaaaagaccagtccatgagggaggtttttaagggttt
    gtggaaaatgaaaattaggatttcatgatttttttttttcagtccccgtgaaggagagcccttcatttgg
    agattatgttctttcggggagaggctgaggacttaaaatattcctgcatttgtgaaatgatggtgaaagt
    aagtggtagcttttcccttctttttcttctttttttgtgatgtcccaacttgtaaaaattaaaagttatg
    gtactatgttagccccataattttttttttccttttaaaacacttccataatctggactcctctgtccag
    gcactgctgcccagcctccaagctccatctccactccagattttttacagctgcctgcagtactttacct
    cctatcagaagtttctcagctcccaaggctctgagcaaatgtggctcctgggggttctttcttcctctgc
    tgaaggaataaattgctccttgacattgtagagcttctggcacttggagacttgtatgaaagatggctgt
    gcctctgcctgtctcccccaccgggctgggagctctgcagagcaggaaacatgactcgtatatgtctcag
    gtccctgcagggccaagcacctagcctcgctcttggcaggtactcagcgaatgaatgctgtatatgttgg
    gtgcaaagttccctacttcctgtgacttcagctctgttttacaataaaatcttgaaaatgcctaaaaaaa
    aaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaa.
  • Human IL-1Ra, transcript 2, is encoded by the following amino acid sequence (NCBI Accession No. NM173841 and SEQ ID NO: 8):
  • MALADLYEEGGGGGGEGEDNADSKETICRPSGRKSSKMQAFRIWDVNQKTFYLRNNQ
    LVAGYLQGPNVNLEEKIDVVPIEPHALFLGIHGGKMCLSCVKSGDETRLQLEAVNITDLS
    ENRKQDKRFAFIRSDSGPTTSFESAACPGWFLCTAMEADQPVSLTNMPDEGVMVTKFY
    FQEDE.
  • Human IL-1Ra, transcript 3, is encoded by the following mRNA sequence (NCBI Accession No. NM000577 and SEQ ID NO: 9):
  • gggcagctccaccctgggagggactgtggcccaggtactgcccgggtgctactttatgggcagcagctca
    gttgagttagagtctggaagacctcagaagacctcctgtcctatgaggccctccccATGgctttagagac
    gatctgccgaccctctgggagaaaatccagcaagatgcaagccttcagaatctgggatgttaaccagaag
    accttctatctgaggaacaaccaactagttgctggatacttgcaaggaccaaatgtcaatttagaagaaa
    agatagatgtggtacccattgagcctcatgctctgttcttgggaatccatggagggaagatgtgcctgtc
    ctgtgtcaagtctggtgatgagaccagactccagctggaggcagttaacatcactgacctgagcgagaac
    agaaagcaggacaagcgcttcgccttcatccgctcagacagcggccccaccaccagttttgagtctgccg
    cctgccccggttggttcctctgcacagcgatggaagctgaccagcccgtcagcctcaccaatatgcctga
    cgaaggcgtcatggtcaccaaattctacttccaggaggacgagtagtactgcccaggcctgcctgttccc
    attcttgcatggcaaggactgcagggactgccagtccccctgccccagggctcccggctatgggggcact
    gaggaccagccattgaggggtggaccctcagaaggcgtcacaagaacctggtcacaggactctgcctcct
    cttcaactgaccagcctccatgctgcctccagaatggtctttctaatgtgtgaatcagagcacagcagcc
    cctgcacaaagcccttccatgtcgcctctgcattcaggatcaaaccccgaccacctgcccaacctgctct
    cctcttgccactgcctcttcctccctcattccaccttcccatgccctggatccatcaggccacttgatga
    cccccaaccaagtggctcccacaccctgttttacaaaaaagaaaagaccagtccatgagggaggttttta
    agggtttgtggaaaatgaaaattaggatttcatgatttttttttttcagtccccgtgaaggagagccctt
    catttggagattatgttctttcggggagaggctgaggacttaaaatattcctgcatttgtgaaatgatgg
    tgaaagtaagtggtagcttttcccttctttttcttctttttttgtgatgtcccaacttgtaaaaattaaa
    agttatggtactatgttagccccataattttttttttccttttaaaacacttccataatctggactcctc
    tgtccaggcactgctgcccagcctccaagctccatctccactccagattttttacagctgcctgcagtac
    tttacctcctatcagaagtttctcagctcccaaggctctgagcaaatgtggctcctgggggttctttctt
    cctctgctgaaggaataaattgctccttgacattgtagagcttctggcacttggagacttgtatgaaaga
    tggctgtgcctctgcctgtctcccccaccgggctgggagctctgcagagcaggaaacatgactcgtatat
    gtctcaggtccctgcagggccaagcacctagcctcgctcttggcaggtactcagcgaatgaatgctgtat
    atgttgggtgcaaagttccctacttcctgtgacttcagctctgttttacaataaaatcttgaaaatgcct
    aaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaa.
  • Human IL-1Ra, transcript 3, is encoded by the following amino acid sequence (NCBI Accession No. NM000577 and SEQ ID NO: 10):
  • MALETICRPSGRKSSKMQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVVPIE
    PHALFLGIHGGKMCLSCVKSGDETRLQLEAVNITDLSENRKQDKRFAFIRSDSGPTTSFE
    SAACPGWFLCTAMEADQPVSLTNMPDEGVMVTKFYFQEDE.
  • Human IL-1Ra, transcript 4, is encoded by the following mRNA sequence (NCBI Accession No. NM173843 and SEQ ID NO: 11):
  • gggcagctccaccctgggagggactgtggcccaggtactgcccgggtgctactttatgggcagcagctca
    gttgagttagagtctggaagacctcagaagacctcctgtcctatgaggccctccccatggctttaggggg
    attataaaactaatcatcaaagccaagaaggcaagagcaagcatgtaccgctgaaaacacaagataactg
    cataagtaatgactttcagtgcagattcatagctaacccataaactgctggggcaaaaatcatcttggaa
    ggctctgaacctcagaaaggattcacaagacgatctgccgaccctctgggagaaaatccagcaagATGca
    agccttcagaatctgggatgttaaccagaagaccttctatctgaggaacaaccaactagttgctggatac
    ttgcaaggaccaaatgtcaatttagaagaaaagatagatgtggtacccattgagcctcatgctctgttct
    tgggaatccatggagggaagatgtgcctgtcctgtgtcaagtctggtgatgagaccagactccagctgga
    ggcagttaacatcactgacctgagcgagaacagaaagcaggacaagcgcttcgccttcatccgctcagac
    agcggccccaccaccagttttgagtctgccgcctgccccggttggttcctctgcacagcgatggaagctg
    accagcccgtcagcctcaccaatatgcctgacgaaggcgtcatggtcaccaaattctacttccaggagga
    cgagtagtactgcccaggcctgcctgttcccattcttgcatggcaaggactgcagggactgccagtcccc
    ctgccccagggctcccggctatgggggcactgaggaccagccattgaggggtggaccctcagaaggcgtc
    acaagaacctggtcacaggactctgcctcctcttcaactgaccagcctccatgctgcctccagaatggtc
    tttctaatgtgtgaatcagagcacagcagcccctgcacaaagcccttccatgtcgcctctgcattcagga
    tcaaaccccgaccacctgcccaacctgctctcctcttgccactgcctcttcctccctcattccaccttcc
    catgccctggatccatcaggccacttgatgacccccaaccaagtggctcccacaccctgttttacaaaaa
    agaaaagaccagtccatgagggaggtttttaagggtttgtggaaaatgaaaattaggatttcatgatttt
    tttttttcagtccccgtgaaggagagcccttcatttggagattatgttctttcggggagaggctgaggac
    ttaaaatattcctgcatttgtgaaatgatggtgaaagtaagtggtagcttttcccttctttttcttcttt
    ttttgtgatgtcccaacttgtaaaaattaaaagttatggtactatgttagccccataattttttttttcc
    ttttaaaacacttccataatctggactcctctgtccaggcactgctgcccagcctccaagctccatctcc
    actccagattttttacagctgcctgcagtactttacctcctatcagaagtttctcagctcccaaggctct
    gagcaaatgtggctcctgggggttctttcttcctctgctgaaggaataaattgctccttgacattgtaga
    gcttctggcacttggagacttgtatgaaagatggctgtgcctctgcctgtctcccccaccgggctgggag
    ctctgcagagcaggaaacatgactcgtatatgtctcaggtccctgcagggccaagcacctagcctcgctc
    ttggcaggtactcagcgaatgaatgctgtatatgttgggtgcaaagttccctacttcctgtgacttcagc
    tctgttttacaataaaatcttgaaaatgcctaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaaa
    aaaaaaaaaaaaa.
  • Human IL-1Ra, transcript 4, is encoded by the following amino acid sequence (NCBI Accession No. NM173843 and SEQ ID NO: 12):
  • MQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVVPIEPHALFLGIHGGKMCL
    SCVKSGDETRLQLEAVNITDLSENRKQDKRFAFIRSDSGPTTSFESAACPGWFLCTAMEA
    DQPVSLTNMPDEGVMVTKFYFQEDE.
  • Human intracellular IL-1Ra, icIL-1Ra, is encoded by the following mRNA sequence (NCBI Accession No. M55646 and SEQ ID NO: 13):
  • agctccaccctgggagggactgtggcccaggtactgcccgggtgctactttatgggcagcagctcagttg
    agttagagtctggaagacctcagaagacctcctgtcctatgaggccctccccATGgctttagagacgatc
    tgccgaccctctgggagaaaatccagcaagatgcaagccttcagaatctgggatgttaaccagaagacct
    tctatctgaggaacaaccaactagttgctggatacttgcaaggaccaaatgtcaatttagaagaaaagat
    agatgtggtacccattgagcctcatgctctgttcttgggaatccatggagggaagatgtgcctgtcctgt
    gtcaagtctggtgatgagaccagactccagctggaggcagttaacatcactgacctgagcgagaacagaa
    agcaggacaagcgcttcgccttcatccgctcagacagtggccccaccaccagttttgagtctgccgcctg
    ccccggttggttcctctgcacagcgatggaagctgaccagcccgtcagcctcaccaatatgcctgacgaa
    ggcgtcatggtcaccaaattctacttccaggaggacgagtag.
  • Human intracellular IL-1Ra, icIL-1Ra, is encoded by the following amino acid sequence (NCBI Accession No. M55646 and SEQ ID NO: 14):
  • MALETICRPSGRKSSKMQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVVPIE
    PHALFLGIHGGKMCLSCVKSGDETRLQLEAVNITDLSENRKQDKRFAFIRSDSGPTTSFE
    SAACPGWFLCTAMEADQPVSLTNMPDEGVMVTKFYFQEDE.
  • Human Recombinant IL-1Ra:
  • A recombinant form of human IL-1Ra (rHuIL-1Ra) was developed and tested in animal models for arthritis. This form of rHuIL-1Ra is also known as Anakinra or Kineret® differs from the native nonglycosylated IL-1Ra by the addition of an N-terminal methionine. It binds to IL-1R type I with the same affinity as IL-113. Kineret® consists of 153 amino acids and has a molecular weight of 17.3 kilodaltons. It is produced by recombinant DNA technology using an E. coli bacterial expression system.
  • Anakinra has been investigated in several conditions considered mediated at least in part via IL-1. Some evidence suggests involvement of IL-1 in the pathogenesis of rheumatoid arthritis and septic shock (Jiang, Y. et al. 2000. Arthritis Rheum. 43:1001-1009; Fisher, C. J. et al. 1994. JAMA. 271:1836-1843; Okusawa, S. et al. 1988. J Clin Invest. 81:1162-1172; Bresnihan, B. et al. 1998. Rheum Dis Clin North Am. 24(3):615-628; Dayer, J. M. et al. 2001. Curr Opin Rheumatol. 13:170-176; Edwards, C. K. 2001. J Clin Rheumatol. 7:S17-S24). Anakinra has been approved by the FDA for the reduction in signs and symptoms of moderately to severely active rheumatoid arthritis in patients 18 years of age or older who have failed one or more disease-modifying antirheumatic drugs. Considering its high safety profile, administration of Anakinra has also been used in the treatment of arthritis in patients with Juvenile rheumatoid arthritis (Reiff, A. 2005. Curr Rheumatol Rep. 7:434-40). Other indications include prevention of graft-versus-host disease (GVHD) after bone marrow transplantation (Antin, J. H. et al. 1994. Blood. 84:1342-8), uveitis (Teoh, S. C. et al. 2007. Br J Opthalmol. 91: 263-4) osteoarthritis (Caron, J. P. et al. 1996. Arthritis Rheum. 39:1535-44), asthma, inflammatory bowel disease, acute pancreatitis (Hynninen, M. et al. 1999. J Crit Care. 14:63-8), psoriasis, and type II diabetes mellitus (Larsen, C. M. et al. 2007. N Engl J. Med. 356:1517-26). The systemic safety profile of IL-1Ra is extremely favorable, especially in comparison to other immunosuppressive treatments such as TNF-α blockers, cytotoxic agents, or even steroids.
  • Topical human recombinant IL-1Ra has been successfully used for prevention of corneal transplant rejection (Yamada, J. et al. 2000. Invest Opthalmol Vis Sci. 41:4203-8) and allergic conjunctivitis (Keane-Myers, A. M. et al. 1999. Invest Opthalmol Vis Sci. 40:3041-6) in experimental animal models. Similarly, using topical IL-1Ra significantly decreases corneal inflammation and leads to enhanced corneal transparency in the rat model of corneal alkali injury (Yamada, J. et al. 2003. Exp Eye Res. 76:161-7).
  • A recombinant form of human IL-1Ra (rHuIL-1Ra) was developed and approved for use in humans by subcutaneous injection for the treatment of arthritis. This form of rHuIL-1Ra, also known as Anakinra or Kineret® (Amgen Inc.), differs from the native nonglycosylated IL-1Ra by the addition of an N-terminal methionine. It binds to human IL-1R, type 1, (IL-1R1) with the same affinity as IL-1β. Kineret® consists of 153 amino acids (see SEQ ID NO: 16) and has a molecular weight of 17.3 kilodaltons. It is produced by recombinant DNA technology using an E. coli bacterial expression system. This composition comprises one or more regions of SEQ ID NO: 15 or SEQ ID NO: 16. Furthermore, this composition comprises the entire sequence of either SEQ ID NO: 15 or SEQ ID NO: 16.
  • Anakinra/Kineret® is encoded by the following mRNA sequence (NCBI Accession No. M55646 and SEQ ID NO: 15):
  • agctccaccctgggagggactgtggcccaggtactgcccgggtgctactttatgggcagcagctcagttg
    agttagagtctggaagacctcagaagacctcctgtcctatgaggccctccccATGgctttagagacgatc
    tgccgaccctctgggagaaaatccagcaagatgcaagccttcagaatctgggatgttaaccagaagacct
    tctatctgaggaacaaccaactagttgctggatacttgcaaggaccaaatgtcaatttagaagaaaagat
    agatgtggtacccattgagcctcatgctctgttcttgggaatccatggagggaagatgtgcctgtcctgt
    gtcaagtctggtgatgagaccagactccagctggaggcagttaacatcactgacctgagcgagaacagaa
    agcaggacaagcgcttcgccttcatccgctcagacagtggccccaccaccagttttgagtctgccgcctg
    ccccggttggttcctctgcacagcgatggaagctgaccagcccgtcagcctcaccaatatgcctgacgaa
    ggcgtcatggtcaccaaattctacttccaggaggacgagtag.
  • Anakinra/Kineret® is encoded by the following polypeptide sequence (DrugBank Accession No. BTD00060 and SEQ ID NO: 16):
  • MRPSGRKSSKMQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVVPIEPHALF
    LGIHGGKMCLSCVKSGDETRLQLEAVNITDLSENRKQDKRFAFIRSDSGPTTSFESAACP
    GWFLCTAMEADQPVSLTNMPDEGVMVTKFYFQEDE
  • IL-1 Receptors:
  • The composition of the present invention comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding the IL-1 receptor, either type 1 or 2. In the present application the IL-1 Receptor, type 1 (IL-1R1), is defined by the polynucleotide sequence of SEQ ID NO: 17 or the polypeptide sequence of SEQ ID NO: 18. In the present application the IL-1 Receptor, type 2 (IL-1R2), transcript variants 1 and 2, are defined by the polynucleotide sequences of SEQ ID NO: 19 and 20, or the polypeptide sequence of SEQ ID NO: 21. IL-1R2 can function as a “decoy” receptor which binds IL-1 cytokines and inhibits IL-1R1. Polynucleotide or polypeptide compositions bind to one or more region(s) of IL-1R1 or IL-1R2, and associated isoforms, comprised by SEQ ID NO: 17-21.
  • IL-1R1 is encoded by the following mRNA sequence (NCBI Accession No. NM000877 and SEQ ID NO: 17):
  • tagacgcaccctctgaagatggtgactccctcctgagaagctggaccccttggtaaaagacaaggccttc
    tccaagaagaatATGaaagtgttactcagacttatttgtttcatagctctactgatttcttctctggagg
    ctgataaatgcaaggaacgtgaagaaaaaataattttagtgtcatctgcaaatgaaattgatgttcgtcc
