US20140308239A1 - Chimeric cytokine formulations for ocular delivery - Google Patents

Chimeric cytokine formulations for ocular delivery Download PDF

Info

Publication number
US20140308239A1
US20140308239A1 US14/209,605 US201414209605A US2014308239A1 US 20140308239 A1 US20140308239 A1 US 20140308239A1 US 201414209605 A US201414209605 A US 201414209605A US 2014308239 A1 US2014308239 A1 US 2014308239A1
Authority
US
United States
Prior art keywords
formulation
concentration
weeks
container
months
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
US14/209,605
Other languages
English (en)
Inventor
Gregory Zarbis-Papastoitsis
Patricia Lowden
Byeong Chang
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.)
Integrity Bio Inc
Carisma Therapeutics Inc
Original Assignee
Eleven Biotherapeutics 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 Eleven Biotherapeutics Inc filed Critical Eleven Biotherapeutics Inc
Priority to US14/209,605 priority Critical patent/US20140308239A1/en
Assigned to ELEVEN BIOTHERAPEUTICS, INC. reassignment ELEVEN BIOTHERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LOWDEN, Patricia, ZARBIS-PAPASTOITSIS, GREGORY
Assigned to Integrity Bio, Inc. reassignment Integrity Bio, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANG, BYEONG
Assigned to ELEVEN BIOTHERAPEUTICS, INC. reassignment ELEVEN BIOTHERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Integrity Bio, Inc.
Publication of US20140308239A1 publication Critical patent/US20140308239A1/en
Priority to US15/670,840 priority patent/US10799589B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/04Artificial tears; Irrigation solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to therapeutic compositions and formulations, e.g., for IL-1 inhibitors.
  • Interleukin-1 alpha (IL-1 ⁇ ) and beta (IL-1 ⁇ ) are members of the IL-1 family of immunoregulatory cytokines. At least eleven human members of the interleukin-1 cytokine family have been identified, nine putative or demonstrated agonists (IL-1 ⁇ , IL-1 ⁇ , IL-18, IL-36 ⁇ (IL-1F6), IL-36 ⁇ (IL-1F8), IL-36 ⁇ (IL-1F9), IL-33, IL-1F7 and IL-1F10) and two natural antagonists (IL-1Ra and IL36Ra (IL-1F5)).
  • IL-1 ⁇ and IL-1 ⁇ have roles in regulating the immune system, and have been implicated in inflammatory ophthalmic diseases, including significant inflammatory ocular disorders.
  • IL-1 inhibitory agents have roles in regulating the immune system, and have been implicated in inflammatory ophthalmic diseases, including significant inflammatory ocular disorders.
  • Anakinra (Kineret®, Amgen, Thousand Oaks, Calif.), a recombinant IL1-Ra molecule, is approved for use in treating rheumatoid arthritis and Cryopyrin-Associated Periodic Syndromes (CAPS) called Neonatal-Onset Multisystem Inflammatory Disease (NOMID). It is supplied as a single use, glass syringe with 27 gauge needles as a sterile, clear, colorless-to-white, preservative-free solution for daily subcutaneous administration.
  • CPS Cryopyrin-Associated Periodic Syndromes
  • NOMID Neonatal-Onset Multisystem Inflammatory Disease
  • Kineret® is formulated in 0.67 ml solution with pH 6.5, containing 100 mg anakinra, 1.29 mg sodium citrate, 5.48 mg sodium chloride, 0.12 mg disodium EDTA, and 0.70 mg polysorbate 80 in water for injection. Storage at 2-8° C. is advised. Kineret® is not approved for ocular administration.
  • stable formulations e.g., stable aqueous formulations
  • chimeric cytokines e.g., chimeric cytokines or chimeric cytokine domains as described in WO2012/016203 or in WO 2012/103240
  • a pharmaceutical formulation that includes 1 mg/ml to 50 mg/ml of an IL-1 ⁇ /IL-1Ra chimeric cytokine protein.
  • the pharmaceutical formulation comprises 1 mg/ml to 50 mg/ml of an IL-1 ⁇ /IL-1Ra chimeric cytokine protein; a surfactant; a tonicity agent; and a buffering agent.
  • the chimeric cytokine protein is P05 or another chimeric cytokine such as, e.g., those described in WO2012/016203 or WO 2012/103240.
  • the formulation is for topical administration.
  • the formulation is for administration to the eye.
  • the formulation is for topical administration to the eye.
  • the formulation has a pH of 5.5 to 7.5, for example a pH of 6.0 to 7.0.
  • the formulation does not contain a viscosity agent.
  • a formulation that also contains a viscocity agent is a sodium carboxymethyl cellulose; an hydroxy ethyl cellulose; an hydroxypropyl methylcellulose; a polyvinyl alcohol; and/or a glycerin.
  • sodium carboxymethyl cellulose is included in the formulation at a concentration of 0.1-6% w/v.
  • the formulation typically has a purity of at least 90%, 93%, 95%, or 98% after storage for at least 60 days at 25° C. In some aspects, the formulation has a purity of at least 90%, 93%, 95%, or 98% after storage for at least four months at 25° C. In other aspects, the formulation has a purity of at least 90% after storage for a period of at least 2 weeks at 40° C. In some cases, the formulation is stable for at least two years when stored at 2° C. to 8° C. In some cases the formulation is stable for at least six months when stored at 25° C. In some cases the formulation is stable for at least eight months when stored at 25° C.
  • the surfactant is a non-ionic surfactant, for example, pluronic acid F-68 (poloxamer 188), polysorbate-20, or polysorbate-80.
  • the surfactant is pluronic acid F-68 (poloxamer 188), and the surfactant is present in a concentration of about 0.1% w/v. In some cases, the surfactant is pluronic acid F-68 (poloxamer 188), and the surfactant is present in a concentration of 0.1% w/v.
  • the tonicity agent in a formulation can be, for example, sodium chloride, sorbitol, mannitol, sucrose, or trehalose.
  • the tonicity agent is sorbitol, and the sorbitol is present in a concentration of about 5% w/v.
  • the tonicity agent is sorbitol, and the sorbitol is present in a concentration of 5% w/v.
  • the buffering agent in the formulation is generally a weak buffering agent.
  • the buffering agent is a phosphate a citrate, an acetate, a borate, and/or a succinate.
  • the buffering agent can be a pharmaceutically acceptable salt of a phosphate, a citrate, an acetate, a borate, or a succinate.
  • the buffering agent is present in an amount of from about 10 mM to about 50 mM.
  • the buffering agent is present at a concentration of 20 mM or less.
  • the buffering agent, e.g., the sodium citrate and/or sodium phosphate is present at a total concentration of 5-15 mM.
  • the buffering agent e.g., sodium citrate
  • the buffering agent is sodium citrate and is present in the formulation at a concentration of about 10 mM. In some cases, the buffering agent is sodium citrate and is present in the formulation at a concentration of 10 mM.
  • the formulation includes an IL-1 ⁇ /IL-1Ra chimeric cytokine protein such as P05; 10 mM sodium citrate; 5% w/v sorbitol; and 0.1% w/v poloxamer 188 (poloxamer F-68), and the pH of the formulation is 6.0.
  • an IL-1 ⁇ /IL-1Ra chimeric cytokine protein such as P05; 10 mM sodium citrate; 5% w/v sorbitol; and 0.1% w/v poloxamer 188 (poloxamer F-68)
  • the pH of the formulation is 6.0.
  • the formulation further includes an amino acid.
  • the amino acid is arginine, glutamic acid, histidine, or methionine.
  • a chimeric cytokine e.g., a chimeric cytokine polypeptide
  • a chimeric cytokine polypeptide e.g., a chimeric cytokine containing sequences derived from an Il-1 ⁇ and an IL-1Ra
  • concentrations of 5 mg/ml to 20 mg/ml e.g., at a concentration of 1 mg/ml, 5 mg/ml, or 20 mg/ml
  • 10 mM sodium citrate pH 6.0 containing 5% w/v sorbitol and 0.1% w/v poloxamer, e.g., poloxamer 188 (also referred to as, for example, Lutrol® F-68 (also referred to herein as Lutrol®), Kolliphor® P 188, and poly(ethylene glycol)-block-poly(ethylene glycol)).
  • the chimeric cytokine is selected from one or more of P01, P02, P03, P04, P
  • the components of a formulation described herein are present in amounts that may vary around the values provided herein by up to 5%, 10%, 15%, 20%, 25%, 30%, 40%, or 50%. In embodiments, the components of a formulation are present in amounts that vary around the values provided herein by 10%. In embodiments, the formulation comprises 9.5-10.5 mM, 9-11 mM, 8.5-11.5 mM, 8-12 mM, 7.5-12.5 mM, 7-13 mM, 6-14 mM, or 5-15 mM sodium citrate.
  • the formulation comprises 4.75-5.25%, 4.5-5.5%, 4.25-5.75%, 4-6%, 3.75-6.25%, 3.5-6.5%, 3-7%, or 2.5-7.5% w/v sorbitol.
  • the formulation comprises 0.095-0.105%, 0.09-0.11%, 0.085-0.115%, 0.08-0.12%, 0.075-0.125%, 0.07-0.13%, 0.06-0.14%, or 0.05-0.15% w/v poloxamer 188.
  • the concentration of the therapeutic protein e.g., the chimeric cytokine, e.g., P05
  • the concentration of the therapeutic protein is 1-50 mg/ml, 1-25 mg/ml, or 1-20 mg/ml.
  • the concentration of the therapeutic protein is 4.75-5.25 mg/ml, 4.5-5.5 mg/ml, 4.25-5.75 mg/ml, 4-6 mg/ml, 3.75-6.25 mg/ml, 3.5-6.5 mg/ml, 3-7 mg/ml, or 2.5-7.5 mg/ml.
  • the pH of the formulation is 5.5 to 7.5, or 5.5 to 6.5.
  • the formulation comprises 8-12 mM sodium citrate, 4-6% w/v sorbitol, 0.08-0.12% w/v poloxamer 188, and 4-6 mg/ml P05.
  • the pH of the formulation is 5.5 to 7.5. In embodiments, the pH is 5.5 to 6.5. In embodiments, the pH is 6 to 7.
  • the formulation comprises 9-11 mM sodium citrate, 4.5-5.5% w/v sorbitol, 0.09-0.11% w/v poloxamer 188, and 4.5-5.5 mg/ml P05.
  • the pH of the formulation is 5.5 to 7.5. In embodiments, the pH is 5.5 to 6.5. In embodiments, the pH is 6 to 7.
  • an IL-1 inhibitor e.g., anakinra
  • an appropriate concentration e.g., at a concentration of 5 mg/ml to 100, e.g., 5 to 50 mg/ml, e.g., 5 to 20 mg/me in 10 mM sodium citrate, pH 6.0 containing 5% w/v sorbitol and 0.1% w/v poloxamer, e.g., poloxamer 188.
  • the amounts of the components of the formulation may vary around the values provided herein by up to 5%, 10%, 15%, 20%, 25%, 30%, 40%, or 50%.
  • the pH is 5.5 to 7.5.
  • the pH is 5.5 to 6.5.
  • the pH is 6 to 7.
  • a formulation described herein further comprises a viscosity agent, e.g., sodium carboxymethyl cellulose (CMC).
  • CMC sodium carboxymethyl cellulose
  • the formulation comprises CMC, e.g., CMC at a concentration of 0.1-1% w/v, 0.1-0.5% w/v, or 0.2-0.3 w/v %.
  • the method includes administering to the subject a therapeutically effective amount of a composition comprising a formulation described herein.
  • the method includes identifying a subject having an IL-1-related disorder such as, e.g., a dry eye disorder; and administering to the subject a therapeutically effective amount of a composition comprising a formulation as described herein.
  • Also described herein is a method of inhibiting IL-1 activity in a subject.
  • the method includes administering to the subject a formulation as described herein.
  • the subject has an IL-1-related disorder, e.g., a dry eye disorder.
  • the invention relates to a drug delivery device comprising a formulation as described herein.
  • the medicament is for administration to the eye, e.g., for topical administration to the eye.
  • the medicament is a vehicle formulation, e.g., an aqueous formulation comprising or consisting of sorbitol, sodium citrate, and poloxamer 188 as described herein.
  • the vehicle formulation is substantially free of (e.g., does not comprise) a therapeutic protein.
  • the subject treated as described herein is a human or other mammal such as a dog or cat.
  • the invention relates to a container or device comprising a formulation as described herein.
  • the container is a blow fill seal container.
  • a pharmaceutical formulation e.g., a vehicle formulation
  • a surfactant e.g., Pluronic F68 (poloxamer 188)
  • the buffering agent is citrate
  • the tonicity agent is sorbitol.
  • the formulation comprises a chimeric cytokine protein, e.g., a chimeric cytokine protein as described herein or in WO 2012/103240, e.g., P05.
  • the pharmaceutical formulation also includes a viscocity agent, e.g., CMC.
  • the pharmaceutical formulation is suitable for use in the eye (i.e., suitable for ocular delivery), e.g., for treating ocular disease such as signs and/or symptoms of dry eye.
  • Naturally occurring proteins referenced herein specifically include human forms of such proteins, and as well as forms from other mammalian species.
  • An aqueous formulation comprising sodium citrate or sodium phosphate at a concentration of 8 to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v); and optionally sodium carboxymethyl cellulose, wherein the formulation has a pH of 5.5 to 7.5 and wherein the formulation is effective for treating an ocular disorder.
  • An aqueous formulation comprising sodium citrate at a concentration of 9-11 mM; sorbitol at 4.5-5.5% (w/v); and poloxamer 188 at a concentration of 0.09-0.11%, wherein the formulation has a pH of 5.7 to 6.3, wherein the formulation is substantially free of therapeutic protein, and wherein the formulation is effective for treating an ocular disorder (e.g., an ocular disorder described herein).
  • an ocular disorder e.g., an ocular disorder described herein.
  • An aqueous formulation comprising 1-50 mg/ml of an IL-1 ⁇ /IL-1Ra chimeric cytokine protein (e.g., P01, P02, P03, P04, P05, P06, or P07); a buffering agent selected from sodium citrate and sodium phosphate; sorbitol, e.g., at a concentration of 3.5-6.5% (w/v); poloxamer 188, e.g., at a concentration of 0.07-0.13% (w/v); and optionally sodium carboxymethyl cellulose (CMC), wherein the formulation has a pH of 5.5 to 7.5.
  • an IL-1 ⁇ /IL-1Ra chimeric cytokine protein e.g., P01, P02, P03, P04, P05, P06, or P07
  • a buffering agent selected from sodium citrate and sodium phosphate
  • sorbitol e.g., at a concentration of 3.5-6.5% (w/v)
  • invention 13 comprising sodium citrate and/or sodium phosphate at a total concentration of 5 mM to 15 mM.
  • An aqueous formulation comprising 1-25 mg/ml P05; sodium citrate or sodium phosphate at a concentration of 8 mM to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v); and, optionally, sodium carboxymethyl cellulose, wherein the formulation has a pH of 5.5 to 7.5.
  • An aqueous formulation comprising or consisting of 1 mg/ml to 25 mg/ml P05; sodium citrate at a concentration of 8-12 mM; sorbitol at 4% to 6% (w/v); and poloxamer 188 at a concentration of 0.08% to 0.12% (w/v), wherein the formulation has a pH of 5.5 to 7.5.
  • An aqueous formulation comprising or consisting of 1 mg/ml to 25 mg/ml P05; sodium citrate at a concentration of 9 mM toll mM; sorbitol at 4.5% to 5.5% (w/v); and poloxamer 188 at a concentration of 0.09% to 0.11% (w/v); wherein the formulation has a pH of 5.7 to 6.3.
  • An aqueous formulation comprising or consisting of 4-6 mg/ml P05; sodium citrate at a concentration of 9-11 mM; sorbitol at 4.5-5.5% (w/v); and poloxamer 188 at a concentration of 0.09-0.11% (w/v); wherein the formulation has a pH of 5.7-6.3.
  • composition of embodiment 30, wherein the formulation is suitable for topical administration to the eye.
  • formulation of any one of embodiments 12 to 34 wherein the formulation further comprises an amino acid, e.g., arginine, glutamic acid, histidine, or methionine.
  • amino acid e.g., arginine, glutamic acid, histidine, or methionine.
  • any one of embodiments 12 to 40 wherein the formulation has less than or equal to 50 particles per ml for particles ⁇ 10 ⁇ m and less than or equal to 5 particles per ml for particles ⁇ 25 ⁇ m, as assessed using a light obscuration particle count test.
  • any one of embodiments 12 to 44 wherein the formulation is stable after storage for at least 5 months under ambient conditions, e.g., at room temperature, e.g., at 25° C.
  • any one of embodiments 12 to 46 wherein the formulation is stable after storage for at least 4 months under ambient conditions, e.g., at room temperature, e.g., at 25° C.
  • any one of embodiments 12 to 48 wherein the formulation is stable after storage for at least 3 months under ambient conditions, e.g., at room temperature, e.g., at 25° C.
  • any one of embodiments 12 to 50 wherein the formulation is stable after storage for at least 2 months under ambient conditions, e.g., at room temperature, e.g., at 25° C.
  • any one of embodiments 12 to 52 wherein the formulation is stable after storage for at least 1 month under ambient conditions, e.g., at room temperature, e.g., at 25° C.
  • any one of embodiments 12 to 53 wherein the formulation is stable as indicated by the presence of less than or equal to 50 particles per ml for particles ⁇ 10 ⁇ m, less than or equal to 5 particles per ml for particles ⁇ 25 ⁇ m, and less than or equal to 2 particles per ml for particles ⁇ 50 ⁇ m, e.g., as assessed using a microscopic particle count test.
  • IL-1 related disorder is a dry eye disorder.
  • a method of treating an ocular disorder comprising administering to a subject having the ocular disorder, e.g., the dry eye disorder, an aqueous formulation comprising sodium citrate or sodium phosphate at a concentration of 8 mM to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v); and optionally sodium carboxymethyl cellulose; wherein the formulation has a pH of 5.5 to 7.5 and is substantially free of therapeutic protein, thereby treating the dry eye disorder.
  • an aqueous formulation comprising sodium citrate or sodium phosphate at a concentration of 8 mM to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v); and optionally sodium carboxymethyl cellulose; wherein the formulation has a pH of 5.5 to 7.5 and is substantially free of therapeutic protein, thereby treating the dry eye disorder
  • aqueous formulation comprises sodium citrate at a concentration of 8 mM to 11 mM, sorbitol at 4.5% to ⁇ 5.5% (w/v) and poloxamer 188 at a concentration of 0.09% to 0.11%.
  • aqueous formulation consists of sodium citrate at a concentration of 8-11 mM, sorbitol at 4.5-5.5% (w/v) and poloxamer 188 at a concentration of 0.09-0.11%.
  • a method of treating a dry eye disorder comprising administering to a subject having a dry eye disorder an aqueous formulation comprising 1 to 25 mg/ml P05; sodium citrate or sodium phosphate at a concentration of 8 mM to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v); and optionally sodium carboxymethyl cellulose, wherein the formulation has a pH of 5.5 to 7.5, thereby treating the dry eye disorder.
  • a method of treating a dry eye disorder comprising administering to a subject having a dry eye disorder an aqueous formulation consisting of 1 mg/ml to 25 mg/ml P05; sodium citrate at a concentration of 8 mM to 12 mM; sorbitol at 4% to 6% (w/v); poloxamer 188 at a concentration of 0.08% to 0.12% (w/v), wherein the formulation has a pH of 5.5 to 7.5, thereby treating the dry eye disorder.
  • a method of treating a dry eye disorder comprising administering to a subject having a dry eye disorder an aqueous formulation comprising or consisting of 1 mg/ml to 25 mg/ml P05; sodium citrate at a concentration of 9 mM to 11 mM; sorbitol at 4.5% to 5.5% (w/v); and poloxamer 188 at a concentration of 0.09% to 0.11% (w/v), wherein the formulation has a pH of 5.7 to 6.3, thereby treating the dry eye disorder.
  • a container or device comprising the formulation of any one of embodiments 1 to 61.
  • the container or device of embodiment 77 wherein the container or device has been stored at 25° C. for at least two weeks, e.g., for at least four weeks, and is substantially free of particulates.
  • a blow fill seal container comprising the formulation of any one of embodiments 1 to 61.
