WO2014113358A1 - Lyophilization process - Google Patents

Lyophilization process Download PDF

Info

Publication number
WO2014113358A1
WO2014113358A1 PCT/US2014/011399 US2014011399W WO2014113358A1 WO 2014113358 A1 WO2014113358 A1 WO 2014113358A1 US 2014011399 W US2014011399 W US 2014011399W WO 2014113358 A1 WO2014113358 A1 WO 2014113358A1
Authority
WO
WIPO (PCT)
Prior art keywords
temperature
protein
hours
pharmaceutical composition
per minute
Prior art date
Application number
PCT/US2014/011399
Other languages
French (fr)
Inventor
Qinghai Zhao
Xia Luo
Jason Bock
Original Assignee
Teva Pharmaceutical Industries Ltd.
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 Teva Pharmaceutical Industries Ltd. filed Critical Teva Pharmaceutical Industries Ltd.
Priority to EP14740889.2A priority Critical patent/EP2945593A4/en
Priority to MX2015008944A priority patent/MX2015008944A/en
Priority to CA2897062A priority patent/CA2897062A1/en
Publication of WO2014113358A1 publication Critical patent/WO2014113358A1/en
Priority to HK16105645.0A priority patent/HK1217633A1/en

Links

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
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • 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
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • 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
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • 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
    • A61K38/38Albumins
    • A61K38/385Serum albumin
    • 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
    • A61K47/26Carbohydrates, 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • 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
    • C07K14/53Colony-stimulating factor [CSF]
    • 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
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • 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
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/01Carboxylic ester hydrolases (3.1.1)
    • C12Y301/01008Cholinesterase (3.1.1.8), i.e. butyrylcholine-esterase
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F26DRYING
    • F26BDRYING SOLID MATERIALS OR OBJECTS BY REMOVING LIQUID THEREFROM
    • F26B5/00Drying solid materials or objects by processes not involving the application of heat
    • F26B5/04Drying solid materials or objects by processes not involving the application of heat by evaporation or sublimation of moisture under reduced pressure, e.g. in a vacuum
    • F26B5/06Drying solid materials or objects by processes not involving the application of heat by evaporation or sublimation of moisture under reduced pressure, e.g. in a vacuum the process involving freezing
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • Lyophilization is widely used to produce and distribute pharmaceutical products, including proteins. However, as the concentration of protein in a lyophilate increases, the time required to reconstitute it increases as well.
  • the present invention provides a process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of: (i) obtaining a solution comprising the protein in one or more containers;
  • the present invention further provides a product produced by the process.
  • the present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes, thereby producing an injectable pharmaceutical composition.
  • the present invention further provides a method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
  • reconstituted solution means a solution produced by dissolving a lyophilized substance in an amount of solvent.
  • the solvent is water for injection (WFI) .
  • the volume of solvent used is the volume of pre-lyophilization solution used to make the lyophilized substance.
  • the volume of solvent used is more than the volume of pre-lyophilization solution used to make the lyophilized substance.
  • the volume of solvent used is 90 percent of than the volume of pre-lyophilization solution used to make the lyophilized substance.
  • the volume of solvent used is less than the volume of pre-lyophilization solution used to make the lyophilized substance.
  • purity refers to the relative amount of a protein that is not disintegrated, monomeric, and in its native conformation. Purity may be measured by size exclusion high performance liquid chromatography (SE-HPLC) , hydrophobic interaction high performance liquid chromatography (HI-HPLC) , sodium dodecylsylfate polyacramide gel electrophoresis (SDS- PAGE) , or any other method known in the art, and may be expressed as a percentage.
  • SE-HPLC size exclusion high performance liquid chromatography
  • HI-HPLC hydrophobic interaction high performance liquid chromatography
  • SDS- PAGE sodium dodecylsylfate polyacramide gel electrophoresis
  • “recommended conditions,” or “recommended storage conditions” as in a sample stored at the recommended conditions means the storage conditions determined to keep the characteristics of the composition within acceptable parameters for the duration of storage. In a specific embodiment, the recommended storage conditions are a temperature of 2-8°C, in an upright position, and/or with
  • any range disclosed herein it is meant that all hundredth, tenth and integer unit amounts within the range are specifically disclosed as part of the invention.
  • 0.01 mg to 50 mg means that 0.02, 0.03 ... 0.09; 0.1, 0.2 ... 0.9; and 1, 2 ... 49 mg unit amounts are included as embodiments of this invention.
  • the present invention provides a process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of:
  • step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 to 10 °C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
  • step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 to 5°C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
  • the initial shelf temperature is from -5 to 10 °C. In an embodiment, the initial shelf temperature is - 5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C, 3°C, 4°C, 5°C,
  • the initial shelf temperature is from -5 to 5°C. In an embodiment, the initial shelf temperature is - 5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C, 3°C, 4°C or 5°C.