    ctgtcctcttaacccaaatgaacacaaaggcactataacttggtataaagatgacagcaagacacctgta
    tctacagaacaagcctccaggattcatcaacacaaagagaaactttggtttgttcctgctaaggtggagg
    attcaggacattactattgcgtggtaagaaattcatcttactgcctcagaattaaaataagtgcaaaatt
    tgtggagaatgagcctaacttatgttataatgcacaagccatatttaagcagaaactacccgttgcagga
    gacggaggacttgtgtgcccttatatggagttttttaaaaatgaaaataatgagttacctaaattacagt
    ggtataaggattgcaaacctctacttcttgacaatatacactttagtggagtcaaagataggctcatcgt
    gatgaatgtggctgaaaagcatagagggaactatacttgtcatgcatcctacacatacttgggcaagcaa
    tatcctattacccgggtaatagaatttattactctagaggaaaacaaacccacaaggcctgtgattgtga
    gcccagctaatgagacaatggaagtagacttgggatcccagatacaattgatctgtaatgtcaccggcca
    gttgagtgacattgcttactggaagtggaatgggtcagtaattgatgaagatgacccagtgctaggggaa
    gactattacagtgtggaaaatcctgcaaacaaaagaaggagtaccctcatcacagtgcttaatatatcgg
    aaattgaaagtagattttataaacatccatttacctgttttgccaagaatacacatggtatagatgcagc
    atatatccagttaatatatccagtcactaatttccagaagcacatgattggtatatgtgtcacgttgaca
    gtcataattgtgtgttctgttttcatctataaaatcttcaagattgacattgtgctttggtacagggatt
    cctgctatgattttctcccaataaaagcttcagatggaaagacctatgacgcatatatactgtatccaaa
    gactgttggggaagggtctacctctgactgtgatatttttgtgtttaaagtcttgcctgaggtcttggaa
    aaacagtgtggatataagctgttcatttatggaagggatgactacgttggggaagacattgttgaggtca
    ttaatgaaaacgtaaagaaaagcagaagactgattatcattttagtcagagaaacatcaggcttcagctg
    gctgggtggttcatctgaagagcaaatagccatgtataatgctcttgttcaggatggaattaaagttgtc
    ctgcttgagctggagaaaatccaagactatgagaaaatgccagaatcgattaaattcattaagcagaaac
    atggggctatccgctggtcaggggactttacacagggaccacagtctgcaaagacaaggttctggaagaa
    tgtcaggtaccacatgccagtccagcgacggtcaccttcatctaaacaccagttactgtcaccagccact
    aaggagaaactgcaaagagaggctcacgtgcctctcgggtagcatggagaagttgccaagagttctttag
    gtgcctcctgtcttatggcgttgcaggccaggttatgcctcatgctgacttgcagagttcatggaatgta
    actatatcatcctttatccctgaggtcacctggaatcagattattaagggaataagccatgacgtcaata
    gcagcccagggcacttcagagtagagggcttgggaagatcttttaaaaaggcagtaggcccggtgtggtg
    gctcacgcctataatcccagcactttgggaggctgaagtgggtggatcaccagaggtcaggagttcgaga
    ccagcccagccaacatggcaaaaccccatctctactaaaaatacaaaaatgagctaggcatggtggcaca
    cgcctgtaatcccagctacacctgaggctgaggcaggagaattgcttgaaccggggagacggaggttgca
    gtgagccgagtttgggccactgcactctagcctggcaacagagcaagactccgtctcaaaaaaagggcaa
    taaatgccctctctgaatgtttgaactgccaagaaaaggcatggagacagcgaactagaagaaagggcaa
    gaaggaaatagccaccgtctacagatggcttagttaagtcatccacagcccaagggcggggctatgcctt
    gtctggggaccctgtagagtcactgaccctggagcggctctcctgagaggtgctgcaggcaaagtgagac
    tgacacctcactgaggaagggagacatattcttggagaactttccatctgcttgtattttccatacacat
    ccccagccagaagttagtgtccgaagaccgaattttattttacagagcttgaaaactcacttcaatgaac
    aaagggattctccaggattccaaagttttgaagtcatcttagctttccacaggagggagagaacttaaaa
    aagcaacagtagcagggaattgatccacttcttaatgctttcctccctggcatgaccatcctgtcctttg
    ttattatcctgcattttacgtctttggaggaacagctccctagtggcttcctccgtctgcaatgtccctt
    gcacagcccacacatgaaccatccttcccatgatgccgctcttctgtcatcccgctcctgctgaaacacc
    tcccaggggctccacctgttcaggagctgaagcccatgctttcccaccagcatgtcactcccagaccacc
    tccctgccctgtcctccagcttcccctcgctgtcctgctgtgtgaattcccaggttggcctggtggccat
    gtcgcctgcccccagcactcctctgtctctgctcttgcctcgacccttcctcctcctttgcctaggaggc
    cttctcgcattttctctagctgatcagaattttaccaaaattcagaacatcctccaattccacagtctct
    gggagactttccctaagaggcgacttcctctccagccttctctctctggtcaggcccactgcagagatgg
    tggtgagcacatctgggaggctggtctccctccagctggaattgctgctctctgagggagaggctgtggt
    ggctgtctctgtccctcactgccttccaggagcaatttgcacatgtaacatagatttatgtaatgcttta
    tgtttaaaaacattccccaattatcttatttaatttttgcaattattctaattttatatatagagaaagt
    gacctattttttaaaaaaatcacactctaagttctattgaacctaggacttgagcctccatttctggctt
    ctagtctggtgttctgagtacttgatttcaggtcaataacggtcccccctcactccacactggcacgttt
    gtgagaagaaatgacattttgctaggaagtgaccgagtctaggaatgcttttattcaagacaccaaattc
    caaacttctaaatgttggaattttcaaaaattgtgtttagattttatgaaaaactcttctactttcatct
    attctttccctagaggcaaacatttcttaaaatgtttcattttcattaaaaatgaaagccaaatttatat
    gccaccgattgcaggacacaagcacagttttaagagttgtatgaacatggagaggacttttggtttttat
    atttctcgtatttaatatgggtgaacaccaacttttatttggaataataattttcctcctaaacaaaaac
    acattgagtttaagtctctgactcttgcctttccacctgctttctcctgggcccgctttgcctgcttgaa
    ggaacagtgctgttctggagctgctgttccaacagacagggcctagctttcatttgacacacagactaca
    gccagaagcccatggagcagggatgtcacgtcttgaaaagcctattagatgttttacaaatttaattttg
    cagattattttagtctgtcatccagaaaatgtgtcagcatgcatagtgctaagaaagcaagccaatttgg
    aaacttaggttagtgacaaaattggccagagagtgggggtgatgatgaccaagaattacaagtagaatgg
    cagctggaatttaaggagggacaagaatcaatggataagcgtgggtggaggaagatccaaacagaaaagt
    gcaaagttattccccatcttccaagggttgaattctggaggaagaagacacattcctagttccccgtgaa
    cttcctttgacttattgtccccactaaaacaaaacaaaaaacttttaatgccttccacattaattagatt
    ttcttgcagtttttttatggcatttttttaaagatgccctaagtgttgaagaagagtttgcaaatgcaac
    aaaatatttaattaccggttgttaaaactggtttagcacaatttatattttccctctcttgcctttctta
    tttgcaataaaaggtattgagccattttttaaatgacatttttgataaattatgtttgtactagttgatg
    aaggagttttttttaacctgtttatataattttgcagcagaagccaaattttttgtatattaaagcacca
    aattcatgtacagcatgcatcacggatcaatagactgtacttattttccaataaaattttcaaactttgt
    actgttaaa.
  • IL-1R1 is encoded by the following amino acid sequence (NCBI Accession No. NM000877 and SEQ ID NO: 18):
  • MKVLLRLICFIALLISSLEADKCKEREEKIILVSSANEIDVRPCPLNPNEHKGTITWYKDDS
    KTPVSTEQASRIHQHKEKLWFVPAKVEDSGHYYCVVRNSSYCLRIKISAKFVENEPNLC
    YNAQAIFKQKLPVAGDGGLVCPYMEFFKNENNELPKLQWYKDCKPLLLDNIHFSGVKD
    RLIVMNVAEKHRGNYTCHASYTYLGKQYPITRVIEFITLEENKPTRPVIVSPANETMEVD
    LGSQIQLICNVTGQLSDIAYWKWNGSVIDEDDPVLGEDYYSVENPANKRRSTLITVLNIS
    EIESRFYKHPFTCFAKNTHGIDAAYIQLIYPVTNFQKHMIGICVTLTVIIVCSVFIYKIFKIDI
    VLWYRDSCYDFLPIKASDGKTYDAYILYPKTVGEGSTSDCDIFVFKVLPEVLEKQCGYK
    LFIYGRDDYVGEDIVEVINENVKKSRRLIIILVRETSGFSWLGGSSEEQIAMYNALVQDGI
    KVVLLELEKIQDYEKMPESIKFIKQKHGAIRWSGDFTQGPQSAKTRFWKNVRYHMPVQ
    RRSPSSKHQLLSPATKEKLQREAHVPLG.
  • IL-1R2, transcript variant 1, is encoded by the following mRNA sequence (NCBI Accession No. NM004633 and SEQ ID NO: 19):
  • cccgtgaggaggaaaaggtgtgtccgctgccacccagtgtgagcgggtgacaccacccggttaggaaatc
    ccagctcccaagagggtataaatccctgctttactgctgagctcctgctggaggtgaaagtctggcctgg
    cagccttccccaggtgagcagcaacaaggccacgtgctgctgggtctcagtcctccacttcccgtgtcct
    ctggaagttgtcaggagcaATGttgcgcttgtacgtgttggtaatgggagtttctgccttcacccttcag
    cctgcggcacacacaggggctgccagaagctgccggtttcgtgggaggcattacaagcgggagttcaggc
    tggaaggggagcctgtagccctgaggtgcccccaggtgccctactggttgtgggcctctgtcagcccccg
    catcaacctgacatggcataaaaatgactctgctaggacggtcccaggagaagaagagacacggatgtgg
    gcccaggacggtgctctgtggcttctgccagccttgcaggaggactctggcacctacgtctgcactacta
    gaaatgcttcttactgtgacaaaatgtccattgagctcagagtttttgagaatacagatgctttcctgcc
    gttcatctcatacccgcaaattttaaccttgtcaacctctggggtattagtatgccctgacctgagtgaa
    ttcacccgtgacaaaactgacgtgaagattcaatggtacaaggattctcttcttttggataaagacaatg
    agaaatttctaagtgtgagggggaccactcacttactcgtacacgatgtggccctggaagatgctggcta
    ttaccgctgtgtcctgacatttgcccatgaaggccagcaatacaacatcactaggagtattgagctacgc
    atcaagaaaaaaaaagaagagaccattcctgtgatcatttcccccctcaagaccatatcagcttctctgg
    ggtcaagactgacaatcccgtgtaaggtgtttctgggaaccggcacacccttaaccaccatgctgtggtg
    gacggccaatgacacccacatagagagcgcctacccgggaggccgcgtgaccgaggggccacgccaggaa
    tattcagaaaataatgagaactacattgaagtgccattgatttttgatcctgtcacaagagaggatttgc
    acatggattttaaatgtgttgtccataataccctgagttttcagacactacgcaccacagtcaaggaagc
    ctcctccacgttctcctggggcattgtgctggccccactttcactggccttcttggttttggggggaata
    tggatgcacagacggtgcaaacacagaactggaaaagcagatggtctgactgtgctatggcctcatcatc
    aagactttcaatcctatcccaagtgaaataaatggaatgaaataattcaaacacaaaaaaaaaaaaaaaa
    aaaaaaaaaaaaaa.
  • IL-1R2, transcript variant 2, is encoded by the following mRNA sequence (NCBI Accession No. NM173343 and SEQ ID NO: 20):
  • gggatgggagatactgttgtggtcacctctggaaaatacattctgctactcttaaaaactagtgacgctc
    atacaaatcaacagaaagagcttctgaaggaagactttaaagctgcttctgccacgtgctgctgggtctc
    agtcctccacttcccgtgtcctctggaagttgtcaggagcaATGttgcgcttgtacgtgttggtaatggg
    agtttctgccttcacccttcagcctgcggcacacacaggggctgccagaagctgccggtttcgtgggagg
    cattacaagcgggagttcaggctggaaggggagcctgtagccctgaggtgcccccaggtgccctactggt
    tgtgggcctctgtcagcccccgcatcaacctgacatggcataaaaatgactctgctaggacggtcccagg
    agaagaagagacacggatgtgggcccaggacggtgctctgtggcttctgccagccttgcaggaggactct
    ggcacctacgtctgcactactagaaatgcttcttactgtgacaaaatgtccattgagctcagagtttttg
    agaatacagatgctttcctgccgttcatctcatacccgcaaattttaaccttgtcaacctctggggtatt
    agtatgccctgacctgagtgaattcacccgtgacaaaactgacgtgaagattcaatggtacaaggattct
    cttcttttggataaagacaatgagaaatttctaagtgtgagggggaccactcacttactcgtacacgatg
    tggccctggaagatgctggctattaccgctgtgtcctgacatttgcccatgaaggccagcaatacaacat
    cactaggagtattgagctacgcatcaagaaaaaaaaagaagagaccattcctgtgatcatttcccccctc
    aagaccatatcagcttctctggggtcaagactgacaatcccgtgtaaggtgtttctgggaaccggcacac
    ccttaaccaccatgctgtggtggacggccaatgacacccacatagagagcgcctacccgggaggccgcgt
    gaccgaggggccacgccaggaatattcagaaaataatgagaactacattgaagtgccattgatttttgat
    cctgtcacaagagaggatttgcacatggattttaaatgtgttgtccataataccctgagttttcagacac
    tacgcaccacagtcaaggaagcctcctccacgttctcctggggcattgtgctggccccactttcactggc
    cttcttggttttggggggaatatggatgcacagacggtgcaaacacagaactggaaaagcagatggtctg
    actgtgctatggcctcatcatcaagactttcaatcctatcccaagtgaaataaatggaatgaaataattc
    aaacacaaaaaaaaaaaaaaaaaaaaaaaaaaaaaa.
  • IL-1R2, transcript variants 1 and 2, are encoded by the following amino acid sequence (NCBI Accession No. NM004633, NM173343, and SEQ ID NO: 21):
  • MLRLYVLVMGVSAFTLQPAAHTGAARSCRFRGRHYKREFRLEGEPVALRCPQVPYWL
    WASVSPRINLTWHKNDSARTVPGEEETRMWAQDGALWLLPALQEDSGTYVCTTRNAS
    YCDKMSIELRVFENTDAFLPFISYPQILTLSTSGVLVCPDLSEFTRDKTDVKIQWYKDSLL
    LDKDNEKFLSVRGTTHLLVHDVALEDAGYYRCVLTFAHEGQQYNITRSIELRIKKKKEE
    TIPVIISPLKTISASLGSRLTIPCKVFLGTGTPLTTMLWWTANDTHIESAYPGGRVTEGPRQ
    EYSENNENYIEVPLIFDPVTREDLHMDFKCVVHNTLSFQTLRTTVKEASSTFSWGIVLAP
    LSLAFLVLGGIWMHRRCKHRTGKADGLTVLWPHHQDFQSYPK.
  • Interleukin-1 Receptor (Type 2) Antagonist (IL-1Ra3):
  • The present invention comprises compositions with means to inhibit or enhance the activity of the human IL-1R2. Compositions that comprise the IL-1R2 antagonist, IL-1Ra3, have either agonist or antagonist activity regarding the efficacy of IL-1R1 function. The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding IL-1Ra3. The inhibitory polynucleotide or polypeptide composition binds to one or more region(s) of IL-1Ra3 comprised by SEQ ID NO: 22 and SEQ ID NO: 23.
  • IL-1Ra3 is encoded by a region or the entirety of the following mRNA sequence (NCBI Accession No. AF057168 and SEQ ID NO: 22): (for this sequence, the bolded and capitalized codon does not encode methionine, but rather represents the codon that encodes the first amino acid of the corresponding polypeptide)
  • cagaagacctcctgtcctatgaggccctccccatggctttaggtaagctccttccactctcattttttca
    cctgagaaatgagagaggaaaatgtctacaattggtgtttatcaaatgctttcaggctctggtgagcaag
    cgtccaggaaaatgtcaagcgcatggagctccaggcctgtctgggggatctgggcacggggaggcatcca
    tgggagaccatgcaggcactctgaggcaggggctgcaagctagtgcctgctggggcagcaggtgaacaga
    gaggtgtaactgctgtgacagaagtcatggagtccttggagtgtgagggtcattttccactgttgataga
    atagggaaattggtgaaatagccctgttaaatgagagaaagaacagtgtgagctcaatgagaaatactaa
    tagaatgtggcactgagccacaaggtctgagggttgattgataaggaagggtggggactgtggagaatta
    agggcttggcacaggtcagttccaccagttgtcacaagagaatgcaggctcaggtggccagaacttctcg
    cttttccagaagagtccgatattctgatttcattatatatagtattctgattaaaccagacaataaagca
    agcagataaaatatttaaagtataagctgccagtttgcaacctccggttaggatttgtgtggggcaaaga
    aaaaaactctcaggatcattggtatgtagactctaattttaagtttctaatttaaaattggcccctgagg
    ctgggcgtggtggctcacacctgtaatcccagcattttgggaggccaaggtgggtggatctcttgaggtc
    aagagttcaaggcctgcctggccaacatggtgaaaccctgtctctattaaaaatacaaaaattagctggg
    catggtggtgcatgtctgcaatcttagctacttgggtagctaaggcaggagaattgctggaacccgggag
    gtagaggttgcagtgaatggagatcacaccactgcactccagtctgggcaatagagagagacgctctctc
    taaaaaaaaatatgtaaagataaataaaatgaaataaaataggcctctaatgagcaggccattctccttt
    ctgggtcttactttccttgcactcctttctgggtgttaagaggaggtctagaggaagctggacaactctt
    agcttgtagtaagcacagtggaagtatcagctcttaatgggtcatggacacgttacgaagctaggcgccg
    tgctgagcactttacatggtttatcccactgaaccctctcaataaccctatgaggaagggctattattgc
    tcacattttcagaagaggaaatggatatagagagattagataatttgcccatggccagacagctagtata
    agaggaggaggtggattgactgcagacattctgtcttcaaaccactacactatgctatggaggcacagag
    acttaatgaaatcatggagaggggaattgctttgtcaaccacaagcagttattccgggggcagcagatcc
    tcccctgtcccccagtggtacaatggtccctggtgggttgtgctacaatgttagcccatggtcttatgtg
    tttttcaaatgtgtaaagtaggatgctggaaccactcttagaaccagataccaatacattgtgaagaaat
    aaatctctgtgcttaaaactggttcatcccaaaatattttgaactgacacacaataggtgctaaataaat
    gtgtgttaacttgaattggattgaattcgggaaaaaagtgcaataagcttagtgaagacaccatgttccc
    tgggtagaggaaccacattctccatctaaggccaggagtatgggaggtatcaatgtttgcccagcacaga
    acagggtgccaagaagagaaaagttgacggggtgcatactctgactggaaactggaagggtgagaacaga
    gggtaaaggatagagatggaaccatgtgcatacactttgtgttaccttggacaagtcattcatttctctg
    gacctctgctttctctctacacaatggggtcccaccacttcccttacagctgacttgtatgaagaaggag