  • a multidose container comprising the formulation of any one of embodiments 1 to 61.
  • a multidose container comprising the formulation of embodiments 35 to 40.
  • a drug delivery device comprising a formulation of any one of embodiments 1 to 61.
  • the drug delivery device of embodiment 82 wherein the drug delivery device is a blow fill seal container.
  • the container or device of any one of embodiments 77 to 83 wherein the container or device is sealed in a pouch, optionally containing an inert gas, e.g., nitrogen or argon.
  • an inert gas e.g., nitrogen or argon.
  • a kit comprising a container or device comprising the formulation of any one of embodiments 1 to 61, and optionally, instructions for use.
  • FIG. 1 is a reproduction of an SDS-PAGE gel showing exemplary samples of protein purified from E. coli expressing receptor binding agents. The 15 and 20 kDa molecular weight markers are indicated at left. Lanes are as follows: molecular weight marker (lanes 1 and 6), extract (lanes 2 and 7), material purified by cation exchange chromatography (lanes 3 and 8), material additionally purified by anion exchange chromatography (lanes 4 and 9), and reduced samples of such material (lanes 5 and 10). Lanes 2-5 are of P05 purification, and Lanes 6-10 are of P04 purification. See also Example 2.
  • FIG. 2A is a table and accompanying bar graph illustrating the results of an experiment testing the ability of the P06, P07, and P01 proteins to agonize signaling relative to IL-1 ⁇ and a negative control, [3-glucuronidase (GUS) protein.
  • FIG. 2B is a graph depicting the results of an experiment testing the ability of P01 to antagonize IL-1 ⁇ activity at various IL-1 ⁇ concentrations.
  • FIG. 3A is a graph depicting the results of an experiment testing antagonism of IL-1 ⁇ by P03 (hexa-histidine tagged (SEQ ID NO: 23)), P04 (hexa-histidine tagged (SEQ ID NO: 23)), P05 (hexa-histidine tagged (SEQ ID NO: 23)), and IL-1Ra in the presence of 0.1 ng/ml IL-1 ⁇ (human).
  • FIG. 3B is a graph depicting the results of an experiment testing antagonism of IL-1 ⁇ by lysates containing untagged forms of P01, P02, P03, P04, and P05, and IL-1Ra in the presence of 0.1 ng/ml IL-1 ⁇ (human) and using estimates of the concentration of protein in the respective lysates.
  • FIG. 4A is a graph showing the dynamic light scattering (DLS) results for P05 in a phosphate formulation (P05 at 20 mg/ml, 10 mM phosphate, 5% w/v sorbitol, 0.1% w/v poloxamer 188, pH 6.5).
  • DLS dynamic light scattering
  • FIG. 4B is a graph showing DLS results for P05 in a citrate formulation (P05 at 20 mg/ml, 10 mM citrate, 5% w/v sorbitol, 0.1% w/v poloxamer 188, pH 6.0).
  • FIG. 5A is a graph depicting thermal denaturation of IL-1Ra, IL-1 ⁇ , P03, P04, and P05 as described in Example 8.
  • FIG. 5B is a graph depicting the negative first derivative of the graph in FIG. 5A (the negative first derivative provides improved visualization of the melting temperature).
  • FIG. 6A is a bar graph depicting the mean corneal staining score ⁇ SEM as tested by fluorescein staining of the cornea per eye of two independent studies, on days 0, 3, 7, 9, and 11 for mice in a dry eye model.
  • Asterisks indicate statistical significance of P05 relative to vehicle as follows: * (P ⁇ 0.05) and ** (P ⁇ 0.005).
  • FIG. 6B is a bar graph representing data showing mean corneal staining score ⁇ SEM of the cornea per eye, on days 0, 3, 7, 9, and 11 for mice in a dry eye model.
  • MSA murine serum albumin
  • Asterisks indicate statistical significance of P05 relative to murine serum albumin as follows: * (P ⁇ 0.05) and *** (P ⁇ 0.0005).
  • Asterisks indicate statistical significance of P05 relative to cyclosporine (Restasis®) as follows: ** (P ⁇ 0.005) and *** (P ⁇ 0.0005).
  • FIG. 7 depicts the design of the clinical trial described in Example 16.
  • FIG. 8 is a graph showing the mean change from baseline in the OSDI score for the groups of subjects who received EBI-005 formulations (combined data for the groups that received 5 mg/ml and 20 mg/ml treatments) and vehicle formulation.
  • FIG. 9 is a graph showing the mean change from baseline in pain for the groups of subjects who received EBI-005 formulations (combined data for the groups that received 5 mg/ml and 20 mg/ml treatments) and vehicle formulation.
  • FIG. 10 is a graph showing the mean change from baseline in corneal fluorescein staining (CFS) score for the groups of subjects who received EBI-005 formulations (combined data for the groups that received 5 mg/ml and 20 mg/ml treatments) and vehicle formulation.
  • CFS corneal fluorescein staining
  • formulations that are useful for providing a protein, e.g., a chimeric cytokine polypeptide such as an IL-1 ⁇ /IL-1Ra chimera to a subject in need of treatment with such a formulation.
  • the formulations are generally useful for formulation of protein compositions requiring stability, e.g., proteins that are susceptible to agitation, are susceptible to oxidation, e.g., due to methionine residues, or susceptible to deamindation, e.g., due to asparagine or arginine residues.
  • methods of preparing and administering such formulations are also disclosed herein.
  • a formulation comprises a chimeric cytokine polypeptide, e.g., a chimeric polypeptide containing selected sequences derived from an IL-1 ⁇ and an IL-1Ra sequence, that are suitable for pharmaceutical use, for example, for ophthalmic use, including effective topical treatment for an IL-1-related disorder.
  • formulations described herein are surprisingly stable, even at relatively high concentrations of the polypeptide, e.g., at concentrations suitable for storage of bulk drug substance as well as at concentrations suitable for for treating a subject.
  • An advantage of this feature is that it is not necessary to remove undesirable agents from the bulk drug substance in order to formulate the drug for patient use.
  • Applicants have successfully achieved an effective aqueous formulation of chimeric cytokine polypeptides that is suitable for topical administration, e.g., in the eye, e.g., to the front or corneal surface of the eye.
  • aqueous formulation of chimeric cytokine polypeptides that is suitable for topical administration, e.g., in the eye, e.g., to the front or corneal surface of the eye.
  • biologic drugs biologicals
  • applicants were able to formulate such a polypeptide at a pH that is compatible with administration to the eye (e.g., a pH of 4.5 to 7.0, 5.5 to 7.0, 5.5 to 6.5, or 6.0 to 7.0), and contains a components that render the formulation comfortable for subjects being treated with the polypeptide.
  • the formulation is comfortable, e.g., does not cause irritation.
  • the formulation does not cause one or more symptoms of irritation such as, e.g., eye redness, tearing, mucous discharge, or subjective discomfort.
  • Topical ophthalmic drugs are generally self-administered by patients. Because the patient may be storing a drug for a relatively long period of time, the formulation may be subjected to higher temperatures and greater levels of agitation stress than a formulation that is typically stored only by a physician or pharmacist prior to administration. As is known in the art, proteins are more sensitive to agitation and temperature than small molecules. Agitation stress can lead to precipitation and heat stress can lead to precipitation and to chemical degradation. In addition, during loading of a compound into a delivery device, there can be exposure to heat stress. Applicants have achieved a formulation that successfully provides excellent stability when exposed to agitation stress and heat.
  • Some manufacturing processes require at least brief exposure of a formulation to relatively high temperature.
  • loading a formulation into a blow fill seal (BFS) container can result in exposure of the formulation to elevated temperatures, in addition to agitation associated with the filling process.
  • Applicants have loaded a formulation into such a device (a BFS container) and demonstrated stability of the formulation immediately following loading and over an extended period of time.
  • a formulation as provided herein is suitable for use with BFS.
  • a formulation that is suitable for use with BFS shows stability immediately following loading into a BFS container and/or after storage in a BFS container, e.g., after storage for periods of time and under conditions described herein.
  • a formulation described herein is stable.
  • the formulation exhibits stability under conditions (e.g., storage at particular temperatures, or agitation stress) described herein.
  • stability is assessed using one or more methods described herein (e.g., based on visual appearance, content by spectrophotometry (A280), SDS-PAGE non-reduced, SDS-PAGE reduced; size exclusion HPLC (SE HPLC); reverse phase HPLC (RP-HPLC); weak cation exchange HPLC (WCEX-HPLC); potency; a light obscuration particle count test (e.g., a light obscuration particle count test as described in USP ⁇ 788>); or a microscopic particle count test (e.g., a microscopic particle count test as described in USP ⁇ 788>)) and/or methods known in the art.
  • A280 visual appearance, content by spectrophotometry
  • SE HPLC size exclusion HPLC
  • RP-HPLC reverse phase HPLC
  • Stability can be assessed based on visual appearance.
  • a formulation is stable if it is a clear to slightly opalescent colorless solution essentially free from visible particulates.
  • the formulation is stable at about 25° C. to about 40° C., for example, about 27° C., about 28° C., about 29° C., about 30° C., about 31° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., or about 40° C.
  • the formulations are stable for long periods of time during storage at temperatures of from about 2° C. to about 8° C., such as at about 4° C., about 5° C., about 6° C., from 2° C. to 8° C., at 4° C., at 5° C., or at 6° C.
  • the formulations are stable at such storage temperatures for a period of at least two weeks; four weeks; six weeks; two months; three months; six months, one year, two years, three years, or four years.
  • Stability of a formulation can be assessed, e.g., after storage for at least 2, 4, 6, 8, 12, or 18 months, e.g., at 2-8° C., or after storage under ambient conditions, e.g., at room temperature (RT), e.g. at about 25° C. for, e.g., at least 2 weeks, 1 month, 2 months, 3 months 5 months, 6 months, 12 months, or 18 months.
  • RT room temperature
  • the formulation is stable after storage at 2-8° C. for at least 8 months.
  • the formulation is stable after exposure to room temperature for at least 5 months.
  • the formulation is stable after storage, e.g., for at least 5 months, in a BFS container.
  • Stability can be assessed, e.g., based on methods and criteria described herein or known in the art. For example, stability can be assessed based on physical purity (e.g., lack of aggregation, e.g., as assessed using size exclusion HPLC, also referred to herein as size exclusion, SE HPLC, or SEC HPLC), chemical purity (e.g., as assessed using weak cation exchange HPLC, reverse phase HPLC, and/or SDS PAGE (e.g., reduced or nonreduced SDS PAGE)), and/or the levels of particulates (e.g., as assessed visually or by particle count using an HIAC liquid particle counter (Beckman Coulter, Brea, Calif.)).
  • physical purity e.g., lack of aggregation, e.g., as assessed using size exclusion HPLC, also referred to herein as size exclusion, SE HPLC, or SEC HPLC
  • chemical purity e.g., as assessed using weak cation exchange HPLC
  • stability is demonstrated based on compliance with guidelines for particulate matter in opthalmic solutions, e.g., as set forth in USP ⁇ 789> (U.S. Pharmacopeia, Particulate Matter in Opthalmic Solutions).
  • the formulation has less than or equal to 50 particles per ml for particles ⁇ 10 ⁇ m and/or less than or equal to 5 particles per ml for particles ⁇ 25 ⁇ m, e.g., as assessed using a light obscuration particle count test (e.g., a light obscuration particle count test as described in USP ⁇ 788>).
  • a light obscuration particle count test e.g., a light obscuration particle count test as described in USP ⁇ 788>.
  • the formulation has less than or equal to 50 particles per ml for particles ⁇ 10 ⁇ m, less than or equal to 5 particles per ml for particles ⁇ 25 ⁇ m, and/or less than or equal to 2 particles per ml for particles ⁇ 50 ⁇ m, e.g., as assessed using a microscopic particle count test (e.g., a microscopic particle count test as described in USP ⁇ 788>).
  • a microscopic particle count test e.g., a microscopic particle count test as described in USP ⁇ 788>.
  • stability is demonstrated based on compliance with guidelines for particulate matter in injections, e.g., as set forth in USP ⁇ 788> (U.S. Pharmacopeia, Particulate Matter in Injections).
  • the formulation has less than or equal to 6000 particles per container (for containers with a volume of 100 ml or less) for particles ⁇ 10 ⁇ m, and/or less than or equal to 600 particles per container (for containers with a volume of 100 ml or lower) for particles ⁇ 25 um, e.g., as assessed using a light obscuration particle count test (e.g., a light obscuration particle count test as described in USP ⁇ 788>).
  • a light obscuration particle count test e.g., a light obscuration particle count test as described in USP ⁇ 788>.
  • the formulation has less than or equal to 3000 particles per 5 ml for particles ⁇ 10 m and/or less than or equal to 300 particles per 5 ml for particles ⁇ 25 um, e.g., as assessed using a microscopic particle count test (e.g., a microscopic particle count test as described in USP ⁇ 788>).
  • a microscopic particle count test e.g., a microscopic particle count test as described in USP ⁇ 788>.
  • the protein in a formulation is protected from agitation stress as demonstrated, e.g., by lack of aggregation (lack of aggregation may be demonstrated, e.g., if the formulation contains contains >90%, >91%, >92%, >93%, >94%, >95%, >96%, >97%, >98%, or >99% of the monomeric form of the protein relative to aggregated form) after vortexing the protein solution, e.g., for 1-8 hours at room temperature (RT), e.g., for 4 hours at RT.
  • Aggregation can be assessed, e.g., using methods described herein or methods known in the art. For example, aggregation can be assessed using ultracentrifugation, size-exclusion chromatography, gel electrophoresis, dynamic light scattering, and/or turbidity measurements.
  • stability is assayed by physical or chemical methods known in the art. For example, physical purity or lack of aggregation can be determined using size exclusion HPLC or other methods that determine the relative amount of monomeric polypeptide in a formulation.
  • a formulation with acceptable stability contains >90% of the monomeric form of therapeutic protein (e.g., the chimeric cytokine, e.g., P05) relative to aggregated forms of the protein.
  • the formulation contains >90% (e.g., >91%, >92%, >93%, >94%, >95%, >96%, >97%, >98%, or >99%) of the monomeric form of the therapeutic protein (e.g., the chimeric cytokine, e.g., P05), relative to aggregated forms of the protein.
  • >90% e.g., >91%, >92%, >93%, >94%, >95%, >96%, >97%, >98%, or >99%
  • Chemical purity can be determined, for example, using weak cation exchange HPLC or reverse phase HPLC.
  • a formulation with acceptable stability contains >80% of the native molecule, relative to chemically modified forms of the molecule, e.g., as assessed using weak cation exchange HPLC.
  • the formulation contains >80% (e.g., >85%, >87%, >90%, or >95%) of the native molecule, relative to chemically modified forms of the molecule (e.g., oxidized or acetylated forms).
  • Particulates may be identified visually.
  • the formulation is one that is essentially free of particulates that can be identified visually.
  • anakinra an IL-1Ra, formulated for delivery by injection states that the product has a shelf life of three years, is to be stored at 3-8° C., and “For the purpose of ambulatory use, Kineret® may be removed from the refrigerator for 12 hours at temperature not above 25° C., without exceeding the expiry date. At the end of this period, the product must not be put back in the refrigerator and must be disposed of.” (See: medicines.org.uk/EMC/medicine/23104/SPC/Kineret+100+mg+solution+for+injection+in+a+pre-filled+syringe#SHELF_LIFE). This provides a contrast to the surprising stability of, for example, the P05 formulation provided herein.
  • Biologic treatments can be problematic to administer because they can have a relatively short shelf life or require special storage conditions that can create obstacles for storage, transport, and patient use as well as assuring a sufficient supply of the biologic.
  • An advantage of certain formulations provided herein is that the formulations are surprisingly stable not only under conditions of refrigeration, but also at temperatures that are in accord with room temperature (e.g., 25° C.) and above (e.g., 40° C.). Accordingly, the cytokine protein or polypeptide formulations (e.g.
  • heterologous cytokine protein or polypeptide formulations are, in some embodiments, provided in a liquid form that is stable at RT (e.g., at 25° C.) for a period of at least three days, five days, one week, ten days, two weeks, three weeks, six weeks, eight weeks, 16 weeks, 20 weeks, 25 weeks, 30 weeks, 35 weeks, 40 weeks, 45 weeks, one month, two months, three months, four months, five months, six months, seven months, eight months, twelve months, or more.
  • a month is determined on date to date basis, e.g., from the first of the month to the first of the second month.
  • the formulations are stable at about 25° C. to about 40° C., for example, about 27° C., about 28° C., about 29° C., about 30° C., about 31° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., or about 40° C.
  • a formulation is stable for one month at 25° C. and 1 week at 40° C. when the protein component of the formulation, e.g., P05, is at a concentration of 20 mg/ml.
  • the formulation, loaded into a blow fill seal vial or blow fill delivery device is stable at 25° C. for at least three months for a formulation comprising protein, e.g., P05, at a concentration of 1 mg/ml, 5 mg/ml, or 10 mg/ml. In some embodiments, this formulation is stable for at least eight months.
  • a formulation comprising 4.5-5.5 mg/ml P05, 9-11 mM sodium citrate; 4.5-5.5% w/v sorbitol, and 0.09-0.11% w/v poloxamer 188 is stable for at least five months at 2° C. to 8° C. and/or at room temperature, e.g., at 25° C.
  • a formulation consisting of 10 mM Na citrate, pH 6.0, 5% sorbitol, 0.1% poloxamer, and 5 mg/ml or 20 mg/ml P05 is stable for at least five months at 2° C. to 8° C. and/or at room temperature, e.g., at 25° C. for at least 5 months.
  • a further problem in administering biologics is providing a sufficient concentration of the biologic. This is a particular problem in ophthalmic applications in which it is desirable to provide a relatively high concentration of the biologic so as to achieve a therapeutic effect with a minimum number of doses.
  • Applicants have been able to achieve a formulation that can deliver an effective dose of a chimeric cytokine formulation containing a high concentration, or a therapeutically effective concentration, of the polypeptide that does not appreciably aggregate, precipitate, or lose chemical purity when stored under conditions such as those described supra and elsewhere within this specification.