  • the shelf is held at the initial shelf temperature for 1.1 to 5 hours. In an embodiment, the shelf is held at the initial shelf temperature for 2 to 5 hours. In an embodiment, the shelf is held at the initial shelf temperature for 2, 3, 4 or 5 hours.
  • the shelf is held at the initial shelf temperature for 2 hours or more. In an embodiment, the shelf is held at the initial shelf temperature for 3 to 5 hours.
  • the temperature in steps (iii) to (viii) is the shelf temperature. In an embodiment, the temperature in steps (iii) to (viii) is the chamber temperature. In an embodiment, step (ii) further comprises pre-cooling the one or more containers. In an embodiment, the pre-cooling is by liquid nitrogen.
  • the containers are pre-cooled to a temperature from -5 to 5°C. In an embodiment, the containers are pre-cooled to -5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C,
  • step (iii) the temperature is reduced at a rate of 0.3°C per minute. In an embodiment, in step (iii) the temperature is reduced at a rate of 0.2°C per minute. In an embodiment, in step (iii) the temperature is reduced at a rate of 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C, 1.8°C, 1.9°C or 2.0°C per minute.
  • step (iii) the temperature is held at the initial freezing temperature for 2.1 to 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for 2.5 to 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5 or 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for more than 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5 or 5.5 hours. In an embodiment, in step (iv) the temperature is increased at a rate of 0.8°C per minute.
  • step (iv) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C, 1.8°C, 1.9°C or 2.0°C per minute.
  • step (iv) the temperature is held at the annealing temperature for 2.1 to 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 3 to 10 hours.
  • step (iv) the temperature is held at the annealing temperature for 5 to 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 1, 1.5, 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for more than 1, 1.5, 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9 or 9.5 hours .
  • step (iv) the temperature is held at the annealing temperature for 5 hours.
  • step (v) the temperature is reduced at a rate of 0.3°C per minute.
  • step (v) the temperature is held at the refreezing temperature for 1.1 to 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for 2 to 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5 or 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for more than 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5 or 5.5 hours. In an embodiment, in step (vi) the temperature is held at the refreezing temperature for 1 hour.
  • step (vii) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C,
  • step (vii) the temperature is held at the primary drying temperature for 36 hours or more.
  • step (vii) the temperature is held at the primary drying temperature for 36 hours.
  • step (vii) the temperature is held at the primary drying temperature for 10 to 29 hours.
  • step (vii) the temperature is held at the primary drying temperature for 29 to 42 hours. In an embodiment, in step (vii) the primary drying temperature is -30°C to -5°C.
  • the process further comprises measuring the temperature of the frozen solution within one or more of the containers during step (vii) , wherein in step (vii) the temperature is held at the primary drying temperature for three hours beyond the time at which the temperature of each measured container is equal to or greater than the primary drying temperature.
  • step (viii) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C,
  • step (viii) the temperature is held at the secondary drying temperature for 4.1, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 or more hours. In an embodiment, in step (viii) the temperature is held at the secondary drying temperature for 15 hours.
  • step (ix) the partial atmospheric pressure is 810 mBar. In an embodiment, in step (ix) the partial atmospheric pressure is 600 T.
  • step (ix) the restoring to partial atmospheric pressure is adding sterile filtered nitrogen to the chamber.
  • the process further comprises the step:
  • the sealing comprises inserting a stopper.
  • the initial freezing temperature is -49 °C to -25°C. In an embodiment, the initial freezing temperature is -47°C to -40°C. In an embodiment, the initial freezing temperature is -45°C to -35°C.
  • the initial freezing temperature is -45°C.
  • the annealing temperature is -19 to -10 °C.
  • the annealing temperature is -30 to -20 °C. In an embodiment, the annealing temperature is -25 to -15°C.
  • the annealing temperature is -19 to -15°C.
  • the annealing temperature is -15 to -10 °C.
  • the annealing temperature is -19°C, -18 °C, -17°C, -16°C, -15°C, -14°C, -13°C, -12°C, -11°C or -10°C.
  • the refreezing temperature is -49 to -25°C.
  • the refreezing temperature is -45 °C. In an embodiment, the refreezing temperature is the same as the initial freezing temperature.
  • the primary drying temperature is -19 °C. to 0°C. In an embodiment, the primary drying temperature is -19°C, -18°C, -17°C, -16°C, -15°C, -14°C, -13°C, -12°C, -11°C, -10°C, -9°C, -8°C, -7°C, -6°C, -5°C, -4°C, -3°C, -2°C, -1°C or 0°C.
  • the primary drying temperature is -10 °C.
  • the secondary drying temperature is 5 to 30 °C. In an embodiment, the secondary drying temperature is 20°C to 30°C. In an embodiment, the secondary drying temperature is 25 °C.
  • step (vi) the pressure is reduced to 100 mT .
  • the solution comprising a protein has a protein concentration from 2 to 250 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration greater than 65 mg/ml.