    gtggaggaggaggagaaggtgaagacaatgCTGactcaaagggtaaattatttttaggatccaagtttga
    aaacaattttaggctactagatatgaacaacatcttgattatgtagttgaaggaaattaaagatgaatgg
    tttaattaaaaattaatcagaatgaaaacgattgattactaatatatctgcaatggtttattttcctgag
    tggcagactcactaaggtttttgaatactcctgtgtgattgctctatgtatgtatgtatgtatgtatgta
    tgcatgtatctatctatctgttgtctaatagaatggatcacatctctgctaataaaaacactacactggc
    agggtacaattataatcattaactgtgcctggaatttgcagcagcagccaccagaggtaccagtgccctt
    taagggttcataatttagaataatccaattatctgagtttttcagggactgaggggtttggcaaggtgta
    gaactttcagtaataaagtcaagaaagtcctggacaaaccaaggtagttggtcactctagtccataacca
    ggtaaagagctttccctgtaacctgtgtaaggttttagaatcatttctttccttattaccaaaaatcctc
    cccaaattttcaagaaattatgaactaaatagttactctatgagataggagttcagcccaaaagaaacac
    cataagaacaaatataattcttgcttatgttaaccatgcaatgaagcagagagaaaaagtcagtggcctc
    tttaggaggactgtagtgtgggaagaaataactaaactgggtttcaatcctggcctggccaggatctgga
    gcaagtgagttaatctttctaagccttgagtagtttataaaagaatggccactccatagacagagtagcc
    tgaaccttgagttcttctataaagtcactatgaatttatactcattttgaaagtgggtgtcaatatgtct
    gtccactttgcacagctgttatgtggacaaaaggagatctgtgtgaaagtgtaacacagagcctaaacta
    taacaggtaagcaacacagttgtccct.
  • One or more isoforms of IL-1Ra3 comprise the following amino acid sequence (NCBI Accession No. AF057168 and SEQ ID NO: 23):
  • DLYEEGGGGGGEGEDNADSK.
  • Interleukin-1 Receptor Accessory Protein (IL-1RAP):
  • Compositions that inhibit the activity of human IL-1RAP inhibit IL-1RAP binding to an IL-1 cytokine or an IL-1 receptor, and subsequent transduction of downstream intracellular signals. Compositions that comprise an inhibitor of IL-1RAP function antagonize the activity of an IL-1 receptor. The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding IL-1RAP. The inhibitory polynucleotide or polypeptide composition binds to one or more region(s) of IL-1RAP, and associated isoforms, comprised by SEQ ID NO: 24-27.
  • IL-1RAP, transcript variant 1, is encoded by the following mRNA sequence (NCBI Accession No. NM002182 and SEQ ID NO: 24):
  • tgccgggatccaggtctccggggtccgctttggccagaggcgcggaaggaagcagtgcccggcgacactg
    cacccatcccggctgcttttgctgcgccctctcagcttcccaagaaaggcatcgtcatgtgatcatcacc
    taagaactagaacatcagcaggccctagaagcctcactcttgcccctccctttaatatctcaaaggATGa
    cacttctgtggtgtgtagtgagtctctacttttatggaatcctgcaaagtgatgcctcagaacgctgcga
    tgactggggactagacaccatgaggcaaatccaagtgtttgaagatgagccagctcgcatcaagtgccca
    ctctttgaacacttcttgaaattcaactacagcacagcccattcagctggccttactctgatctggtatt
    ggactaggcaggaccgggaccttgaggagccaattaacttccgcctccccgagaaccgcattagtaagga
    gaaagatgtgctgtggttccggcccactctcctcaatgacactggcaactatacctgcatgttaaggaac
    actacatattgcagcaaagttgcatttcccttggaagttgttcaaaaagacagctgtttcaattccccca
    tgaaactcccagtgcataaactgtatatagaatatggcattcagaggatcacttgtccaaatgtagatgg
    atattttccttccagtgtcaaaccgactatcacttggtatatgggctgttataaaatacagaattttaat
    aatgtaatacccgaaggtatgaacttgagtttcctcattgccttaatttcaaataatggaaattacacat
    gtgttgttacatatccagaaaatggacgtacgtttcatctcaccaggactctgactgtaaaggtagtagg
    ctctccaaaaaatgcagtgccccctgtgatccattcacctaatgatcatgtggtctatgagaaagaacca
    ggagaggagctactcattccctgtacggtctattttagttttctgatggattctcgcaatgaggtttggt
    ggaccattgatggaaaaaaacctgatgacatcactattgatgtcaccattaacgaaagtataagtcatag
    tagaacagaagatgaaacaagaactcagattttgagcatcaagaaagttacctctgaggatctcaagcgc
    agctatgtctgtcatgctagaagtgccaaaggcgaagttccaaagcagccaaggtgaagcagaaagtgcc
    agctccaagatacacagtggaactggcttgtggttttggagccacagtcctgctagtggtgattctcatt
    gttgtttaccatgtttactggctagagatggtcctattttaccgggctcattttggaacagatgaaacca
    ttttagatggaaaagagtatgatatttatgtatcctatgcaaggaatgcggaagaagaagaatttgtatt
    actgaccctccgtggagttttggagaatgaatttggatacaagctgtgcatctttgaccgagacagtctg
    cctgggggaattgtcacagatgagactttgagcttcattcagaaaagcagacgcctcctggttgttctaa
    gccccaactacgtgctccagggaacccaagccctcctggagctcaaggctggcctagaaaatatggcctc
    tcggggcaacatcaacgtcattttagtacagtacaaagctgtgaaggaaacgaaggtgaaagagctgaag
    agggctaagacggtgctcacggtcattaaatggaaaggggaaaaatccaagtatccacagggcaggttct
    ggaagcagctgcaggtggccatgccagtgaagaaaagtcccaggcggtctagcagtgatgagcagggcct
    ctcgtattcatctttgaaaaatgtatgaaaggaataatgaaaagggtaaaaagaacaaggggtgctccag
    gaagaaagagtccccccagtcttcattcgcagtttatggtttcataggcaaaaataatggtctaagcctc
    ccaatagggataaatttagggtgactgtgtggctgactattctgcttcctcaggcaacactaaagtttag
    aaagatatcatcaacgttctgtcaccagtctctgatgccactatgttctttgcaggcaaagacttgttca
    atgcgaatttccccttctacattgtctatccctgtttttatatgtctccattctttttaaaatcttaaca
    tatggagcagcctttcctatgaatttaaatatgcctttaaaataagtcactgttgacagggtcatgagtt
    tccgagtatagttttctttttatcttatttttactcgtccgttgaaaagataatcaaggcctacatttta
    gctgaggataatgaacttttttcctcattcggctgtataatacataaccacagcaagactgacatccact
    taggatgatacaaagcagtgtaactgaaaatgtttcttttaattgatttaaaggacttgtcttctatacc
    acccttgtcctcatctcaggtaatttatgaaatctatgtaaacttgaaaaatatttcttaatttttgttt
    ttgctccagtcaattcctgattatccacaggtcaacccacattttttcattccttctccctatctgctta
    tatcgcattgctcatttagagtttgcaggaggctccatactaggttcagtctgaaagaaatctcctaatg
    gtgctatagagagggaggtaacagaaagactcttttagggcatttttctgactcatgaaaagagcacaga
    aaaggatgtttggcaatttgtcttttaagtcttaaccttgctaatgtgaatactgggaaagtgatttttt
    ctcactcgtttttgttgctccattgtaaagggcggaggtcagtcttagtggccttgagagttgcttttgg
    cattaatattctaagagaattaactgtatttcctgtcacctattcactagtgcaggaaatatacttgctc
    caaataagtcagtatgagaagtcactgtcaatgaaagttgttttgtttgttttcagtaatattttgctgt
    ttttaagacttggaaaactaagtgcagagtttacagagtggtaaatatctatgttacatgtagattatac
    atatatatacacacgtgtatatgagatatatatcttatatctccacaaacacaaattatatatatacata
    tccacacacatacattacatatatctgtgtatataaatccacatgcacatgaaatatatatatatatata
    atttgtgtgtgtgtatgtgtatgtatatgactttaaatagctatgggtacaatattaaaaaccactggaa
    ctcttgtccagtttttaaattatgtttttactggaatgtttttgtgtcagtgttttctgtacatattatt
    tgttaattcacagctcacagagtgatagttgtcatagttcttgccttccctaagtttatataaataactt
    aagtattgctacagtttatctaggttgcagtggcatctgctgtgcacagagcttccatggtcactgctaa
    gcagtagccagccatcgggcattaattgatttcctactatattcccagcagacacatttagaaactaagc
    tatgttaacctcagtgctcaactatttgaactgttgagtgataaaggaaacaaatataactgtaaatgaa
    tcttggtatcctgtgaaacagaataattcgtaatttaagaaagcccttatcccggtaacatgaatgttga
    tgaacaaatgtaaaattatatcctatatttaagtacccataataaatcatttccctctataagtgttatt
    gattattttaaattgaaaaaagtttcacttggatgaaaaaagtagaaaagtaggtcattcttggatctac
    ttttttttagccttattaatatttttccctattagaaaccacaattactccctctattaacccttcactt
    actagaccagaaaagaacttattccagataagctttgaatatcaattcttacataaactttaggcaaaca
    gggaatagtctagtcaccaaaggaccattctcttgccaatgctgcattccttttgcacttttggattcca
    tatttatcccaaatgctgttgggcacccctagaaataccttgatgttttttctatttatatgcctgcctt
    tggtacttaattttacaaatgctgtaatataaagcatatcaagtttatgtgatacgtatcattgcaagag
    aatttgtttcaagatttttttttaatgttccagaagatggccaatagagaacattcaagggaaatgggga
    aacataatttagagaacaagaacaaaccatgtctcaaatttttttaaaaaaaattaatggttttaaatat
    atgctatagggacgttccatgcccaggttaacaaagaactgtgatatatagagtgtctaattacaaaatc
    atatacgatttatttaattctcttctgtattgtaacttagatgattcccaaggactctaataaaaaatca
    cttcattgtatttggaaacaaaaacatcattcattaattacttattttctttccataggttttaatattt
    tgagagtgtcttttttatttcattcatgaacttttgtatttttcatttttcatttgatttgtaaatttac
    ttatgttaaaaataaaccatttattttcagctttg.
  • IL-1RAP, transcript variant 1, is encoded by the following amino acid sequence (NCBI Accession No. NM002182 and SEQ ID NO: 25):
  • MTLLWCVVSLYFYGILQSDASERCDDWGLDTMRQIQVFEDEPARIKCPLFEHFLKFNYS
    TAHSAGLTLIWYWTRQDRDLEEPINFRLPENRISKEKDVLWFRPTLLNDTGNYTCMLRN
    TTYCSKVAFPLEVVQKDSCFNSPMKLPVHKLYIEYGIQRITCPNVDGYFPSSVKPTITWY
    MGCYKIQNFNNVIPEGMNLSFLIALISNNGNYTCVVTYPENGRTFHLTRTLTVKVVGSP
    KNAVPPVIHSPNDHVVYEKEPGEELLIPCTVYFSFLMDSRNEVWWTIDGKKPDDITIDVT
    INESISHSRTEDETRTQILSIKKVTSEDLKRSYVCHARSAKGEVAKAAKVKQKVPAPRYT
    VELACGFGATVLLVVILIVVYHVYWLEMVLFYRAHFGTDETILDGKEYDIYVSYARNAE
    EEEFVLLTLRGVLENEFGYKLCIFDRDSLPGGIVTDETLSFIQKSRRLLVVLSPNYVLQGT
    QALLELKAGLENMASRGNINVILVQYKAVKETKVKELKRAKTVLTVIKWKGEKSKYPQ
    GRFWKQLQVAMPVKKSPRRSSSDEQGLSYSSLKNV.
  • IL-1RAP, transcript variant 2, is encoded by the following mRNA sequence (NCBI Accession No. NM134470 and SEQ ID NO: 26):
  • tgccgggatccaggtctccggggtccgctttggccagaggcgcggaaggaagcagtgcccggcgacactg
    cacccatcccggctgcttttgctgcgccctctcagcttcccaagaaaggcatcgtcatgtgatcatcacc
    taagaactagaacatcagcaggccctagaagcctcactcttgcccctccctttaatatctcaaaggATGa
    cacttctgtggtgtgtagtgagtctctacttttatggaatcctgcaaagtgatgcctcagaacgctgcga
    tgactggggactagacaccatgaggcaaatccaagtgtttgaagatgagccagctcgcatcaagtgccca
    ctctttgaacacttcttgaaattcaactacagcacagcccattcagctggccttactctgatctggtatt
    ggactaggcaggaccgggaccttgaggagccaattaacttccgcctccccgagaaccgcattagtaagga
    gaaagatgtgctgtggttccggcccactctcctcaatgacactggcaactatacctgcatgttaaggaac
    actacatattgcagcaaagttgcatttcccttggaagttgttcaaaaagacagctgtttcaattccccca
    tgaaactcccagtgcataaactgtatatagaatatggcattcagaggatcacttgtccaaatgtagatgg
    atattttccttccagtgtcaaaccgactatcacttggtatatgggctgttataaaatacagaattttaat
    aatgtaatacccgaaggtatgaacttgagtttcctcattgccttaatttcaaataatggaaattacacat
    gtgttgttacatatccagaaaatggacgtacgtttcatctcaccaggactctgactgtaaaggtagtagg
    ctctccaaaaaatgcagtgccccctgtgatccattcacctaatgatcatgtggtctatgagaaagaacca
    ggagaggagctactcattccctgtacggtctattttagttttctgatggattctcgcaatgaggtttggt
    ggaccattgatggaaaaaaacctgatgacatcactattgatgtcaccattaacgaaagtataagtcatag
    tagaacagaagatgaaacaagaactcagattttgagcatcaagaaagttacctctgaggatctcaagcgc
    agctatgtctgtcatgctagaagtgccaaaggcgaagttgccaaagcagccaaggtgaagcagaaaggta
    atagatgcggtcagtgatgaatctctcagctccaaattaacattgtggtgaataaggacaaaaggagaga
    ttgagaacaagagagctccagcacctagcccgacggcatctaacccatagtaatgaatcaaacttaaatg
    aaaaatatgaaagttttcatctatgtaagatactcaaaatattgtttctgatattgttagtaccgtaatg
    cccaaatgtagctaaaaaaatcgacgtgagtacagtgagacacaattttgtgtctgtacaattatgaaaa
    attaaaaacaaagaaaatattcaaagctaccaaagatagaaaaaactggtagagccacatattgttggtg
    aattattaagacccttttaaaaatcattcatggtagagtttaagagtcataaaaaagattgcatcatctg
    acctaagactttcggaatttttcctgaacaaataacagaaagggaattatataccttttaatattattag
    aagcattatctgtagttgtaaaacattattaatagcagccatccaattgtatgcaactaattaaggtatt
    gaatgtttattttccaaaaatgcataattataatattattttaaacactatgtatcaatatttaagcagg
    tttataatataccagcagccacaattgctaaaatgaaaatcatttaaattatgattttaaatggtataaa
    catgatttctatgttgatagtactatattattctacaataaatggaaattataaagccttcttgtcagaa
    gtgctgctcctaaaaaaaaaaaaaaaaaaaaaa.
  • IL-1RAP, transcript variant 2, is encoded by the following amino acid sequence (NCBI Accession No. NM134470 and SEQ ID NO: 27):
  • MTLLWCVVSLYFYGILQSDASERCDDWGLDTMRQIQVFEDEPARIKCPLFEHFLKFNYS
    TAHSAGLTLIWYWTRQDRDLEEPINFRLPENRISKEKDVLWFRPTLLNDTGNYTCMLRN
    TTYCSKVAFPLEVVQKDSCFNSPMKLPVHKLYIEYGIQRITCPNVDGYFPSSVKPTITWY
    MGCYKIQNFNNVIPEGMNLSFLIALISNNGNYTCVVTYPENGRTFHLTRTLTVKVVGSP
    KNAVPPVIHSPNDHVVYEKEPGEELLIPCTVYFSFLMDSRNEVWWTIDGKKPDDITIDVT
    INESISHSRTEDETRTQILSIKKVTSEDLKRSYVCHARSAKGEVAKAAKVKQKGNRCGQ.
  • Interleukin-1 Receptor Associated Kinase 1 (IRAK1):
  • The invention also comprises compositions and methods to inhibit the activity of human IRAK1, defined as the ability of this protein to bind an IL-1 receptor following ligation of this receptor with IL-1, as well as to transduce downstream signals leading to an inflammatory response. Compositions that comprise an inhibitor of IRAK1 antagonize downstream signaling from an IL-1 receptor. The composition comprises a polynucleotide, a polypeptide, an antibody, a compound, or a small molecule with means to inhibit the transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding IRAK1. The inhibitory polynucleotide or polypeptide composition binds to one or more region(s) of IRAK1, and associated isoforms, comprised by SEQ ID NO: 28-33.
  • IRAK1, transcript variant 1, is encoded by the following mRNA sequence (NCBI Accession No. NM001569 and SEQ ID NO: 28):
  • cgcggacccggccggcccaggcccgcgcccgccgcggccctgagaggccccggcaggtcccggcccggcg
    gcggcagccATGgccggggggccgggcccgggggagcccgcagcccccggcgcccagcacttcttgtacg
    aggtgccgccctgggtcatgtgccgcttctacaaagtgatggacgccctggagcccgccgactggtgcca
    gttcgccgccctgatcgtgcgcgaccagaccgagctgcggctgtgcgagcgctccgggcagcgcacggcc
    agcgtcctgtggccctggatcaaccgcaacgcccgtgtggccgacctcgtgcacatcctcacgcacctgc
    agctgctccgtgcgcgggacatcatcacagcctggcaccctcccgccccgcttccgtccccaggcaccac
    tgccccgaggcccagcagcatccctgcacccgccgaggccgaggcctggagcccccggaagttgccatcc
    tcagcctccaccttcctctccccagcttttccaggctcccagacccattcagggcctgagctcggcctgg
    tcccaagccctgcttccctgtggcctccaccgccatctccagccccttcttctaccaagccaggcccaga
    gagctcagtgtccctcctgcagggagcccgcccctttccgttttgctggcccctctgtgagatttcccgg
    ggcacccacaacttctcggaggagctcaagatcggggagggtggctttgggtgcgtgtaccgggcggtga
    tgaggaacacggtgtatgctgtgaagaggctgaaggagaacgctgacctggagtggactgcagtgaagca
    gagcttcctgaccgaggtggagcagctgtccaggtttcgtcacccaaacattgtggactttgctggctac
    tgtgctcagaacggcttctactgcctggtgtacggcttcctgcccaacggctccctggaggaccgtctcc
    actgccagacccaggcctgcccacctctctcctggcctcagcgactggacatccttctgggtacagcccg
    ggcaattcagtttctacatcaggacagccccagcctcatccatggagacatcaagagttccaacgtcctt