  • cytokine formulation at protein concentrations of up to 80 mg/ml, e.g., 50 mg/ml in a formulation comprising a tonicity agent, a surfactant, and a buffering agent.
  • a formulation featured in the invention contains a chimeric cytokine polypeptide stably present in the formulation in a concentration of from 0.1 mg/ml to 100 mg/ml, 0.1-80 mg/ml, 0.1 to 50 mg/ml, 0.1 mg/ml to 20 mg/ml, 0.1 mg/ml to 5 mg/ml, 0.1 mg/ml to 1 mg/ml, 1 mg/ml to 100 mg/ml; 5 mg/ml to 100 mg/ml; 5 mg/ml to 30 mg/ml; 10 mg/ml to 100 mg/ml; 10 mg/ml to 30 mg/ml; 20 mg/ml to 100 mg/ml; 30 mg/ml to 100 mg/ml; 40 mg/ml to 100 mg/ml; 50 mg/ml to 100 mg/ml; 60 mg/ml to 100 mg/ml; 1 mg/ml to 80 mg/ml; 5 mg/ml to 80 mg/ml; 10 mg/ml to 100
  • Viscocity agents are frequently used in formulations, e.g., for ophthalmic use. Such agents are generally included to increase the residence time of an ophthalmic treatment that would otherwise be rapidly cleared by blinking and drainage through the conjunctival sac. However, such agents can have deleterious effects, e.g., allergic symptoms, damage protein components of a formulation, or cause blurry vision. While such agents can be used in certain formulations described herein, in some embodiments Applicants have achieved formulations that do not require a viscocity agent for the active component, i.e., a chimeric cytokine, to be used as an effective therapeutic.
  • a viscocity agent for the active component i.e., a chimeric cytokine
  • the formulations featured in the invention contain one or more surfactants.
  • a surfactant can be useful, e.g., for reducing adhesion of a molecule to a container, reducing aggregation of a protein particularly under conditions of agitation, addition of a surfactant can also render a therapeutic agent unusable because of foaming, disruption of natural membranes and other barriers, and unacceptable discomfort caused by treatment.
  • Applicants have succeeded in providing a formulation that includes a surfactant, but does not incur such disadvantages.
  • the surfactant is a non-ionic surfactant.
  • Surfactants suitable for use in the disclosed formulations can include, but are not limited to: poloxamers, such as poloxamer 188.
  • a surfactant is a polysorbate, such as polysorbate-20 and polysorbate-80.
  • Other surfactants that can be useful include Cremophor® EL, tyloxapol, octoxynol 40 (Triton® X405, and polyoxyl 40 stearate.
  • a formulation contains a surfactant (e.g., poloxamer 188) in a concentration of about 0.05%, 0.06%, 0.1% to 1.0%, 0.1% to 0.5%, 0.2% to 0.5%, or 0.1% to 0.2% w/v, for example, 0.1% w/v poloxamer 188.
  • Suitable surfactants and concentrations of such surfactants can be determined by testing whether the surfactant prevents aggregation in agitation studies. Methods of conducting such studies are known in the art. For example, it can be determined whether surfactant is needed to prevent precipitation from agitation stress. In such experiments, typically, a screen is performed using agitation and analysis. Examples of concentrations used for such studies are 0.01%, 0.02%, 0.06%, and 0.1% w/v surfactant, e.g., poloxamer 188.
  • aggregation and/or precipitation are assessed using analysis by spectrophotometry (A 280 ), visual inspection, size exclusion chromatography (SEC), light obscuration (e.g., using a HIAC device), or Micro-Flow ImagingTM (MFI, ProteinSimple, Santa Clara, Calif.).
  • a surfactant is generally selected for use in a formulation that is associated with the least amount of precipitation, e.g., no visible precipitation, or particle count that meets guidelines for particulate matter in injections (see, e.g., USP ⁇ 788>) or guidelines for particulate matter in ophthalmic solutions (see, e.g., USP ⁇ 789>).
  • the formulations featured in the invention contain one or more tonicity agents.
  • Suitable tonicity agents include, but are not limited to: sodium chloride, sorbitol; mannitol, sucrose, trehalose, or other sugars. Without committing to any theory, such agents may contribute to the surprising stability of a chimeric cytokine polypeptide.
  • a tonicity agent e.g., a sugar such as, e.g., sorbitol, provides or contributes to thermal stability.
  • the formulations featured in the invention are isotonic for the eye (e.g., having an osmolality of about 270-330 mOsm per kg).
  • the formulation has an osmolality of from about 250 to about 450 mOsm per kg, 300 to 400 mOsm per kg, 350 to 400 mOsm per kg, 200 to 375 mOsm per kg, or 350 to 375 mOsm per kg. In embodiments, the formulation has an osmolality of 270-330 mOsm per kg, e.g., about 320 mOsm per kg. Depending upon the tonicity agent, certain embodiments featured in the invention contain from about 1% to about 15% w/v; 2% to 12% w/v; 5% to 12% w/v; or 5% to 10% w/v.
  • a concentration is about 5% w/v, e.g., the concentration is 5% w/v.
  • sucrose or trehalose an example of a concentration is about 9% w/v.
  • a formulation featured herein contains one or more buffering agents.
  • buffering agents include, but are not limited to, phosphates; citrates; acetates; borates; succinates; and TRIS.
  • a salt of the buffering agents is a sodium salt or a potassium salt.
  • the buffering agent is present in an amount of from about 10 mM to about 50 mM, for example from about 20 mM to about 40 mM, to provide a weak buffering effect. This allows the formulation to be quickly neutralized at the administration site, e.g., on the surface of the eye, in the event of stinging or discomfort.
  • the buffering agent is present in an amount of about 10 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM or about 50 mM.
  • the buffering agent is a citrate, e.g., sodium citrate.
  • the buffering agent is citrate present at 10 mM.
  • the buffering agent is a weak buffering agent.
  • a suitable buffer is selected by conducting a stability study in which the polypeptide of interest is exposed to various buffers at various pH's, concentrations, temperatures, and for various times.
  • Buffers can be selected, for example by placing the polypeptide of interest in the buffer and subjecting the samples to elevated temperatures (accelerated stability testing) then test for physical stability (precipitation by visual inspection) or chemical stability, for example, by monitoring deamidation by weak cation exchange chromatography or oxidation by reversed phase chromatography. Additional assays can include monitoring of A 280 , SDS-PAGE, pH, and osmolality.
  • a buffer that provides the best physical and chemical stability is selected.
  • a formulation featured in the invention contains one or more amino acids.
  • Suitable amino acids include, but are not limited to: arginine, glutamic acid, histidine, or methionine.
  • the amino acid is typically selected to enhance the stability and/or the solubility of the protein. Methods of identifying such amino acids are known in the art.
  • a formulation such as a P05 formulation contains histidine or methionine.
  • a formulation contains an oxygen scavenger, e.g. methionine.
  • the formulation is in a plastic container.
  • the plastic container is sterilized using a method that generates free radicals, e.g., the container is sterilized using gamma radiation or ethylene oxide.
  • the formulation includes methionine, e.g., methionine at a concentration of 1-20 mM.
  • methionine is present at a concentration of 1-5 mM, 5-10 mM, 10-15 mM, 15-20 mM, or 5-15 mM.
  • methionine is present at about 1 mM, 5 mM, 10 mM, 15, mM, or 20 mM.
  • the formulation comprises methionine at a concentration of about 5 mM, e.g., at a concentration of 2.5-7.5 mM, 3-7 mM, or 4-6 mM.
  • a formulation comprising methionine in a sterilized plastic container and the amount of oxidation is less than that of a corresponding formulation that does not contain methionine.
  • formulations featured in the invention may contain one or more viscosity agents.
  • Suitable viscosity agents include, but are not limited to, methylcelluloses, including sodium carboxymethyl cellulose (also referred to herein as carboxymethyl cellulose or CMC); hydroxy celluloses, including ethyl cellulose; hydroxypropyl methylcellulose (hypromellose); carbomers, such as 934P, 971P and 974P; polyvinyl alcohol; xanthan gum; guar gum; gellan gum; and glycerin.
  • the formulations featured in the invention may also contain other pharmaceutically acceptable excipients. See e.g., Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott Williams & Wilkins Publishers (1999) (ISBN: 0683305727); Kibbe (ed.), Handbook of Pharmaceutical Excipients, 3rd ed. (2000) (ISBN: 091733096X); Protein Formulation and Delivery , McNally and Hastedt (eds.), Informa Health Care (ISBN: 0849379490) (2007).
  • Gennaro ed.
  • Remington The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472)
  • Ansel et al. Pharmaceutical Dosage Form
  • Penetration enhancers and bioadhesives may include, for example, chitosan, cytochalasin B, aminated gelatin, poly- ⁇ -caprolectone (carbopol 941P); poly(butylcyanoacrylate); poly-L-arginine; cyclodextrins; gellan; poly(acrylic acid); hyaluronic acid; mucin; alginate; a carbophil, and poloxamers (e.g., see Nagarwal et al., J Controlled Release, 136:2-13 (2009); Ding, PSTT 1:328-35 (1998); and Sahoo et al., Drug Discovery Today, 13:144-51(2008).
  • excipients may be useful as stabilizers, and can include, for example, glycerin, potassium chloride, potassium phosphate, propylene glycol, sodium acetate, sodium bisulfite, sodium borate, sodium borate decahydrate, sodium chloride, sodium citrate, sodium phosphate, sodium phosphate (including sodium phosphate monobasic and dibasic); zinc chloride, phenol, benzoate, derivatives of castor oil and ethylene oxides, and Cremophor® (BASF Corp., Germany)
  • Pharmaceutical compositions featured in the invention can be formulated in a variety of forms.
  • liquid, semi-solid, and solid dosage forms such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, including nanoparticles and liposomes.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions including nanoparticles and liposomes.
  • the form will generally depend on the intended mode of administration and therapeutic application.
  • Compositions for the agents described herein are typically in the form of injectable or infusible solutions, or are formulated for topical delivery, e.g., topical ocular delivery.
  • a pharmaceutical composition described herein is sterile and stable under the conditions of manufacture and storage.
  • a pharmaceutical composition can also be tested to ensure it meets regulatory and industry standards for administration.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug (e.g., a biologic) concentration.
  • Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating an agent described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • exemplary methods of preparation include vacuum drying and freeze-drying that yields a powder of an agent described herein plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be engineered by inclusion of an agent that delays absorption, for example, monostearate salts and gelatin. Such an agent may be particularly useful in a low-dose formulation.
  • the formulation comprises ⁇ 1 mg/ml of a therapeutic protein (e.g., a chimeric cytokine, e.g., P05) and gelatin is included in the formulation.
  • a formulation is prepared with a carrier, e.g., to extend the pharmacokinetics (PK) of a chimeric cytokine polypeptide (e.g., as assessed based on its half-life in the body, e.g., in the eye, e.g., on the cornea).
  • the chimeric cytokine polypeptide can be delivered, for example, as a controlled release formulation, delivered by an implant or a microencapsulated delivery system.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. See e.g., Sustained and Controlled Release Drug Delivery Systems , J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • preservatives can affect the chimeric cytokine polypeptide, e.g., causing changes to the structure of the polypeptide.
  • preservatives can cause in a subject, for example, an inflammatory response, which is antithetical to the desired treatment effect.
  • Formulations are sterile, stored, and filled into their final containers under sterile conditions.
  • a formulation as described herein that does not contain a therapeutic protein is useful for treating one or more signs or symptoms of ocular disease, e.g., dry eye disease, e.g., signs or symptoms of dry eye disease described herein.
  • a vehicle formulation comprises a surfactant, a tonicity agent, and a buffering agent.
  • the formulation is effective to decrease pain (e.g., pain as assessed using the pain question score or a visual analog scale), the OSDI, or a subscale of the OSDI.
  • the formulation decreases corneal fluorescein staining (CFS).
  • vehicle only formulation specifically refers to a formulation described herein that is substantially free of protein or peptide components, e.g., does not contain a therapeutic protein. It is to be understood that a vehicle formulation described herein can contain any therapeutic protein, e.g., a therapeutic polypeptide.
  • the vehicle formulation e.g., the vehicle only formulation, substantially comprises a surfactant (e.g., poloxamer 188), a tonicity agent (e.g., sorbitol), and a buffering agent (e.g., sodium citrate).
  • a surfactant e.g., poloxamer 188
  • a tonicity agent e.g., sorbitol
  • a buffering agent e.g., sodium citrate
  • the vehicle formulation is substantially free of protein.
  • the vehicle formulation is substantially free of a therapeutic protein or peptide.
  • the vehicle formulation does not contain a viscosity agent.
  • surfactants useful surfactants, tonicity agents, and buffering agents include those disclosed herein.
  • the surfactant is poloxamer 188
  • the tonicity agent is sorbitol
  • the buffering agent is sodium citrate and/or sodium phosphate.
  • the buffering agent is present at a concentration of 20 mM or less.
  • the vehicle formulation comprises about 0.1% w/v poloxamer 188, about 5% w/v sorbitol, and about 10 mM w/v sodium citrate.
  • the components of a vehicle formulation described herein are present in amounts that may vary around the values provided herein by up to 5%, 10%, 15%, 20%, 25%, 30%, 40%, or 50%.
  • the components of a vehicle formulation are present in amounts that vary around the values provided herein by 10%.
  • the vehicle formulation is an aqueous formulation consisting of 10 mM sodium citrate, pH 6.0, 5% sorbitol (w/v), and 0.1% poloxamer 188.
  • the vehicle formulation comprises 9.5-10.5 mM, 9-11 mM, 8.5-11.5 mM, 8-12 mM, 7.5-12.5 mM, 7-13 mM, 6-14 mM, or 5-15 mM sodium phosphate or sodium citrate. In embodiments, the vehicle formulation comprises 9.5-10.5 mM, 9-11 mM, 8.5-11.5 mM, 8-12 mM, 7.5-12.5 mM, 7-13 mM, 6-14 mM, or 5-15 mM sodium citrate.
  • the vehicle formulation comprises 4.75-5.25%, 4.5-5.5%, 4.25-5.75%, 4-6%, 3.75-6.25%, 3.5-6.5%, 3-7%, or 2.5-7.5% w/v sorbitol.
  • the vehicle formulation comprises 0.095-0.105%, 0.09-0.11%, 0.085-0.115%, 0.08-0.12%, 0.075-0.125%, 0.07-0.13%, 0.06-0.14%, or 0.05-0.15% w/v poloxamer 188.
  • the pH of the vehicle formulation is 5.5 to 7.5. In embodiments, the pH is 5.5 to 6.5. In embodiments, the pH is 6 to 7. In embodiments, the formulation comprises 8-12 mM sodium citrate, 4-6% w/v sorbitol, 0.08-0.12% w/v poloxamer 188, and has a pH of 5.5. to 7.5, e.g., a pH of 5.5 to 6.5.
  • the vehicle formulation comprises 9-11 mM sodium citrate, 4.5-5.5% w/v sorbitol, 0.09-0.11% w/v poloxamer 188, and has a pH of 5.5. to 7.5, e.g., a pH of 5.5 to 6.5.
  • the vehicle formulation comprises 7-13 mM sodium citrate, 3.5-5.5 w/v % sorbitol, 0.07-0.13% w/v poloxamer 188, and has a pH of 5.5. to 7.5, e.g., a pH of 5.5 to 6.5.
  • the vehicle formulation does not contain a viscosity agent.
  • the vehicle formulation comprises a viscosity agent, e.g., sodium carboxymethyl cellulose (CMC).
  • CMC carboxymethyl cellulose
  • the vehicle formulation comprises CMC, e.g., CMC at a concentration of 0.1-1% w/v, 0.1-0.5% w/v, or 0.2-0.3 w/v %.
  • the vehicle formulation comprises 0.1% w/v poloxamer 188, 5% w/v sorbitol, 0.25% w/v sodium carboxymethyl cellulose and 10 mM sodium phosphate.
  • the vehicle formulation has a pH of about 6.5.
  • the components of the formulation are present in amounts that may vary around the values provided by up to 5%, 10%, 15%, 20%, 25%, or 30%.
  • the vehicle formulation consists of 0.1% w/v poloxamer 188, 5% w/v sorbitol, 0.25% w/v sodium carboxymethyl cellulose and 10 mM sodium phosphate.
  • the formulation comprises 0.08-0.12% w/v poloxamer 188, 4-6% w/v sorbitol, 0.2-0.3% w/v sodium carboxymethyl cellulose and 8-10 mM sodium phosphate.
  • the formulation has a pH of 5.5-7.5, e.g., a pH of 5.5-6.5.
  • a formulation featured herein e.g., a formulation containing a therapeutic protein such as a chimeric IL-1 inhibitor or a vehicle formulation, is administered topically to a subject, e.g., a human or other mammal such as a dog, cat, or horse, and, for example administered to the eye.
  • a formulation described herein can be administered to a subject, by any suitable method, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, intrasynovial, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural injection, intrasternal injection and infusion.
  • suitable modes of administration include topical (e.g., dermal or mucosal) or inhalation (e.g., intranasal or intrapulmonary) routes.
  • the route of administration is one of: intravenous injection or infusion, subcutaneous injection, or intramuscular injection.
  • the mode of administration for a formulation featured herein e.g., a chimeric cytokine formulation described herein
  • topical administration to the eye e.g., in the form of drops.
  • Examples of devices that may contain the formulation and/or be used for adminstration of the formulation include simple eye droppers, squeeze bottles with or without metering function, and blow/fill/seal (BFS) devices such as those manufactured by Catalent (Somerset, N.J.), multi-use devices using, for example tip-seal technology, silver/oligodynamic technology, sterile filters, collapsing primary containers, and the like.
  • BFS blow/fill/seal
  • Another consideration for a formulation is minimizing sticking to the delivery device or container.
  • the addition of surfactant, e.g., poloxamer 188 can minimize sticking of P05 to a container.
  • the container is suitable to provide a shelf-life of at least two years, e.g., at least 3 years, at least 4 years, or at least 5 years, e.g., at 5° C. In embodiments, the container is suitable to provide a shelf-life of at least 3 years at 5° C. In embodiments, the container is suitable to provide a shelf-life of at least 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, 10 months, or 12 months at RT.
  • the container is suitable to provide a shelf-life of at least 5 months at RT.
  • suitable container materials are known in the art, for example certain plastics, for example, low density polyethylene (LDPE), high densidy polyethylene (HDPE), or polypropylene.
  • the formulation can be prepared for single use application in a container or can be prepared for use in a multiuse container.
  • a formulation featured herein can be delivered intravitreally, e.g., to treat disorders that are associated with, for example, the posterior segment of the eye.
