US20200316158A1 - Collagen 7 compositions and methods of using the same - Google Patents

Collagen 7 compositions and methods of using the same Download PDF

Info

Publication number
US20200316158A1
US20200316158A1 US16/875,223 US202016875223A US2020316158A1 US 20200316158 A1 US20200316158 A1 US 20200316158A1 US 202016875223 A US202016875223 A US 202016875223A US 2020316158 A1 US2020316158 A1 US 2020316158A1
Authority
US
United States
Prior art keywords
composition
arginine
polysorbate
collagen
certain embodiments
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
US16/875,223
Inventor
Igor Quinones-Garcia
Lin Guey
Kris Lowe
Vinh Nguyen
Bing He
Amey Bandekar
Sujit Basu
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.)
Novatec Inc
Phoenix Tissue Repair Inc
Original Assignee
Novatec Inc
Phoenix Tissue Repair 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 Novatec Inc, Phoenix Tissue Repair Inc filed Critical Novatec Inc
Priority to US16/875,223 priority Critical patent/US20200316158A1/en
Assigned to PHOENIX TISSUE REPAIR, INC. reassignment PHOENIX TISSUE REPAIR, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHIRE HUMAN GENETIC THERAPIES, INC.
Assigned to SHIRE HUMAN GENETIC THERAPIES, INC. reassignment SHIRE HUMAN GENETIC THERAPIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: QUINONES-GARCIA, IGOR, BANDEKAR, Amey, BASU, SUJIT, GUEY, Lin, LOWE, KRIS, NGUYEN, VINH, HE, BING
Publication of US20200316158A1 publication Critical patent/US20200316158A1/en
Assigned to NOVATEC, INC. reassignment NOVATEC, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZINSKI, JAMES
Assigned to U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT reassignment U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADRENAS THERAPEUTICS, INC., BRIDGEBIO PHARMA, INC., Eidos Therapeutics, Inc., ORIGIN BIOSCIENCES, INC., PHOENIX TISSUE REPAIR, INC., QED THERAPEUTICS, INC.
Priority to US18/169,368 priority patent/US20230414698A1/en
Assigned to PHOENIX TISSUE REPAIR, INC., ORIGIN BIOSCIENCES, INC., QED THERAPEUTICS, INC., BRIDGEBIO PHARMA, INC., ADRENAS THERAPEUTICS, INC., Eidos Therapeutics, Inc. reassignment PHOENIX TISSUE REPAIR, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides 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
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/60Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/01Hydrolysed proteins; Derivatives thereof
    • A61K38/012Hydrolysed proteins; Derivatives thereof from animals
    • A61K38/014Hydrolysed proteins; Derivatives thereof from animals from connective tissue peptides, e.g. gelatin, collagen
    • 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
    • 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
    • A61K47/06Organic 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
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/19Cosmetics or similar toiletry preparations characterised by the composition containing inorganic ingredients
    • A61K8/20Halogens; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/19Cosmetics or similar toiletry preparations characterised by the composition containing inorganic ingredients
    • A61K8/24Phosphorous; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/40Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing nitrogen
    • A61K8/44Aminocarboxylic acids or derivatives thereof, e.g. aminocarboxylic acids containing sulfur; Salts; Esters or N-acylated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4993Derivatives containing from 2 to 10 oxyalkylene groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • A61K8/65Collagen; Gelatin; Keratin; Derivatives or degradation products thereof
    • 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • 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
    • C07K14/78Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]