    ctggatgagaggctgacacccaagctgggagactttggcctggcccggttcagccgctttgccgggtcca
    gccccagccagagcagcatggtggcccggacacagacagtgcggggcaccctggcctacctgcccgagga
    gtacatcaagacgggaaggctggctgtggacacggacaccttcagctttggggtggtagtgctagagacc
    ttggctggtcagagggctgtgaagacgcacggtgccaggaccaagtatctgaaagacctggtggaagagg
    aggctgaggaggctggagtggctttgagaagcacccagagcacactgcaagcaggtctggctgcagatgc
    ctgggctgctcccatcgccatgcagatctacaagaagcacctggaccccaggcccgggccctgcccacct
    gagctgggcctgggcctgggccagctggcctgctgctgcctgcaccgccgggccaaaaggaggcctccta
    tgacccaggtgtacgagaggctagagaagctgcaggcagtggtggcgggggtgcccgggcattcggaggc
    cgccagctgcatccccccttccccgcaggagaactcctacgtgtccagcactggcagagcccacagtggg
    gctgctccatggcagcccctggcagcgccatcaggagccagtgcccaggcagcagagcagctgcagagag
    gccccaaccagcccgtggagagtgacgagagcctaggcggcctctctgctgccctgcgctcctggcactt
    gactccaagctgccctctggacccagcacccctcagggaggccggctgtcctcagggggacacggcagga
    gaatcgagctgggggagtggcccaggatcccggcccacagccgtggaaggactggcccttggcagctctg
    catcatcgtcgtcagagccaccgcagattatcatcaaccctgcccgacagaagatggtccagaagctggc
    cctgtacgaggatggggccctggacagcctgcagctgctgtcgtccagctccctcccaggcttgggcctg
    gaacaggacaggcaggggcccgaagaaagtgatgaatttcagagctgatgtgttcacctgggcagatccc
    ccaaatccggaagtcaaagttctcatggtcagaagttctcatggtgcacgagtcctcagcactctgccgg
    cagtgggggtgggggcccatgcccgcgggggagagaaggaggtggccctgctgttctaggctctgtgggc
    ataggcaggcagagtggaaccctgcctccatgccagcatctgggggcaaggaaggctggcatcatccagt
    gaggaggctggcgcatgttgggaggctgctggctgcacagacccgtgaggggaggagaggggctgctgtg
    caggggtgtggagtagggagctggctcccctgagagccatgcagggcgtctgcagcccaggcctctggca
    gcagctctttgcccatctctttggacagtggccaccctgcacaatggggccgacgaggcctagggccctc
    ctacctgcttacaatttggaaaagtgtggccgggtgcggtggctcacgcctgtaatcccagcactttggg
    aggccaaggcaggaggatcgctggagcccagtaggtcaagaccagccagggcaacatgatgagaccctgt
    ctctgccaaaaaattttttaaactattagcctggcgtggtagcgcacgcctgtggtcccagctgctgggg
    aggctgaagtaggaggatcatttatgcttgggaggtcgaggctgcagtgagtcatgattgtatgactgca
    ctccagcctgggtgacagagcaagaccctgtttcaaaaagaaaaaccctgggaaaagtgaagtatggctg
    taagtctcatggttcagtcctagcaagaagcgagaattctgagatcctccagaaagtcgagcagcaccca
    cctccaacctcgggccagtgtcttcaggctttactggggacctgcgagctggcctaatgtggtggcctgc
    aagccaggccatccctgggcgccacagacgagctccgagccaggtcaggcttcggaggccacaagctcag
    cctcaggcccaggcactgattgtggcagaggggccactacccaaggtctagctaggcccaagacctagtt
    acccagacagtgagaagcccctggaaggcagaaaagttgggagcatggcagacagggaagggaaacattt
    tcagggaaaagacatgtatcacatgtcttcagaagcaagtcaggtttcatgtaaccgagtgtcctcttgc
    gtgtccaaaagtagcccagggctgtagcacaggcttcacagtgattttgtgttcagccgtgagtcacact
    acatgcccccgtgaagctgggcattggtgacgtccaggttgtccttgagtaataaaaacgtatgttgcaa
    taaaaaaaaaaaaaaaaaa.
  • IRAK1, transcript variant 1, is encoded by the following amino acid sequence (NCBI Accession No. NM001569 and SEQ ID NO: 29):
  • MAGGPGPGEPAAPGAQHFLYEVPPWVMCRFYKVMDALEPADWCQFAALIVRDQTELR
    LCERSGQRTASVLWPWINRNARVADLVHILTHLQLLRARDIITAWHPPAPLPSPGTTAPR
    PSSIPAPAEAEAWSPRKLPSSASTFLSPAFPGSQTHSGPELGLVPSPASLWPPPPSPAPSST
    KPGPESSVSLLQGARPFPFCWPLCEISRGTHNFSEELKIGEGGFGCVYRAVMRNTVYAV
    KRLKENADLEWTAVKQSFLTEVEQLSRFRHPNIVDFAGYCAQNGFYCLVYGFLPNGSL
    EDRLHCQTQACPPLSWPQRLDILLGTARAIQFLHQDSPSLIHGDIKSSNVLLDERLTPKLG
    DFGLARFSRFAGSSPSQSSMVARTQTVRGTLAYLPEEYIKTGRLAVDTDTFSFGVVVLET
    LAGQRAVKTHGARTKYLKDLVEEEAEEAGVALRSTQSTLQAGLAADAWAAPIAMQIY
    KKHLDPRPGPCPPELGLGLGQLACCCLHRRAKRRPPMTQVYERLEKLQAVVAGVPGHS
    EAASCIPPSPQENSYVSSTGRAHSGAAPWQPLAAPSGASAQAAEQLQRGPNQPVESDES
    LGGLSAALRSWHLTPSCPLDPAPLREAGCPQGDTAGESSWGSGPGSRPTAVEGLALGSS
    ASSSSEPPQIIINPARQKMVQKLALYEDGALDSLQLLSSSSLPGLGLEQDRQGPEESDEFQ
    S.
  • IRAK1, transcript variant 2, is encoded by the following mRNA sequence (NCBI Accession No. NM001025242 and SEQ ID NO: 30):
  • cgcggacccggccggcccaggcccgcgcccgccgcggccctgagaggccccggcaggtcccggcccggcg
    gcggcagccATGgccggggggccgggcccgggggagcccgcagcccccggcgcccagcacttcttgtacg
    aggtgccgccctgggtcatgtgccgcttctacaaagtgatggacgccctggagcccgccgactggtgcca
    gttcgccgccctgatcgtgcgcgaccagaccgagctgcggctgtgcgagcgctccgggcagcgcacggcc
    agcgtcctgtggccctggatcaaccgcaacgcccgtgtggccgacctcgtgcacatcctcacgcacctgc
    agctgctccgtgcgcgggacatcatcacagcctggcaccctcccgccccgcttccgtccccaggcaccac
    tgccccgaggcccagcagcatccctgcacccgccgaggccgaggcctggagcccccggaagttgccatcc
    tcagcctccaccttcctctccccagcttttccaggctcccagacccattcagggcctgagctcggcctgg
    tcccaagccctgcttccctgtggcctccaccgccatctccagccccttcttctaccaagccaggcccaga
    gagctcagtgtccctcctgcagggagcccgcccctttccgttttgctggcccctctgtgagatttcccgg
    ggcacccacaacttctcggaggagctcaagatcggggagggtggctttgggtgcgtgtaccgggcggtga
    tgaggaacacggtgtatgctgtgaagaggctgaaggagaacgctgacctggagtggactgcagtgaagca
    gagcttcctgaccgaggtggagcagctgtccaggtttcgtcacccaaacattgtggactttgctggctac
    tgtgctcagaacggcttctactgcctggtgtacggcttcctgcccaacggctccctggaggaccgtctcc
    actgccagacccaggcctgcccacctctctcctggcctcagcgactggacatccttctgggtacagcccg
    ggcaattcagtttctacatcaggacagccccagcctcatccatggagacatcaagagttccaacgtcctt
    ctggatgagaggctgacacccaagctgggagactttggcctggcccggttcagccgctttgccgggtcca
    gccccagccagagcagcatggtggcccggacacagacagtgcggggcaccctggcctacctgcccgagga
    gtacatcaagacgggaaggctggctgtggacacggacaccttcagctttggggtggtagtgctagagacc
    ttggctggtcagagggctgtgaagacgcacggtgccaggaccaagtatctgaaagacctggtggaagagg
    aggctgaggaggctggagtggctttgagaagcacccagagcacactgcaagcaggtctggctgcagatgc
    ctgggctgctcccatcgccatgcagatctacaagaagcacctggaccccaggcccgggccctgcccacct
    gagctgggcctgggcctgggccagctggcctgctgctgcctgcaccgccgggccaaaaggaggcctccta
    tgacccaggagaactcctacgtgtccagcactggcagagcccacagtggggctgctccatggcagcccct
    ggcagcgccatcaggagccagtgcccaggcagcagagcagctgcagagaggccccaaccagcccgtggag
    agtgacgagagcctaggcggcctctctgctgccctgcgctcctggcacttgactccaagctgccctctgg
    acccagcacccctcagggaggccggctgtcctcagggggacacggcaggagaatcgagctgggggagtgg
    cccaggatcccggcccacagccgtggaaggactggcccttggcagctctgcatcatcgtcgtcagagcca
    ccgcagattatcatcaaccctgcccgacagaagatggtccagaagctggccctgtacgaggatggggccc
    tggacagcctgcagctgctgtcgtccagctccctcccaggcttgggcctggaacaggacaggcaggggcc
    cgaagaaagtgatgaatttcagagctgatgtgttcacctgggcagatcccccaaatccggaagtcaaagt
    tctcatggtcagaagttctcatggtgcacgagtcctcagcactctgccggcagtgggggtgggggcccat
    gcccgcgggggagagaaggaggtggccctgctgttctaggctctgtgggcataggcaggcagagtggaac
    cctgcctccatgccagcatctgggggcaaggaaggctggcatcatccagtgaggaggctggcgcatgttg
    ggaggctgctggctgcacagacccgtgaggggaggagaggggctgctgtgcaggggtgtggagtagggag
    ctggctcccctgagagccatgcagggcgtctgcagcccaggcctctggcagcagctctttgcccatctct
    ttggacagtggccaccctgcacaatggggccgacgaggcctagggccctcctacctgcttacaatttgga
    aaagtgtggccgggtgcggtggctcacgcctgtaatcccagcactttgggaggccaaggcaggaggatcg
    ctggagcccagtaggtcaagaccagccagggcaacatgatgagaccctgtctctgccaaaaaatttttta
    aactattagcctggcgtggtagcgcacgcctgtggtcccagctgctggggaggctgaagtaggaggatca
    tttatgcttgggaggtcgaggctgcagtgagtcatgattgtatgactgcactccagcctgggtgacagag
    caagaccctgtttcaaaaagaaaaaccctgggaaaagtgaagtatggctgtaagtctcatggttcagtcc
    tagcaagaagcgagaattctgagatcctccagaaagtcgagcagcacccacctccaacctcgggccagtg
    tcttcaggctttactggggacctgcgagctggcctaatgtggtggcctgcaagccaggccatccctgggc
    gccacagacgagctccgagccaggtcaggcttcggaggccacaagctcagcctcaggcccaggcactgat
    tgtggcagaggggccactacccaaggtctagctaggcccaagacctagttacccagacagtgagaagccc
    ctggaaggcagaaaagttgggagcatggcagacagggaagggaaacattttcagggaaaagacatgtatc
    acatgtcttcagaagcaagtcaggtttcatgtaaccgagtgtcctcttgcgtgtccaaaagtagcccagg
    gctgtagcacaggcttcacagtgattttgtgttcagccgtgagtcacactacatgcccccgtgaagctgg
    gcattggtgacgtccaggttgtccttgagtaataaaaacgtatgttgcaataaaaaaaaaaaaaaaaaa.
  • IRAK1, transcript variant 2, is encoded by the following amino acid sequence (NCBI Accession No. NM001025242 and SEQ ID NO: 31):
  • MAGGPGPGEPAAPGAQHFLYEVPPWVMCRFYKVMDALEPADWCQFAALIVRDQTELR
    LCERSGQRTASVLWPWINRNARVADLVHILTHLQLLRARDIITAWHPPAPLPSPGTTAPR
    PSSIPAPAEAEAWSPRKLPSSASTFLSPAFPGSQTHSGPELGLVPSPASLWPPPPSPAPSST
    KPGPESSVSLLQGARPFPFCWPLCEISRGTHNFSEELKIGEGGFGCVYRAVMRNTVYAV
    KRLKENADLEWTAVKQSFLTEVEQLSRFRHPNIVDFAGYCAQNGFYCLVYGFLPNSGL
    EDRLHCQTQACPPLSWPQRLDILLGTARAIQFLHQDSPSLIHGDIKSSNVLLDERLTPKLG
    DFGLARFSRFAGSSPSQSSMVARTQTVRGTLAYLPEEYIKTGRLAVDTDTFSFGVVVLET
    LAGQRAVKTHGARTKYLKDLVEEEAEEAGVALRSTQSTLQAGLAADAWAAPIAMQIY
    KKHLDPRPGPCPPELGLGLGQLACCCLHRRAKRRPPMTQENSYVSSTGRAHSGAAPWQ
    PLAAPSGASAQAAEQLQRGPNQPVESDESLGGLSAALRSWHLTPSCPLDPAPLREAGCP
    QGDTAGESSWGSGPGSRPTAVEGLALGSSASSSSEPPQIIINPARQKMVQKLALYEDGAL
    DSLQLLSSSSLPGLGLEQDRQGPEESDEFQS.
  • IRAK1, transcript variant 3, is encoded by the following mRNA sequence (NCBI Accession No. NM001025243 and SEQ ID NO: 32):
  • cgcggacccggccggcccaggcccgcgcccgccgcggccctgagaggccccggcaggtcccggcccggcg
    gcggcagccATGgccggggggccgggcccgggggagcccgcagcccccggcgcccagcacttcttgtacg
    aggtgccgccctgggtcatgtgccgcttctacaaagtgatggacgccctggagcccgccgactggtgcca
    gttcgccgccctgatcgtgcgcgaccagaccgagctgcggctgtgcgagcgctccgggcagcgcacggcc
    agcgtcctgtggccctggatcaaccgcaacgcccgtgtggccgacctcgtgcacatcctcacgcacctgc
    agctgctccgtgcgcgggacatcatcacagcctggcaccctcccgccccgcttccgtccccaggcaccac
    tgccccgaggcccagcagcatccctgcacccgccgaggccgaggcctggagcccccggaagttgccatcc
    tcagcctccaccttcctctccccagcttttccaggctcccagacccattcagggcctgagctcggcctgg
    tcccaagccctgcttccctgtggcctccaccgccatctccagccccttcttctaccaagccaggcccaga
    gagctcagtgtccctcctgcagggagcccgcccctttccgttttgctggcccctctgtgagatttcccgg
    ggcacccacaacttctcggaggagctcaagatcggggagggtggctttgggtgcgtgtaccgggcggtga
    tgaggaacacggtgtatgctgtgaagaggctgaaggagaacgctgacctggagtggactgcagtgaagca
    gagcttcctgaccgaggtggagcagctgtccaggtttcgtcacccaaacattgtggactttgctggctac
    tgtgctcagaacggcttctactgcctggtgtacggcttcctgcccaacggctccctggaggaccgtctcc
    actgccagacccaggcctgcccacctctctcctggcctcagcgactggacatccttctgggtacagcccg
    ggcaattcagtttctacatcaggacagccccagcctcatccatggagacatcaagagttccaacgtcctt
    ctggatgagaggctgacacccaagctgggagactttggcctggcccggttcagccgctttgccgggtcca
    gccccagccagagcagcatggtggcccggacacagacagtgcggggcaccctggcctacctgcccgagga
    gtacatcaagacgggaaggctggctgtggacacggacaccttcagctttggggtggtagtgctagagacc
    ttggctggtcagagggctgtgaagacgcacggtgccaggaccaagtatctggtgtacgagaggctagaga
    agctgcaggcagtggtggcgggggtgcccgggcattcggaggccgccagctgcatccccccttccccgca
    ggagaactcctacgtgtccagcactggcagagcccacagtggggctgctccatggcagcccctggcagcg
    ccatcaggagccagtgcccaggcagcagagcagctgcagagaggccccaaccagcccgtggagagtgacg
    agagcctaggcggcctctctgctgccctgcgctcctggcacttgactccaagctgccctctggacccagc
    acccctcagggaggccggctgtcctcagggggacacggcaggagaatcgagctgggggagtggcccagga
    tcccggcccacagccgtggaaggactggcccttggcagctctgcatcatcgtcgtcagagccaccgcaga
    ttatcatcaaccctgcccgacagaagatggtccagaagctggccctgtacgaggatggggccctggacag
    cctgcagctgctgtcgtccagctccctcccaggcttgggcctggaacaggacaggcaggggcccgaagaa
    agtgatgaatttcagagctgatgtgttcacctgggcagatcccccaaatccggaagtcaaagttctcatg
    gtcagaagttctcatggtgcacgagtcctcagcactctgccggcagtgggggtgggggcccatgcccgcg
    ggggagagaaggaggtggccctgctgttctaggctctgtgggcataggcaggcagagtggaaccctgcct
    ccatgccagcatctgggggcaaggaaggctggcatcatccagtgaggaggctggcgcatgttgggaggct
    gctggctgcacagacccgtgaggggaggagaggggctgctgtgcaggggtgtggagtagggagctggctc
    ccctgagagccatgcagggcgtctgcagcccaggcctctggcagcagctctttgcccatctctttggaca
    gtggccaccctgcacaatggggccgacgaggcctagggccctcctacctgcttacaatttggaaaagtgt
    ggccgggtgcggtggctcacgcctgtaatcccagcactttgggaggccaaggcaggaggatcgctggagc
    ccagtaggtcaagaccagccagggcaacatgatgagaccctgtctctgccaaaaaattttttaaactatt
    agcctggcgtggtagcgcacgcctgtggtcccagctgctggggaggctgaagtaggaggatcatttatgc
    ttgggaggtcgaggctgcagtgagtcatgattgtatgactgcactccagcctgggtgacagagcaagacc
    ctgtttcaaaaagaaaaaccctgggaaaagtgaagtatggctgtaagtctcatggttcagtcctagcaag
    aagcgagaattctgagatcctccagaaagtcgagcagcacccacctccaacctcgggccagtgtcttcag
    gctttactggggacctgcgagctggcctaatgtggtggcctgcaagccaggccatccctgggcgccacag
    acgagctccgagccaggtcaggcttcggaggccacaagctcagcctcaggcccaggcactgattgtggca
    gaggggccactacccaaggtctagctaggcccaagacctagttacccagacagtgagaagcccctggaag
    gcagaaaagttgggagcatggcagacagggaagggaaacattttcagggaaaagacatgtatcacatgtc
    ttcagaagcaagtcaggtttcatgtaaccgagtgtcctcttgcgtgtccaaaagtagcccagggctgtag
    cacaggcttcacagtgattttgtgttcagccgtgagtcacactacatgcccccgtgaagctgggcattgg
    tgacgtccaggttgtccttgagtaataaaaacgtatgttgcaataaaaaaaaaaaaaaaaaa.
  • IRAK1, transcript variant 3, is encoded by the following amino acid sequence (NCBI Accession No. NM001025243 and SEQ ID NO: 33):
  • MAGGPGPGEPAAPGAQHFLYEVPPWVMCRFYKVMDALEPADWCQFAALIVRDQTELR
    LCERSGQRTASVLWPWINRNARVADLVHILTHLQLLRARDIITAWHPPAPLPSPGTTAPR
    PSSIPAPAEAEAWSPRKLPSSASTFLSPAFPGSQTHSGPELGLVPSPASLWPPPPSPAPSST
    KPGPESSVSLLQGARPFPFCWPLCEISRGTHNFSEELKIGEGGFGCVYRAVMRNTVYAV
    KRLKENADLEWTAVKQSFLTEVEQLSRFRHPNIVDFAGYCAQNGFYCLVYGFLPNGSL
    EDRLHCQTQACPPLSWPQRLDILLGTARAIQFLHQDSPSLIHGDIKSSNVLLDERLTPKLG
    DFGLARFSRFAGSSPSQSSMVARTQTVRGTLAYLPEEYIKTGRLAVDTDTFSFGVVVLET
    LAGQRAVKTHGARTKYLVYERLEKLQAVVAGVPGHSEAASCIPPSPQENSYVSSTGRA
    HSGAAPWQPLAAPSGASAQAAEQLQRGPNQPVESDESLGGLSAALRSWHLTPSCPLDP
    APLREAGCPQGDTAGESSWGSGPGSRPTAVEGLALGSSASSSSEPPQIIINPARQKMVQK
    LALYEDGALDSLQLLSSSSLPGLGLEQDRQGPEESDEFQS.