  • Methods of intravitreal administration are known in the art and include, for example, intraocular injection, implantable devices.
  • the formulation is administered intravitreally using an implantable device.
  • the formulation comprises a thermal stabilizer, e.g., sorbitol.
  • the sorbitol is present at a concentration of ⁇ 5% w/v.
  • Implantable devices can be, for example, nonbiodegradable devices such as polyvinyl alcohol-ethylene vinyl acetate polymers and polysulfone capillary fibers, biodegradable devices such as polylactic acid, polyglycolic acid, and polylactic-co-glycolic acid, polycaprolactones, and polyanhydrides. Devices can be delivered in forms such as nanoparticles, liposomes, or microspheres.
  • nonbiodegradable devices such as polyvinyl alcohol-ethylene vinyl acetate polymers and polysulfone capillary fibers
  • biodegradable devices such as polylactic acid, polyglycolic acid, and polylactic-co-glycolic acid, polycaprolactones, and polyanhydrides.
  • Devices can be delivered in forms such as nanoparticles, liposomes, or microspheres.
  • a formulation featured in the invention can be administered as a fixed dose, as weight determined dose (e.g., mg/kg), or as an age determined dose.
  • the formulations e.g., a vehicle formulation or a therapeutic formulation (a formulation that includes a therapeutic such as a therapeutic protein) can be administered, for example, four times a day; three times a day; twice a day; once every day; every other day; every third, fourth or fifth day; every week; every two weeks; every three weeks; every four weeks; every five weeks; monthly; every two months; every three months; every four months; every six months; or as needed (ad libitum).
  • the formulation is administered once, twice, or three times a day. In some such embodiments, the formulation is administered topically, e.g., to the surface of the eye.
  • a pharmaceutical composition can include a “therapeutically effective amount” of an agent described herein.
  • a therapeutically effective amount of an agent can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, e.g., amelioration of at least one disorder parameter (e.g., sign), or amelioration of at least one symptom of the disorder (and optionally the effect of any additional agents being administered).
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.
  • a “therapeutically effective amount” is determined in a population of individuals and the amount is effective in ameliorating at least one symptom or indication of a cytokine-related disorder, e.g., an IL-1-related disorder in at least 5%, 10%, 25%, 50%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of an affected population.
  • a formulation is typically administered in a therapeutically effective amount.
  • a therapeutically effective formulation is a vehicle formulation.
  • a therapeutically effective formulation comprises a therapeutic protein.
  • the formulation is administered to a subject having an IL-1-related disorder and the chimeric cytokine polypeptide comprises fragments of IL-1 ⁇ and Il-1Ra sequences.
  • a formulation contains, for example, 5 mg/ml to 20 mg/ml, 5 mg/ml or 20 mg/ml of the polypeptide.
  • the formulation is administered topically to the eye once, twice, three, four, five, or six times per day.
  • Pharmaceutical compositions can be administered using medical devices as described herein and as known in the art, e.g., implants, infusion pumps, hypodermic needles, and needleless hypodermic injection devices.
  • a device can include, e.g., one or more housings for storing pharmaceutical compositions, and can be configured to deliver unit doses of the chimeric cytokine polypeptide, and optionally a second agent.
  • the doses can be fixed doses, i.e., physically discrete units suited as unitary dosages for the subjects to be treated; each unit can contain a predetermined quantity of chimeric cytokine polypeptide calculated to produce the desired therapeutic effect in association with a pharmaceutical carrier and optionally in association with another agent, e.g., Restasis® or artificial tears such as those available as over the counter or prescribed products.
  • the formulation is administered to a subject having the disorder in an amount and for a time sufficient to induce a sustained improvement in at least one sign or symptom of the disorder.
  • An improvement is considered “sustained” if the subject exhibits the improvement over a prolonged period, e.g., on at least two occasions separated by one to four weeks.
  • the degree of improvement can be determined based on signs or symptoms, and can also employ questionnaires that are administered to the subject, such as quality-of-life questionnaires.
  • the chimeric cytokine polypeptide comprises fragments of an IL-1 ⁇ and an IL-1Ra and is topically administered at least once per week, e.g., at least once per day, at least twice per day, or at least three times per day.
  • Improvement can be induced by repeatedly administering a dose of the formulation until the subject manifests an improvement over baseline for selected signs and/or symptoms.
  • the amount of improvement can be evaluated by repeated administration over a period of at least a month or more, e.g., for one, two, or three months or longer, or indefinitely.
  • the agent can be administered for a period of one to six weeks or even as a single dose.
  • treatment can be continued indefinitely at the same level or at a reduced dose or frequency. Treatment can also be discontinued, e.g., upon improvement or disappearance of signs or symptoms. Once treatment has been reduced or discontinued, it may be resumed if symptoms should reappear.
  • formulations featured herein comprise a therapeutic protein.
  • the formulations comprise a chimeric receptor binding agent (e.g., a chimeric cytokine) such as one that can bind to an IL-1R and that can antagonize IL-1 signaling, and therefore can be used to treat an “IL-1 related disorder,” which includes any disease or medical condition that is (i) caused at least in part by IL-1 agonism, (ii) is associated with elevated levels or activity of an IL-1 signaling component (such as IL-1 ⁇ , IL-1 ⁇ , or IL-1RI) or elevated IL-1 signaling, and/or (iii) is ameliorated by decreasing IL-1 activity.
  • IL-1 related disorders include acute and chronic disorders, including autoimmune disorders and inflammatory disorders.
  • IL-1 related disorders include systemic and non-systemic disorders. It is well established that IL-1 ⁇ and IL-1 ⁇ are potent pro-inflammatory cytokines implicated in infectious responses as well as in inflammatory disease, including, e.g., rheumatoid arthritis. Increased IL-1 production has been observed in patients with certain autoimmune disorders, ischemia, and various cancers, therefore implicating IL-1 in these and related diseases (for example, see Sims and Smith, Nature Rev Immunol, 10:89-102 (2010)).
  • the term “treat” refers to the administration of an agent described herein to a subject, e.g., a patient, in an amount, manner, and/or mode effective to improve a condition, symptom, or parameter associated with a disorder, e.g., a disorder described herein, or to prevent the onset or progression of a disorder, to either a statistically significant degree or to a degree detectable to one skilled in the art.
  • the treatment can be to cure, heal, alleviate, relieve, alter, remedy, ameliorate, palliate, improve or affect the disorder, the symptoms of the disorder or the predisposition toward the disorder.
  • An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject. Exemplary subjects include humans, primates, and other non-human mammals.
  • a formulation featured in the invention can also be given prophylactically to reduce the risk of the occurrence of a disorder or symptom or sign thereof.
  • the IL-1-related disorder can be an autoimmune disorder.
  • IL-1-related autoimmune disorders include rheumatoid arthritis, ankylosing spondylitis, Behçet's syndrome, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis), asthma, psoriasis, type I diabetes, some forms of acne, and other disorders identified herein.
  • the formulations described herein can be administered to a subject having or at risk for such IL-1 mediated autoimmune disorders.
  • the IL-1 mediated disorder can be an inflammatory disorder such as described below.
  • the formulations described herein can be administered to a subject having or at risk for such IL-1 mediated inflammatory disorders.
  • IL-1-related ocular disorders include Sjögren's syndrome (e.g., keratoconjunctivitis sicca associated with Sjögren's syndrome), dry eye disorders including keratoconjunctivitis sicca (Sjögren's-associated or non-Sjögren's-associated), keratitis sicca, sicca syndrome, xerophthalmia, tear film disorder, decreased tear production, aqueous tear deficiency, dry eye associated with graft-versus-host disease, and Meibomian gland dysfunction.
  • Subjects having a dry eye disorder can exhibit inflammation of the eye, and can experience scratchy, stingy, itchy, burning or pressured sensations, irritation, pain, and redness. Dry eye disorders can be associated with excessive eye watering and insufficient tear production.
  • a formulation featured in the invention can be administered to such a subject to ameliorate or prevent the onset or worsening of one or more such symptoms.
  • a formulation featured in the invention can also be used to mitigate pain, e.g., ocular pain, such as pain due to neuroinflammation, in a subject.
  • the embodiments described herein include methods of treating animals having IL-1-related disorders, for example, dry eye disorders.
  • Dry eye can be a serious disorder in, for example canines.
  • disorders in dogs associated with dry eye include congenital disorders, infections (e.g., canine distemper virus), drug induction (e.g., by sulfa antibiotics), and removal of the tear gland of the third eyelid (“cherry eye”).
  • Dry eye disorders are also commonly seen in certain dog breeds, for example, Cocker Dog, Shih Tzu, Lhasa Apso, Bulldog, Schnauzer, and West Highland White Terrier.
  • Other non-limiting examples of animals that can be treated include cats and horses.
  • the formulations featured herein can also be used to treat other disorders affecting the surface of the eye, such as the cornea.
  • disorders include corneal ocular surface inflammatory conditions, corneal neovascularization, keratitis, including peripheral ulcerative keratitis and microbial keratitis.
  • the formulations can be used to treat a subject undergoing corneal wound healing (e.g., a subject having a corneal wound).
  • the formulation can be administered to a subject who is about to receive, undergoing, or recovering from a procedure involving the eye, e.g., corneal transplantation/keratoplasty, keratoprosthesis surgery, lamellar transplantation, selective endothelial transplantation.
  • the formulation can be used to treat disorders affecting the conjunctiva, including conjunctival scarring disorders and conjunctivitis, e.g., allergic conjunctivitis, for example, severe allergic conjunctivitis.
  • the formulation can be used to treat still other disorders such as pemphigoid syndrome and Stevens-Johnson syndrome.
  • the formulations featured in the invention can be administered to a subject to modulate neovascularization in or around the eye. See, e.g., Dana (2007) Trans Am Ophthalmol Soc 105: 330-43.
  • the formulations of the present invention can be administered to a subject having an allergic reaction affecting the eye, e.g., a subject experiencing severe allergic (atopic) eye disease such as, e.g., allergic conjunctivitis.
  • the formulation can be administered topically. See also, e.g., Keane-Myers et al. (1999) Invest Ophthalmol Vis Sci, 40(12): 3041-6.
  • the formulations featured in the invention can be administered to a subject having an autoimmune disorder affecting the eye.
  • autoimmune ocular disorders include sympathetic ophthalmia, Vogt-Koyanagi Harada (VKH) syndrome, birdshot retinochoriodopathy, ocular cicatricial pemphigoid, Fuchs' heterochronic iridocyclitis, and various forms of uveitis.
  • the formulations can be administered to a subject to treat any of the foregoing disorders.
  • the formulations featured in the invention can be administered to a subject who has or is at risk for diabetic retinopathy. See, e.g., Demircan et al. (2006) Eye 20:1366-1369 and Doganay et al. (2006) Eye, 16:163-170
  • Uveitis includes acute and chronic forms and includes inflammation of one or more of the iris, the ciliary body, and the choroid. Chronic forms may be associated with systemic autoimmune disease, e.g., Behçet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease.
  • systemic autoimmune disease e.g., Behçet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease.
  • systemic autoimmune disease e.g., Behçet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease.
  • anterior uveitis inflammation is primarily in the iris (also ulceris).
  • Anterior uveitis can affect subjects who have systemic autoimmune disease, but also subjects
  • Pan planitis results from inflammation of the pars plana between the iris and the choroid.
  • Posterior uveitis involves the uveal tract and primarily the choroid, and is also referred to as choroiditis.
  • Posterior uveitis can be associated with a systemic infection or an autoimmune disease. It can persist for months and even years.
  • the formulations featured in the invention can be administered to a subject to treat any of the foregoing forms of uveitis. See also e.g., Tsai et al. (2009) Mol Vis 15:1542-1552 and Trittibach et al. (2008) Gene Ther. 15(22): 1478-88.
  • the formulations featured in the invention are used to treat a subject having or at risk for age-related macular degeneration (AMD).
  • AMD age-related macular degeneration
  • the formulations can be applied topically to the eye, injected (e.g., intravitreally) or provided systemically. See, e.g., Olson et al. (2009) Ocul Immunol Inflamm 17(3):195-200.
  • a formulation described herein can be administered by any mode to treat an ocular disease.
  • the agent can be delivered by a parenteral mode.
  • the formulation can be delivered directly to the eye or in the vicinity of the eye.
  • the formulation can be administered topically or intraocularly, e.g., as described herein.
  • Ophthalmic formulations featured in the invention can be delivered for topical administration, e.g., for administration as a liquid drop, an ointment, or a gel, or for implantation, e.g., into an anterior chamber of the eye or the conjunctival sac.
  • Drops, such as liquid drops, can be delivered using an eye dropper.
  • Gels and ointments can also be administered using a dropper.
  • an active agent e.g., the chimeric cytokine protein or receptor binding agent
  • an active agent can be present at 0.0001% to 0.1%, 0.001% to 5%, e.g., 0.005% to 0.5%, 0.05% to 0.5%, 0.01% to 5%, 0.1% to 2% or 1% to 5% concentration.
  • the concentration is 2%, e.g., of P05. In other embodiments, the concentration is 0.5%, e.g., of P05.
  • the receptor binding agent e.g., P05 is formulated on a mg/ml basis, e.g., as described supra.
  • the active agent e.g., the receptor binding agent
  • the active agent is present at a concentration of 1 mg/ml, 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 8 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, or 50 mg/ml.
  • the active agent e.g., the IL-1 inhibitor, is present at a concentration of up to 100 mg/ml.
  • the ophthalmic formulation is applied directly to the eye including onto the cornea, the eyelid or instillation into the space (cul-de-sac) between the eyeball and the eyelids.
  • the ophthalmic formulation can be designed to mix readily with the lacrimal fluids and spread over the surfaces of the cornea and conjunctiva.
  • the major portion of the drug is typically deposited in the lower fornix. Capillarity, diffusional forces, and the blinking reflex drive incorporation of the drug in the precorneal film from which it penetrates into and through the cornea.
  • Ophthalmic formulations featured in the invention can also include one or more other agents, e.g., an anti-inflammatory steroid such as rimexolone, loteprednol, medrysone and hydrocortisone, or a non-steroidal anti-inflammatory.
  • an anti-inflammatory steroid such as rimexolone, loteprednol, medrysone and hydrocortisone, or a non-steroidal anti-inflammatory.
  • the steroid can be present at a concentration of 0.001% to 1%. In some embodiments, no steroid is present.
  • the receptor binding agent is the only active agent in the formulation.
  • the formulation can also include one or more of the following components as described herein: surfactants, tonicity agents, buffers, preservatives, co-solvents and viscosity building agents.
  • Tonicity agents can be used to adjust the tonicity of the composition, e.g., to that of natural tears.
  • Tonicity agents, particularly sugars, may also function as thermal stabilizers.
  • potassium chloride, sodium chloride, magnesium chloride, calcium chloride, dextrose and/or mannitol may be added to achieve an appropriate tonicity, e.g., physiological tonicity.
  • Tonicity agents can be added in an amount sufficient to provide an appropriate osmolality as described herein.
  • a tonicity agent is added to provide an osmolality of about 150 mOsm per kg to 450 mOsm per kg or 250 mOsm per kg to 350 mOsm per kg. In embodiments, a tonicity agent is added to provide an osmolality that is isotonic in the eye. In embodiments, a tonicity agent, e.g., sorbitol, is added to provide an osmolality of 270-330 mOsm per kg.
  • the formulation can also include buffering suitable for ophthalmic delivery and as described herein.
  • the buffer can include one or more buffering components such as a citrate, phosphate, borate, boric acid, succinate, acetate or a pharmaceutically acceptable salt thereof (e.g., sodium phosphate, sodium acetate, sodium citrate, sodium borate, sodium succinate, or sodium acetate), to changes in pH.
  • the buffering component can be used especially under storage conditions, e.g., when the formulation will be subjected to prolonged storage.
  • the buffer can be selected to provide a target pH within the range of pH 5.5-6.5, pH 5.5-6.0, pH 6.0 to 7.5, or pH 6.5 to 7.5.
  • the buffering agent is a weak buffering agent, wherein the concentration of the buffering components is below 20 mM. In embodiments, the concentration of the buffering components is between about 5 to 20 mM, e.g., 5 to 15 mM, e.g., 5 to 10 mM.
  • the formulation comprising a therapeutic protein can include an aqueous or phospholipid carrier.
  • the formulation can include agents to provide short-term relief, e.g., compounds that lubricate the eye and assist in tear formation.
  • phospholipid carriers which include one or more phospholipids
  • artificial tears compositions useful as artificial tears carriers include commercial products such as Tears Naturale® (Alcon Labs, Inc., TX USA).
  • the formulation can include: 1 mg dextran, 70 and 3 mg hydroxypropyl methylcellulose, and optionally a preservative such POLYQUAD® (polyquaternium-1) 0.001% (m/v).
  • phospholipid carrier formulations include those disclosed in U.S. Pat. No. 4,804,539, U.S. Pat. No. 4,883,658, U.S. Pat. No. 5,075,104, U.S. Pat. No. 5,278,151, and U.S. Pat. No. 5,578,586.