Definitions

  • the present invention relates to compositions (e.g., pharmaceutical compositions) and more particularly, to collagen 7 compositions and methods of using the same to treat dystrophic epidermolysis bullosa.
  • the collagen VII (C7) protein is a structural protein in which C7 is both a structural and functional precursor for the formation of anchoring fibrils which enable the attachment of the epidermis and the dermis.
  • the C7 monomer assembles as a homotrimer of approximately 900 kDa, containing one NC-1 and one NC-2 binding region. The homotrimer is held together via an alpha helical coil. From a homotrimer, these C7 protein precursors align into an anti-parallel dimer with an increase in molecular weight to 1.8 mDa. Lateral assembly of anti-parallel dimers leads to the formation of the anchoring fibrils.
  • the rC7 protein demonstrates “stickiness,” and the propensity to precipitate at higher protein concentrations, particularly in PBS buffer.
  • EB Dystrophic Epidermolysis Bullosa
  • Dystrophic epidermolysis bullosa is a form of EB characterized by development of scarring with healing. (3) In patients with DEB, blistering can be triggered by even minor mechanical trauma owing to the extremely fragile nature of the skin. This leads to a chronic cycle of blistering, healing, and reblistering that causes patients to suffer from painful wounds and debilitating scarring of epithelial tissue. There is no cure for DEB and management focuses on supportive care.
  • Dystrophic EB is caused by mutations in the Col7a1 gene, which encodes the alpha chain of collagen VII (C7), a protein essential for the formation of the anchoring fibrils that anchor the basement membrane to the underlying dermis.
  • C7 alpha chain of collagen VII
  • the condition is inherited as either a dominant form (DDEB) or a recessive form (RDEB), with RDEB typically having a more severe phenotype.
  • the DEB mouse model (Col7a1 ⁇ / ⁇ ) has a targeted inactivation of the Col7a1 gene, resulting in severe blistering that mimics the histological and ultrastructural presentation of severe human RDEB.
  • (6) These mice have no detectable collagen VII at the dermal-epidermal basement membrane zone (BMZ), completely lack anchoring fibrils, and are born with extensive cutaneous blisters on their ventral surface and hemorrhagic blisters on their paws and neck, with mortality typically observed within the first week of life.
  • BMZ dermal-epidermal basement membrane zone
  • compositions comprising collagen 7 and methods of preparing such compositions.
  • the composition comprises recombinant collagen 7, sodium phosphate, sodium citrate, sodium chloride, arginine, a sugar, and a surfactant. It is possible to exclude one or more of the sodium phosphate, sodium citrate, sodium chloride, arginine, sugar, or surfactant from the composition.
  • the sugar is sucrose.
  • the surfactant is a polysorbate, such as polysorbate 20 or polysorbate 80.
  • the sodium phosphate is present in an amount ranging from 100 mM to 1 mM, 50 mM to 5 mM, 25 mM to 5 mM, 10 mM to 5 mM, 10 mM to 1 mM or about 10 mM.
  • the sodium citrate is present in an amount ranging from 100 mM to 1 mM, 50 mM to 1 mM, 25 mM to 1 mM, 10 mM to 1 mM, 5 mM to 1 mM or about 5 mM.
  • the sodium chloride is present in an amount ranging from 200 mM to 10 mM, 100 mM to 20 mM, 80 mM to 50 mM, or about 70 mM.
  • the arginine is present in an amount ranging from 200 mM to 5 mM, 150 mM to 50 mM, 100 mM to 75 mM, or about 100 mM.
  • the sugar is present in an amount ranging from 100 mM to 10 mM, 100 mM to 50 mM, 75 mM to 35 mM, 60 mM to 40 mM, or about 50 mM. In certain embodiments, the sucrose is present in an amount ranging from 100 mM to 1 mM, 100 mM to 50 mM, 75 mM to 35 mM, 60 mM to 40 mM, or about 50 mM.
  • the surfactant is present in an amount ranging from 1% to 0.01%, 0.5% to 0.05%, 0.025% to 0.05%, 0.1% to 0.05%, 0.05% to 0.01% or about 0.05% (w/v).
  • the polysorbate e.g., polysorbate 20
  • the polysorbate is present in an amount ranging from 1% to 0.01%, 0.5% to 0.05%, 0.025% to 0.05%, 0.1% to 0.05%, 0.05% to 0.01% or about 0.05% (w/v).
  • the composition has a pH of between 8.0 to 6.0, 7.5 to 6.5, 7.5 to 7.0, 7.4 to 7.0, 7.3 to 7.0, 7.2 to 7.0, 7.1 to 7.0, 7.2 to 7.1, 7.2 ⁇ 0.3, about 7.1, or about 7.2.
  • the composition comprises recombinant collagen 7, 100 mM to 10 mM sodium phosphate, 100 mM to 1 mM sodium citrate, 200 mM to 10 mM sodium chloride, 200 mM to 10 mM arginine, 100 mM to 10 mM sucrose, and 1.0% to 0.01% (w/v) polysorbate 20 and optionally a pH of 7.2 to 7.1.
  • the composition comprises recombinant collagen 7, 25 mM to 5 mM sodium phosphate, 25 mM to 1 mM sodium citrate, 100 mM to 20 mM sodium chloride, 150 mM to 50 mM arginine, 75 mM to 35 mM sucrose, and 0.5% to 0.05% polysorbate 20 (w/v) and optionally a pH of7.2 to 7.1.
  • the composition comprises recombinant collagen 7, about 10 mM sodium phosphate, about 5 mM sodium citrate, about 70 mM sodium chloride, about 100 mM arginine, about 50 mM sucrose, and about 0.05% polysorbate 20 (w/v) and has a pH of about 7.1.
  • the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.1.
  • the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.2 ⁇ 0.3.
  • the recombinant collagen 7 is a human collagen 7.
  • the collagen 7 compositions have increased stability.
  • the composition is stable for 3 months following storage at ⁇ 65 ⁇ 5° C. or ⁇ 20 ⁇ 5° C. in a 1 liter polycarbonate bottle (500 mL of the composition).
  • the composition is stable for 6 months following storage at ⁇ 65 ⁇ 5° C. or ⁇ 20 ⁇ 5° C. in a 10 cc glass vial (5.5 mL of composition).
  • the composition is stable following storage for 2 weeks at 25 ⁇ 3° C.
  • the composition is stable following storage for up to 1 month at 5 ⁇ 3° C.
  • compositions may be pharmaceutical compositions comprising one or more additional pharmaceutical excipients that can be used, for example, to treat DEB.
  • Another aspect is directed to a method of treating DEB, the method comprising administering to a subject in need thereof an effective amount of a recombinant collagen 7 composition, as described herein.
  • FIG. 1 shows representative immunofluorescent images in tongue and front leg from Col7a1 ⁇ / ⁇ mice surviving 5-7 days following administration of rC7 compositions or a control.
  • FIG. 2 shows representative H&E staining in tongue from Col7a1 ⁇ / ⁇ mice following administration of rC7 compositions or a control.
  • FIG. 3 is a Kaplan-Meier Curve showing the probability of survival of Col7a1 ⁇ / ⁇ mice following administration of rC7 compositions or a control.
  • FIG. 4 shows SEC-HPLC profiles of a rC7 formulation after exposure to freeze/thaw stress.
  • FIG. 5 shows SEC-HPLC for 0.5 mg/mL rC7 diluted in D-PBS and 10% serum after incubation for 1 hour at 37° C.
  • FIG. 6 shows % HMW species of rC7 formulations containing different amounts of arginine after storage at various temperatures.
  • FIG. 7 shows fibronectin binding of rC7 formulations containing different amounts of arginine after storage at various temperatures.
  • Anchoring fibrils are structural elements that serve as attachment complexes at the interface between the epithelial and mesenchymal layers of several tissues, including the skin, oral mucosa, and cervix (Chung et al. Dermatol Clin 28(1): 93-105 (2010)). In the skin, anchoring fibrils extend from the lower portion of the epidermal basement membrane to the underlying papillary dermis, securing the association between the epidermal basement membrane and the papillary dermis (Varki et al. J Med Genet 44:181-192 (2007)).
  • Collagen 7 produced by recombinant methods described herein may be recovered from the host cell culture system according to standard protocols known in the art (e.g., precipitation, centrifugation, etc.). Recombinant collagen 7 described herein may be secreted into the host cell medium and recovered by ammonium sulfate precipitation and subsequent centrifugation; as demonstrated in the following reference, (Chen et al. J Bio Chem 277(18): 2118-2124 (2002)).
  • Collagen 7 produced and recovered by recombinant and molecular biology methods described herein may be purified according to standard protocols known in the art (e.g., dialysis, ion exchange chromatography, affinity chromatography, SDS gel electrophoresis, etc.).
  • the recombinant collagen 7 described herein may be purified to homogeneity by ion exchange chromatography; as demonstrated in the following reference, (Chen et al. J. Bio. Chem. 277(18): 2118-2124 (2002)).
  • rC7 recombinant C7
  • a buffer containing a non-essential amino acid, a cryoprotectant, a surfactant, and a chelating agent.
  • the non-essential amino acid is arginine, preferably L-arginine.
  • the cryoprotectant is a sugar.
  • the buffer comprises a phosphate buffer, such as sodium phosphate.
  • the chelating agent is citrate.
  • the rC7 formulation comprises 10 mM sodium phosphate, 5 mM sodium citrate, 100 mM L-arginine, 50 mM sucrose, 70 mM NaCl, 0.05% (w/v) polysorbate-20, and pH of about 7.2.
  • the rC7 compositions described herein are stable for 3 months at both ⁇ 65 ⁇ 5° C. and ⁇ 20 ⁇ 5° C. In certain embodiments, the rC7 compositions described herein are stable for 6 months at both ⁇ 65 ⁇ 5° C. and ⁇ 20 ⁇ 5° C. In certain embodiments, the rC7 composition is stable for 2 weeks at 25 ⁇ 3° C. and up to 1 month at 5 ⁇ 3° C.
  • the rC7 composition described herein is stable for up to 8 hours at 25 ⁇ 3° C. when stored in disposable syringes or for up to 6 hours at 25 ⁇ 3° C. when diluted down to 0.1 mg/mL into saline.