  • the solution comprising a protein has a protein concentration from 65 to 250 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration from 80 to 120 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration of 100, 150, 200, or 250 mg/ml.
  • the solution comprising a protein has a protein concentration from 100 to 110 mg/ml.
  • each of the one or more containers contains from 0.5 to 2.0 ml of the solution.
  • each of the one or more containers contains 1.0 to 1.2 ml of the solution.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection in 15 minutes or less.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection in 6 minutes or less.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection after one month of storage at recommended conditions in 15 minutes or less.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection after one month of storage at recommended conditions in 6 minutes or less.
  • the solution comprising the protein further comprises 40 to 60 mM phosphate.
  • the solution comprising the protein further comprises 100 to 150 mM mannitol, 20 to 40 mM trehalose, or 0.02 to 0.05 percent polysorbate 80. In an embodiment, the solution comprising the protein further comprises one or more of 100 to 150 mM mannitol, 20 to 40 mM trehalose, or 0.02 to 0.05 percent polysorbate 80. In an embodiment, the solution comprising the protein comprises 50 mM sodium phosphate, 115 mM mannitol, 35 mM trehalose, and 0.03 percent polysorbate 80.
  • the solution comprising the protein comprises 60 mM sodium phosphate, 100 mM mannitol, 30 mM trehalose, and 0.03 percent polysorbate 80.
  • the sodium phosphate comprises 16 mM sodium phosphate monobasic and 34 mM sodium phosphate dibasic.
  • the solution comprising the protein further comprises mannitol and trehalose in a molar ratio of about 3.3 to 1.
  • the process produces a lyophiliz pharmaceutical composition containing a protein, which has residual moisture of 3.0 weight percent or less.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which has a residual moisture of 0.3 weight percent or less.
  • the residual moisture is 3 percent or less.
  • the residual moisture is 0.1, 0.3, 0.4, 0.5, 1 or 2 percent or less.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which is stable under recommended storage conditions for at least six months .
  • the process produces a lyophilized pharmaceutical composition containing a protein, which is stable under recommended storage conditions for at least 18 months .
  • the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 99.0% or more after storage for six months at 2-8°C.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 96.0% or more after storage for six months at 25°C.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 89.0% or more after storage for six months at 40 °C.
  • the process produces a lyophilized pharmaceutical composition containing a protein, which has a 9.6% or less loss in purity after storage for six months.
  • the present invention further provides a product produced by the process.
  • the product reconstitutes in water for injection within 15 minutes.
  • the product reconstitutes in water for injection within 7, 8, 9, 10, 11, 12, 13 or 14 minutes. In an embodiment, the product reconstitutes in water for injection within 6 minutes.
  • the product reconstitutes to a protein concentration from 2 to 250 mg/ml.
  • the product reconstitutes to a protein concentration from 65 to 250 mg/ml. In an embodiment, the product reconstitutes to a protein concentration from 80 to 120 mg/ml.
  • the product reconstitutes to a protein concentration from 100 to 110 mg/ml.
  • the present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes, thereby producing an injectable pharmaceutical composition.
  • the present invention further provides a method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
  • the present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injecting within 6 minutes, thereby producing an injectable pharmaceutical composition.
  • the present invention further provides a method of treating a patient with a therapeutic protein composition, comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 6 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
  • the osmolality of the reconstituted solution is from 250 to 350 mOsm/kg. In an embodiment, the osmolality of the reconstituted solution is from 275 to 325 mOsm/kg. In an embodiment, the osmolality of the reconstituted solution is
  • the reconstituted solution has a pH of 6.9- 7.5. In an embodiment, the reconstituted solution has a pH of 7.1-7.3. In an embodiment, the reconstituted solution has a pH of 7.2.
  • the present invention further provides a sealed package comprising the lyophilized pharmaceutical composition.
  • the sealed package comprises 80-120 mg of protein. In an embodiment, the sealed package comprises 100- 110 mg of protein.
  • the pharmaceutical composition is stable under recommended storage conditions for at least 6-36 months. In an embodiment, the pharmaceutical composition is stable under recommended storage conditions for at least 6 months . In an embodiment, the pharmaceutical composition is stable under recommended storage conditions for at least 9, 12, 18, 24, 30, or 36 months. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 17. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 18. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 19.
  • the container is a vial.
  • the vial is made of glass. In an embodiment, the vial is made of USP Type 1 glass. In an embodiment, the container is made of flint glass.
  • the vial is closed by a stopper.
  • the stopper is sealed by an aluminum seal.
  • the stopper has a FLUROTECTM coating.
  • the volume of the vial is from 1.5 to 5 ml. In an embodiment, the volume of the vial is 3 ml.
  • the sealing comprises inserting a stopper.
  • the stopper is elastomeric.
  • the stopper comprises rubber.
  • the stopper comprises butyl rubber.
  • the stopper is halogenated.
  • the stopper comprises chlorobutyl rubber.
  • the stopper is coated with a coating. In an embodiment, the coating is FLUROTECTM.