  • the formulation can also include other compounds that act as a lubricant or wetting agent.
  • viscosity agents such as: monomeric polyols, such as, glycerol, propylene glycol, ethylene glycol; polymeric polyols, such as polyethylene glycol, various polymers of the cellulose family: hydroxypropylmethyl cellulose (“HPMC”), sodium carboxymethyl cellulose, hydroxy propylcellulose (“HPC”), dextrans, such as dextran 70; water soluble proteins, such as gelatin; and vinyl polymers, such as polyvinyl alcohol, polyvinylpyrrolidone, povidone and carbomers, such as carbomer 934P, carbomer 941; carbomer 940, carbomer 974P.
  • HPMC hydroxypropylmethyl cellulose
  • HPC hydroxy propylcellulose
  • dextrans such as dextran 70
  • water soluble proteins such as gelatin
  • vinyl polymers such as polyvinyl alcohol, polyvinylpyrrol
  • Still additional examples include polysaccharides, such as hyaluronic acid and its salts and chondroitin sulfate and its salts, and acrylic acid polymers.
  • the formulation has a viscosity between 1 cP to 400 cP.
  • the formulation e.g., a vehicle formulation
  • the formulation can include one or more preservatives, e.g., to prevent microbial and fungal contamination during use, and/or one or more detergents, or surfactants, e.g., to solubilize proteins.
  • preservatives include: benzalkonium chloride, chlorobutanol, benzododecinium bromide, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, and polyquaternium-1, and can be included at a concentration of from 0.001 w/v to 1.0% w/v.
  • a formulation containing a therapeutic protein as described herein is sterile yet free of preservatives.
  • Exemplary detergents/surfactants include Pluronics®, such as F-68; Triton® surfactants, such as Triton X-100, polysorbates, such as Tween-20 and Tween-80, ElugentTM, and Cremophor® polyethoxylated castor oil, as well as tyloxapol, octoxynol 40 and polyoxyl 40 stearate.
  • Pluronics® such as F-68
  • Triton® surfactants such as Triton X-100
  • polysorbates such as Tween-20 and Tween-80, ElugentTM, and Cremophor® polyethoxylated castor oil, as well as tyloxapol, octoxynol 40 and polyoxyl 40 stearate.
  • detergents and/or surfactants can be included at a concentration of from 0.001% w/v to 1.0% w/v. In some aspects, the formulation is free of detergents.
  • Ophthalmic packs may be used to give prolonged contact of an ophthalmic formulation with the eye.
  • a cotton pledget is saturated with the formulation and then inserted into the superior or inferior fornix.
  • the formulation may also be administered by the way of iontophoresis. This procedure keeps the solution in contact with the cornea in an eyecup bearing an electrode. Diffusion of the drug is effected by difference of electrical potential.
  • Iontophoretic systems which have been used include Ocuphor®1 (Iomed Inc., USA); Eyegate® II Delivery Systeml (EyeGate Pharma, USA); and Visulex®1 (Aciont Inc., USA). See Amo and Urtti, Drug Discovery Today, 13:143 (2008).
  • Another strategy for sustained ocular delivery is the use of gelifying agents. These materials can be delivered in a liquid form, as an eye drop or intraocular injection. After instillation the polymer undergoes a phase change and forms a semi-solid or solid matrix that releases the drug over prolonged period. The phase transition can be induced by changes in the temperature, ion concentration, or pH.
  • the gel forming solutions such as Timoptic®-XE1 (Merck and Co. Inc., USA), which contains Gelrite® (purified anionic heteropolysaccharide from gellan gum); Pilogel®1 (Alcon, Inc., Switzerland) eye drops contain poly(acrylic acid); and Azasite®1 (Insite Vision, USA) have been tested clinically. These materials enhance the drug retention relative to the conventional eye drops and lead to increased drug absorption into the eye and reduced dosing frequency. See Amo and Urtti, Drug Discovery Today, 13:135-143 (2008).
  • a formulation featured in the invention can be delivered by injection, e.g., intravitreal, periocular, or subconjunctival injection.
  • the formulation can be injected underneath the conjunctiva facilitating passage through the sclera and into the eye by simple diffusion.
  • the formulation can also be injected underneath the conjunctiva and the underlying Tenon's capsule in the more posterior portion of the eye to deliver the agent to the ciliary body, choroid, and retina.
  • the formulation may also be administered by retrobulbar injection.
  • a formulation provided herein is administered intravitreally. In embodiments, the formulation does not comprise CMC.
  • subjects can be evaluated using one or more of the approaches known in the art, for example, the Ocular Surface Disease Index (OSDI), corneal and conjunctival staining, and the Schirmer test.
  • OSDI Ocular Surface Disease Index
  • corneal and conjunctival staining corneal and conjunctival staining
  • Schirmer test a negative change from baseline indicates an improvement in vision-related function and the ocular inflammatory disorders.
  • 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 can be graded, e.g., according to the NEI scale, the Oxford Schema, or a modified Oxford Schema.
  • staining is graded according to the NEI scale, which is a 15 point scale where the cornea is divided into 5 sections (a central circular section, and 4 quadrants surrounding the central corneal section which are referred to as inferior, superior, nasal and temporal quadrants) each of which is scored from 0-3 for punctate staining to yield a maximum possible score of 15.
  • NEI scale is a 15 point scale where the cornea is divided into 5 sections (a central circular section, and 4 quadrants surrounding the central corneal section which are referred to as inferior, superior, nasal and temporal quadrants) each of which is scored from 0-3 for punctate staining to yield a maximum possible score of 15.
  • Conjunctival staining is likewise a measure of epithelial disease or break in the epithelial barrier of the ocular surface. 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 two minutes later. Using white light of moderate intensity, only the interpalpebral region of the nasal and temporal conjunctival staining is graded, e.g., using the Oxford Schema.
  • the Schirmer test is performed in the presence or 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.
  • Efficacy of the formulations featured in the invention can be evaluated in a mouse model for dry eye disease.
  • Dry eye can be induced in mice by subcutaneous injection of scopolamine and then placement of the mice in controlled-environment chambers.
  • normal healthy 6 to 10 weeks old female C57BL/6 mice can be induced to have dry eye by continuous exposure to dry environment in a controlled environmental chamber.
  • the chamber has low relative humidity of less than 30% (generally about 19%), high airflow (15 liters/minute) and constant temperature (about 22° C.).
  • the mice placed in the chamber are also treated with scopolamine to inhibit tear secretion.
  • Sustained-release transdermal scopolamine patches can be obtained from Novartis (Summit, N.J.).
  • One-fourth of a patch is applied to the depilated mid-tail of mice every 48 hours.
  • the combination of the controlled environmental chamber and scopolamine produces severe dry eye in a relative short period of time (about 2-4 days).
  • the controlled environmental chamber can be prepared as described in Barbino et al. (Invest Ophthal Vis Sci, 46: 2766-2711 (2005)), and enables control of air flow, humidity, and temperature.
  • Mice can be monitored for signs of dry eye, e.g., by performing: a) cotton thread test to measure aqueous tear production, which is generally decreased in patients with dry eye; b) corneal fluorescein staining which is a marker of corneal surface damage; and general ophthalmic examination.
  • Cotton Thread Test Tear production can be measured with cotton thread test, impregnated with phenol red (Zone-Quick, Lacrimedics, Eastsound, Wash.). Under a magnifying fluorescent lamp, the thread is held with jeweler forceps and placed in the lateral cantus of the conjunctival fornix of the right eye for 30 or 60 seconds. The tear distance in mm is read under a microscope using the scale of a hemacytometer.
  • Corneal Fluorescein Staining can be evaluated by applying 1.0 ml of 5% fluorescein by a micropipette into the inferior conjunctival sac of the eye. The cornea is examined with a slit lamp biomicroscope using cobalt blue light 3 minutes after the fluorescein instillation. Punctuate staining is recorded in a masked fashion using a standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas in which the corneal surface has been divided.
  • NKI National Eye Institute
  • Therapeutic Proteins e.g., Chimeric Proteins
  • Nucleic acids encoding the proteins with the amino acid sequences listed in Table 1 were constructed in a pET vector containing a T7 promoter and ampicillin (pET31 series) or kanamycin resistance genes (pET28 series) (EMD Chemicals, Gibbstown, N.J., USA), and expressed. Examples of coding sequences that can be used for expression are provided in Table 2.
  • the nucleic acid sequence further includes an ATG prior to the first nucleotide listed below. In some embodiments, the nucleic acid sequence further includes a stop codon (such as TAA, TAG, or TGA) after the last nucleotide listed below.
  • a stop codon such as TAA, TAG, or TGA
  • the cytokine domains can have 48-70% residues from IL-1 ⁇ and 55-78% residues from IL-1Ra. Because a number of amino acid residues are conserved between the two proteins, the sum of the percentage identity to IL-1 ⁇ and to IL-1Ra can be greater than 100%.
  • IL-1 ⁇ Total residues IL-1RA residues residues % IL-1 ⁇ % IL-1RA P06 130 62 152 85.5 40.8 P07 113 80 153 73.9 52.3 P05 108 85 153 70.6 55.6 P04 104 89 153 68.0 58.2 P03 94 99 153 61.4 64.7 P02 85 108 153 55.6 70.6 P01 740 119 153 48.4 77.8
  • Other examples of therapeutic proteins include IL-1Ra (e.g., anakinra), canakinumab, gevokizumab, rilanacept, or an anti-IL-1R antibody (e.g., as produced by Amgen).
  • Proteins that contain a hexa-histidine tag were expressed in E. coli cells BL21(DES) strain by induction with 1 mM isopropyl ⁇ -D-1-thiogalactopyranoside (IPTG) at 37° C. for 3 hours in LB broth media.
  • the cells were lysed in 20-50 mM Tris, 0.5 M NaCl, 2.5 mM EDTA, 0.1% Triton X-100, pH 8.0.
  • Lysate was dialyzed against 1.25 ⁇ PBS containing 0.1% polysorbate 80, then sterile filtered through a 0.8/0.2 ⁇ m filter before being subjected to immobilized ion affinity chromatography (IMAC) using a HisTrap HP® pre-packed column (GE Healthcare, Piscataway N.J., USA).
  • the column was equilibrated in 50 mM phosphate, 500 mM NaCl, pH 7.1, loaded, and washed with same buffer. It was pre-eluted with 25 mM imidazole and eluted with 125 mM imidazole in same buffer. Eluted protein was dialyzed extensively against 1.25 ⁇ PBS, 0.1% polysorbate 80, pH 7.4.
  • the protein was loaded in 20 mM sodium phosphate, 0.5 M NaCl 10 mM imidazole, pH 7.4 buffer. It was eluted with 200 mM imidazole, 20 mM sodium phosphate, 0.5 M NaCl pH 7.4 buffer. Eluted protein was dialyzed extensively against PBS, 0.1% polysorbate 80, pH 7.4, concentrated using an Amicon Ultra® (10K) filter, and stored at 4° or ⁇ 80° C.
  • Proteins lacking a hexa-histidine tag were purified by ion exchange chromatography.
  • a 5 ml fraction containing the eluted protein was then diluted with 5 ml of H 2 O and 5 ml of 20 mM Tris pH 8) and then applied to CaptoTM Q resin (GE Healthcare, Piscataway N.J., USA) and eluted with a 0 mM to 250 mM NaCl gradient in 20 mM Tris pH 8.0.
  • the eluted protein was dialyzed extensively against 1.25 ⁇ PBS 0.1% TWEEN® 80 or 1.25 ⁇ PBS lacking TWEEN® and stored. See FIG. 1 .
  • P03 and P04 proteins were purified using similar methods.
  • Cells expressing P05 were also grown in TEKNOVATM Terrific Broth with animal free soytone (# T7660) supplemented with 10 g/L glucose, 10 mM MgSO 4 , trace elements (1 mg/ml TEKNOVATM 1000 ⁇ Trace Elements, #T1001), and antibiotic in a Sartorius 2L BIOSTATTM A+ and were induced at OD 35-40 with 1 mM IPTG for about 6 hours.
  • Cells were grown at 37° C. with 30% dissolved oxygen at pH 7.0, and agitation at 200-800 rpm with oxygen sparge at 2 L/min Cells were fed 9 g glucose/L/hr when glucose was depleted as detected by a pH increase. Feed was reduced to 6 g glucose/L/hr when the pH decreased (about 2.5 hrs after induction).
  • lysis buffer (20 mM Tris, 10 mM EDTA, 0.1% Triton, pH 8.0; 20 mM Tris, 10 mM EDTA, 0.1% Triton, pH 7.0; 50 mM MOPS, 10 mM EDTA, 0.1% Triton, pH 6.5; or 50 mM MOPS, 10 mM EDTA, 0.1% Triton, pH 6.0). Lysate is loaded onto Poros® XS cation ion exchange media (Life Technologies Corp., Carlsbad Calif. USA) at pH 5.3 and 3 mS/cm (35 mg product per ml column resin).
  • P05 protein is eluted by a step to pH 7.0 using buffer containing 100 mM MOPS 25 mM NaCl pH 7.0. The first eluting peak was discarded, and the second eluting peak was collected in pools and contained P05 protein. Early pools are enriched for intact P05 protein relative to a des-Ala species. This eluted material is then flowed over Capto®Q anion exchange resin. The flow through, which contains intact P05 protein, is collected.
  • the media is washed with 100 mM MOPS 20 mM NaCl pH 6.0.
  • P05 protein is eluted by a step to pH 6.0 using buffer containing 100 mM MOPS 50-58 mM NaCl pH 6.0.
  • the first eluting peak was separated from subsequent peaks and contained intact P05 protein.
  • This eluted material is then flowed over Capto®Q anion exchange resin. The flow through, which contains intact P05 protein, is collected.
  • HEK-BlueTM IL-1 ⁇ responsive cells were used to monitor IL-1 ⁇ activity (available from InvivoGen Inc., San Diego Calif., USA). These cells include a secreted embryonic alkaline phosphatase (SEAP) reporter gene under the control of the IFN- ⁇ minimal promoter fused to five NF-kB and five AP-1 binding sites. IL-1 ⁇ engagement of IL-1 receptors on the cell surface led to NF-kB activation and SEAP production.
  • SEAP secreted embryonic alkaline phosphatase
  • the SEAP report can be detected, e.g., using QUANTI-BlueTM (InvivoGen Inc., San Diego Calif., USA) and spectrophotometric analysis.
  • a HEK-Blue IL-1 ⁇ cell suspension was prepared from cells cultured to 70-80% confluence. The resuspended cells were adjusted to ⁇ 330,000 cells/ml in fresh growth medium (DMEM, 4.5 g/l glucose, 2 mM L-glutamine, 10% (v/v) heat-inactivated fetal bovine serum (30 minutes at 56° C.), 50 U/mL penicillin, 50 mg/ml streptomycin, 100 mg/ml Normocin®T).
  • DMEM fresh growth medium
  • penicillin 50 mg/ml streptomycin
  • 100 mg/ml Normocin®T 100 mg/ml Normocin®T.
  • Reagents were added to wells of a flat-bottom 96-well cell culture plate: 10 ⁇ l of IL-1 ⁇ at 20 ng/ml, 10 ⁇ l of the agent of interest, and 30 ⁇ l of cell culture medium to a final volume of 50 ⁇ l. Positive and negative control samples were prepared in parallel. Then 150 ⁇ l of HEK-Blue IL-1 ⁇ cell suspension (50,000 cells) was added to each well and the plate was cultured overnight at 37° C. in 5% CO 2 tissue culture incubator. Generally, the final IL-1 ⁇ concentration (in the 200 ⁇ l final volume) was 0.1 ng/ml. IL-1 ⁇ activity was evaluated the next day (12-15 hours later).
  • the QUANTI-BlueTM reagent was prepared according to the manufacturer's instructions.
  • a flat bottomed 96-well assay plate was prepared in which 150 ⁇ l of QUANTI-BlueTM solution was added to each well.
  • 50 ⁇ l of conditioned media from the wells of the 96 well tissue culture plate was added to each well of the assay plate.
  • the plate was incubated at 37° C. for approximately 15-20 minutes. SEAP levels were then measured using a spectrophotometer at 620-655 nm.
  • FIG. 2A shows antagonism of IL-1 ⁇ activity by P01 at a range of IL-1 ⁇ protein concentrations using the HEKBlueTM cell assay described herein. Antagonism increased with increasing amounts of P01 (x-axis reflects microliters of supernatant containing P01).
  • the proteins P01, P02, P03, P04, and P05 each antagonized IL-1 ⁇ activity. See FIG. 3A and FIG. 3B , for example.
  • the IC50 of P05 was less than about 5 ng/ml.
  • P05 was test for ability to agonize IL-1RI in this assay and was not observed to have any detectable agonistic activity even at the highest concentrations tested, 1 mg/ml.
  • P01, P02, P03, P04, and P05 also inhibited IL-1 ⁇ induced IL-6 expression in MG-63 cells, a human osteosarcoma cell line that is responsive to IL-1 ⁇ .
  • hexa-histidine tagged (SEQ ID NO:23) P05 was observed to have biological activity. See also Example 9 below regarding untagged P05.
  • binding properties of proteins for soluble recombinant human IL-1RI were evaluated using surface plasmon resonance with a Reichert SR7000DC Dual Channel SPR system. Binding was evaluated in phosphate buffered saline with 0.005% Tween 20.
  • IL-1 ⁇ was observed to have a K D of between 8-9 nM and a dissociation constant (K d ) of between 2-3 ⁇ 10 ⁇ 3 s ⁇ 1 , and in another experiment a K D of about 2 nM, an association constant of 1.3-1.5 ⁇ 10 6 M ⁇ 1 s ⁇ 1 , and a dissociation constant (K d ) of about 2.9-3.0 ⁇ 10 ⁇ 3 s ⁇ 1 .
  • the P01 protein bound with similar association kinetics as IL-1 ⁇ , but did not dissociate during of the dissociation phase of the binding experiment (about 180 seconds). Thus, the P01 protein bound to IL-1RI with a greater affinity than did IL-1 ⁇ under similar conditions.
  • Binding of IL-1Ra was observed to have a K D of about 0.33 nM, an association constant (K a ) of about 2 ⁇ 10 5 M ⁇ 1 s ⁇ 1 , and a dissociation constant (K d ) of about 6.6 ⁇ 10 ⁇ 5 s ⁇ 1 .
  • Chimeric cytokine domains P01, P02, P03, P04, and P05 were observed to have K D ranging from about 12-1700 pM, an association constant (K a ) ranging from about 3 ⁇ 10 4 M ⁇ 1 s ⁇ 1 to 3 ⁇ 10 6 M ⁇ 1 s ⁇ 1 , and a dissociation constant (K d ) ranging from about 2 ⁇ 10 ⁇ 5 to 1 ⁇ 10 ⁇ 3 s ⁇ 1 . See Table 3 below.
  • Additional exemplary chimeric IL-1 family proteins also include the following:
  • the polypeptide below is a chimeric domain that includes at least two segments from IL-1 ⁇ and at least two segments from IL-1Ra.
  • the formulation has a pH of 6.5.
  • the formulation is tested for stability at two weeks at room temperature and up to at least twelve months storage stability at 2-8° C. as measured using one or more of reverse phase HPLC (RP-HPLC); weak cation exchange HPLC (WCEX-HPLC); spectrophotometry (A280); and visual assays.
  • RP-HPLC reverse phase HPLC
  • WCEX-HPLC weak cation exchange HPLC
  • A280 spectrophotometry
  • Formulations of P05 utilized in Phase 1 clinical studies were aqueous formulations that contained sodium carboxymethylcellulose in a concentration of 0.25% w/v; poloxamer 188 in a concentration of 0.1% w/v; sorbitol in a concentration of 5% w/v; sodium phosphate in a concentration of 10 mM, and P05 in a concentration of either 5 or 20 mg/mL.
  • the formulation has a pH of about 6.5.
  • Proteins P03, P04, P05, mIL-1Ra (methionyl IL-1Ra), and IL-1 ⁇ were prepared in phosphate-buffered saline (PBS), pH 7.4, at 0.5 mg/ml.
  • the proteins were combined with SYPRO® orange dye (Invitrogen, Calif.) at a 1:500 dilution of the stock concentration and subject to differential scanning fluorimetry. See, e.g., He et al. (2010) J Pharm Sciences, 99 1707-1720. Fluorescence measurements were monitored using an Agilent Mx3005 QPCR machine as the temperature was increased from 25° C. to 95° C. at a rate of 1° C. per minute.