  • “stable” means that after the indicated storage conditions, there is no significant difference in the amount of protein, as measured by absorbance at 280 nm, no significant difference in the amount of high molecular weight species, dimers, homotrimers, and low molecular weight species as measured by SEC-HPLC, and no significant loss in potency, as measured in a fibronectin binding assay.
  • Sugars that may be used in the compositions include, but are not limited, to non-reducing disaccharides (e.g., sucrose, trehalose) or a sugar alcohol, such as sorbitol.
  • the sugar in the composition is sucrose.
  • the composition may also include one or more buffers, such as a phosphate buffer (e.g., sodium phosphate, a histidine buffer, a Tris(hydroxymethyl)aminomethane buffer.
  • a phosphate buffer e.g., sodium phosphate, a histidine buffer, a Tris(hydroxymethyl)aminomethane buffer.
  • the composition may also include one or more surfactants and/or bulking agents.
  • Surfactants include but are not limited poloxamers (e.g., Pluronic®), polysorbates (e.g., polysorbate 80, polysorbate 20), CTAB (hexadecyltrimethylammonium bromide), or SDS (sodium dodecyl sulfate or sodium lauryl sulfate).
  • the surfactant in the composition is a polysorbate (e.g., polysorbate 20).
  • Bulking agents include but are not limited to PVP, PEG, dextran, mannitol and glycine.
  • composition may also include one more additional pharmaceutically acceptable excipients.
  • pharmaceutically acceptable excipients include, but are not limited to salts, proteins (e.g., albumin), chelating agents (e.g., citrate, EDTA), gelling polymers, urea (or its derivatives), and combinations thereof.
  • compositions described herein to treat DEB.
  • a method of treating DEB comprising administering to a subject in need thereof a composition comprising recombinant collagen 7, a buffer, anon-essential amino acid, a cryoprotectant, a surfactant, and a chelating agent.
  • the buffer is a phosphate buffer (e.g., sodium phosphate).
  • the non-essential amino acid is arginine.
  • cryoprotectant is a sugar, such as sucrose.
  • the surfactant is a polysorbate, such as polysorbate 20 or polysorbate 80.
  • the chelating agent is citrate.
  • the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.1 or 7.2.
  • L-arginine and sucrose and preferably 75-100 mM L-arginine and 1.7% (w/v) sucrose, improved solubility.
  • a surfactant e.g., 0.05% polysorbate-20
  • NaCl and preferably about 70 mM NaCl, in the formulation improved the appearance.
  • PBS rC7
  • Phase I formulation A formulation comprising 1 mg/mL rC7, 10 mM phosphate, 5 mM citrate, 100 mM L-arginine, 1.7% (w/v) sucrose, 70 mM NaCl, 0.05% (v/v) polysorbate-20 at pH of about 7.2 (“Phase I formulation”) was able to achieve approximately ten-fold higher protein concentration (upwards of about 4 mg/mL) using conventional spin columns with a 100 kDa PES molecular weight cutoff.
  • the Phase I formulation was also subjected to freezing and thawing stress conditions, using a robust, small scale freeze/thaw model.
  • the Phase I formulation was exposed to three freeze/thaw cycles (see Table 2) to reflect freeze/thaw stress that may be encountered at manufacturing scale (scalability).
  • the arginine-containing formulations were subjected to 2 months of storage at temperatures of ⁇ 6S° C., ⁇ 20° C., S ⁇ 3° C., and 2S° C. There were no changes in appearance after 2 months at any storage condition.
  • the same materials showed no significant change in protein concentration (A280 nm) or SEC-HPLC profile (see Table 4) with the exception of the formulations stored either at 5 ⁇ 3° C. (minimally) or 25° C. After 2 months storage at 25° C., an increase ( ⁇ 2%) in high molecular weight (HMW) species was seen.
  • the Phase I formulation at a 1 mg/mL dose was also assessed for compatibility with the animal delivery devices and dosing syringes.
  • the 1 mg/mL drug product was injected into the infusion set(s) with the appropriate syringe(s), and the drug held for up to 60 minutes in each of the devices. There were no visible particles or pH shifts after 60 minutes in the devices.
  • the SEC-HPLC profiles also demonstrated no significant changes.
  • rC7 is injected IV into the bloodstream. It is possible, however, to assess the potential for product quality changes by monitoring for any changes when the protein is exposed to more physiologically relevant conditions such as buffers containing 10% bovine serum and held at 37° C.
  • the rC7 protein is prone to soluble aggregation and precipitation. These events can be monitored through common analytical techniques.
  • the A280 absorbance is used to monitor for changes in protein content.
  • the A320 absorbance is used to monitor for changes in turbidity due to insoluble particulate formation.
  • SEC-HPLC is used to monitor for changes in the soluble protein where HMW and/or LMW species may form.
  • the buffers were diluted with equal parts serum and incubated in the presence of rC7 (in the Phase I formulation) at a final protein concentration of 0.5 mg/mL. To simulate body temperature, all samples were held for 1 hour at 37° C. The summary of these results is listed in Table 5. The results demonstrated no changes in protein content (A280), or turbidity (A320), after exposure to physiological media for 1 hour at 37° C. Thus, the data suggest there are no apparent immediate solubility limitations upon dilution ofrC7 delivery into serum.
  • D-PBS Dulbecco's phosphate buffered saline
  • the SEC-HPLC profiles confirmed that after 1 hour exposure to the various test buffer matrices at 37° C., there were no increases in insoluble aggregates.
  • FIG. 5 The exposure to the various buffers triggers an initial change in the lower molecular weight region of the peak, likely due to an increase in the flexibility of the a-helix.
  • Recombinant human collagen 7 was produced in CHO cells.
  • rC7 was purified from the CHO cells using two different manufacturing processes.
  • a rC7 lot produced from each manufacturing process was formulated in a composition comprising 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, 0.05% polysorbate 20, and pH of about 7.1.
  • test article 1 and test article 2 were administered intravenously at a dosage of 0.4 mg/mL (16 ⁇ g in 40 ⁇ L) to 26 neonatal Col7a1 ⁇ / ⁇ mice.
  • the compositions were administered as a single IV bolus via the superficial temporal vein on postnatal day 2-3.
  • a control composition comprising 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, 0.05% polysorbate 20, and pH of about 7.1-but without rC7 was also intravenously administered to neonatal Col7a1 ⁇ / ⁇ mice.
  • test article 1 and test article 2 resulted in the distribution of rC7 to the dermal-epidermal BMZ in tongue and front leg skin in the neonatal Col7a1 ⁇ / ⁇ mice.
  • FIG. 1 In contrast, C7 was not detected in any mouse injected with vehicle control.
  • FIG. 1 In contrast, C7 was not detected in any mouse injected with vehicle control.
  • Intravenous administration of the test compositions resulted in correction of dermal-epidermal separation in Col7a1 ⁇ / ⁇ mice compared to vehicle control, as demonstrated by representative hematoxylin and eosin (H&E) images of tongue tissue sections.
  • FIG. 2 The tongue is the optimal tissue to examine dermal-epidermal separation due to the lack of hair, which can serve to anchor the epidermis to the dermis and prevent the formation of newly induced blisters.
  • mice with complete closure i.e., no separation
  • the proportion of mice with complete closure was 50% (12 out of 26), 63% (15 out of 26), and 10% (1 out of 10) for Col7a1 ⁇ / ⁇ mice administered test article 1, test article 2, and vehicle control, respectively.
  • test article 1 or 2 A single IV administration of test article 1 or 2 to neonatal Col7a1 ⁇ / ⁇ mice resulted in a statistically significant improvement in survival compared to vehicle controls, as shown in Table 7 below and FIG. 3 .
  • the median (interquartile-range) survival time was 2.5, 9, and 14 days for Col7a1 ⁇ / ⁇ mice administered vehicle control, test article 1, and test article 2, respectively.
  • Drug Substance Frozen liquid with 500 mL fill in 1 L polycarbonate (PC) bottle with rC7 at 1.0 mg/mL
  • Drug Product DP—Frozen liquid with 5.5 mL fill in 10 cc glass vial with rC7 at 1.0 mg/mL.
  • the formulation comprises 10 mM sodium phosphate, 5 mM sodium citrate, 100 mM L-arginine, 50 mM sucrose, 70 mM NaCl, 0.05% (w/v) polysorbate-20, pH of about 7.2.
  • GMP material is stable for 3 months at both ⁇ 65 ⁇ 5° C. and ⁇ 20 ⁇ 5° C., with no significant change in the protein concentration, as measured by absorbance at 280 nm, no significant change in the amount of high molecular weight species, dimers, homotrimers, and low molecular weight species as measured by SEC-HPLC, and no significant change in potency, as measured in a fibronectin binding assay.
  • development lots are stable for 6 months at both ⁇ 65 ⁇ 5° C. and ⁇ 20 ⁇ 5° C.
  • the formulation is stable for 2 weeks at 25 ⁇ 3° C. and up to 1 month at 5 ⁇ 3° C.
  • the formulation is stable for up to 8 hours at 25 ⁇ 3° C. when stored in disposable syringes or for up to 6 hours at 25 ⁇ 3° C. when rC7 is diluted down to 0.1 mg/mL into saline. Furthermore, no changes in protein content were observed in the presence of PBS containing 10% bovine serum after storage for 1 hour at 37° C. This indicates physiological compatibility of the drug product upon administration.