  • the protein is a fusion protein.
  • the fusion protein is a fusion of human serum albumin and a therapeutic protein.
  • the therapeutic protein is one of: Interferon alpha (Interferon alfa-2b; Interferon alfa-2a; recombinant; Interferon alfa-nl; Interferon alfan3; Peginterferon alpha-2b; Ribavirin and interferon alfa-2b; Interferon alfacon-1; interferon consensus; YM 643; CIFN; interferonalpha consensus; recombinant methionyl consensus interferon; recombinant consensus interferon; CGP 35269; RO 253036; RO 258310; Intron A; Pegintron; Oif; Omniferon; Pegomniferon; Veldona; Pegrebetron; Roferon A; Wellferon; Alferon N/Ldo; Rebetron; Altemol; Viraferonpeg
  • BNP brain natriuretic peptide
  • LPP Long-acting natriuretic peptide
  • VDP Vessel Dialator
  • KUP Kaliuretic Peptide
  • CNP C-type Natriuretic Peptide
  • DNP Dendroaspis natriuretic peptide
  • Beta defensin-2 beta defensin 4; SAP1; DEFB2; HBD-2;
  • the fusion protein is a fusion of human serum albumin and butyryl-cholinesterase .
  • the fusion protein is Composition 1.
  • the fusion protein is a fusion of human serum albumin and human growth hormone. Examples of such proteins which may be used in embodiments of the invention are disclosed in U.S. Patent Application Publication Nos. US 2011/0002888 and US 2009/0029914, and U.S. Patents Nos.
  • the fusion protein is Composition 2. In an embodiment, the fusion protein is Composition 3. In an embodiment, the fusion protein is Composition 4. In an embodiment, the fusion protein is Composition 5. In an embodiment, the protein is a therapeutic protein. In an embodiment, the protein is an antibody. In an embodiment, the protein is not an antibody.
  • the protein is one of: Insulin; Humulin;
  • Insulin lispro Insulin lispro; Humalog (lispro) ; Isophane insulin; NPH;
  • Insulin detemir Levemir (detemir) ; Insulin glargine; Lantus
  • Pramlintide acetate Symlin; Growth hormone (GH) ; somatotropin; genotropin; humatrope; norditropin;
  • Recominate ReFacto; Factor IX; Benefix; Antithromin III (AT- III); Thrombate III; Protein C concentrate; Ceprotin; ⁇ -
  • Proteinase inhibitor Aralast; Prolastin; Lactase; Lactaid;
  • Pancreatic enzymes lipase, amylase, protease
  • Arco-Lase Arco-Lase
  • Adenosine deaminase (pegademase bovine, PEG-ADA) ; Adagen; Pooled immunoglobulins; Octagam; Human albumin; Albumarc;
  • Darbepoetin- Aranesp; Filgrastim (granulocyte colony stimulating factor; G-CS F) ; Neupogen; Pegfilgrastim (Peg-G- CSF) ; Neulasta; Sargramostim (granulocytemacrophage colony stimulating factor; GM-CS F) ; Leukine; Oprelvekin (interleukinll ; IL11); Neumega; Human follicle-stimulating hormone (FSH) ; Gonal-F; Follistim; Human chorionic gonadotropin (HCG) ; Ovidrel; Luveris; Type I alpha-interferon; interferon alfacon 1; consensus interferon; Infergen; Interferon- 2a (IFN 2a); Roferon-A; Peglnterferon- 2a;
  • Pegasys Interferon- 2b (IFN 2b); Intron A; Peglnterferon- 2b;
  • Interferon-ylb Actimmune; Aldesleukin (interleukin 2 (IL2); epidermal thymocyte activating factor; ETAF) ;
  • Alteplase tissue plasminogen activator; tPA
  • Reteplase (deletion mutein of tPA) ; Retavase;
  • Tenecteplase TNKase; Urokinase; Abbokinase; Factor Vila;
  • Drotrecogin- activate protein C
  • Xigris Salmon calcitonin
  • Fortical Miacalcin
  • Teriparatide human parathyroid hormone residues 1-34
  • Forteo Exenatide; Byetta;
  • Octreotide Sandostatin; Dibotermin- (recombinant human bone morphogenic protein 2; rhBMP2 ) ; Infuse; Recombinant human bone morphogenic protein 7 (rhBMP7); Osteogenic protein 1; Histrelin acetate (gonadotropin releasing hormone; GnRH) ;
  • KGF KGF
  • kepivance kepivance
  • Becaplermin platelet-derived growth factor
  • PDGF PDGF
  • Streptokinase Streptase; Streptase; Anistreplase (anisoylated plasminogen streptokinase activator complex; APSAC) ; Eminase;
  • Bevacizumab Avastin; Cetuximab; Erbitux; Panitumumab;
  • Vectibix Alemtuzumab; Campath; Rituximab; Rituxan; Trastuzumab; Herceptin; Abatacept; Orencia; Anakinra; Antril;
  • HPV vaccine Gardasil; OspA; LYMErix; Anti-Rhesus (Rh) immunoglobulin G; Rhophylac; Recombinant purified protein derivative (DPPD) ; Glucagon; GlucaGen; Growth hormone releasing hormone (GHRH) ; Geref ; Secretin; ChiRhoStim (human peptide) , SecreFlo (porcine peptide) ; Thyroid stimulating hormone (TSH) ; thyrotropin; Capromab pendetide; ProstaScint; Indium-lll-octreotide; OctreoScan; Satumomab pendetide; OncoScint; Arcitumomab; CEA-scan; Nofetumomab; Verluma; Apcitide; Acutect; Imciromab pentetate; Myoscint; Technetium fanolesomab; NeutroSpec; HIV
  • Composition 1 a recombinant protein composed of the mature form of recombinant human serum albumin (rHSA) fused at its amino terminus to the carboxy-terminus of a mutated human butyrylcholinesterase (BChE) , was used to develop a novel lyophilization process and a suitable formulation.