  • T m Melting temperature
  • P04 has a T m that is about four degrees higher than mIL-1Ra and IL-1 ⁇ and exhibits an onset of unfolding about three degrees higher than mIL-1Ra and about ten degrees higher than IL-1 ⁇ .
  • P03 and P05 have a T m that is about nine degrees higher than mIL-1Ra and IL-1 ⁇ and exhibit an onset of unfolding about 11 degrees higher than mIL-1Ra and about 18 degrees higher than IL-1 ⁇ .
  • Purified P05 (lacking a hexa-histidine tag (SEQ ID NO:23)) was prepared in 1.25 ⁇ PBS and tested in a murine model of dry eye disease.
  • female C57BL/6 mice 6 to 10 weeks of age from Jackson Laboratories (acclimated for 1 to 2 weeks in an animal holding room with ⁇ 30% relative humidity, hydrogel food supplement, and Enviro-DriTM environment enrichment) were pre-screened for fluorescein staining on Day 0.
  • fluorescein staining freshly made fluorescein diluted in WFI H 2 O at 10 mg/mL was administered at 0.4 ⁇ L to each eye. Approximately 8-13 minutes after administration, eyes were scored using an Olympus fluorescent dissecting microscope.
  • Punctuate staining was recorded using the standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas into which the corneal surface has been divided (score range 0-15/eye). Using a teaching bridge, two masked scorers evaluated mice at the same time to give a single collective score for each eye.
  • NKI National Eye Institute
  • mice with scores ⁇ 7 for each eye were placed in a dry eye chamber (20% ⁇ 2% humidity and constant air flow ⁇ 21 L/min/cage) on day 1 and were maintained in this chamber during the course of the experiment (except for examination).
  • mice were scored again and randomized into treatment groups with 8 to 10 mice/group. Mice were randomized such that each cage of 4 to 5 mice had approximately the same mean disease score.
  • mice were topically administered P05 or vehicle (1.25 ⁇ PBS) in an eye drop at 3 ⁇ L/eye BID. Mice were examined and scored on days 7, 9, and 11 for corneal fluorescein staining as described above. Scorers were blinded as to the treatment groups during the course of the experiment.
  • FIG. 6A is a bar graph of mean corneal staining score ⁇ SEM at day 0, 3, 7, 9, and 11 for mice from two identical experiments under the following bid treatments: no treatment, vehicle (1.25 ⁇ PBS), and 10 mg/ml (1%) P05. 10 mg/ml P05 significantly reduced corneal staining on days 7, 9, and 11 of the experiment. Efficacy as evaluated by a reduction in corneal staining was also observed with doses as low as 0.1 mg/ml P05. Recombinant IL-1Ra produced in E. coli also moderately reduced corneal staining in the animal model.
  • the effect of 10 mg/ml P05 was specific based on a comparison to 10 mg/ml murine serum albumin in the same vehicle. No effect was seen with 10 mg/ml murine serum albumin (MSA) relative to vehicle, and the effect of 10 mg/ml P05 was statistically significant relative to 10 mg/ml murine serum albumin.
  • 10 mg/ml P05 was also compared to 0.05% cyclosporine in an ophthalmic emulsion (Restasis®). Whereas P05 reduced corneal staining, no effect was observed for the 0.05% cyclosporine ophthalmic emulsion after about 1 week of b.i.d. dosing.
  • P05 at 1 mg/ml and at 50 mg/ml was prepared in solutions of either (i) PBS, 0.5% w/v CMC, pH 7.4 or (ii) 10 mM sodium citrate, pH 6.0 containing various surfactants. Agitation was performed by vortexing the protein at room temperature for four hours. The samples were analyzed by micro-fluid imaging (MGI), SEC, A 280 , and visual inspection.
  • MMI micro-fluid imaging
  • 0.1% w/v poloxamer 188 compared with other surfactants (including, e.g., polysorbate 20, polysorbate 80, or no surfactant) protected the protein from precipitation (assessed using visual inspection) and significant subvisible particle accumulation during agitation.
  • the 0.1% w/v poloxamer reduced particle counts for particles ⁇ 10 microns and for particles ⁇ 25 microns.
  • poloxamer 188 is a suitable surfactant for formulating a polypeptide such as P05. Furthermore, it demonstrates that as little as 0.1% w/v surfactant can be effective for limiting and even decreasing the amount of precipitation. These experiments also demonstrate a method of determining the suitability of a formulation described herein.
  • P05 was provided as a frozen liquid containing 52.8 mg/mL P05 in 1 ⁇ PBS, pH 6.5.
  • the polypeptide was dialyzed against a buffer containing 10 mM sodium citrate and 5% w/v sorbitol at pH 6.0 using 3500 molecular weight cutoff dialysis cassettes in an approximately 10,000-fold exchange over about 24 hours at 2-8° C. Following dialysis, the concentration was determined by measuring A280/A320.
  • formulations were prepared with various concentrations of P05 as follows, 100 ⁇ poloxamer 188 surfactant was added to a 1 ⁇ concentration to a stock solution of dialyzed P05.
  • the protein concentrations were adjusted to approximately 1 mg/mL, 5 mg/mL and 20 mg/mL by adding formulation buffer (10 mM Na citrate, 5% w/v sorbitol, pH 6.0).
  • formulation buffer (10 mM Na citrate, 5% w/v sorbitol, pH 6.0).
  • the final concentration of the formulation components was about 10 mM sodium citrate, 5% w/v sorbitol, and 0.1% w/v poloxamer 188.
  • Samples were then mixed and sterile filtered under aseptic conditions then were filled (at 250 ⁇ L) into 2 cc glass vials under aseptic conditions. After preparation, samples were placed in stability studies to confirm stability of the formulation prepared under the foregoing conditions.
  • Dynamic light scattering or DLS measures the diffusion of an analyte (e.g., P05) in a well plate by focusing laser light on the sample, and monitoring the rate of fluctuation of the scattered light as measured by a fast photon counter.
  • a mathematical technique known as a correlation function, is used to quantify the rate of fluctuation to determine the diffusion coefficient.
  • the diffusion coefficient is used to obtain radius of hydration (Rh) by the Stokes-Einstein equation.
  • the radius of P05 was measured as a function of increasing temperature in a DLS plate reader (Wyatt DynaProTM, Wyatt Technologies, Santa Barbara, Calif.). The acquisition time was 5 seconds and 5 scans were performed for each measurement. The ramp rate was 0.17° C./min. As the protein unfolded, the radius increased. The temperature at which the radius increased is referred as T on (temperature of onset for unfolding).
  • the reference standard for the 1 mg/ml and 5 mg/ml P05 formulations had an assayed purity of 99.1% and the purity of 1 mg/ml and 5 mg/ml samples of the P05 formulation were 99.1%-99.2%, respectively, regardless of whether they were stored at 25° C. or 40° C.
  • the reference had a purity of 99.2%.
  • the 20 mg/ml samples had a purity of 99.2%, regardless of whether they were stored at 25° C. or 40° C.
  • the 20 mg/ml P05 formulation had a purity of 99.2%.
  • a weak cation exchange HPLC method was used to assess the formulations.
  • a Dionex ProPac® WCX-10, 4 ⁇ 250 mm (PN 054993) column was used.
  • Mobile phase A was 10 mM Na acetate, pH 5.5 and mobile phase B was mobile A+0.25 M NaCl.
  • the assay was performed using an Agilent 1100 HPLC system with a flow rate of 1.2 mL/minute and a total run time of 35 minutes at ambient temperature. Detection was performed by assaying at 214 nm and 280 nm. Sample size was 25 ⁇ g/injection.
  • Table 6 A summary of the results after storage for 2 weeks at 25° C. is shown in Table 6, after storage for 2 weeks at 40° C. in Table 7, and after storage for 4 weeks at 25° C. in Table 8.
  • the weak cation exchange assay is another method of assessing purity by monitoring charge heterogeneity.
  • Analysis of P05 formulation samples resolves the main product peak from several product related impurities based on charge.
  • a typical preparation of P05 consists of approximately >85% main peak and several pre- and post-peaks.
  • Pre-peak 1 is unknown
  • Pre-peak 2 is a form of deamidated P05
  • Pre-peak 3 is a form of P05 with an N-terminal methionine.
  • Post-peak 1 is a form of P05 missing the N-terminal alanine
  • Post-peak-2 is a form of P05 missing both the N-terminal alanine and proline
  • Post-peak 3 is unknown.
  • Formulations were also assessed using RP-HPLC.
  • the RP-HPLC assay is another method of assessing purity by monitoring product heterogeneity based on hydrophobicity. The method is capable of separating the native molecule from product related impurities that contain oxidized methionines. Pre-peaks 2 and 3 are oxidized forms of the P05, and post-peaks 1 and 2 are acetylated forms of the molecule.
  • a Waters Symmetry® C4 (4.6 ⁇ 150 mm; 3.5 ⁇ m; PN 186000283) was used with a mobile phase A of 0.05% trifluoroacetic acid (TFA) in water and mobile phase B was 0.05% TFA in 95% acetonitrile (ACN).
  • Assays were run using an Agilent 1200 HPLC system with a flow rate of 1 mL/minute for a total run time of 35 minutes and a column temperature of 55° C. Detection was performed at 280 nm. The amount of sample loaded for 1 mg/ml and 5 mg/ml samples was 25 ⁇ g and the amount of sample loaded was 50 ⁇ g. A summary of the results after storage for 2 weeks at 25° C.
  • the invention relates to a formulation stored for at least 2 weeks, e.g., at least 4 weeks at 25° C., e.g., 40° C. at a concentration of at least 1 mg/ml, at least 5 mg/ml, or at least 20 mg/ml and has a purity of at least 92%, e.g., at least 94%, or at least 96%.
  • BFS blow fill seal
  • the stability of the formulation may suffer during long term storage (e.g., due to evaporation of water from the container and/or protein oxidation).
  • Sealing such containers in an aluminum foil pouch or other suitable package may protect the formulation from light-induced degradation.
  • Sealing the containers in such aluminum foil pouches with an inert gas, e.g., nitrogen or argon can protect against oxidation. Accordingly, experiments were conducted to investigate the effects of packaging and subsequent storage of P05 in blow fill seal (BFS) containers, with or without aluminum foil pouches with an inert gas overlay.
  • BFS blow fill seal
  • the formulation was cooled to about 2° C.-8° C. and filled into containers. Approximately 1000 containers were filled and the target fill volume of the containers was 0.32 mL. A portion of the containers were pouched in foil packages with a nitrogen overlay. An initial characterization was performed following the packaging into BFS containers and further stability evaluations were conducted following storage at two temperatures (2° C. to 8° C. and 25° C.), with or without pouching.
  • the initial characterization analysis included: concentration by A 280 , SDS-PAGE, SEC-HPLC, wCEX-HPLC, RP-HPLC, osmolality and particle analysis by light obscuration.
  • concentration by A 280 SDS-PAGE
  • SEC-HPLC SEC-HPLC
  • wCEX-HPLC wCEX-HPLC
  • RP-HPLC osmolality and particle analysis by light obscuration.
  • the stability of P05 was monitored monthly by SEC-HPLC, wCEX-HPLC and RP-HPLC, with A 280 evaluation performed at months 4 and 5, and pH and osmolality at month 5.
  • the subvisible particle counts were within the USP specifications for topical ophthalmics (having less than or equal to 50 particles per ml for particles ⁇ 10 ⁇ m and less than or equal to 5 particles per ml for particles ⁇ 25 ⁇ m), consistent with the lack of visible precipitation in the BFS containers (data not shown).
  • osmolality and concentration by A 280 were measured.
  • the average osmolality measurement was 317 mOsm, and the average concentration by A 280 was 5.0 mg/mL.
  • the P05 formulation in BFS containers was stored in an incubator at 25° C. with 60% relative humidity or at 2 to 8° C.
  • samples were analyzed by SEC, RP-HPLC, and wCEX-HPLC.
  • concentration was measured by A 280 .
  • osmolality to assess evaporation
  • pH and concentration by A 280 were also measured.
  • EBI-005 exhibited excellent physical stability after prolonged storage in blow fill seal vials at at 2 to 8° C. and at ambient temperature (RT).
  • the invention relates to a formulation as described herein containing methionine.
  • antioxidant for P05 formulated in 10 mM sodium citrate, 5% w/v sorbitol, 0.1% w/v poloxamer 188, pH 6.0 was studied using two different stress conditions: temperature (storage at 40° C.) and forced oxidation using hydrogen peroxide.
  • Hydrogen peroxide is a stressor that causes oxidation by free radicals and thus was used to emulate the oxidation effect that may occur after storage in multidose containers that have been gamma irradiated (see below).
  • P05 exhibits increases in oxidation levels when stored at high temperatures for prolonged periods.
  • the use of either 10 mM methionine or 7 mM bisulfate added to the formulation was tested for the P05 formulation stored 3 weeks at 40° C. Additionally, the same formulation was tested using forced oxidation, where 10% v/v of 0.02% hydrogen peroxide was added to the samples. The samples were tested by RP-HPLC to assess the levels of oxidation. Table 22 provides a data summary.
  • EBI-005 also referred to herein as P05
  • the formulation employed in this study contained EBI-005 (also referred to herein as P05) at a concentration of either 20 mg/ml or 5 mg/ml (see below), sodium carboxymethyl cellulose in a concentration of 0.25% w/v; poloxamer 188 in a concentration of 0.1% w/v; sorbitol in a concentration of 5% w/v; and sodium phosphate in a concentration of 10 mM.
  • the trial was conducted in 74 patients at eight centers in the United States. The trial was conducted in a natural environment (a controlled adverse environment chamber was not used).
  • CFS Corneal Fluorescein Staining
  • OSDI Ocular Surface Disease Index
  • Eligible subjects were at least 18 years of age, with moderate to severe dry eye disease. Additional eligibility criteria included the following: (i) OSDI score greater than or equal to 23 and less than 90 at the time of screening; (ii) OSDI score greater than or equal to 19 at randomization; (iii) CFS score greater than or equal to six and less than 15 on the NEI scale at the time of screening; and (iv) CFS score greater than or equal to five at randomization.
  • Treatments for the three groups in this trial were as follows: (i) in the first group, 22 patients received topical administration in each eye three times per day of EBI-005 formulation containing EBI-005 at a concentration of 20 mg/ml, (ii) in the second group, 22 patients received topical administration in each eye three times per day of EBI-005 formulation, containing EBI-005 at a concentration of 5 mg/ml, (iii) in the third group, 30 patients received topical administration in each eye three times per day of vehicle formulation (vehicle formulation was an aqueous formulation containing the same components as the EBI-005 formulation, except that EBI-005 was not in the vehicle formulation).
  • results are shown in FIGS. 8 to 10 . These results show that signs and symptoms of dry eye disease, as assessed using OSDI score ( FIG. 8 ), pain ( FIG. 9 ), and corneal fluorescein staining (CFS) score ( FIG. 10 ), improved during the course of treatment with the EBI-005 formulations. Surprisingly, treatment with the vehicle only formulation also resulted in notable improvements in OSDI score, pain, and CFS score.
  • a vehicle formulation (10 mM sodium citrate, pH 6.0, 5% sorbitol (w/v), and 0.1% poloxamer 188 (w/v)
  • a therapeutic formulation (10 mM sodium citrate, pH 6.0, 5% sorbitol (w/v), and 0.1% poloxamer 188 (w/v) containing 5 mg/ml P05
  • Subjects are assessed for DED and inclusion criteria include having a history of dry eye disease (DED) in both eyes supported by a previous clinical diagnosis or have a self-reported history of subjective complaints for at least 6 months prior to screening (Visit 1), have ongoing DED, in the same eye or both eyes, as defined by the following criteria at Visit 1: an OSDI score of ⁇ 23 and ⁇ 75 and have scored the painful or sore eye question of the OSDI and a Total Corneal Fluorescein Staining Score of ⁇ 6 (NEI scale) and ⁇ 15. Screened subjects then undergo a five to eight day treatment with masked vehicle formulation, and are then rescreened (Visit 2) to confirm they meet the randomization criteria at this visit.
  • DED dry eye disease
  • Visit 1 an OSDI score of ⁇ 23 and ⁇ 75 and have scored the painful or sore eye question of the OSDI and a Total Corneal Fluorescein Staining Score of ⁇ 6 (NEI scale) and ⁇ 15.
  • the randomization criteria include having a total OSDI score of ⁇ 19 and ⁇ 50, having a total corneal fluorescein staining score of ⁇ 5 (NEI scale) in the same qualifying eye as in Visit 1 and CFS ⁇ 15 in at least one eye, and having complied with the five to eight day masked vehicle formulation period. Compliance is defined as administering at least 80% of the doses.
  • Subjects are then randomized to treatment with vehicle formulation or therapeutic formulation and are provided with enough vehicle formulation or therapeutic formulation to administer one drop in each eye 3 times daily through visit 4 (week 3/note that the week numbering starts after subjects are randomized and assigned to receive vehicle or therapeutic formulation). Additional drug is dispensed at each subsequent visit through Visit 6 (week 9). Subjects are evaluated at Visit 3 (week 1), Visit 4 (week 3), Visit 5 (week 6), Visit 6 (week 9), Visit 7 (week 12) and Visit 8 (week 15). The last dose of study drug and final treatment visit are completed at Visit 7 (Week 12). The final evaluation is three weeks later at Visit 8 (follow-up, Week 15).
  • Formulations are provided as a 2 to 8° C. solution in a low-density polyethylene (LDPE) blow fill unit. Subjects are provided with ReFresh Plus® tears to be used if required, over the 21 day period preceding the final evaluation. Subjects do not use any such artificial tears during other parts of the study. Additional study information is available at clinicaltrials.gov, trial no. NCT01998802.
  • LDPE low-density polyethylene
  • Evaluation of subjects during the study includes Total Corneal Fluorescein Staining (a sign), painful or sore eye question of the OSDI questionnaire (a symptom), total OSDI and individual questions and domains of the OSDI, inferior and central region CFS, global assessment (investigator and subject), subject-rated severity of individual symptoms of dry eye, and Schirmer test without anesthesia. Improvements in at least one of these criteria at the end of the first week (i.e., after the three to eight day masked vehicle run-in) and/or during subsequent study visits for those subjects assigned to the vehicle formulation group compared to their initial evaluation, further demonstrate the efficacy of a vehicle formulation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Ophthalmology & Optometry (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Inorganic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
US14/209,605 2013-03-13 2014-03-13 Chimeric cytokine formulations for ocular delivery Abandoned US20140308239A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/209,605 US20140308239A1 (en) 2013-03-13 2014-03-13 Chimeric cytokine formulations for ocular delivery
US15/670,840 US10799589B2 (en) 2013-03-13 2017-08-07 Chimeric cytokine formulations for ocular delivery