    Silencing Expression with MicroRNAs
  • The present invention comprises compositions with means to inhibit the activity of IL-1α, IL-1b, IL-1R1, IL-1R2, Il-1Ra3, IL-1RAP, or IRAK1, by delivering microRNA (miRNA) molecules to an ocular or adnexal tissue with an appropriate pharmaceutical carrier. Compositions that comprise an miRNA targeted to either IL-1a, IL-1b, IL-1R1, IL-1R2, Il-1Ra3, IL-1RAP, or IRAK1 antagonize the function of IL-1R1. The composition comprises one or more miRNA(s) that bind to one or more regions of IL-1a, IL-1b, IL-1R1, IL-1R2, Il-1Ra3, IL-1RAP, or IRAK1. The following table contains exemplary miRNAs that have been shown to partially or completely silence the expression of human IL-1α or IL-1R1.
  • TABLE 1
    Summary of miRNAs, their human target genes, 
    nucleotide sequences, and their sequence
    identifier numbers.
    Polynucleotide SEQ
    Target sequence ID
    Gene miRNA (5′ to 3′) NO:
    IL-1α miR-30c UGUAAACAUCCUACACUCUCAGC 34
    IL-1α miR-30b UGUAAACAUCCUACACUCAGC 35
    IL-1α miR-30a-5p UGUAAACAUCCUCGACUGGAAGC 36
    IL-1α miR-24 UGGCUCAGUUCAGCAGGAACAG 37
    IL-1R1 miR-135b UAUGGCUUUUCAUUCCUAUGUG 38
    IL-1R1 miR-326 CCUCUGGGCCCUUCCUCCAG 39
    IL-1R1 miR-184 UGGACGGAGAACUGAUAAGGGU 40
    IL-1R1 miR-214 ACAGCAGGCACAGACAGGCAG 41
    IL-1R1 miR-203 GUGAAAUGUUUAGGACCACUAG 42
    IL-1R1 miR-331 GCCCCUGGGCCUAUCCUAGAA 43
    IL-1R1 miR-205 UCCUUCAUUCCACCGGAGUCUG 44
  • IL-1 and IL-1R-Mediated Signaling
  • As used herein, the term “inhibit an activity of an inflammatory interleukin-1 cytokine” is meant to describe the inhibition, prevention, diminution, reduction, decrease, repression, or interruption intracellular signaling initiated, communicated, or transduced from an IL-1 receptor. In one aspect of the invention, inhibition, prevention, diminution, reduction, decreases, repression, or interruption of intracellular signaling initiated, communicated, or transduced from an IL-1 receptor is achieved by preventing or decreasing binding of an IL-1 cytokine to an IL-1R. Alternatively, or in addition, transduction of intracellular signaling from an IL-1R is prevented by removing, silencing, or mutating a downstream effector or target within a signaling cascade. The expression and/or function or activity of downstream effectors and/or targets are removed (e.g. deleted, knocked-out, sequestered, denatured, degraded, etc.), silenced (degraded, transcriptionally or translationally repressed), or mutated (nucleotide or amino acid sequence encoding the active product is altered to encode a non-functional product) by genetic modification or administration of a therapeutic compound.
  • Exemplary downstream effectors and/or targets include, but are not limited to, one or more isoforms or homologs of an IL-1 (interleukin 1), an IL-1α (interleukin 1 alpha), an IL-1β (interleukin 1 beta), an IL-1R (interleukin 1 receptor, type I), an IL-1Ra (IL-1R antagonist), an IL-1RAcP (IL-1R accessory protein), a TOLLIP (TOLL interacting protein), an IRAK1 (IL-1R associated kinase 1), an IRAK2 (IL-1R associated kinase 2), an IRAK 3 (IL-1R associated kinase 3), a MYD88 (myeloid differentiation primary response gene 88), an ECSIT (evolutionarily conserved signaling intermediate in Toll pathways), a TRAF6 (TNF-receptor associated factor 6), a MEKK1 (MAP ERK kinase kinase 1), a TAB1 (TAK1 binding protein 1), a TAK1 (transforming growth factor b activated kinase 1), a NIK (NFkB Inducing Kinase), a RKIP (Raf kinase inhibitor protein), a MEK3 (Mitogen-Activated Protein Kinase Kinase 3; MEK3 or MKK3), a MEK6 (Mitogen-Activated Protein Kinase Kinase 6; MEK6 or MKK6), a MAPK14 (mitogen activated protein kinase 14), a MAPK8 (mitogen activated protein kinase 8), a MEKK1 (mitogen activated protein kinase kinase kinase 1), a MAP3K14 (mitogen activated protein kinase kinase kinase 14), a MEKK7 (mitogen activated protein kinase kinase kinase 7 or MKK7), a MAP3K7IP1 (mitogen activated protein kinase kinase kinase 7 interacting protein 1), a JNK (Jun N-terminal kinase), p38 (also known as p38 MAPK or p38 mitogen activated protein kinase), cJUN (jun oncogene), AP-1 (activator protein 1; transcription factor), IL-6 (interleukin 6, also known as interferon beta 2), TNFα (tumor necrosis factor-alpha), a TNF (tumor necrosis factor superfamily member), an IFNα (interferon alpha, interferon alpha 1), an IFNβ (interferon beta, interferon beta 1), a TGFβ1 (transforming growth factor beta 1), a TGFβ2 (transforming growth factor beta 2), a TGFβ3 (transforming growth factor beta 3), an IKKα (inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase alpha), an IKKβ (inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta), a IκBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha), a Chuk (conserved helix-loop-helix ubiquitous kinase), and a NFκB (nuclear factor of kappa light polypeptide gene enhancer in B-cells 1; also known as p105). Additional signaling molecules and relationships are defined by O'Neill, L. A. J. and Greene, C. 1998. Journal of Leukocyte Biology. 63: 650-657.
  • The inhibition of an activity of an inflammatory interleukin-1 cytokine is determined by sampling an ocular or adnexal tissue or fluid and determining the abundance of a polynucleotide or polypeptide which encodes for component of an IL-1R signaling cascade. An increase or decrease in the abundance of a polynucleotide or polypeptide which encodes for component of an IL-1R signaling cascade following administration of a therapeutic composition of the invention compared to the abundance of the component of an IL-1R signaling cascade prior to the administration indicates inhibition of an activity of an inflammatory interleukin-1 cytokine.
  • Specifically, FIG. 4 shows the functional interrelationships between components of two exemplary signaling cascades. The arrows between components in this figure indicate that the component preceding the arrow activates the component following the arrow. Conversely, the blunted lines indicated that the component preceding the blunted line inhibits the activity or function of the component following the blunted line.
  • Briefly, the IL-1R, type I, binds IL-1β, however, IL-1R requires the IL-1 receptor accessory protein (IL-1RAcP) to transduce a signal. IL-1 binding causes activation of two kinases, IRAK-1 and IRAK-2, associated with the IL-1 receptor complex. IRAK-1 (IL-1 Receptor Associated Kinase) activates and recruits TRAF6 to the IL-1 receptor complex. TRAF6 activates two pathways, one leading to NF-kB activation and another leading to c-jun activation. The TRAF associated protein ECSIT leads to c-Jun activation through the Map kinase/JNK signaling system. TRAF6 also signals through the TAB1/TAK1 kinases to trigger the degradation of 1-kB, and activation of NF-kB.
  • For instance, in certain embodiments of the invention, a decrease in the abundance or absence of the processed form of MEKK1, a decrease in the abundance or absence of phosphorylated IκBα, a decrease in the abundance or absence of phosphorylated c-JUN, a decrease in the abundance or absence of ICAM-1, or a decrease in the abundance or absence of IL-6, TNFα, IFNα, IFNβ, TGFβ in an ocular or adnexal tissue or fluid is indicative of inhibition of an inflammatory interleukin-1 cytokine. Similarly, a decrease or absence of activity or function of any of the above-listed components is indicative of inhibition of an inflammatory interleukin-1 cytokine.
  • Pharmaceutically-Appropriate Carriers
  • Exemplary compounds incorporated to facilitate and expedite transdermal delivery of topical compositions into ocular or adnexal tissues include, but are not limited to, alcohol (ethanol, propanol, and nonanol), fatty alcohol (lauryl alcohol), fatty acid (valeric acid, caproic acid and capric acid), fatty acid ester (isopropyl myristate and isopropyl n-hexanoate), alkyl ester (ethyl acetate and butyl acetate), polyol (propylene glycol, propanedione and hexanetriol), sulfoxide (dimethylsulfoxide and decylmethylsulfoxide), amide (urea, dimethylacetamide and pyrrolidone derivatives), surfactant (sodium lauryl sulfate, cetyltrimethylammonium bromide, polaxamers, spans, tweens, bile salts and lecithin), terpene (d-limonene, alpha-terpeneol, 1,8-cineole and menthone), and alkanone (N-heptane and N-nonane). Moreover, topically-administered compositions comprise surface adhesion molecule modulating agents including, but not limited to, a cadherin antagonist, a selectin antagonist, and an integrin antagonist.
  • Optionally, the composition further contains a compound selected from the group consisting of a physiological acceptable salt, poloxamer analogs with carbopol, carbopol/hydroxypropyl methyl cellulose (HPMC), carbopol-methyl cellulose, carboxymethylcellulose (CMC), hyaluronic acid, cyclodextrin, and petroleum.
  • Drug Delivery by Contact Lens
  • The invention comprises a contact lens and a composition that inhibits an activity of an inflammatory interleukin-1 cytokine. For example, the composition is incorporated into or coated onto said lens. The composition is chemically bound or physically entrapped by the contact lens polymer. Alternatively, a color additive is chemically bound or physically entrapped by the polymer composition that is released at the same rate as the therapeutic drug composition, such that changes in the intensity of the color additive indicate changes in the amount or dose of therapeutic drug composition remaining bound or entrapped within the polymer. Alternatively, or in addition, an ultraviolet (UV) absorber is chemically bound or physically entrapped within the contact lens polymer. The contact lens is either hydrophobic or hydrophilic.
  • Exemplary materials used to fabricate a hydrophobic lens with means to deliver the compositions of the present invention include, but are not limited to, amefocon A, amsilfocon A, aquilafocon A, arfocon A, cabufocon A, cabufocon B, carbosilfocon A, crilfocon A, crilfocon B, dimefocon A, enflufocon A, enflofocon B, erifocon A, fluorofocon A, flusilfocon A, flusilfocon B, flusilfocon C, flusilfocon D, flusilfocon E, hexafocon A, hofocon A, hybufocon A, itabisfluorofocon A, itafluorofocon A, itafocon A, itafocon B, kolfocon A, kolfocon B, kolfocon C, kolfocon D, lotifocon A, lotifocon B, lotifocon C, melafocon A, migafocon A, nefocon A, nefocon B, nefocon C, onsifocon A, oprifocon A, oxyfluflocon A, paflufocon B, paflufocon C, paflufocon D, paflufocon E, paflufocon F, pasifocon A, pasifocon B, pasifocon C, pasifocon D, pasifocon E, pemufocon A, porofocon A, porofocon B, roflufocon A, roflufocon B, roflufocon C, roflufocon D, roflufocon E, rosilfocon A, satafocon A, siflufocon A, silafocon A, sterafocon A, sulfocon A, sulfocon B, telafocon A, tisilfocon A, tolofocon A, trifocon A, unifocon A, vinafocon A, and wilofocon A.
  • Exemplary materials used to fabricate a hydrophilic lens with means to deliver the compositions of the present invention include, but are not limited to, abafilcon A, acofilcon A, acofilcon B, acquafilcon A, alofilcon A, alphafilcon A, amfilcon A, astifilcon A, atlafilcon A, balafilcon A, bisfilcon A, bufilcon A, comfilcon A, crofilcon A, cyclofilcon A, darfilcon A, deltafilcon A, deltafilcon B, dimefilcon A, droxfilcon A, elastofilcon A, epsilfilcon A, esterifilcon A, etafilcon A, focofilcon A, galyfilcon A, genfilcon A, govafilcon A, hefilcon A, hefilcon B, hefilcon C, hilafilcon A, hilafilcon B, hioxifilcon A, hioxifilcon B, hioxifilcon C, hydrofilcon A, lenefilcon A, licryfilcon A, licryfilcon B, lidofilcon A, lidofilcon B, lotrafilcon A, lotrafilcon B, mafilcon A, mesafilcon A, methafilcon B, mipafilcon A, nelfilcon A, netrafilcon A, ocufilcon A, ocufilcon B, C, ocufilcon D, ocufilcon E, ofilcon A, omafilcon A, oxyfilcon A, pentafilcon A, perfilcon A, pevafilcon A, phemfilcon A, polymacon, senofilcon A, silafilcon A, siloxyfilcon A, surfilcon A, tefilcon A, tetrafilcon A, trilfilcon A, vifilcon A, vifilcon B, and xylofilcon A.
  • EXAMPLES Example 1 The Ocular Surface Disease Index (OSDI)
  • The Ocular Surface Disease Index (OSDI) is a 12-item questionnaire that provides a rapid assessment of the symptoms of ocular irritation consistent with ocular surface disease, including posterior blepharitis and dry eye disease, and their impact on vision-related functioning (FIG. 1). The 12 items of the OSDI questionnaire are graded on a scale of 0 to 4, where 0 indicates none of the time; 1, some of the time; 2, half of the time; 3, most of the time; and 4, all of the time. The total OSDI score is then calculated on the basis of the following formula: OSDI=[sum of scores for all questions answered)×100]/[(total number of questions answered)×4]. Thus, the OSDI is scored on a scale of 0 to 100, with higher scores representing greater disability. A negative change from baseline indicates an improvement in vision-related function and the ocular inflammatory disorders described herein. For the therapeutic method described herein, treatment is considered more effective than control (vehicle) as indicated by a mean change (decrease) from baseline for the OSDI of >10 units compared to control.
  • Therapeutic treatment is considered more effective than the vehicle as indicated by a mean change from baseline of average score (0-100) for the Ocular Surface Disease Index (OSDI) of >10 units better than vehicle.
  • Example 2 Tear Film Break-Up Time
  • The standard TBUT measurement is performed by moistening a fluorescein strip with sterile non-preserved saline and applying it to the inferior tarsal conjunctiva. After several blinks, the tear film is examined using a broad beam of the slit lamp with a blue filter. The time lapse between the last blink and the appearance of the first randomly distributed dark discontinuity in the fluorescein stained tear film is measured three times and the mean value of the measurements is calculated. The tear break-up time is evaluated prior to the instillation of any eye drops and before the eyelids are manipulated in any way. Break-up times less than 10 seconds are considered abnormal. A positive change from baseline indicates improvement in symptoms of the ocular inflammatory disorders described herein. The treatment described herein, leads to an improvement in TBUT significantly greater than that observed from treatment with vehicle alone.
  • Example 3 Corneal and Conjunctival Staining
  • Corneal staining is a measure of epithelial disease, or break in the epithelial barrier of the ocular surface, typically seen with ocular surface disorders such as posterior blepharitis and dry eye, among others. Importantly, corneal staining can exist even without clinically evident dry eye, if there is significant lid disease, such as posterior blepharitis. Corneal staining is highly correlated with ocular discomfort in many, though not all patients; in general corneal staining is associated with high scores in the OSDI, as described above. For corneal fluorescein staining, saline-moistened fluorescein strips or 1% sodium fluorescein solution are used to stain the tear film. The entire cornea is then examined using slit-lamp evaluation with a yellow barrier filter (#12 Wratten) and cobalt blue illumination (staining is more intense when it is observed with a yellow filter). Staining is graded according to the Oxford Schema (FIG. 2).
  • Conjunctival staining is a measure of epithelial disease or break in the epithelial barrier of the ocular surface, typically seen with ocular surface disorders such as posterior blepharitis and dry eye, among others. Importantly, conjunctival staining, similar to corneal staining, can exist even without clinically evident dry eye, if there is significant lid disease, such as posterior blepharitis. Conjunctival staining can also correlate with symptoms of ocular irritation and high OSDI scores as described above. Conjunctival staining is performed under the slit-lamp using lissamine green. Saline-moistened strip or 1% lissamine green solution is used to stain the tear film, and interpalpebral conjunctival staining is evaluated more than 30 seconds, but less than 2 minutes, later. Using white light of moderate intensity, only the interpalpebral region of the nasal and temporal conjunctival staining is graded using the Oxford Schema (above). The treatment described herein leads to decreases in ocular staining scores beyond what is observed with the vehicle alone.
  • Therapeutic treatment is considered more effective than vehicle as indicated by a mean change from baseline in average score (0-5 scale) for corneal and conjunctival staining of >1 unit better than vehicle, e.g. as detected using the Oxford Schema.
  • Example 4 Schirmer Test
  • The Schirmer test is performed in the presence and in the absence of anesthesia by placing a narrow filter-paper strip (5×3 5 mm strip of Whatman #41 filter paper) in the inferior cul-de-sac. This test is conducted in a dimly lit room. The patient gently closes his/her eyes until five minutes have elapsed and the strips are removed. Because the tear front will continue advancing a few millimeters after it has been removed from the eyes, the tear front is marked with a ball-point pen at precisely five minutes. Aqueous tear production is measured by the length in millimeters that the strip wets during 5 minutes. Results of 10 mm or less for the Schirmer test without anesthesia and 5 mm or less for the Schirmer test with anesthesia are considered abnormal. A positive change from baseline indicates improvement of one or more symptoms of an ocular inflammatory disorder described herein.
  • Example 5 Meibomian Gland Evaluation
  • In the center of the lower lid, 10 adjacent central glands are located on both sides and the glands are expressed by applying a firm digital pressure at the base of the glands. The number of glands expressed for each eye is documented. The quality of secretion is described as follows:
  • Clear excreta or clear with small particles (0)
  • Opaque excreta with normal viscosity (1)
  • Opaque excreta with increased viscosity (2)
  • Secretions retain shape after expression (3)
  • Posterior blepharitis is associated with lid inflammation and alterations in the quantity and/or quality of the meibomian gland secretions, with severe disease associated with quality grades 2-3, as described above. The treatment described herein leads to improvement in meibomian secretion characterized by a decrease in this score; for example, from 3 to 2, or from 2 to 1. An improvement is indicated by a mean change from baseline (0-3 scale) for meibomian gland secretion quality of >1 unit better than vehicle.
  • Example 6 Lid and Lid Margin Erythema
  • Lid margin vascular injection (erythema) is defined as a red discoloration, compared to the surrounding eyelid skin and is graded as follows:
  • None (0): none
    Mild (1): redness localized to a small region of the lid margin(s) or skin
    Moderate (2): redness of most of the lid margin(s)
    Severe (3): redness of most or all the lid margin(s) and skin
    Very Severe (4): marked diffuse redness of both lid margins and skin
    The presence or absence of tarsal telangiectasis is also noted. Lid telangiectasia is defined as the presence of at least two blood vessels along the eyelid margin.
  • Example 7 Conjunctiva Hyperemia
  • Bulbar conjunctival hyperemia is graded as follows:
  • None (0): none
    Mild (1): slight localized injection
    Moderate (2): pink color, confined to palpebral or bulbar conjunctiva
    Severe (3): red color of the palpebral and/or bulbar conjunctiva
    Very Severe (4): marked dark redness of the palpebral and/or bulbar conjunctiva
    The presence or absence of tarsal papillary hypertrophy is also noted.
  • Example 8 Topical Administration of IL-1Ra for Treating Posterior Blepharitis
  • The following study evaluates the therapeutic benefit of topically administering a solution comprising a known, and commercially-available, recombinant IL-1 receptor antagonist, Anakinra (Kineret®), versus vehicle to subjects with inflammatory conditions affecting one or more ocular and/or adnexal tissues.
  • The following is a prospective, single-center, randomized, double-masked, vehicle-controlled, parallel-group clinical study. There is one active treatment group and one vehicle treated group. Patients who meet the requirements of the inclusion/exclusion criteria at the screening visit are separated into a moderate stratum and a severe stratum based on the meibomian gland secretion quality. Within each stratum, patients are randomized to either 2.5% topical human recombinant IL-1Ra or vehicle in even allocations. There are a minimum of 20 patients in each treatment group in the moderate stratum and a minimum of 10 patients in each treatment group in the severe stratum. This study consists of 6 scheduled visits over four months (Table 2).
  • Subjects who present signs or reported symptoms consistent with inflammatory disease affecting an ocular or adnexal tissue are further evaluated prior to treatment using the above-described Ocular Surface Disease Index (OSDI). Exemplary subjects are Male and female patients with signs and symptoms of posterior blepharitis (provided that not more than 7 glands are not expressible) with or without aqueous deficiency excluding those patients with end-stage lacrimal gland (Schirmer reading without anesthesia <3 mm/5 min or if their dry eye disease is the result of destruction of conjunctival goblet cells or scarring). A subject is included in the following study if he or she meets the following criteria: male or female; at least 18 years of age; has not worn contact lenses for at least 2 weeks prior to the study and agrees to not wear contact lenses during study; patient is in generally good & stable overall health. patient must have a diagnosis of posterior blepharitis as defined in FIG. 5; a negative urine pregnancy test result for women of childbearing potential; women of childbearing potential must agree to use adequate contraception (hormonal or barrier method of birth control) prior to study entry and for the duration of study participation; normal lid position and closure; ability to understand and provide informed consent to participate in this study; and willingness to follow study instructions and likely to complete all required visits.
  • Subjects are excluded if they had used topical steroids or the commercially-available drug, Restasis, within the past 2 weeks as well as tetracycline compounds (tetracycline, doxycycline, and minocycline) within the last month or isotretinoin (Accutane) within the past 6 months. Subjects who report any previous treatment with Anakinra (Kineret®) or any therapeutic agent targeted at IL-1 blockade are similarly excluded. Furthermore, a subject is excluded from the following study if he or she: has a history of Stevens-Johnson syndrome or ocular pemphigoid; has a history of eyelid surgery; has had intra-ocular surgery or ocular laser surgery within 3 months; has a history of microbial keratitis, including herpes; has active ocular allergies; has a corneal epithelial defect >1 mm2; has used topical steroids or Restasis within the past 2 weeks; has experienced any change in dosage of tetracycline compounds (tetracycline, doxycycline, and minocycline) within the last month; has used isotretinoin (Accutane) within the past 6 months; has had any previous treatment with Anakinra (Kineret®) or any therapeutic agent targeted at IL-1 blockade; is a pregnant or lactating woman; has signs of current infection, including fever and current treatment with antibiotics; has liver, renal, or hematologic disease; has a history of cancer; has used any other investigational drug.
  • Under certain circumstances, subjects are withdrawn from the following study. The following criteria are used to determine when a subject must be removed or permitted to leave the study. Individuals are discontinued early from the study due to the following reasons, including, but not limited to: protocol violations, adverse events, lack of efficacy, pregnancy, and administrative reasons (e.g., inability to continue, lost to follow-up).
  • Screening Visit
  • Prospective patients as defined by inclusion/exclusion criteria are considered for entry into this study. The study design and treatment regimen are discussed with each patient. Those wishing to participate are examined for entry into the study with the following exams:
  • Medical and ophthalmic histories
  • Patient questionnaire (OSDI) (see Example 1 and FIG. 1)
  • Best corrected visual acuity (BCVA) (Snellen chart)
  • Fluorescein tear break-up time (TBUT) (see Example 2)
  • Cornea and conjunctival staining (see Example 3
  • Schirmer test without anesthesia (see Example 4)
  • Schirmer test with anesthesia (see Example 4)
  • Meibomian gland evaluation (see Example 5)
  • Biomicroscopy
  • Intraocular pressure
  • Fundus examination
  • To avoid the influence of one procedure on another, dry eye tests and ocular surface evaluation are done in the following sequence because the Schirmer test can disrupt tear film stability and cause false-positive ocular surface dye staining: measurement of tear film break-up time (TBUT), ocular surface dye staining pattern (fluorescein and lissamine green staining), Schirmer's test without and with anesthesia, and meibomian gland evaluation (FIG. 6).
  • After the eye examinations, each patient who qualifies to continue in the study (according to the inclusion/exclusion criteria) is instructed to instill one drop of the randomly assigned masked treatment three times a day in both eyes. The topical solution of 2.5% (25 mg/ml) concentration of Kineret is formulated and prepared from commercially available preservative-free solution of Kineret® (Amgen, Thousand Oaks, Calif.) by the MEEI hospital pharmacy using aseptic technique. For the control group, vehicle [Refresh Liquigel (1% Carboxymethylcellulose)] is used three times a day in both eyes. During the treatment phase, patients are instructed to instill 1 drop of study medication three times a day for 3 months; 1 drop in each eye upon waking in the morning, at noon, and at bedtime.
  • To reduce the chance of systemic absorption of Kineret, patients are asked to put digital pressure on lacrimal ducts for five minutes. Patients are advised not use artificial tears or other topical medications 30 minutes before or 30 minutes after instilling the study medication to prevent dilution of the study medication. This study does not include blood sampling or any pharmacokinetic measures.
  • Compliance Visit
  • Each patient returns for a compliance visit at 2 weeks (Table 2). At this visit, the investigator asks direct questions about patient compliance with the study treatment and adverse events. Patients can be discontinued from the study because of adverse events and protocol violations, but every effort is made to enhance compliance and maintain subjects on the treatment protocol.
  • Follow-Up Visits
  • To determine the safety and efficacy of topical IL-1Ra for management of posterior blepharitis, 5 follow-up visits (eye exam) are scheduled at day 1, week 2, 6, 12, and 16 (see Table 2). Patients are instructed to strictly follow the study visit schedule. In each follow-up visit, the patient is asked to fill out the OSDI questionnaires (FIG. 1). In addition to a comprehensive eye examination, tear function and ocular surface tests including TBUT, cornea and conjunctival staining tests, meibomian gland evaluation, and Schirmer tests, are performed.
  • TABLE 2
    Schedule of Events and Procedures
    Visit
    1 2 3 4 5 6
    Time Period Screening Day 1 Week 2 Week 6 Week 12 Week 16
    Visit (Safety (Compliance
    Visit) Visit)
    Obtain Informed X
    Consent
    Inclusion/exclusion X
    criteria
    OSDI questionnaire X X X X X
    Medical & X
    Ophthalmic History
    Pregnancy Test X
    BCVA X X X X X
    TBUT X X X X X
    Cornea & Conj. X X X X X
    Staining
    Schirmer without X X X X X
    Anesthesia
    Schirmer with X X X X X
    Anesthesia
    Meibomian Gland X X X X X
    Evaluation
    Biomicroscopy X X X X X X
    Intraocular Pressure X X X X X
    Funduscopy X X X X X
    Compliance Check X X X X
    Adverse Event Query X X X X X
  • Concurrent Therapies
  • Concurrent enrollment in another clinical investigational drug or device study is prohibited. The use of any concurrent medication, prescription or over-the-counter, is recorded along with the reason the medication is taken. During the study, all concomitant medication treatment regimens, ocular hygiene treatments (i.e., lid scrubs and warm compresses), or insertion of punctal plugs are kept as constant as permitted by accepted medical practice.
  • Using artificial tears is permissible at screening visits and during the study. However, concomitant use of artificial tears is monitored. At each study visit, patients are queried about the average number of times they used artificial tears each day during the past week and the number of days during the past week when they did not use any artificial tears.
  • Systemic and topical ophthalmic medications that could interfere with the response to study medications or the interpretation of the study results are prohibited during the study. This includes any systemic or topical steroid, cyclosporine, and tetracycline compound.
  • When necessary for the treatment of patients, administration of a prohibited therapy is done with the safety of the study participant as the primary consideration. The administration of a prohibited medication or procedure is considered a protocol violation and the patient may be discontinued from the study.
  • During the course of study, if the patient complains of mild to moderate form of ocular surface symptoms, more frequent use of artificial tears (Refresh®) is encouraged without unmasking the patient. If treatment with artificial tears is inadequate, or the patient develops a severe form of ocular surface disease and dry eye, for the safety and proper treatment of the subject, the investigator can unmask the subject's treatment assignment to determine which treatment has been assigned and institute appropriate follow-up care. However, the patient is kept in the study so that an “intention to treat” analysis can be performed.
  • Efficacy Measures
  • Several signs of the efficacy of administration of therapeutic compounds of the invention are monitored. The objective signs are meibomian gland secretion quality, meibomian gland occlusion, tear break-up time, corneal and conjunctival staining, and Schirmer test (with and without anesthesia). The subjective endpoints are the OSDI questionnaire score. All these variables are carefully measured at baseline and all visits toward the end of follow up (Table 2).
  • The following are non-limiting examples of primary efficacy variables: Meibomian gland secretion quality; Tear Break-up time; Cornea and Conjunctival staining score; and the OSDI questionnaire score. Alternatively, or in addition, secondary efficacy variables are used to determine the therapeutic value of administration of compositions of the invention. Nonlimiting examples of secondary efficacy variables include: Meibomian gland occlusion; Schirmer test without anesthesia; Schirmer test with anesthesia. Primary and/or secondary efficacy variables are considered.
  • In this study, the treatment with topical IL-1Ra is considered efficacious if the 2.5% solution shows a mean change from baseline (0-3 scale) for meibomian gland secretion quality of >1 unit better than vehicle at the week-12 visit. Alternatively, the treatment with topical IL-1Ra is considered efficacious if the 2.5% solution shows a mean change from baseline in average score (0-5 scale) for corneal and conjunctival staining of >1 unit better than vehicle at the week-12 visit.
  • Safety Measures
  • The primary safety variable monitored is the occurrence of adverse events. The severity of each adverse event observed (ocular and systemic) is rated from mild (awareness of sign or symptom, but easily tolerated) to severe (incapacitating with inability to work or do usual activity). The relationship of the event to the study medication is assessed by the investigator as none, unlikely, possible, probable, or definite. Safety variables are evaluated at baseline and at all study visits.
  • At each visit throughout the study, the investigator begins querying for adverse events by asking each patient a general, non-directed question such as “How have you been feeling since the last visit?” Directed questioning and examination is then done as appropriate. The investigator asks questions to subjects at each visit to determine if they have had any changes to the use of concomitant medications since the previous visit. A comprehensive eye examination including best-corrected visual acuity, measuring intraocular pressure, evaluation of the condition of the lid/lashes, conjunctiva, cornea, anterior chamber, iris/pupil, lens, vitreous, macula and optic nerve is performed. Any changes in the study eye from the baseline visit is recorded.
  • Statistical Procedures Power and Sample Size Considerations:
  • TABLE 3
    Estimated Prevalence for Anticipated Ordered
    Categories of Meibomian Quality Scores
    Scenario Meibomian
    High Moderate Low Score
    0.20 0.28 0.40 0
    0.28 0.32 0.28 1
    0.32 0.25 0.20 2
    0.20 0.20 0.12 3
    Mean (SD) = Mean (SD) = 1.32 Mean (SD) = 1.04
    1.52 (1.09) (1.04)
    (1.02)
  • In order to estimate the power of the study, it is assumed that 50% of the subjects are randomized to topical IL1-Ra and 50% to vehicle. A conservative estimate can then be constructed of study power, according to Sullivan and D'Agostino (Sullivan, L. M. and D'Agostino, R. B. 2003. Stat Med. 22(8):1317-34), using a t-test comparison of the predicted distribution of primary endpoints within ordered categories. Power estimates using this method are conservative because the actual analysis uses data from two eyes while appropriately accounting for their correlation, which has been demonstrated to have greater power as compared to methods based on using the person as the unit of analysis.
  • The work of Sullivan and D'Agostino indicates that the t-test performs well for the comparison of ordinal scales with 3 to 5 ordinal categories. Estimates of power are based on three scenarios (high, moderate, and low) for the anticipated distribution of scores in the placebo group. As displayed in Table 3 above, a mean score between 1.04 and 1.52 is anticipated, with a standard deviation between 1.02 and 1.09. A clinically meaningful difference is a reduction of 1 in the mean score, or approximately 1 standard deviation.
  • Based on the two-sample t-test, with Bonferonni adjustment for multiple comparisons (four primary outcome variables) an observer would have ≧80% power to detect a mean difference of 1 standard deviation for sample sizes of 28, 30, and 28, per group, under the high, moderate, and low scenarios, respectively. As noted, actual power would be enhanced through use of scores from both eyes of study participants, with consideration of the correlation between fellow eyes in the analysis. Therefore, the sample size of N=30 per group was chosen.
  • Statistical Analysis:
  • For efficacy variables and any other variables except for safety, all subjects are analyzed with the treatment to which they were randomized (the intent-to-treat population). For safety variables, subjects are analyzed with the treatment actually received (the safety population).
  • The primary analysis for the proposed study is based on a standard intention-to-treat analysis with each study participant analyzed with respect to the randomized treatment assignment, regardless of eventual compliance. A secondary analysis may include imputation of missing data for select variables. A per-protocol analysis, disqualifying patients or patient visits, might also be done, but is not planned because observational analyses of actual treatment use could introduce bias if the pattern of use is in some way related to the outcome.
  • Despite the randomized nature of the treatment assignments, in this relatively small sample of study subjects there may be imbalances with regard to potential confounding variables. Thus, as an initial step in the analysis, those assigned to active treatment versus those randomized to vehicle are compared with regard to demographic characteristics and potential confounding variables, using the non-parametric Wilcoxon rank sum test for continuous or ordinal variables, and chi-square or Fisher's exact tests for categorical variables.
  • To assess the effect of topical IL-1Ra treatment, the distribution of a) meibomian gland secretion score, and b) corneal and conjunctival staining scores between patients assigned to topical IL-1Ra and those assigned to vehicle are compared using the stratified Wilcoxon rank sum test with variance correction for clustering effects as recently developed by Rosner and Glynn (Rosner, et al. 2003. Biometrics. 59(4): 1089-98). Based on simulation studies, analyses of ophthalmologic data that use the information from two eyes and appropriately account for their correlation have greater power than methods based on a single eye or the average of the two eyes (Rosner, et al. 2003. Biometrics. 59(4): 1089-98; Glynn, R. J. and Rosner, B. 1994. Stat Med. 13(10): 1023-36). This method uses large sample theory to incorporate clustering effects to ordinal outcomes such as the clinical scoring scales used here. It can be implemented with standard software (e.g. SAS PROC RANK), and provides a valid test for either balanced or unbalanced clustered data in the presence or absence of tied rankings in datasets in which there are ≧20 clusters per group. The test may be used in place of the cluster-mean procedure (e.g. as would result from analysis using SAS PROC MIXED), and provides unbiased p-values where the standard Wilcoxon test is inappropriate. These analyses are then extended to control for potential confounding by variables using the stratified version of the test. A logical cut-point for stratification of continuous variables would be the median level among the vehicle treated group.
  • Prior to the development of these methods, studies have generally used the person as the unit of analysis, using either a composite score or data from one eye per subject. However, information is potentially lost by collapsing eye-specific grades into a single person-specific grade.
  • In secondary analyses, the effect of topical IL-1Ra on other outcomes such as the OSDI score and tear break up time is also explored. Similarly, these analyses use the stratified Wilcoxon test with variance correction.
  • In general, a two-sided test with p-value less than or equal to 0.0125 is considered statistically significant. This level of significance was arrived at by dividing the overall type 1 error rate of 0.05 by 4, which is the number of comparisons in the primary analysis.
  • Although patients are evaluated at multiple time points throughout the study, the primary endpoint is on the last observation while on treatment during the 3-month study period.
  • Example 9 Effect of IL-1Ra on Intraocular Pressure (IOP)
  • IL-1Ra was administered to one eye of wild type BALB/c mice and the mean intraocular pressure (IOP) of the both the treated and the non-treated (contralateral eye) were measured. Data from these experiments show that administration of IL-1Ra is sufficient to reduce IOP by a statistically significant amount over the course of one day, and particularly, over the duration of one night (Table 4, below, and FIG. 3). IOP is a risk factor for the development of glaucoma. Importantly, the compositions and methods used were effective on wild type, or normal, subjects. As such, this example is proof-of-concept that the compositions and methods of the invention are effective for treating elevated intraocular pressure or ocular hypertension that has been caused by a variety of mechanisms, including, but not limited to IL-1-mediated inflammation.
  • TABLE 4
    Effect of one drop of topical IL-1Ra (2.5%) on Intraocular pressure
    (IOP) during the Day (after 6 hours) and Overnight (after 12 hours)
    in BALB/c mice. Data are expressed as the mean IOP (mm Hg) ± SEM.
    P value is for treated eye versus the contralateral eye (paired t-test).
    Difference
    Contralateral (Contralateral P
    N Eye Treated Eye Eye − Treated Eye) % Reduction value
    Day
    5  9.1 ± 0.46  8.0 ± 0.27 1.1 ± 0.51 11.5 ± 0.05 0.095
    (after 6 h)
    Overnight 5 13.7 ± 0.90 10.3 ± 0.75 3.4 ± 0.38 24.7 ± 0.02 0.0009
    (After 12 h)
  • OTHER EMBODIMENTS
  • While the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
  • The patent and scientific literature referred to herein establishes the knowledge that is available to those with skill in the art. All United States patents and published or unpublished United States patent applications cited herein are incorporated by reference. All published foreign patents and patent applications cited herein are hereby incorporated by reference. Genbank and NCBI submissions indicated by accession number cited herein are hereby incorporated by reference. All other published references, documents, manuscripts and scientific literature cited herein are hereby incorporated by reference.
  • While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (24)