Abstract

The present disclosure relates to compositions comprising collagen 7 and methods of using the same to treat DEB.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation application of U.S. patent application Ser. No. 16/065,512 filed Jun. 22, 2018, which is a U.S. National Phase entry application of PCT Patent Application No.: PCT/US2016/067995 filed Dec. 21, 2016, which claims the benefit of, and relies on the filing date of, U.S. provisional patent application No. 62/387,241, filed Dec. 23, 2015; U.S. provisional patent application No. 62/278,816, filed Jan. 14, 2016; and U.S. provisional patent application No. 62/293,209, filed Feb. 9, 2016; the contents of each of which are incorporated herein by reference in their entirety.
  • FIELD
  • The present invention relates to compositions (e.g., pharmaceutical compositions) and more particularly, to collagen 7 compositions and methods of using the same to treat dystrophic epidermolysis bullosa.
  • BACKGROUND
  • The collagen VII (C7) protein is a structural protein in which C7 is both a structural and functional precursor for the formation of anchoring fibrils which enable the attachment of the epidermis and the dermis. The C7 monomer assembles as a homotrimer of approximately 900 kDa, containing one NC-1 and one NC-2 binding region. The homotrimer is held together via an alpha helical coil. From a homotrimer, these C7 protein precursors align into an anti-parallel dimer with an increase in molecular weight to 1.8 mDa. Lateral assembly of anti-parallel dimers leads to the formation of the anchoring fibrils. The rC7 protein demonstrates “stickiness,” and the propensity to precipitate at higher protein concentrations, particularly in PBS buffer.
  • Lack of C7 leads to abnormal decrease in these anchoring fibrils and a condition known as Dystrophic Epidermolysis Bullosa (DEB). EB encompasses a group of molecularly diverse diseases characterized by the development of blisters after minor mechanical trauma to the skin. (1) Severity, extracutaneous manifestations and clinical course depend on the type of EB; however, almost all patients experience blistering or ruptured skin. (2)
  • Dystrophic epidermolysis bullosa (DEB) is a form of EB characterized by development of scarring with healing. (3) In patients with DEB, blistering can be triggered by even minor mechanical trauma owing to the extremely fragile nature of the skin. This leads to a chronic cycle of blistering, healing, and reblistering that causes patients to suffer from painful wounds and debilitating scarring of epithelial tissue. There is no cure for DEB and management focuses on supportive care.
  • Dystrophic EB is caused by mutations in the Col7a1 gene, which encodes the alpha chain of collagen VII (C7), a protein essential for the formation of the anchoring fibrils that anchor the basement membrane to the underlying dermis. (4) The condition is inherited as either a dominant form (DDEB) or a recessive form (RDEB), with RDEB typically having a more severe phenotype. (5)
  • The DEB mouse model (Col7a1−/−) has a targeted inactivation of the Col7a1 gene, resulting in severe blistering that mimics the histological and ultrastructural presentation of severe human RDEB. (6) These mice have no detectable collagen VII at the dermal-epidermal basement membrane zone (BMZ), completely lack anchoring fibrils, and are born with extensive cutaneous blisters on their ventral surface and hemorrhagic blisters on their paws and neck, with mortality typically observed within the first week of life.
  • SUMMARY
  • The present disclosure relates to compositions comprising collagen 7 and methods of preparing such compositions. In certain embodiments, the composition comprises recombinant collagen 7, sodium phosphate, sodium citrate, sodium chloride, arginine, a sugar, and a surfactant. It is possible to exclude one or more of the sodium phosphate, sodium citrate, sodium chloride, arginine, sugar, or surfactant from the composition.
  • In certain embodiments, the sugar is sucrose. In certain embodiments the surfactant is a polysorbate, such as polysorbate 20 or polysorbate 80.
  • In certain embodiments, the sodium phosphate is present in an amount ranging from 100 mM to 1 mM, 50 mM to 5 mM, 25 mM to 5 mM, 10 mM to 5 mM, 10 mM to 1 mM or about 10 mM.
  • In certain embodiments, the sodium citrate is present in an amount ranging from 100 mM to 1 mM, 50 mM to 1 mM, 25 mM to 1 mM, 10 mM to 1 mM, 5 mM to 1 mM or about 5 mM.
  • In certain embodiments, the sodium chloride is present in an amount ranging from 200 mM to 10 mM, 100 mM to 20 mM, 80 mM to 50 mM, or about 70 mM.
  • In certain embodiments, the arginine is present in an amount ranging from 200 mM to 5 mM, 150 mM to 50 mM, 100 mM to 75 mM, or about 100 mM.
  • In certain embodiments, the sugar is present in an amount ranging from 100 mM to 10 mM, 100 mM to 50 mM, 75 mM to 35 mM, 60 mM to 40 mM, or about 50 mM. In certain embodiments, the sucrose is present in an amount ranging from 100 mM to 1 mM, 100 mM to 50 mM, 75 mM to 35 mM, 60 mM to 40 mM, or about 50 mM.
  • In certain embodiments, the surfactant is present in an amount ranging from 1% to 0.01%, 0.5% to 0.05%, 0.025% to 0.05%, 0.1% to 0.05%, 0.05% to 0.01% or about 0.05% (w/v). In certain embodiments, the polysorbate (e.g., polysorbate 20) is present in an amount ranging from 1% to 0.01%, 0.5% to 0.05%, 0.025% to 0.05%, 0.1% to 0.05%, 0.05% to 0.01% or about 0.05% (w/v).
  • In certain embodiments, the composition has a pH of between 8.0 to 6.0, 7.5 to 6.5, 7.5 to 7.0, 7.4 to 7.0, 7.3 to 7.0, 7.2 to 7.0, 7.1 to 7.0, 7.2 to 7.1, 7.2±0.3, about 7.1, or about 7.2.
  • In certain embodiments, the composition comprises recombinant collagen 7, 100 mM to 10 mM sodium phosphate, 100 mM to 1 mM sodium citrate, 200 mM to 10 mM sodium chloride, 200 mM to 10 mM arginine, 100 mM to 10 mM sucrose, and 1.0% to 0.01% (w/v) polysorbate 20 and optionally a pH of 7.2 to 7.1.
  • In certain embodiments, the composition comprises recombinant collagen 7, 25 mM to 5 mM sodium phosphate, 25 mM to 1 mM sodium citrate, 100 mM to 20 mM sodium chloride, 150 mM to 50 mM arginine, 75 mM to 35 mM sucrose, and 0.5% to 0.05% polysorbate 20 (w/v) and optionally a pH of7.2 to 7.1.
  • In one embodiment, the composition comprises recombinant collagen 7, about 10 mM sodium phosphate, about 5 mM sodium citrate, about 70 mM sodium chloride, about 100 mM arginine, about 50 mM sucrose, and about 0.05% polysorbate 20 (w/v) and has a pH of about 7.1.
  • In another embodiment, the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.1.
  • In another embodiment, the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.2±0.3.
  • In certain embodiments, the recombinant collagen 7 is a human collagen 7.
  • The collagen 7 compositions have increased stability. In certain embodiments, the composition is stable for 3 months following storage at −65±5° C. or −20±5° C. in a 1 liter polycarbonate bottle (500 mL of the composition). In certain embodiments, the composition is stable for 6 months following storage at −65±5° C. or −20±5° C. in a 10 cc glass vial (5.5 mL of composition). In certain embodiments, the composition is stable following storage for 2 weeks at 25±3° C. In certain embodiments, the composition is stable following storage for up to 1 month at 5±3° C.
  • The compositions may be pharmaceutical compositions comprising one or more additional pharmaceutical excipients that can be used, for example, to treat DEB.
  • Another aspect is directed to a method of treating DEB, the method comprising administering to a subject in need thereof an effective amount of a recombinant collagen 7 composition, as described herein.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows representative immunofluorescent images in tongue and front leg from Col7a1−/− mice surviving 5-7 days following administration of rC7 compositions or a control.
  • FIG. 2 shows representative H&E staining in tongue from Col7a1−/− mice following administration of rC7 compositions or a control.
  • FIG. 3 is a Kaplan-Meier Curve showing the probability of survival of Col7a1−/− mice following administration of rC7 compositions or a control.
  • FIG. 4 shows SEC-HPLC profiles of a rC7 formulation after exposure to freeze/thaw stress.
  • FIG. 5 shows SEC-HPLC for 0.5 mg/mL rC7 diluted in D-PBS and 10% serum after incubation for 1 hour at 37° C.
  • FIG. 6 shows % HMW species of rC7 formulations containing different amounts of arginine after storage at various temperatures.
  • FIG. 7 shows fibronectin binding of rC7 formulations containing different amounts of arginine after storage at various temperatures.
  • The foregoing and other objects, features, and advantages of the rC7 compositions will become more apparent from the following detailed description.
  • DETAILED DESCRIPTION
  • As a major component of anchoring fibrils, collagen 7 functions in maintaining tissue integrity. Anchoring fibrils are structural elements that serve as attachment complexes at the interface between the epithelial and mesenchymal layers of several tissues, including the skin, oral mucosa, and cervix (Chung et al. Dermatol Clin 28(1): 93-105 (2010)). In the skin, anchoring fibrils extend from the lower portion of the epidermal basement membrane to the underlying papillary dermis, securing the association between the epidermal basement membrane and the papillary dermis (Varki et al. J Med Genet 44:181-192 (2007)). This association aids to provide and maintain cohesion between the epidermis and dermis, contributing to the integrity to the skin, which is critical for its proper structure, function, and homeostasis (Villone et al. J Biol Chem 283(36): 24506-24513 (2008)). Nucleic acid sequences encoding collagen 7 and polypeptides encoding thereby are known in the art. In one embodiment the recombinant collagen 7 is a human collagen 7.
  • Collagen 7 produced by recombinant methods described herein may be recovered from the host cell culture system according to standard protocols known in the art (e.g., precipitation, centrifugation, etc.). Recombinant collagen 7 described herein may be secreted into the host cell medium and recovered by ammonium sulfate precipitation and subsequent centrifugation; as demonstrated in the following reference, (Chen et al. J Bio Chem 277(18): 2118-2124 (2002)).