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361779974P 2013-03-13 2013-03-13
US14/209,605 US20140308239A1 (en) 2013-03-13 2014-03-13 Chimeric cytokine formulations for ocular delivery

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/670,840 Continuation US10799589B2 (en) 2013-03-13 2017-08-07 Chimeric cytokine formulations for ocular delivery

Publications (1)

Publication Number Publication Date
US20140308239A1 true US20140308239A1 (en) 2014-10-16

Family

ID=50680140

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/209,605 Abandoned US20140308239A1 (en) 2013-03-13 2014-03-13 Chimeric cytokine formulations for ocular delivery
US15/670,840 Active US10799589B2 (en) 2013-03-13 2017-08-07 Chimeric cytokine formulations for ocular delivery

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/670,840 Active US10799589B2 (en) 2013-03-13 2017-08-07 Chimeric cytokine formulations for ocular delivery

Country Status (17)

Country Link
US (2) US20140308239A1 (zh)
EP (1) EP2968468B1 (zh)
JP (1) JP6450364B2 (zh)
KR (1) KR102236926B1 (zh)
CN (2) CN107693781B (zh)
AU (2) AU2014243839B2 (zh)
BR (1) BR112015022587A2 (zh)
CA (1) CA2903706C (zh)
DK (1) DK2968468T3 (zh)
ES (1) ES2884813T3 (zh)
HK (1) HK1246692A1 (zh)
IL (1) IL241171A0 (zh)
MX (1) MX2015012404A (zh)
PL (1) PL2968468T3 (zh)
RU (1) RU2015143521A (zh)
SG (2) SG11201507447PA (zh)
WO (1) WO2014160371A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015138844A1 (en) 2014-03-13 2015-09-17 Eleven Biotherapeutics, Inc. Methods for production of stable chimeric cytokine protein formulations in blow fill seal containers
WO2017007957A1 (en) * 2015-07-07 2017-01-12 Mast Therapeutics, Inc. Reduced sodium poloxamer-188 formulations and methods for use
WO2022226177A1 (en) * 2021-04-22 2022-10-27 Altavant Sciences Gmbh Compositions of interleukin-1 receptor antagonist
US20230201387A1 (en) * 2020-05-21 2023-06-29 Coherus Biosciences, Inc. Systems and methods for producing sterile injection devices

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2598526T3 (en) 2010-07-29 2018-11-19 Eleven Biotherapeutics Inc CHEMICAL IL-1 RECEPTOR TYPE-I AGONISTS AND ANTAGONISTS
WO2014160371A1 (en) 2013-03-13 2014-10-02 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery
ES2584534B1 (es) * 2015-03-27 2017-03-13 Retinset, S.L. Formulación tópica oftálmica de bosentan
US11324807B2 (en) * 2015-04-28 2022-05-10 Swedish Orphan Biovitrum Ab (Publ) Compositions comprising anakinra
WO2017094506A1 (ja) * 2015-11-30 2017-06-08 ロート製薬株式会社 眼科組成物
CN115089547A (zh) * 2016-10-12 2022-09-23 Ps治疗有限公司 人工泪液、隐形眼镜和药物载体组合物及其使用方法
WO2018142514A1 (ja) * 2017-02-01 2018-08-09 協和発酵キリン株式会社 ダルベポエチンを含む液体医薬組成物
WO2023169686A1 (en) 2022-03-10 2023-09-14 Buzzard Pharmaceuticals AB Inhibitor of interleukin-1 receptor type 1 for use in the treatment of cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030104996A1 (en) * 2001-08-30 2003-06-05 Tiansheng Li L-methionine as a stabilizer for NESP/EPO in HSA-free formulations