  • rHSA human serum albumin
  • BChE mutated human butyrylcholinesterase
  • Composition 1 50 mg/mL in PMTT (which comprises 10 mM phosphate, 200 mM mannitol, 60 mM trehalose and 0.01% PS80, at pH 7.2)) at six target sodium chloride concentrations (5 mM, 10 mM, 20 mM, 50 mM, 80 mM, 120 mM) .
  • PMTT which comprises 10 mM phosphate, 200 mM mannitol, 60 mM trehalose and 0.01% PS80, at pH 7.2
  • Vials of each sample were incubated at 25 °C for 5 days. Samples were removed from incubation after 5 days. The samples were compared to the 0 day and 0 mM sodium chloride controls by visual inspection and SE-HPLC.
  • Buffer controls containing 5 mM, 10 mM, 20 mM, 50 mM, 80 mM, and 120 mM sodium chloride were measured for conductivity.
  • Buffer controls containing 10 mM, 20 mM, 30 mM, 40 mM, 50 mM, and 60 mM phosphate were measured for conductivity.
  • Composition 1 100 mg/mL in 200 mM mannitol, 60 mM trehalose, 0.03% PS80, pH 7.2
  • target phosphate concentrations 10 mM, 20 mM, 30 mM, 40 mM, 50 mM, 60 mM
  • Vials of each sample were incubated at 25 °C for 5 days. Samples were removed from incubation after 3 and 5 days. The samples were compared to the 0 day controls by visual inspection and SE-HPLC. Buffer controls were measured for conductivity.
  • composition 1 100 mg/mL in 10 mM phosphate, 200 mM mannitol, 60 mM trehalose, pH 7.2
  • target PS80 concentrations 0.01%, 0.05%, 0.1%, and 0.2%) .
  • the samples were incubated at 2-8°C and 25°C for 1,
  • PS80 concentration (% ) PS80 concentration (°/ '>)
  • Formulation buffers containing varying concentrations of phosphate (40 mM, 50 mM and 60mM) , mannitol (60-200 mM) , trehalose (18-60 mM) and 0.03% PS80 were made by combining varying amounts of 500 mM phosphate (pH 7.2) stock solution, 500 mM mannitol stock solution and 200 mM trehalose stock solution, while keeping the ratio of trehalose to mannitol the same as PMTT. The osmolality of each buffer was tested and compared to the osmolality of PMTT (Table 6) .
  • Buffers with osmolality approximately equal to 300 mOsm/kg were made, and conductivity and osmolality were measured for the buffers and for Composition 1 (100 mg/mL in PMTT) (Table 7) .
  • Table 7 Proto-formulation Buffer Measurements (Bolded lines indicate P50MTT and P60MTT)
  • Composition 1 100 mg/mL in PMTT
  • PMTT PMTT alone
  • Composition 1 at 100 mg/mL contributes approximately 31.5 mOsm/kg to osmolality.
  • Targeting an osmolality of 300 mOsm/kg two formulations were selected: P50MTT (267 mOsm/kg) , and P60MTT (268 mOsm/kg) .
  • P50MTT comprises 50 mM sodium phosphate, 115 mM mannitol, 35 mM trehalose and 0.03% PS80, at pH 7.2
  • P60MTT comprises 60 mM phosphate, 100 mM mannitol, and 30 mM trehalose and 0.03% PS80, at pH 7.2. Measurements were performed for Composition 1 (100 mg/mL) in the new P50MTT and P60MTT formulations (Table 8) .
  • the pre-formulation studies were executed to determine potential formulation candidates for the lyophilization formulation of the concentrated product. Previous studies showed that Composition 1 was affected by concentration dependent aggregation, suggesting that aggregation is a major degradation pathway. In response, the ionic strength study was conducted to determine if increasing the ionic strength of the formulation buffer would have an effect on reducing aggregation. The results of the study demonstrate that there is a significant ionic strength effect, and in the higher ionic strength formulation there was a significant reduction in dose dependent aggregation at a protein concentration of 100 mg/ml.