  • Collagen 7 produced and recovered by recombinant and molecular biology methods described herein, may be purified according to standard protocols known in the art (e.g., dialysis, ion exchange chromatography, affinity chromatography, SDS gel electrophoresis, etc.). The recombinant collagen 7 described herein may be purified to homogeneity by ion exchange chromatography; as demonstrated in the following reference, (Chen et al. J. Bio. Chem. 277(18): 2118-2124 (2002)).
  • This disclosure provides a recombinant C7 (rC7) formulation containing a buffer, a non-essential amino acid, a cryoprotectant, a surfactant, and a chelating agent. In certain embodiments the non-essential amino acid is arginine, preferably L-arginine. In certain embodiments, the cryoprotectant is a sugar. In certain embodiments, the buffer comprises a phosphate buffer, such as sodium phosphate. In certain embodiments the chelating agent is citrate. In a preferred embodiment, the rC7 formulation comprises 10 mM sodium phosphate, 5 mM sodium citrate, 100 mM L-arginine, 50 mM sucrose, 70 mM NaCl, 0.05% (w/v) polysorbate-20, and pH of about 7.2.
  • As compared to the previously used PBS buffer, these improved formulations enhance rC7 solubility, increasing rC7 protein concentration about 8-10 fold as compared to previously used PBS buffers, and increase robustness/stability against freeze and thaw, as well as thermal stresses during storage and handling. In certain embodiments, the rC7 compositions described herein are stable for 3 months at both −65±5° C. and −20±5° C. In certain embodiments, the rC7 compositions described herein are stable for 6 months at both −65±5° C. and −20±5° C. In certain embodiments, the rC7 composition is stable for 2 weeks at 25±3° C. and up to 1 month at 5±3° C. In terms of delivery device compatibility, in certain embodiments, the rC7 composition described herein is stable for up to 8 hours at 25±3° C. when stored in disposable syringes or for up to 6 hours at 25±3° C. when diluted down to 0.1 mg/mL into saline. As used herein, “stable” means that after the indicated storage conditions, there is no significant difference in the amount of protein, as measured by absorbance at 280 nm, no significant difference in the amount of high molecular weight species, dimers, homotrimers, and low molecular weight species as measured by SEC-HPLC, and no significant loss in potency, as measured in a fibronectin binding assay.
  • Sugars that may be used in the compositions include, but are not limited, to non-reducing disaccharides (e.g., sucrose, trehalose) or a sugar alcohol, such as sorbitol. In certain embodiments, the sugar in the composition is sucrose.
  • The composition may also include one or more buffers, such as a phosphate buffer (e.g., sodium phosphate, a histidine buffer, a Tris(hydroxymethyl)aminomethane buffer.
  • The composition may also include one or more surfactants and/or bulking agents. Surfactants include but are not limited poloxamers (e.g., Pluronic®), polysorbates (e.g., polysorbate 80, polysorbate 20), CTAB (hexadecyltrimethylammonium bromide), or SDS (sodium dodecyl sulfate or sodium lauryl sulfate). In certain embodiments, the surfactant in the composition is a polysorbate (e.g., polysorbate 20). Bulking agents include but are not limited to PVP, PEG, dextran, mannitol and glycine.
  • The composition may also include one more additional pharmaceutically acceptable excipients. Examples of pharmaceutically acceptable excipients include, but are not limited to salts, proteins (e.g., albumin), chelating agents (e.g., citrate, EDTA), gelling polymers, urea (or its derivatives), and combinations thereof.
  • One aspect is directed to the use of the compositions described herein to treat DEB. Also provided is a method of treating DEB comprising administering to a subject in need thereof a composition comprising recombinant collagen 7, a buffer, anon-essential amino acid, a cryoprotectant, a surfactant, and a chelating agent. In certain embodiment, the buffer is a phosphate buffer (e.g., sodium phosphate). In certain embodiments, the non-essential amino acid is arginine. In certain embodiments, the cryoprotectant is a sugar, such as sucrose. In certain embodiments the surfactant is a polysorbate, such as polysorbate 20 or polysorbate 80. In certain embodiments, the chelating agent is citrate. The amount of each component can vary as described throughout the application. In one embodiment, the composition comprises recombinant collagen 7, 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, and 0.05% polysorbate 20 (w/v) and has a pH of 7.1 or 7.2.
  • EXAMPLES Example 1
  • Five formulations were selected to compare freeze/thaw profiles and thermal stability, including the previously used PBS formulation. These formulations are listed in Table 1.
  • TABLE 1
    Formulation Buffer Ingredients
    1 PBS, pH 7.3 0.5 mg/mL C7, 150 mm NaCl
    2 10 mM Phos pH 7.3 0.5 mg/mL C7,
    70 mM NaCl, 50 mM Arg,
    50 mM Gln;
    1.7% sucrose,
    0.05% (v/v) Polysorbate-20,
    5 mM citrate
    3 10 mM Phos pH 7.3 0.5 mg/mL C7,
    100 mM NaCl, 50 mM Arg,
    1.7% sucrose,
    0.05% (v/v) Polysorbate-20,
    5 mM citrate
    4 10 mM Phos pH 7.3 0.5 mg/mL C7,
    0 mM NaCl, 100 mM Arg,
    1.7% sucrose,
    0.05% (v/v) Polysorbate-20,
    5 mM citrate
    5 20 mM Histidine 0.5 mg/mL C7,
    pH 7.3 70 mM NaCl, 50 mM Arg,
    1.7% sucrose
    0.05% (v/v) Polysorbate-20,
    5 mM citrate
    6 10 mMTRIS pH 7.3 0.5 mg/mL C7,
    70 mM NaCl, 50 mM Arg,
    0.05% (v/v) Polysorbate-20,
    5 mM citrate
  • By testing these formulations, it was discovered that L-arginine and sucrose, and preferably 75-100 mM L-arginine and 1.7% (w/v) sucrose, improved solubility. The inclusion of a surfactant (e.g., 0.05% polysorbate-20) improved filterability and is expected to lower potential non-specific adsorption. The inclusion of NaCl, and preferably about 70 mM NaCl, in the formulation improved the appearance.
  • The previously used rC7 formulation (PBS) only achieved solubility of upwards of 0.4 mg/mL with significant viscosity challenges above this concentration. In some cases, the PBS formulation could be pushed to upwards of 0.8 mg/mL, but only under very specific lab conditions that were not scalable or compatible with conventional protein purification processes, such as UF/DF. A formulation comprising 1 mg/mL rC7, 10 mM phosphate, 5 mM citrate, 100 mM L-arginine, 1.7% (w/v) sucrose, 70 mM NaCl, 0.05% (v/v) polysorbate-20 at pH of about 7.2 (“Phase I formulation”) was able to achieve approximately ten-fold higher protein concentration (upwards of about 4 mg/mL) using conventional spin columns with a 100 kDa PES molecular weight cutoff.
  • Improvements in the viscosity were seen as well when compared to the PBS formulation. The PBS formulation was not able to support protein concentrations above 0.8 mg/mL. Therefore, head-to-head comparisons with higher concentration protein formulations were not possible. However, the viscosity readings for the 3 mg/mL rC7 protein formulations were all around 4 centipoise (cp), which is not considered highly viscous. These data demonstrate that in general, the increase in viscosity correlates to the increase in rC7 protein concentration. If the viscosity is compared to the protein concentration (cP/protein concentration), it becomes evident that the arginine containing formulations have lower values than those in the PBS formulation. Thus, the arginine formulations support higher protein content while better controlling increases in viscosity.
  • Under the intensity of a Tyndall light, the refraction of the protein and protein precipitates was significantly less for the Phase I formulation at the higher protein concentrations (upwards of 3-4 mg/mL). This gave the Phase I formulation a clearer and less turbid appearance.
  • The Phase I formulation was also subjected to freezing and thawing stress conditions, using a robust, small scale freeze/thaw model. The Phase I formulation was exposed to three freeze/thaw cycles (see Table 2) to reflect freeze/thaw stress that may be encountered at manufacturing scale (scalability).
  • TABLE 2
    Freeze/Thaw (F/T) Stress Cycle for 100 mL in PETG Bottles
    Step Ramp Target T Time
    1 Hold C. 30 min
    2 Ramp −5° C. 0.2° C./min
    3 Hold −5° C. 6 Hours
    4 Ramp −20° C. 0.2° C./min
    5 Hold −20° C. 6 Hours
    6 Ramp −20° C. 0.2° C./min
    7 Hold 25° C. 3 Hours
    8 Ramp C. 30 min
  • After the 3 cycles were completed, there were no changes in either the protein content (as measured by absorbance at A280 nm; see Table 3) or in association state (as measured by SEC-HPLC; see FIG. 4).
  • TABLE 3
    Changes After Freeze/Thaw Stress
    A280 nm Size Exclusion Chromatography (SEC-HPLC)
    mg/mL % HMW % Dimer % Homotrimer % LMW
    T0 1.0 1.4 10.7 78.7 9.2
    Post 3 1.0 1.8 11.2 78.1 8.9
    FIT
    Cycles
  • The arginine-containing formulations were subjected to 2 months of storage at temperatures of −6S° C., −20° C., S±3° C., and 2S° C. There were no changes in appearance after 2 months at any storage condition. The same materials showed no significant change in protein concentration (A280 nm) or SEC-HPLC profile (see Table 4) with the exception of the formulations stored either at 5±3° C. (minimally) or 25° C. After 2 months storage at 25° C., an increase (˜2%) in high molecular weight (HMW) species was seen.
  • TABLE 4
    Changes After Storage for Two Months at Various Temperatures
    A280 nm Size Exclusion Chromatography (SEC-HPLC)
    Sample mg/mL % HMW % Dimer % Homotrimer % LMW
    T0
    <−65° C. 1.1 1.6 9.9 77.3 11.2
    −20° C. 1.1 1.7 10.2 76.8 11.4
    C. 1.1 2.3 12.4 74.4 10.9
    25° C. 1.1 4.1 18.0 67.5 10.4
  • The Phase I formulation at a 1 mg/mL dose was also assessed for compatibility with the animal delivery devices and dosing syringes. The 1 mg/mL drug product was injected into the infusion set(s) with the appropriate syringe(s), and the drug held for up to 60 minutes in each of the devices. There were no visible particles or pH shifts after 60 minutes in the devices. The SEC-HPLC profiles also demonstrated no significant changes.