Family Cites Families (131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL188266C (nl) 1975-07-29 1992-05-18 Merck & Co Inc Werkwijze ter bereiding van een oogheelkundig inplantaat.
ZA811368B (en) 1980-03-24 1982-04-28 Genentech Inc Bacterial polypedtide expression employing tryptophan promoter-operator
US5693489A (en) 1984-03-30 1997-12-02 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US5998578A (en) 1984-05-18 1999-12-07 New England Medical Center Hospitals, Inc. Biologically active fragments of IL-1β
EP0569687B1 (en) 1984-05-18 2002-08-21 New England Medical Center Hospitals, Inc. Human IL-1 cDNA sequences encoding biologically-active human IL-1 proteins
US5484887A (en) 1984-06-19 1996-01-16 Immunex Corporation Homogeneous interleukin 1
DK172052B1 (da) 1984-12-21 1997-09-29 Otsuka Pharma Co Ltd Antitumor-aktivt stof, fremgangsmåde til dets fremstilling, lægemiddel indeholdende stoffet, gen kodende for stoffet, vektor indeholdende genet samt rekombinant mikroorganisme transformet med en sådan vektor
US6107465A (en) 1984-12-21 2000-08-22 Otsuka Pharmaceutical Co., Ltd. IL-1β and derivatives thereof and drugs
US5122459A (en) 1984-12-31 1992-06-16 Immunex Corporation Gene encoding biologically active human interleukin 1
US4770781A (en) 1986-03-03 1988-09-13 Merck & Co., Inc. Purification of human interleukin-1 species
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4883658A (en) 1986-04-28 1989-11-28 Holly Frank J Ophthalmic solution for treatment of dry-eye syndrome
US4804539A (en) 1986-07-28 1989-02-14 Liposome Technology, Inc. Ophthalmic liposomes
US5278151A (en) 1987-04-02 1994-01-11 Ocular Research Of Boston, Inc. Dry eye treatment solution
KR0148009B1 (ko) 1988-05-27 1998-08-01 그래고리 비. 아보트 인터루킨-1 억제제
US6858409B1 (en) 1988-05-27 2005-02-22 Amgen Inc. Nucleic acids encoding interleukin-1 inhibitors and processes for preparing interleukin-1 inhibitors
US6159460A (en) 1988-05-27 2000-12-12 Amgen Inc. Method for treating interleukin-1 mediated diseases
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
AU4005289A (en) 1988-08-25 1990-03-01 Smithkline Beecham Corporation Recombinant saccharomyces
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
EP0460052B1 (en) 1989-02-27 1998-04-15 New England Medical Center Il-1 biological activity inhibitors
US5075104A (en) 1989-03-31 1991-12-24 Alcon Laboratories, Inc. Ophthalmic carboxy vinyl polymer gel for dry eye syndrome
CA2068320C (en) 1989-11-29 2000-10-17 Robert Hageman Production of recombinant human interleukin-1 inhibitor
WO1991017184A1 (en) 1990-04-27 1991-11-14 The Upjohn Company Modified interleukin-1 inhibitors
ZA912797B (en) 1990-05-29 1992-12-30 Boston Ocular Res Dry eye treatment process and solution
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
AU1270592A (en) 1991-02-05 1992-09-07 University Of Maryland Modified interleukin-1beta
CA2103059C (en) 1991-06-14 2005-03-22 Paul J. Carter Method for making humanized antibodies
JP3122139B2 (ja) 1991-10-15 2001-01-09 マラーキー,ミッシェル,エフ. 後期段階炎症反応の治療用組成物
ES2173871T3 (es) 1991-10-31 2002-11-01 Univ Manchester Tratamiento de condiciones neurologicas por un compuesto inhibidor de la interleukina-1.
PL175705B1 (pl) 1992-09-17 1999-01-29 Synergen Inc Kompozycja farmaceutyczna
DE69419721T2 (de) 1993-01-12 2000-04-27 Biogen Inc Rekombinante anti-vla4 antikörpermoleküle
US5340572A (en) 1993-02-08 1994-08-23 Insite Vision Incorporated Alkaline ophthalmic suspensions
WO1995010298A1 (en) 1993-10-12 1995-04-20 Mary Lake Polan Method of contraception
US5861476A (en) 1994-02-02 1999-01-19 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
IT1269989B (it) 1994-09-21 1997-04-16 Dompe Spa Antagonisti recettoriali di il-1 ad aumentata attivita' inibitoria
JP3269770B2 (ja) 1995-03-02 2002-04-02 参天製薬株式会社 再分散性の優れたイオン性高分子配合水性懸濁点眼剤
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc HUMAN ANTIBODIES DERIVED FROM IMMUNIZED XENO MOUSES
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
WO1998022130A1 (en) 1996-11-19 1998-05-28 The Schepens Eye Research Institute, Inc. Local use of il-1ra in corneal transplant rejection or disorders of the eye
EP0942968B1 (en) 1996-12-03 2008-02-27 Amgen Fremont Inc. Fully human antibodies that bind EGFR
ES2312179T3 (es) 1996-12-06 2009-02-16 Amgen Inc. Terapia combinada que utiliza un inhibidor del il-1 para el tratamiento de enfermedades mediadas por el il-1.
JP2002501496A (ja) 1997-04-21 2002-01-15 シェーリング コーポレイション 哺乳動物サイトカイン;関連の試薬および方法
UA72189C2 (uk) 1997-11-17 2005-02-15 Янссен Фармацевтика Н.В. Фармацевтична композиція, що містить водну суспензію субмікронних ефірів 9-гідроксирисперидон жирних кислот
US6337072B1 (en) 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US6927044B2 (en) 1998-09-25 2005-08-09 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps
US7906481B2 (en) 1998-09-25 2011-03-15 Sciaticon Ab Specific TNF-A inhibitors for treating spinal disorders mediated by nucleous pulposus
MXPA01007170A (es) 1999-01-15 2002-07-30 Genentech Inc Variantes de polipeptidos con funcion efectora alterada.
US6419944B2 (en) 1999-02-24 2002-07-16 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
US6471961B1 (en) 1999-02-24 2002-10-29 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
JP3363832B2 (ja) 1999-05-13 2003-01-08 アルプス電気株式会社 薄膜構造体およびその製造方法
MXPA02005729A (es) 1999-12-10 2002-09-18 Amgen Inc Moleculas tipo antagonista del receptor de inteleucina-1 y usos de las mismas.
AU778676B2 (en) 1999-12-10 2004-12-16 Amgen, Inc. Interleukin-1 receptor antagonist-like molecules and uses thereof
CA2395417A1 (en) 1999-12-10 2001-06-14 Amgen Inc. Interleukin-1 receptor antagonist-related molecules and uses thereof
US20030007971A1 (en) 2000-01-31 2003-01-09 Hideaki Hara Remedies for ophthalmic diseases
AU2001236632A1 (en) 2000-02-03 2001-08-14 Regeneration Technologies, Inc. Extraction of growth factors from tissue
US20040028872A1 (en) 2000-04-17 2004-02-12 Edwards Mark Stephen Method and apparatus for making bristle subassemblies
US6623736B2 (en) 2000-05-02 2003-09-23 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
EP2241328A1 (en) 2000-05-12 2010-10-20 Immunex Corporation Interleukin-1 inhibitors in the treatment of diseases
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
US20030166069A1 (en) 2000-11-28 2003-09-04 Amgen, Inc. Interleukin-1 receptor antagonist-like molecules and uses thereof
EP2357187A1 (en) 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
JP2004519230A (ja) 2001-02-06 2004-07-02 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング 低減された免疫原性を有する修飾されたインターロイキン−1受容体アンタゴニスト(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)
AU2002332931A1 (en) 2001-09-06 2003-03-24 Board Of Regents, The University Of Texas System Therapeutic treatments for spinal cord injury via blockade of interleukin-1 receptor
FR2833268B1 (fr) * 2001-12-12 2005-07-08 Fabre Pierre Dermo Cosmetique Nouvelle association contenant un poloxamer et de l'acide chondroitine sulfurique et/ou une glycoproteine et son utilisation
ATE509032T1 (de) 2002-02-13 2011-05-15 Ludwig Inst For Cancer Res Ltd Chimerizierte gm-csf antikörper
WO2003090255A2 (en) 2002-04-18 2003-10-30 Northwestern University Encapsulation of nanotubes via self-assembled nanostructures
US7029712B1 (en) 2002-07-17 2006-04-18 Biosyntrx Inc Treatment for dry eye syndrome
MXPA05007019A (es) 2002-12-30 2005-08-18 Amgen Inc Terapia de combinacion con factores co-estimuladores.
US20050033694A1 (en) 2003-06-03 2005-02-10 United States Postal Service. System and method for fleet card management
KR100610003B1 (ko) * 2003-06-10 2006-08-08 주식회사 엘지생명과학 혈청 알부민을 함유하지 않는 안정한 인 에리쓰로포이에틴용액 제형
US7261882B2 (en) 2003-06-23 2007-08-28 Reagents Of The University Of Colorado Methods for treating neuropathic pain by administering IL-10 polypeptides
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
ES2418830T3 (es) 2003-10-27 2013-08-16 Wyeth Llc Retirada de agregados de alto peso molecular usando cromatografía de hidroxiapatita
KR100543466B1 (ko) 2003-11-13 2006-01-20 삼성전자주식회사 영상 보간 장치 및 방법
CA2551510C (en) * 2003-12-23 2013-07-30 Pharmacia Corporation Stable growth hormone liquid formulation
US7482323B2 (en) 2004-03-04 2009-01-27 The University Of Tennessee Research Foundation Intracellular interleukin-1 receptor antagonist and uses thereof
US7674464B2 (en) 2004-03-04 2010-03-09 The University Of Tennessee Research Foundation Intracellular interleukin-1 receptor antagonists
US10765747B2 (en) * 2004-04-02 2020-09-08 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
JP5022216B2 (ja) 2004-06-04 2012-09-12 リジェネロン・ファーマシューティカルズ・インコーポレイテッド 自己炎症性疾患を処置するためのil−1アンタゴニストを使用する方法
CA2576519C (en) 2004-08-17 2013-05-21 Regeneron Pharmaceuticals, Inc. Il-1 antagonist formulations
US7700318B2 (en) 2004-08-25 2010-04-20 Amprotein Corporation Chimeric polypeptide and use thereof
CN101132800A (zh) 2004-11-24 2008-02-27 席拉坎有限责任公司 一种用于眼内药物释出的植入体
JP2008521426A (ja) 2004-12-02 2008-06-26 ドマンティス リミテッド ドメイン抗体への複合体化により血清半減期が延長されたpladドメインペプチド
ES2390354T3 (es) 2005-07-22 2012-11-12 Amgen, Inc Liofilizados de proteínas concentrados, procedimientos y usos
WO2007027761A2 (en) 2005-09-01 2007-03-08 Schering Corporation Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for the diagnosis of preeclampsia
EP1948220A2 (en) 2005-10-21 2008-07-30 Amgen Inc. Methods of decreasing vascular calcification using il-1 inhibitors
US20100047204A1 (en) 2006-04-14 2010-02-25 Dana Sue Yoo Use of organic compounds
US20080026485A1 (en) 2006-04-18 2008-01-31 Wolfgang Hueber Antibody profiling for determination of patient responsiveness
BRPI0621761A2 (pt) 2006-06-12 2011-12-20 Therakine Ltd solução oftálmica e uso de uma solução salina, e uma quantidade eficaz de pelo menos um agente terapêutico
US20080095754A1 (en) 2006-10-18 2008-04-24 Burke Susan E Ophthalmic compositions comprising diglycine
US8399421B2 (en) 2007-03-30 2013-03-19 The Board Of Regents Of The University Of Texas System Treatment for neuropathic pain due to spinal cord injury
GB0708376D0 (en) 2007-05-01 2007-06-06 Alligator Bioscience Ab Novel polypeptides and uses thereof
EP2468301A1 (en) 2007-05-29 2012-06-27 Novartis AG New indications for anti-IL-1-beta therapy
DE602008005596D1 (de) 2007-06-21 2011-04-28 Univ Muenchen Tech Biologisch aktive proteine mit erhöhter in-vivo- und/oder in-vitro-stabilität
KR101430584B1 (ko) 2007-07-05 2014-08-14 삼성전자주식회사 이동 통신 시스템에서 심 어플리케이션 툴킷을 제공하는장치 및 방법
US8933197B2 (en) 2007-08-15 2015-01-13 Amunix Operating Inc. Compositions comprising modified biologically active polypeptides
WO2009025763A2 (en) 2007-08-16 2009-02-26 Schepens Eye Research Institute Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
CN101889022A (zh) 2007-10-08 2010-11-17 阿纳福公司 三聚IL-1Ra
WO2009080699A2 (en) * 2007-12-20 2009-07-02 Merck Serono S.A. Peg-interferon-beta formulations
CA2711696C (en) 2008-01-09 2021-10-26 Reza Dana Therapeutic compositions for treatment of ocular inflammatory disorders
EP2249809A1 (en) * 2008-01-15 2010-11-17 Abbott GmbH & Co. KG Powdered protein compositions and methods of making same
KR20110076918A (ko) * 2008-09-10 2011-07-06 제넨테크, 인크. 단백질의 산화성 분해를 방지하기 위한 조성물 및 방법
US8298533B2 (en) 2008-11-07 2012-10-30 Medimmune Limited Antibodies to IL-1R1
EP2350649A4 (en) 2008-11-28 2012-11-14 Abbott Lab STABLE ANTIBODY COMPOSITIONS AND METHODS FOR STABILIZING THE SAME
US20120014970A1 (en) 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders
EP2385839B1 (en) 2009-01-09 2014-11-26 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of corneal disorders
BRPI1008602A8 (pt) 2009-02-18 2018-01-02 Eyeon Particle Sciences Llc uso de co-polímero bi-funcional para as aplicações oftálmicas e outras aplicações tópicas e locais
EP2248518B1 (en) 2009-04-17 2013-01-16 Merz Pharma GmbH & Co. KGaA Formulation for stabilizing proteins, peptides or mixtures thereof.
CA2726232A1 (en) 2009-07-16 2010-08-12 Arecor Limited Stable formulation of a therapeutic protein
US20120269765A1 (en) 2009-07-24 2012-10-25 Garcia K Christopher Cytokine compositions and methods of use thereof
WO2011028344A2 (en) 2009-08-25 2011-03-10 Amunix Operating Inc. Interleukin-1 receptor antagonist compositions and methods of making and using same
BR112012008444A2 (pt) 2009-10-10 2019-09-24 Eleven Biotherapeutics Inc proteína isolada, composição farmacêutica, métodos para modular uma resposta imune ou inflamatória em um sujeito, para tratar um distúrbio mediado po ir-17 em um sujeito e para preparar uma proteína recombinante, ácido nucleico isolado, e, célula hospedeira recombinante
WO2011063195A2 (en) 2009-11-20 2011-05-26 Dr. Reddy's Laboratories Ltd. Purification of modified cytokines
EP2538965B1 (en) 2010-02-25 2017-04-12 Schepens Eye Research Institute Therapeutic compositions for the treatment of dry eye disease
EP2552967A4 (en) 2010-04-02 2014-10-08 Amunix Operating Inc BINDING FUSION PROTEINS, BINDING FUSION PROTEIN MEDICINAL CONJUGATES, XTEN MEDICAMENT CONJUGATES, AND METHOD FOR THEIR PREPARATION AND USE
WO2011163452A2 (en) 2010-06-24 2011-12-29 Eleven Biotherapeutics, Inc. Treating surface of the eye disorders
DK2598526T3 (en) 2010-07-29 2018-11-19 Eleven Biotherapeutics Inc CHEMICAL IL-1 RECEPTOR TYPE-I AGONISTS AND ANTAGONISTS
EP2637684A4 (en) * 2010-11-08 2014-05-07 Healor Ltd BUFFERED OPHTHALMIC COMPOSITIONS AND USER METHOD THEREFOR
WO2012103240A2 (en) 2011-01-25 2012-08-02 Eleven Biotherapeutics, Inc. Receptor binding agents
WO2012122985A1 (en) 2011-03-14 2012-09-20 University Of Copenhagen Antagonists of the interleukin- 1 receptor
SG10201606218WA (en) * 2011-07-29 2016-09-29 Eleven Biotherapeutics Inc Purified Proteins
WO2014160371A1 (en) 2013-03-13 2014-10-02 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030104996A1 (en) * 2001-08-30 2003-06-05 Tiansheng Li L-methionine as a stabilizer for NESP/EPO in HSA-free formulations

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015138844A1 (en) 2014-03-13 2015-09-17 Eleven Biotherapeutics, Inc. Methods for production of stable chimeric cytokine protein formulations in blow fill seal containers
WO2017007957A1 (en) * 2015-07-07 2017-01-12 Mast Therapeutics, Inc. Reduced sodium poloxamer-188 formulations and methods for use
US20230201387A1 (en) * 2020-05-21 2023-06-29 Coherus Biosciences, Inc. Systems and methods for producing sterile injection devices
US11752225B2 (en) * 2020-05-21 2023-09-12 Coherus Biosciences, Inc. Systems and methods for producing sterile injection devices
WO2022226177A1 (en) * 2021-04-22 2022-10-27 Altavant Sciences Gmbh Compositions of interleukin-1 receptor antagonist

Also Published As

Publication number Publication date
US20180177877A1 (en) 2018-06-28
HK1246692A1 (zh) 2018-09-14
BR112015022587A2 (pt) 2017-10-24
RU2015143521A (ru) 2017-04-19
IL241171A0 (en) 2015-11-30
EP2968468B1 (en) 2021-07-14
CN107693781B (zh) 2021-11-09
AU2018282269A1 (en) 2019-01-17
CN107693781A (zh) 2018-02-16
RU2015143521A3 (zh) 2018-03-22
WO2014160371A1 (en) 2014-10-02
KR102236926B1 (ko) 2021-04-06
MX2015012404A (es) 2016-02-03
SG10201707477SA (en) 2017-10-30
CA2903706C (en) 2024-01-23
KR20150126688A (ko) 2015-11-12
EP2968468A1 (en) 2016-01-20
CN105392494A (zh) 2016-03-09
AU2014243839A1 (en) 2015-09-17
JP6450364B2 (ja) 2019-01-09
PL2968468T3 (pl) 2021-12-20
AU2014243839B2 (en) 2019-01-03
US10799589B2 (en) 2020-10-13
AU2018282269B2 (en) 2020-06-11
DK2968468T3 (en) 2021-07-26
JP2016521252A (ja) 2016-07-21
CA2903706A1 (en) 2014-10-02
SG11201507447PA (en) 2015-10-29
ES2884813T3 (es) 2021-12-13

Similar Documents

Publication Publication Date Title
US10799589B2 (en) Chimeric cytokine formulations for ocular delivery
US20230043641A1 (en) Ophthalmic composition for treatment of dry eye disease
US20220241414A1 (en) Il-6 antagonist formulations and uses thereof
US11285163B2 (en) Ophthalmic solution
JP2008543877A (ja) Lkktetおよび/またはlkktnt組成物および組織の悪化、傷害または損傷を処置または予防するための方法
SA521430043B1 (ar) صياغات من المركب 4-(7- هيدروكسي-2- أيزوبروبيل-4- أوكسو-4h- كوينازولين-3- يل) - بنزونيتريل
KR20190137788A (ko) 안정한 펩타이드 조성물
US20220017602A1 (en) Ocular compositions and methods
WO2024006653A2 (en) Stable peptide compositions and methods of use thereof for treatment of moderate/severe dry eye-related ocular symptoms
US20230233687A1 (en) Hyaluronic acid-based formulations for treatment and prevention of ocular hypertension and glaucoma

Legal Events

Date Code Title Description
AS Assignment

Owner name: INTEGRITY BIO, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHANG, BYEONG;REEL/FRAME:033847/0875

Effective date: 20140912

Owner name: ELEVEN BIOTHERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INTEGRITY BIO, INC.;REEL/FRAME:033847/0901

Effective date: 20140912

Owner name: ELEVEN BIOTHERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZARBIS-PAPASTOITSIS, GREGORY;LOWDEN, PATRICIA;REEL/FRAME:033847/0852

Effective date: 20140910

STCB Information on status: application discontinuation

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