  • Mannitol and trehalose concentrations in the candidate formulations were modified to target an osmolality of 300 mOsm/kg, while maintaining the ratio between mannitol and trehalose as established during development of the previous PMTT formulation.
  • Two proto-formulations , P50MTT and P60MTT, were selected for additional studies .
  • composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) . Samples were frozen for 2-16 hours at ⁇ -65°C and then thawed for 3 hours at room temperature. Samples were collected after 1, 2, 4, 6 and 10 complete cycles of freezing and thawing. Samples were compared to the 0 point by visual inspection and SE-HPLC. Select samples were also tested by SDS-PAGE and potency analysis.
  • composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) . Samples were shaken horizontally at 150 rpm. Samples were incubated at 2-8°C and 25°C from 0 to 24 hours. Samples were compared to the 0 point by visual inspection, SE-HPLC and HI-HPLC.
  • Composition 1 (101.6 mg/mL P50MTT and 100.8 mg/mL in P60MTT) was used for this study. Samples were incubated at 2-8°C and 25 °C for 6 days. Samples were removed from incubation after 1,
  • composition 1 in both P50MTT and P60MTT had no change in SE-HPLC and HI-HPLC purity (after incubation in 2-8°C and 25°C for 6 days. This is a significant change from the prior formulation (PMTT) , which had an approximate
  • composition 1 at 100 mg/mL is stable at 2-8°C and 25°C for up to 6 days in both P50MTT and P60MTT formulations.
  • Composition 1 in P50MTT and in P60MTT was not sensitive to freeze-thaw or shaking effects.
  • TBU Lyophilization Cycle An initial lyophilization cycle evaluation was carried out using Composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) .
  • the TBU lyophilization cycle is summarized in Table 12.
  • Post-lyophilization tests include visual inspection pre- and post-reconstitution and residual moisture content analysis. 0-12 hour post-reconstitution samples were analyzed by SE-HPLC and HI-HPLC. Selected samples were also tested by potency analysis. Table 12: TBU Lyophilization Cycle
  • the lyophilization products were pharmaceutically acceptable cakes (white to off-white in color and intact) .
  • TBU lyophilization cycle conditions are summarized as follows :
  • Composition 1 (100 mg/mL in P50MTT) was lyophilized using the TBU cycle and used for the long term stability study.
  • lyophilization cycle evaluation was carried out using Composition 1 (103 mg/mL in P50MTT) .
  • samples were analyzed by visual inspection, moisture content analysis, HI- HPLC and SE-HPLC.
  • the lyophilization cycle evaluation was carried out using Composition 1 (103 mg/mL in P50MTT) . Visual inspection, residual moisture content measurement, SE-HPLC and HI-HPLC purity analysis were performed. See Table 13 for detailed information pertaining to the various lyophilization cycle parameters .
  • the results of the lyophilization cycle evaluation further confirm that the TBU lyophilization cycle is more appropriate for Composition 1.
  • the data suggests that the TBU cycle produces pharmaceutically acceptable cakes, with the lowest residual moisture (0.3%) compared to the other lyophilization cycles tested during the evaluation (Table 14) .
  • Composition 1 (100 mg/ml in P50MTT after reconstitution with 1.1 ml of WFI ) 0 month was used for the pre- and post- lyophilization analysis. Time points were 0, 4, 8 and 12 hours. Visual inspection was performed prior to reconstitution. Reconstitution time was recorded. Post- reconstitution, samples were analyzed by visual inspection, pH, osmolality, concentration measurement, SE-HPLC, SDS-PAGE, potency analysis and free thiol content (Table 15) .
  • the formulation containing a lower concentration of salt for the lyophilization process. Therefore, the P50MTT formulation was selected as the final concentrated product formulation and was used for the lyophilization formulation evaluation and long term stability program.
  • the TBU lyophilization cycle is appropriate for the lyophilization of Composition 1.
  • the results of the low thermal analysis study indicate that the parameters of the TBU lyophilization cycle meet the minimum temperature requirements and the pre and post lyophilization results suggest that there is no change in protein quality.
  • Cakes produced using the TBU lyophilization cycle are white to off-white in color and are intact, which is considered to be pharmaceutically acceptable.
  • composition 1 drug substances at 100 mg/mL with these two formulations are stable at 2-8°C and 25°C for up to 6 days, and are neither sensitive to freeze-thaw nor shaking effects, which could support the lyophilization process. There is no significant impact on the product quality by post-lyophilization . Overall, the two formulations are comparable in terms of the product quality and stability.
  • the P50MTT formulation was selected as a formulation candidate for an additional lyophilization cycle evaluation and long term stability study, due to its lower ionic strength compared to P60MTT, which might negatively impact lyophilization process and lyophilization product.
  • Composition 1 (100 mg/ml in P50MTT after reconstitution with 1.1 ml of WFI) was used for the stability program study.