  • It is difficult to know what happens when rC7 is injected IV into the bloodstream. It is possible, however, to assess the potential for product quality changes by monitoring for any changes when the protein is exposed to more physiologically relevant conditions such as buffers containing 10% bovine serum and held at 37° C. The rC7 protein is prone to soluble aggregation and precipitation. These events can be monitored through common analytical techniques. The A280 absorbance is used to monitor for changes in protein content. The A320 absorbance is used to monitor for changes in turbidity due to insoluble particulate formation. SEC-HPLC is used to monitor for changes in the soluble protein where HMW and/or LMW species may form. To maximize the impact of the serum effect, the buffers were diluted with equal parts serum and incubated in the presence of rC7 (in the Phase I formulation) at a final protein concentration of 0.5 mg/mL. To simulate body temperature, all samples were held for 1 hour at 37° C. The summary of these results is listed in Table 5. The results demonstrated no changes in protein content (A280), or turbidity (A320), after exposure to physiological media for 1 hour at 37° C. Thus, the data suggest there are no apparent immediate solubility limitations upon dilution ofrC7 delivery into serum.
  • TABLE 5
    A280 and A320 Absorbance after 1 hour at 37° C. exposure
    Initial
    1 hr at Initial 1 hr at
    (A280/mm) 37° C. (A320/m) 37° C.
    Water 0.00145 0.00134 0.00090 0.00097
    Placebo in PBS + Ca 0 0.00215 0.00127 0.00138
    Placebo in PBS no Ca 0.00375 0.00225 0.00221 0.00108
    Placebo in PBS + Ca 0.32405 0.31336 0.01319 0.0131
    and 10% serum
    Placebo in PBS no Ca 0.32068 0.31129 0.01363 0.01267
    and 10% serum
    0.5 mg/mL C7 in 0.02896 0.03234 0.00188 0.00224
    PBS + Ca
    0.5 mg/mL C7 in PBS 0.03134 0.03139 0.00210 0.00184
    no Ca
    0.5 mg/mL C7 in 0.33182 0.33807 0.01321 0.01345
    PBS + Ca and 10%
    serum
    0.5 mg/mL C7 in PBS 0.34846 0.33424 0.01386 0.01338
    no Ca and 10% serum
    90% PBS + 10% serum n/a 0.31847 n/a 0.01343
  • To see if calcium and magnesium had an effect on changes in product quality, the starting Dulbecco's phosphate buffered saline (D-PBS) was obtained without the inclusion of Ca2+ and Mg2+ cations in the buffer. This solution was tested and compared to D-PBS where physiological levels of Ca2+ and Mg2+ were added. These were found to have no effect on the results (Table 5).
  • The SEC-HPLC profiles confirmed that after 1 hour exposure to the various test buffer matrices at 37° C., there were no increases in insoluble aggregates. FIG. 5. The exposure to the various buffers triggers an initial change in the lower molecular weight region of the peak, likely due to an increase in the flexibility of the a-helix.
  • Thermal stability studies were conducted on rC7 formulations containing different amounts of L-arginine (25 mM to 100 mM). The results show that lowering the L-arginine level leads to a slight increase in high molecular weight species and decreased potency (as measured by a fibronectin binding assay) after storage for up to one month at both 5±3° C. and 25±3° C. FIGS. 6 and 7. Additionally, more opalescence was seen in the samples at 25 mM and 50 mM arginine. Other excipient levels were kept the same in these studies.
  • Example 2
  • Based on initial development studies, a formulation containing 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, 0.05% polysorbate 20, and pH of about 7.1 was used for administration to Col7a1−/− mice in a mouse model of DEB.
  • Recombinant human collagen 7 (rC7) was produced in CHO cells. rC7 was purified from the CHO cells using two different manufacturing processes. A rC7 lot produced from each manufacturing process was formulated in a composition comprising 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, 0.05% polysorbate 20, and pH of about 7.1.
  • The study was conducted in a total of 59 neonatal Col7a1−/− mice across 3 treatment groups to evaluate distribution and histological improvement following a single IV administration of rC7 on postnatal day 2-3. Overall survival was additionally examined in this study. Each composition (test article 1 and test article 2) was administered intravenously at a dosage of 0.4 mg/mL (16 μg in 40 μL) to 26 neonatal Col7a1−/− mice. The compositions were administered as a single IV bolus via the superficial temporal vein on postnatal day 2-3. A control composition comprising 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose, 0.05% polysorbate 20, and pH of about 7.1-but without rC7 was also intravenously administered to neonatal Col7a1−/− mice.
  • Administration of test article 1 and test article 2 resulted in the distribution of rC7 to the dermal-epidermal BMZ in tongue and front leg skin in the neonatal Col7a1−/− mice. FIG. 1. In contrast, C7 was not detected in any mouse injected with vehicle control. FIG. 1.
  • Intravenous administration of the test compositions resulted in correction of dermal-epidermal separation in Col7a1−/− mice compared to vehicle control, as demonstrated by representative hematoxylin and eosin (H&E) images of tongue tissue sections. FIG. 2. The tongue is the optimal tissue to examine dermal-epidermal separation due to the lack of hair, which can serve to anchor the epidermis to the dermis and prevent the formation of newly induced blisters.
  • The proportion of mice with complete closure (i.e., no separation) between the epidermis and dermis in tongue was 50% (12 out of 26), 63% (15 out of 26), and 10% (1 out of 10) for Col7a1−/− mice administered test article 1, test article 2, and vehicle control, respectively. The proportion of Col7a1−/− mice with complete closure of dermal-epidermal separation was higher in rC7 treated mice than vehicle control (test article 1 vs vehicle Fisher p=0.06, test article 2 vs vehicle Fisher p=0.02). The proportion of Col7a1−/− mice with closure of dermal-epidermal separation did not differ between rC7 treated mice (Fisher p=0.58).
  • A single IV administration of test article 1 or 2 to neonatal Col7a1−/− mice resulted in a statistically significant improvement in survival compared to vehicle controls, as shown in Table 7 below and FIG. 3. The median (interquartile-range) survival time was 2.5, 9, and 14 days for Col7a1−/− mice administered vehicle control, test article 1, and test article 2, respectively.
  • TABLE 6
    Hazard Ratios Comparing Treatment Groups
    Hazard 95%
    Comparison Ratio Confidence Interval P Value
    Test Article
    1 versus 0.18 0.08-0.40 <0.001
    Test Article 2 versus 0.11 0.05-0.25 <0.001
    Test Article 1 versus 1.66 0.87-3.15 0.13
    Test Article 2
  • In Table 6, hazard ratios were estimated in a Cox proportional hazards model. The proportional hazards assumption was satisfied as assessed by Schoenfeld residuals (p>0.37).
  • This study demonstrated efficacy for the tested rC7 compositions in a neonatal murine model of DEB. Specifically, both test articles distributed to the dermal-epidermal BMZ in tongue and skin, corrected dermal-epidermal separation, and resulted in a statistically significant improvement in survival compared to vehicle control following a single IV injection in Col7a1−/− mice.
  • To support Phase 1/11 clinical trials of Dystrophic Epidermolysis Bullosa (DEB) program, the following drug substances/drug products will be used: Drug Substance (DS)—Frozen liquid with 500 mL fill in 1 L polycarbonate (PC) bottle with rC7 at 1.0 mg/mL; Drug Product (DP)—Frozen liquid with 5.5 mL fill in 10 cc glass vial with rC7 at 1.0 mg/mL. The formulation comprises 10 mM sodium phosphate, 5 mM sodium citrate, 100 mM L-arginine, 50 mM sucrose, 70 mM NaCl, 0.05% (w/v) polysorbate-20, pH of about 7.2.
  • In terms of long-term storage stability, for DS, to date, GMP material is stable for 3 months at both −65±5° C. and −20±5° C., with no significant change in the protein concentration, as measured by absorbance at 280 nm, no significant change in the amount of high molecular weight species, dimers, homotrimers, and low molecular weight species as measured by SEC-HPLC, and no significant change in potency, as measured in a fibronectin binding assay. For DP, to date, development lots are stable for 6 months at both −65±5° C. and −20±5° C. For handling, the formulation is stable for 2 weeks at 25±3° C. and up to 1 month at 5±3° C. Care should be taken to minimize exposure from light, prevent excessive shaking during product handling. In terms of delivery device compatibility, the formulation is stable for up to 8 hours at 25±3° C. when stored in disposable syringes or for up to 6 hours at 25±3° C. when rC7 is diluted down to 0.1 mg/mL into saline. Furthermore, no changes in protein content were observed in the presence of PBS containing 10% bovine serum after storage for 1 hour at 37° C. This indicates physiological compatibility of the drug product upon administration.
  • While one or more exemplary embodiments have been described in the specification, it will be understood by those of ordinary skill in the art that various changes in form and details may be made therein without departing from the spirit and scope of the inventive concept as defined by the claims that follow.
  • REFERENCES
    • 1. Fine J D, Bauer E A, Gedde-Dahl, T (1999). Inherited epidermolysis bullosa: definition and historical overview. In: Fine J D, Bauer E A, McGuire J, Moshell A, eds. Epidermolysisbullosa: Clinical, epidemiologic, and laboratory advances, and the findings of the National Epidermolysis Bullosa Registry. Baltimore: Johns Hopkins University Press; 1999:1-19.
    • 2. Fine J D, Eady R A, Bauer E A, et al. The classification of inherited epidermolysis bullosa (E B): Report of the Third International Consensus Meeting on Diagnosis and Classification of E B. J Am Acad Dermatol. 2008 June; 58(6):931-50.
    • 3. Bruckner-Tuderman L. Dystrophic epidermolysis bullosa: Pathogenesis and clinical features. Dermatol Clin. 2010 January; 28(1):107-14.
    • 4. Pfendner E G, Lucky A W. Dystrophic Epidermolysis Bullosa. 2006 Aug. 21 [Updated 2010 Nov. 4]. In: Pagon R A, Adam M P, Bird T D, et al., editors. Gene Reviews™ [Internet]. Seattle (Wash.): University of Washington, Seattle; 1993-2013.
    • 5. Dang N, Murrell D F. Mutation analysis and characterization of COL7A1 mutations in dystrophic epidermolysis bullosa. Exp Dermatol. 2008 July; 17(7):553-68.
    • 6. Heinonen S. Targeted inactivation of the type VII collagen gene (Col7a1) in mice results in severe blistering phenotype: a model for recessive dystrophic epidermolysis bullosa. J Cell Sci. 1999 November; 112(21):3641-8.