What is claimed is:
1. A method for inhibiting or reducing the severity of an inflammatory disorder affecting the ocular and adnexal tissues, comprising locally administering to ocular and adnexal tissue of a subject a therapeutically effective amount of a composition that inhibits an activity of an inflammatory interleukin-1 cytokine.
2. The method of claim 1, wherein the therapeutically effective amount of a composition is administered when the inflammatory disorder of the subject is improved after administration of the composition compared to the inflammatory disorder in the subject prior to treatment, wherein the improvement is determined using one or more tests selected from the group consisting of meibomian gland secretion quality, meibomian gland occlusion, tear break-up time, corneal and conjunctival staining, Schirmer test with anesthesia, Schirmer test without anesthesia, or ocular surface disease index (OSDI) score.
3. The method of claim 2, wherein a therapeutically effective amount of the composition is determined
a. by using meibomian gland secretion quality, wherein a mean change in meibomian gland secretion quality from untreated (0-3 scale) of greater than 1 unit by 12 weeks of treatment indicates a therapeutically effective amount was administered; or
b. by using corneal and conjunctival staining, wherein a mean change in average score from untreated (0-5 scale) for corneal and conjunctival staining of greater than 1 unit by 12 weeks after initiation of treatment indicates a therapeutically effective amount was administered.
4. The method of claim 1, wherein the activity comprises binding of an inflammatory IL-1 cytokine to an IL-1 receptor.
5. The method of claim 1, further comprising identifying a subject characterized as suffering from an inflammatory disorder affecting the ocular and adnexal tissues.
6. The method of claim 3, wherein identifying a subject comprises detection of a sign or symptom selected from the group consisting of epithelial overexpression of an inflammatory cytokine, vascular hyperplasia or thickening of lid margin, neovascularization of lid margin or corneal periphery, increase of leukocytes at an ocular surface, or overexpression of a matrix metalloprotease at an ocular surface and wherein the method inhibits or reduces the severity of at least one of the signs or symptoms.
7. The method of claim 1, wherein the inflammatory disorder is characterized as infectious blepharitis, non-infectious blepharitis, meibomian gland dysfunction, dry eye, or Sjogren syndrome.
8. The method of claim 1, wherein the method further comprises administration of an antibiotic compound.
9. The method of claim 1, wherein the composition that inhibits binding of an inflammatory IL-1 cytokine to an IL-1 receptor comprises an amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
10. The method of claim, 1 wherein the composition is present in a concentration of 0.1-10% (mg/ml).
11. The method of claim 1, wherein the form of said composition is a solid, a paste, an ointment, a gel, a liquid, an aerosol, a mist, a polymer, a film, an emulsion, or a suspension.
12. The method of claim 1, wherein the composition is administered topically.
13. The method of claim 1, wherein the method does not comprise systemic administration or substantial dissemination to non-ocular tissue.
14. The method of claim 13, wherein seven hours after a single administration the composition is at a serum concentration less than 1200 ng/ml.
15. The method of claim 1, wherein the composition further comprises a compound selected from the group consisting of a physiological acceptable salt, poloxamer analogs with carbopol, carbopol/HPMC, carbopol-methyl cellulose, carboxymethylcellulose (CMC), hyaluronic acid, cyclodextrin, and petroleum.
16. A formulation comprising a therapeutically effective amount of a composition that inhibits an activity of an inflammatory interleukin-1 cytokine, the formulation being in the form of a solid, a paste, a liquid, an ointment, a gel, an aerosol, a mist, a polymer, a film, an emulsion, or a suspension, and wherein the composition is present at a concentration of 0.1-10% (mg/ml).
17. The formulation of claim 16, wherein said composition comprises a polypeptide comprising the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
18. A method for inhibiting or reducing the severity of an ocular inflammatory disorder, comprising locally administering to the ocular or adnexal tissue of a subject a therapeutically effective amount of a composition that can inhibit transcription, transcript stability, translation, modification, localization, secretion, or function of a polynucleotide or polypeptide encoding an inflammatory interleukin-1 cytokine or an IL-1 receptor.
19. The method of claim 18, wherein the composition comprises a polynucleotide, a polypeptide, an antibody, or a small molecule.
20. The method of claim 18, wherein said composition is administered topically.
21. A device comprising a polymer and a bioactive composition incorporated into or onto said polymer, wherein said bioactive composition inhibits an activity of an inflammatory interleukin-1 cytokine, and wherein said device is incorporated into or onto an ocular or adnexal tissue.
22. The device of claim 21, wherein the device is a contact lens.
23. The method of claim 1, wherein the method comprises administration of a composition that can inhibit binding of an inflammatory IL-1 cytokine to an IL-1 receptor and a second composition comprising one or more inflammatory antagonist(s).
24. The method of claim 23, wherein the second composition can inhibit a tumor necrosis factor alpha (TNFα), one or more interleukin cytokines, one or more member(s) of the vascular epithelial growth factor (VEGF) family, interferon-gamma, or one or more chemokines and their receptors, or is an immunosuppressant.
US13/795,327 2007-08-16 2013-03-12 Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues Abandoned US20130273065A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/795,327 US20130273065A1 (en) 2007-08-16 2013-03-12 Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US96513507P 2007-08-16 2007-08-16
US13068708P 2008-06-02 2008-06-02
PCT/US2008/009776 WO2009025763A2 (en) 2007-08-16 2008-08-15 Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
US29838010A 2010-04-28 2010-04-28
US13/795,327 US20130273065A1 (en) 2007-08-16 2013-03-12 Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/009776 Continuation WO2009025763A2 (en) 2007-08-16 2008-08-15 Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
US29838010A Continuation 2007-08-16 2010-04-28