  • the lyophilized product was stored at 2-8°C, 25°C and 40°C.
  • Samples stored under recommended conditions are stable for 24 months . Samples stored under recommended conditions are stable for 36 months .
  • Composition 2 known as NeugraninTM, is a protein derived from the direct genetic fusion of the genes for Granulocyte Colony
  • GCSF Stimulating Factor
  • human serum albumin The TBU lypholization cycle applied to Composition 2 (15 mg/ml) is summarized in Table 20. The lyophilized product was stored at 2-8°C, 25°C and 40°C. Results
  • Samples stored under recommended conditions are stable for 24 months .
  • Samples stored under recommended conditions are stable for 36 months .
  • Example 4 Long Term Stability Testing of Composition 3 Methods
  • Composition 3 known as AlbuferonTM-Beta, is a product derived from the direct genetic fusion of the genes for human interferon-beta (IFN-beta) and human serum albumin.
  • the TBU lypholization cycle applied to Composition 3 (2.0 mg/ml) is summarized in Table 24.
  • the lyophilized product was stored at 2-8°C, 25°C and 40°C.
  • Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
  • WC White Cake
  • CPF Clear, pale yellow solution, essentially free from foreign particulate matter
  • Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
  • Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
  • Samples stored under recommended conditions are stable under the recommended conditions for 18 months. Samples stored under recommended conditions are stable for 24 months .
  • Samples stored under recommended conditions are stable for 36 months .
  • Composition 4 known as AlbutropinTM, is a contiguous protein comprised of human serum albumin (HSA) and recombinant growth hormone (rHGH) with the mature form of HSA genetically fused at its C-terminus to the N-terminus of the mature form of rHGH.
  • HSA human serum albumin
  • rHGH recombinant growth hormone
  • WC White Cake
  • CPF Clear, pale yellow, essentially free from foreign particulate matter
  • NT 'Result is an average of 3 vials (1 each from beginning, middle, and end)
  • WC White Cake
  • CPF Clear, pale yellow, essentially free from foreign particulate matter
  • NT Not tested 'Result is an average of 3 vials (1 each from beginning, middle, and end)
  • Samples stored under recommended conditions are stable under the recommended conditions for 18 months. Samples stored under recommended conditions are stable for 24 months .
  • Samples stored under recommended conditions are stable for 36 months .
  • Composition 5 known as CardevaTM, is a recombinant human B- type natriuretic peptide (BNP) serum albumin fusion protein.
  • BNP B- type natriuretic peptide
  • a more concentrated formulation can have significant advantages, including increasing convenience (since fewer or smaller vials are required to contain a given dose) and reducing the injection bolus necessary for a given dose. However, it is not always routine and often very difficult to increase the concentration of a peptide formulation.

Abstract

The present invention provides a process for producing a lyophilized pharmaceutical composition containing a protein. The present invention further provides a product produced by the process. The present invention further provides a process for producing an injectable pharmaceutical composition. The present invention further provides a method of treating a patient with a therapeutic protein composition.

Description

LYOPHILIZATION PROCESS
This application claims priority of U.S. Provisional Application Nos . 61/752,797, filed January 15, 2013, and 61/784,538, filed March 14, 2013, the contents of which are hereby incorporated by reference in their entirety.
Throughout this application, various publications are referenced by author and publication date. Full citations for these publications may be found at the end of the specification immediately preceding the claims. The disclosures of these publications are hereby incorporated by reference into this application to describe more fully the art to which this invention pertains.
Background of the Invention
Lyophilization is widely used to produce and distribute pharmaceutical products, including proteins. However, as the concentration of protein in a lyophilate increases, the time required to reconstitute it increases as well.
Summary of the Invention
The present invention provides a process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of: (i) obtaining a solution comprising the protein in one or more containers;
(ii) placing the one or more containers within a chamber of a lyophilizing unit;
(iii) reducing the temperature to an initial freezing temperature of -60°C to -25°C at a rate of 0.2°C to
2.0°C per minute, and holding the temperature at the initial freezing temperature for 1 to 6 hours to form a frozen solution;
(iv) increasing the temperature to an annealing temperature of the frozen solution of -30 °C to -10 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the annealing temperature for 1 to 10 hours;
(v) reducing the temperature to a refreezing temperature of -60 to -25 at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the refreezing temperature for 1 to 6 hours;
(vi) reducing the pressure of the chamber to 50 to 500 mT, and continuing to hold the temperature at the refreezing temperature for an additional 0 to 4 hours;
(vii) increasing the temperature to a primary drying temperature of -30 to -10 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the primary drying temperature for 10 to 72 hours; (viii ) increasing the temperature to a secondary drying temperature of 5 to 30 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the secondary drying temperature for 2 to 25 hours; and (ix) increasing the pressure of the chamber to partial atmospheric pressure.
The present invention further provides a product produced by the process.