Claims (11)

1. A composition comprising at least 0.5 mg/ml recombinant collagen 7 (rCo17) in a formulation comprising arginine.
2. The composition of claim 1, wherein rCol7 is at a concentration of about 1.0 mg/ml to about 4.0 mg/ml.
3. The composition of claim 2, wherein rCol7 is at a concentration of about 1.0 mg/ml to about 2.0 mg/ml.
4. The composition of claim 3, wherein the formulation further comprises 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose and 0.05% (v/v) polysorbate 20.
5. The composition of claim 4 for use of medicine administered to a subject in need through infusion wherein the infusion lasts at least 60 minutes.
6. A method for treating dystrophic epidermolysis bullosa comprising administering to a subject in need thereof an effective amount of a composition that comprises about 1.0 mg/ml to about 4.0 mg/ml recombinant collagen 7.
7. The method of claim 6 wherein the composition is administrated to the subject in need by intravenous administration.
8. A method for increasing closure between epidermis and dermis of the wounded skin in a subject comprising administering to the subject a composition comprising a recombinant collagen 7 in an arginine containing formulation.
9. The method of claim 8 wherein the composition is administrated to the subject in need by intravenous administration.
10. The method of claim 6 wherein the composition comprises about 1.0 mg/ml to about 2.0 mg/ml recombinant collagen 7.
11. The method of claim 10 wherein the composition further comprises 10 mM sodium phosphate, 5 mM sodium citrate, 70 mM sodium chloride, 100 mM arginine, 50 mM sucrose and 0.05% (v/v) polysorbate 20.
US16/875,223 2015-12-23 2020-05-15 Collagen 7 compositions and methods of using the same Abandoned US20200316158A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/875,223 US20200316158A1 (en) 2015-12-23 2020-05-15 Collagen 7 compositions and methods of using the same
US18/169,368 US20230414698A1 (en) 2015-12-23 2023-02-15 Collagen 7 compositions and methods of using the same

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201562387241P 2015-12-23 2015-12-23
US201662278816P 2016-01-14 2016-01-14
US201662293209P 2016-02-09 2016-02-09
PCT/US2016/067995 WO2017112757A1 (en) 2015-12-23 2016-12-21 Collagen 7 compositions and methods of using the same
US201816065512A 2018-06-22 2018-06-22
US16/875,223 US20200316158A1 (en) 2015-12-23 2020-05-15 Collagen 7 compositions and methods of using the same