Publications (1)

Publication Number Publication Date
US20130273065A1 true US20130273065A1 (en) 2013-10-17

Family

ID=40378866

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/298,380 Active 2030-06-30 US10105441B2 (en) 2007-08-16 2008-08-15 Method for inhibiting or reducing dry eye disease by IL-1Ra
US13/795,327 Abandoned US20130273065A1 (en) 2007-08-16 2013-03-12 Therapeutic Compositions For Treatment Of Inflammation Of Ocular And Adnexal Tissues

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/298,380 Active 2030-06-30 US10105441B2 (en) 2007-08-16 2008-08-15 Method for inhibiting or reducing dry eye disease by IL-1Ra

Country Status (5)

Country Link
US (2) US10105441B2 (en)
EP (1) EP2187900B1 (en)
JP (2) JP5894364B2 (en)
AU (1) AU2008289552B2 (en)
WO (1) WO2009025763A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9956195B2 (en) 2014-01-07 2018-05-01 Nanyang Technological University Stable liposomal formulations for ocular drug delivery
US10272040B2 (en) 2010-08-12 2019-04-30 Nanyang Technological University Liposomal formulation for ocular drug delivery
US11612590B2 (en) 2013-03-13 2023-03-28 Santen Pharmaceutical Co., Ltd. Therapeutic agent for meibomian dysfunction

Families Citing this family (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10105441B2 (en) * 2007-08-16 2018-10-23 The Schepens Eye Research Institute, Inc. Method for inhibiting or reducing dry eye disease by IL-1Ra
US20120014970A1 (en) * 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders
AU2010203425B2 (en) 2009-01-09 2015-04-09 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of corneal disorders
US8574277B2 (en) 2009-10-21 2013-11-05 Avedro Inc. Eye therapy
EP2538965B1 (en) 2010-02-25 2017-04-12 Schepens Eye Research Institute Therapeutic compositions for the treatment of dry eye disease
JP6377906B2 (en) 2010-03-19 2018-08-22 アヴェドロ・インコーポレーテッドAvedro,Inc. System for applying and monitoring eye treatment
US9095591B2 (en) * 2010-06-28 2015-08-04 Institut National De La Sante Et De La Recherche Medicale (Inserm) Pharmaceutical composition for use in the treatment of glaucoma
SG186474A1 (en) * 2010-06-30 2013-02-28 Johnson & Johnson Vision Care Ophthalmic devices containing chemokine antagonists
NZ605928A (en) 2010-07-29 2015-02-27 Eleven Biotherapeutics Inc Chimeric il-1 receptor type i agonists and antagonists
WO2012148547A1 (en) * 2011-02-24 2012-11-01 The Schepens Eye Research Institute, Inc. Compositions and methods for treating inflammatory conditions of the ocular surface
US20140147413A1 (en) * 2011-02-28 2014-05-29 Dong Feng Chen Therapies That Target Autoimmunity For Treating Glaucoma And Optic Neuropathy
ES2600616T3 (en) 2011-03-14 2017-02-10 Phlogo Aps Interleukin-1 receptor antagonists
US10451897B2 (en) 2011-03-18 2019-10-22 Johnson & Johnson Vision Care, Inc. Components with multiple energization elements for biomedical devices
WO2012162529A1 (en) 2011-05-24 2012-11-29 Avedro, Inc. Systems and methods for reshaping an eye feature
JP6122845B2 (en) 2011-06-02 2017-04-26 アヴェドロ・インコーポレーテッドAvedro,Inc. System and method for monitoring the delivery of time-based photoactive agents or the presence of photoactive markers
KR20140054002A (en) * 2011-06-29 2014-05-08 인사이트 비젼 인코포레이티드 Methods of treating recurrent meibomian glands disorder and thereby decreasing the frequency of recurrence
US8865685B2 (en) 2011-06-30 2014-10-21 Johnson & Johnson Vision Care, Inc. Esters for treatment of ocular inflammatory conditions
US9812730B2 (en) 2011-08-02 2017-11-07 Johnson & Johnson Vision Care, Inc. Biocompatible wire battery
US8857983B2 (en) 2012-01-26 2014-10-14 Johnson & Johnson Vision Care, Inc. Ophthalmic lens assembly having an integrated antenna structure
WO2013169382A1 (en) * 2012-05-07 2013-11-14 DePuy Synthes Products, LLC Methods and devices for treating intervertebral disc disease
EP2872081B1 (en) 2012-07-16 2022-06-08 Avedro, Inc. Systems for corneal cross-linking with pulsed light
US20150307619A1 (en) * 2012-12-13 2015-10-29 The Schepens Eye Research Institute, Inc. Use of C-C Chemokine Receptor Type 7 (CCR7) Inhibitors
DK2948134T3 (en) 2013-01-24 2020-06-02 Palvella Therapeutics Inc COMPOSITIONS FOR TRANSDERMAL ADMINISTRATION OF MTOR INHIBITORS
DK2968468T3 (en) 2013-03-13 2021-07-26 Buzzard Pharmaceuticals AB Chimeric cytokine formulations for ocular delivery
JP6340673B2 (en) 2013-05-02 2018-06-13 レティナ ファウンデーション オブ ザ サウスウエストRetina Foundation Of The Southwest Double-layered ocular implant
EP2999471B1 (en) * 2013-05-19 2020-12-16 Avedro, Inc. Systems, methods, and compositions for cross-linking
US9498114B2 (en) 2013-06-18 2016-11-22 Avedro, Inc. Systems and methods for determining biomechanical properties of the eye for applying treatment
US9498122B2 (en) 2013-06-18 2016-11-22 Avedro, Inc. Systems and methods for determining biomechanical properties of the eye for applying treatment
US10361405B2 (en) 2014-08-21 2019-07-23 Johnson & Johnson Vision Care, Inc. Biomedical energization elements with polymer electrolytes
US10627651B2 (en) 2014-08-21 2020-04-21 Johnson & Johnson Vision Care, Inc. Methods and apparatus to form biocompatible energization primary elements for biomedical devices with electroless sealing layers
US10361404B2 (en) 2014-08-21 2019-07-23 Johnson & Johnson Vision Care, Inc. Anodes for use in biocompatible energization elements
US9793536B2 (en) 2014-08-21 2017-10-17 Johnson & Johnson Vision Care, Inc. Pellet form cathode for use in a biocompatible battery
US9599842B2 (en) 2014-08-21 2017-03-21 Johnson & Johnson Vision Care, Inc. Device and methods for sealing and encapsulation for biocompatible energization elements
US10381687B2 (en) 2014-08-21 2019-08-13 Johnson & Johnson Vision Care, Inc. Methods of forming biocompatible rechargable energization elements for biomedical devices
US9941547B2 (en) 2014-08-21 2018-04-10 Johnson & Johnson Vision Care, Inc. Biomedical energization elements with polymer electrolytes and cavity structures
US9383593B2 (en) 2014-08-21 2016-07-05 Johnson & Johnson Vision Care, Inc. Methods to form biocompatible energization elements for biomedical devices comprising laminates and placed separators
US9715130B2 (en) 2014-08-21 2017-07-25 Johnson & Johnson Vision Care, Inc. Methods and apparatus to form separators for biocompatible energization elements for biomedical devices
JP6644799B2 (en) 2014-10-27 2020-02-12 アヴェドロ・インコーポレーテッドAvedro,Inc. System and method for ocular crosslinking treatment
WO2016077747A1 (en) 2014-11-13 2016-05-19 Avedro, Inc. Multipass virtually imaged phased array etalon
WO2016145237A1 (en) * 2015-03-11 2016-09-15 Yu Fu-Shin X Composition and methods to promote wound healing
EP3285704B1 (en) 2015-04-24 2020-11-18 Avedro Inc. Systems for photoactivating a photosensitizer applied to an eye
WO2016174082A1 (en) * 2015-04-28 2016-11-03 Swedish Orphan Biovitrum Ab (Publ) Compositions comprising anakinra
US10028657B2 (en) 2015-05-22 2018-07-24 Avedro, Inc. Systems and methods for monitoring cross-linking activity for corneal treatments
EP3324973B1 (en) 2015-07-21 2020-06-03 Avedro, Inc. Treament of an eye with a photosensitizer
US10345620B2 (en) 2016-02-18 2019-07-09 Johnson & Johnson Vision Care, Inc. Methods and apparatus to form biocompatible energization elements incorporating fuel cells for biomedical devices
CN105833234A (en) * 2016-05-20 2016-08-10 刘传彬 Pure natural traditional Chinese medicine composition for treating muscle and bone pain through heating and external applying
KR20190068575A (en) * 2016-10-14 2019-06-18 이콤 메디칼 게엠베하 Establishment of ocular surface homeostasis, recovery and preservation method
CA3049402A1 (en) 2017-01-06 2018-07-12 Palvella Therapeutics Llc Anhydrous compositions of mtor inhibitors and methods of use
CN110997725A (en) 2017-06-12 2020-04-10 蓝鳍生物医药公司 anti-IL 1RAP antibodies and antibody drug conjugates
US20210007670A1 (en) * 2018-03-13 2021-01-14 Menicon Co., Ltd. Determination system, computing device, determination method, and program
CN108707195B (en) * 2018-05-31 2019-03-15 苏州大学 3 gene of Mandarin fish IFN-α, recombinant protein, preparation method and application
EP3817743A4 (en) 2018-07-02 2022-07-06 Palvella Therapeutics, Inc. Anhydrous compositions of mtor inhibitors and methods of use
CN110437043A (en) * 2019-08-24 2019-11-12 黄泳华 The eutectic that is made of resveratrol and prostaglandin analogue agent and its purposes in the preparation of antitumor drugs
US20210308154A1 (en) 2020-03-24 2021-10-07 Hovione Scientia Limited Methods and Compositions for Treating Meibomian Gland Dysfunction

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5895812A (en) * 1995-05-01 1999-04-20 The University Of Virginia Alumni Patent Foundation Diagnostic for Sjogren's syndrome
US20070280924A1 (en) * 2004-03-02 2007-12-06 Julie Daniels Pharmaceutical Preparations For And Treatment Of Ocular Surface and Other Disorders
US20080132475A1 (en) * 2006-12-05 2008-06-05 Charles Gerald Connor Treatment for dry eye
US20080286378A1 (en) * 2005-02-22 2008-11-20 Ashley Behrens Use of Amniotic Fluid (Af) in Treating Ocular Disease and Injury
US20080312194A1 (en) * 2007-02-28 2008-12-18 Ousler Iii George W Methods and compositions for normalizing meibomian gland secretions
US7988294B2 (en) * 2007-06-20 2011-08-02 Tearscience, Inc. Tear film measurement

Family Cites Families (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6159460A (en) 1988-05-27 2000-12-12 Amgen Inc. Method for treating interleukin-1 mediated diseases
KR0148009B1 (en) 1988-05-27 1998-08-01 그래고리 비. 아보트 Interleukin-1 inhibitors
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
US6858409B1 (en) 1988-05-27 2005-02-22 Amgen Inc. Nucleic acids encoding interleukin-1 inhibitors and processes for preparing interleukin-1 inhibitors
BR9007883A (en) 1989-11-29 1992-09-29 Synergen Inc PRODUCTION OF RECOMBINANT HUMAN INTERLEUCIN-1 INHIBITOR
US5770401A (en) * 1991-10-15 1998-06-23 Mullarkey; Michael F. Methods and compositions for treating allergic reactions
US5340572A (en) 1993-02-08 1994-08-23 Insite Vision Incorporated Alkaline ophthalmic suspensions
US6096728A (en) 1996-02-09 2000-08-01 Amgen Inc. Composition and method for treating inflammatory diseases
US6416753B1 (en) 1996-03-15 2002-07-09 The General Hospital Corporation Method for modulating apoptosis
US6974682B1 (en) 1996-08-26 2005-12-13 Human Genome Sciences, Inc. Soluble interleukin-1 receptor accessory molecule
JP2002514194A (en) 1996-11-19 2002-05-14 ザ スキーペンズ アイ リサーチ インスティテュート インコーポレイテッド Topical use of IL-1RA in rejection of corneal transplantation or eye disease
EP2002846B1 (en) 1996-12-06 2017-01-25 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
ES2320579T3 (en) 1998-05-08 2009-05-25 The University Of Miami USE OF SUB-ANTIMICROBIAL TETRACICLINES TO TREAT EYE ROSACEA
US6432934B1 (en) 1998-08-06 2002-08-13 Advanced Vision Research Methods and compositions for topical treatment of eye surface inflammation and related dry eye disease
US6927044B2 (en) 1998-09-25 2005-08-09 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps
US20060262813A1 (en) 1998-12-18 2006-11-23 Digital Networks North America, Inc. Multi-channel video pump
US6471961B1 (en) 1999-02-24 2002-10-29 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
JP3363832B2 (en) 1999-05-13 2003-01-08 アルプス電気株式会社 Thin film structure and method of manufacturing the same
US20030007971A1 (en) * 2000-01-31 2003-01-09 Hideaki Hara Remedies for ophthalmic diseases
JP2002154985A (en) 2000-01-31 2002-05-28 Santen Pharmaceut Co Ltd Therapeutic agent for eye disease
AU2001236632A1 (en) 2000-02-03 2001-08-14 Regeneration Technologies, Inc. Extraction of growth factors from tissue
WO2003070918A2 (en) 2002-02-20 2003-08-28 Ribozyme Pharmaceuticals, Incorporated Rna interference by modified short interfering nucleic acid
US6623736B2 (en) 2000-05-02 2003-09-23 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
US20030026806A1 (en) 2000-10-27 2003-02-06 Amgen Inc. Antibodies and other selective IL-1 binding agents that allow binding to IL-1 receptor but not activation thereof
US7087224B2 (en) 2000-10-31 2006-08-08 Amgen Inc. Method of treating anemia by administering IL-1ra
US20050143333A1 (en) 2001-05-18 2005-06-30 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
EP1432724A4 (en) * 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
AU2003226428A1 (en) 2002-04-18 2003-11-03 Northwestern University Encapsulation of nanotubes via self-assembled nanostructures
US7029712B1 (en) * 2002-07-17 2006-04-18 Biosyntrx Inc Treatment for dry eye syndrome
CA2511823A1 (en) 2002-12-30 2004-07-22 Amgen Inc. Combination therapy with co-stimulatory factors
US20050033694A1 (en) 2003-06-03 2005-02-10 United States Postal Service. System and method for fleet card management
US20050023872A1 (en) 2003-07-28 2005-02-03 Hetzel Thomas R. Modular seat cushion with interlocking human support and base portions and method of creating and using a seat cushion
KR100543466B1 (en) 2003-11-13 2006-01-20 삼성전자주식회사 Image interpolation apparatus and method
AU2005231822B2 (en) 2004-04-02 2011-07-21 Swedish Orphan Biovitrum Ab (Publ) Methods of reducing aggregation of IL-1ra
US8618054B2 (en) 2004-05-05 2013-12-31 Valorisation-Rechereche Société en Commandite Interleukin-1 receptor antagonists, compositions, and methods of treatment
US8541413B2 (en) * 2004-10-01 2013-09-24 Ramscor, Inc. Sustained release eye drop formulations
PL1864666T3 (en) * 2005-03-31 2013-02-28 Agc Inc Protective agent for retinal neuronal cell containing prostaglandin f2 alpha derivative as active ingredient
EP1870099A4 (en) * 2005-04-13 2008-07-16 Ube Industries Protective agent for retinal neuronal cell comprising indazole derivative as active ingredient
WO2006119292A2 (en) * 2005-04-29 2006-11-09 Henkin, Robert Methods for detection of biological substances
EP1948220A2 (en) 2005-10-21 2008-07-30 Amgen Inc. Methods of decreasing vascular calcification using il-1 inhibitors
WO2007056812A1 (en) * 2005-11-16 2007-05-24 Apollo Life Sciences Limited A molecule and chimeric molecules thereof
RU2008144802A (en) 2006-04-14 2010-05-20 Новартис АГ (CH) APPLICATION OF ANTIBODIES AGAINST IL-1 FOR TREATMENT OF EYE DISORDERS
MX2008015976A (en) 2006-06-12 2009-04-14 Therakine Ltd Topical treatment for diseases of eye surface.
US10105441B2 (en) * 2007-08-16 2018-10-23 The Schepens Eye Research Institute, Inc. Method for inhibiting or reducing dry eye disease by IL-1Ra
JP5886523B2 (en) * 2008-01-09 2016-03-16 ザ スキーペンズ アイ リサーチ インスティチュート インコーポレイテッド Therapeutic composition for treating inflammatory disorders of the eye
US20120014970A1 (en) * 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5895812A (en) * 1995-05-01 1999-04-20 The University Of Virginia Alumni Patent Foundation Diagnostic for Sjogren's syndrome
US20070280924A1 (en) * 2004-03-02 2007-12-06 Julie Daniels Pharmaceutical Preparations For And Treatment Of Ocular Surface and Other Disorders
US20080286378A1 (en) * 2005-02-22 2008-11-20 Ashley Behrens Use of Amniotic Fluid (Af) in Treating Ocular Disease and Injury
US20080132475A1 (en) * 2006-12-05 2008-06-05 Charles Gerald Connor Treatment for dry eye
US20080312194A1 (en) * 2007-02-28 2008-12-18 Ousler Iii George W Methods and compositions for normalizing meibomian gland secretions
US7988294B2 (en) * 2007-06-20 2011-08-02 Tearscience, Inc. Tear film measurement

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10272040B2 (en) 2010-08-12 2019-04-30 Nanyang Technological University Liposomal formulation for ocular drug delivery
US11612590B2 (en) 2013-03-13 2023-03-28 Santen Pharmaceutical Co., Ltd. Therapeutic agent for meibomian dysfunction
US11951098B2 (en) 2013-03-13 2024-04-09 Santen Pharmaceutical Co., Ltd. Therapeutic agent for meibomian dysfunction
US9956195B2 (en) 2014-01-07 2018-05-01 Nanyang Technological University Stable liposomal formulations for ocular drug delivery

Also Published As

Publication number Publication date
EP2187900A4 (en) 2011-08-17
JP5894364B2 (en) 2016-03-30
EP2187900B1 (en) 2016-11-09
AU2008289552B2 (en) 2014-10-09
US10105441B2 (en) 2018-10-23
WO2009025763A3 (en) 2009-04-23
JP2011516400A (en) 2011-05-26
US20100203103A1 (en) 2010-08-12
WO2009025763A2 (en) 2009-02-26
AU2008289552A1 (en) 2009-02-26
JP2016065089A (en) 2016-04-28
EP2187900A2 (en) 2010-05-26

Similar Documents

Publication Publication Date Title
US10105441B2 (en) Method for inhibiting or reducing dry eye disease by IL-1Ra
JP2011516400A5 (en)
EP2242504B1 (en) Therapeutic compositions for treatment of ocular inflammatory disorders
Moysidis et al. Mechanisms of inflammation in proliferative vitreoretinopathy: from bench to bedside
Colligris et al. Recent developments on dry eye disease treatment compounds
AU2010203425B2 (en) Therapeutic compositions for treatment of corneal disorders
Ambroziak et al. Immunomodulation on the ocular surface: a review
WO2016060916A1 (en) Histatins as therapeutic agents for ocular surface disease
Sherwood A sequential, multiple-treatment, targeted approach to reduce wound healing and failure of glaucoma filtration surgery in a rabbit model (an American Ophthalmological Society thesis)
US20120014970A1 (en) Therapeutic Compositions for Treatment of Corneal Disorders
CA2640986C (en) Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
AU2014227534B2 (en) Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
CN111918647A (en) NK-1 antagonists for the treatment of ocular pain
US11306140B2 (en) Therapeutics for ocular immunoinflammatory diseases
US20220288028A1 (en) A therapeutic approach for treating non-infectious ocular immunoinflammatory disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE SCHEPENS EYE RESEARCH INSTITUTE INC., MASSACHU

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DANA, REZA;DASTJERDI, MOHAMMAD;SIGNING DATES FROM 20081110 TO 20081118;REEL/FRAME:030804/0001

STCB Information on status: application discontinuation

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