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2016/067995 Continuation WO2017112757A1 (en) 2015-12-23 2016-12-21 Collagen 7 compositions and methods of using the same
US16/065,512 Continuation US10695395B2 (en) 2015-12-23 2016-12-21 Collagen 7 compositions and methods of using the same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/169,368 Continuation US20230414698A1 (en) 2015-12-23 2023-02-15 Collagen 7 compositions and methods of using the same

Publications (1)

Publication Number Publication Date
US20200316158A1 true US20200316158A1 (en) 2020-10-08

Family

ID=59091196

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/065,512 Active US10695395B2 (en) 2015-12-23 2016-12-21 Collagen 7 compositions and methods of using the same
US16/875,223 Abandoned US20200316158A1 (en) 2015-12-23 2020-05-15 Collagen 7 compositions and methods of using the same
US18/169,368 Pending US20230414698A1 (en) 2015-12-23 2023-02-15 Collagen 7 compositions and methods of using the same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/065,512 Active US10695395B2 (en) 2015-12-23 2016-12-21 Collagen 7 compositions and methods of using the same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/169,368 Pending US20230414698A1 (en) 2015-12-23 2023-02-15 Collagen 7 compositions and methods of using the same

Country Status (8)

Country Link
US (3) US10695395B2 (en)
EP (1) EP3394095B1 (en)
JP (3) JP6943858B2 (en)
CN (2) CN114225013A (en)
CA (1) CA3006998A1 (en)
IL (2) IL259583B (en)
MX (1) MX2018007468A (en)
WO (1) WO2017112757A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017112757A1 (en) * 2015-12-23 2017-06-29 Shire Human Genetic Therapies, Inc. Collagen 7 compositions and methods of using the same
US20220177546A1 (en) * 2019-03-27 2022-06-09 Phoenix Tissue Repair, Inc. Systems and methods for producing collagen 7 compositions
WO2023064806A1 (en) 2021-10-12 2023-04-20 Phoenix Tissue Repair, Inc. Collagen 7 protein replacement therapy

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090038753A1 (en) * 2006-02-23 2009-02-12 Sony Chemical & Information Device Corporation Mounting method
US10669327B2 (en) * 2016-03-16 2020-06-02 Phoenix Tissue Repair, Inc. Methods of purifying collagen 7
US10695395B2 (en) * 2015-12-23 2020-06-30 Phoenix Tissue Repair, Inc. Collagen 7 compositions and methods of using the same

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2757874B1 (en) * 1996-12-17 2003-04-25 Biocem RECOMBINANT COLLAGENS AND DERIVED PROTEINS PRODUCED BY PLANTS, THEIR METHODS OF OBTAINING AND THEIR USES
MXPA04008215A (en) * 2002-02-27 2004-11-26 Immunex Corp Polypeptide formulation.
GB0216001D0 (en) * 2002-07-10 2002-08-21 Nat Blood Authority Process and composition
US20080287349A1 (en) * 2003-05-09 2008-11-20 Jeffrey Peterson Fibroblast growth factor 20 and methods of use thereof
MXPA06006886A (en) * 2003-12-19 2006-09-04 Novo Nordisk Healthcare Ag Stabilised compositions of factor vii polypeptides.
AU2007321877B2 (en) * 2006-08-01 2011-11-03 Kineta Two, Llc Pharmaceutical manufacturing methods
US20120121580A1 (en) * 2009-07-28 2012-05-17 Merck Sharp & Dohme Corp. Methods for producing high concentration lyophilized pharmaceutical formulations
US8475824B2 (en) * 2010-01-26 2013-07-02 Warsaw Orthopedic, Inc. Resorbable matrix having elongated particles
WO2011113024A1 (en) * 2010-03-11 2011-09-15 University Of Southern California Compositions and methods for healing of skin wounds
JP2013525502A (en) * 2010-05-06 2013-06-20 ユニヴァーシティー オブ サザン カリフォルニア Methods and agents for enhancing wound healing
RU2614257C2 (en) * 2011-04-20 2017-03-24 Сандоз Аг STABLE LIQUID PHARMACEUTICAL PREPARATIONS OF FUSED PROTEIN TNFR: Fc
CA2854160A1 (en) * 2011-11-02 2013-05-10 Halscion, Inc. Methods and compositions for wound treatment
US20180104186A1 (en) * 2013-03-13 2018-04-19 University Of Miami Methods and compositions for the treatment of epidermolysis bullosa
CN104740612A (en) * 2013-12-31 2015-07-01 上海中信国健药业股份有限公司 Liquid formulation of receptor antibody fusion protein
AU2015243535A1 (en) * 2014-04-08 2016-11-03 University Of Southern California Polypeptide compositions with Type VII collagen fibronectin Type Ill- like repeats and treatment methods for wound closure and healing

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090038753A1 (en) * 2006-02-23 2009-02-12 Sony Chemical & Information Device Corporation Mounting method
US10695395B2 (en) * 2015-12-23 2020-06-30 Phoenix Tissue Repair, Inc. Collagen 7 compositions and methods of using the same
US10669327B2 (en) * 2016-03-16 2020-06-02 Phoenix Tissue Repair, Inc. Methods of purifying collagen 7

Also Published As

Publication number Publication date
JP2023166014A (en) 2023-11-17
MX2018007468A (en) 2018-11-14
US20230414698A1 (en) 2023-12-28
CN114225013A (en) 2022-03-25
US10695395B2 (en) 2020-06-30
US20190000912A1 (en) 2019-01-03
CN108463470B (en) 2021-10-22
IL259583A (en) 2018-07-31
CA3006998A1 (en) 2017-06-29
JP6943858B2 (en) 2021-10-06
EP3394095B1 (en) 2023-09-27
CN108463470A (en) 2018-08-28
EP3394095A4 (en) 2019-08-14
WO2017112757A1 (en) 2017-06-29
JP2019504043A (en) 2019-02-14
IL259583B (en) 2022-03-01
EP3394095A1 (en) 2018-10-31
IL290166A (en) 2022-03-01
JP2021185201A (en) 2021-12-09

Similar Documents

Publication Publication Date Title
US20230414698A1 (en) Collagen 7 compositions and methods of using the same
US11633462B2 (en) Pharmaceutical composition comprising plasminogen and uses thereof
CN106456717B (en) Quick-acting insulin composition
KR102579789B1 (en) C1-inh compositions and methods for the prevention and treatment of disorders associated with c1 esterase inhibitor deficency
US20150150982A1 (en) Pharmaceutical formulation for a therapeutic antibody
JP2018138578A (en) Rapid-acting insulin compositions
JP6525987B2 (en) Stable preparation of insulin glulysin
JP2010513462A (en) Factor VII and Factor VIIa compositions
US20210070852A1 (en) Anti-IL-23p19 Antibody Formulations
JP6869255B2 (en) Frozen pharmaceutical product and its use

Legal Events

Date Code Title Description
AS Assignment

Owner name: PHOENIX TISSUE REPAIR, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SHIRE HUMAN GENETIC THERAPIES, INC.;REEL/FRAME:052736/0733

Effective date: 20170721

Owner name: SHIRE HUMAN GENETIC THERAPIES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:QUINONES-GARCIA, IGOR;GUEY, LIN;LOWE, KRIS;AND OTHERS;SIGNING DATES FROM 20170101 TO 20170116;REEL/FRAME:052736/0687

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: NOVATEC, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZINSKI, JAMES;REEL/FRAME:055643/0394

Effective date: 20161017

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT, MASSACHUSETTS

Free format text: SECURITY INTEREST;ASSIGNORS:BRIDGEBIO PHARMA, INC.;ORIGIN BIOSCIENCES, INC.;EIDOS THERAPEUTICS, INC.;AND OTHERS;REEL/FRAME:058144/0302

Effective date: 20211117

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: PHOENIX TISSUE REPAIR, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117

Owner name: ADRENAS THERAPEUTICS, INC., NORTH CAROLINA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117

Owner name: QED THERAPEUTICS, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117

Owner name: EIDOS THERAPEUTICS, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117

Owner name: ORIGIN BIOSCIENCES, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117

Owner name: BRIDGEBIO PHARMA, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK TRUST COMPANY, NATIONAL ASSOCIATION, AS SUCCESSOR TO U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:066167/0462

Effective date: 20240117