The present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes, thereby producing an injectable pharmaceutical composition.
The present invention further provides a method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
Detailed Description of the Invention
As used herein, and unless stated otherwise, each of the following terms shall have the definition set forth below.
As used herein, "reconstituted solution" means a solution produced by dissolving a lyophilized substance in an amount of solvent. In an embodiment, the solvent is water for injection (WFI) . In an embodiment, the volume of solvent used is the volume of pre-lyophilization solution used to make the lyophilized substance. In an embodiment, the volume of solvent used is more than the volume of pre-lyophilization solution used to make the lyophilized substance. In an embodiment, the volume of solvent used is 90 percent of than the volume of pre-lyophilization solution used to make the lyophilized substance. In an embodiment, the volume of solvent used is less than the volume of pre-lyophilization solution used to make the lyophilized substance.
As used herein, "purity, " as in purity of a pharmaceutical composition, refers to the relative amount of a protein that is not disintegrated, monomeric, and in its native conformation. Purity may be measured by size exclusion high performance liquid chromatography (SE-HPLC) , hydrophobic interaction high performance liquid chromatography (HI-HPLC) , sodium dodecylsylfate polyacramide gel electrophoresis (SDS- PAGE) , or any other method known in the art, and may be expressed as a percentage. As used herein, "recommended conditions," or "recommended storage conditions" as in a sample stored at the recommended conditions, means the storage conditions determined to keep the characteristics of the composition within acceptable parameters for the duration of storage. In a specific embodiment, the recommended storage conditions are a temperature of 2-8°C, in an upright position, and/or with limited exposure to light.
By any range disclosed herein, it is meant that all hundredth, tenth and integer unit amounts within the range are specifically disclosed as part of the invention. Thus, for example, 0.01 mg to 50 mg means that 0.02, 0.03 ... 0.09; 0.1, 0.2 ... 0.9; and 1, 2 ... 49 mg unit amounts are included as embodiments of this invention.
The specific embodiments and examples described herein are illustrative, and many variations can be introduced on these embodiments and examples without departing from the spirit of the disclosure or from the scope of the appended claims. Elements and/or features of different illustrative embodiments and/or examples may be combined with each other and/or substituted for each other within the scope of this disclosure and appended claims.
The present invention provides a process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of:
(i) obtaining a solution comprising the protein in one or more containers;
(ii) placing the one or more containers within a chamber of a lyophilizing unit;
(iii) reducing the temperature to an initial freezing temperature of -60°C to -25°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the initial freezing temperature for 1 to 6 hours to form a frozen solution;
(iv) increasing the temperature to an annealing temperature of the frozen solution of -30 °C to -10 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the annealing temperature for 1 to 10 hours;
(v) reducing the temperature to a refreezing temperature of -60 to -25 at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the refreezing temperature for 1 to 6 hours; (vi) reducing the pressure of the chamber to 50 to 500 mT, and continuing to hold the temperature at the refreezing temperature for an additional 0 to 4 hours;
(vii) increasing the temperature to a primary drying temperature of -30 to -10 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the primary drying temperature for 10 to 72 hours;
(viii) increasing the temperature to a secondary drying temperature of 5 to 30 °C at a rate of 0.2 to 2.0°C per minute, and holding the temperature at the secondary drying temperature for 2 to 25 hours; and
(ix) increasing the pressure of the chamber to partial atmospheric pressure.
In an embodiment, step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 to 10 °C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
In an embodiment, step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 to 5°C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
In an embodiment, the initial shelf temperature is from -5 to 10 °C. In an embodiment, the initial shelf temperature is - 5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C, 3°C, 4°C, 5°C,
6°C, 7°C, 8°C, 9°C or 10°C. In an embodiment, the initial shelf temperature is from -5 to 5°C. In an embodiment, the initial shelf temperature is - 5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C, 3°C, 4°C or 5°C.
In an embodiment, the shelf is held at the initial shelf temperature for 1.1 to 5 hours. In an embodiment, the shelf is held at the initial shelf temperature for 2 to 5 hours. In an embodiment, the shelf is held at the initial shelf temperature for 2, 3, 4 or 5 hours.
In an embodiment, the shelf is held at the initial shelf temperature for 2 hours or more. In an embodiment, the shelf is held at the initial shelf temperature for 3 to 5 hours.
In an embodiment, the temperature in steps (iii) to (viii) is the shelf temperature. In an embodiment, the temperature in steps (iii) to (viii) is the chamber temperature. In an embodiment, step (ii) further comprises pre-cooling the one or more containers. In an embodiment, the pre-cooling is by liquid nitrogen.
In an embodiment, the containers are pre-cooled to a temperature from -5 to 5°C. In an embodiment, the containers are pre-cooled to -5°C, -4°C, -3°C, -2°C, -1°C, 0°C, 1°C, 2°C,
3°C, 4°C, 5°C, 6°C, 7°C, 8°C, 9°C or 10°C.
In an embodiment, in step (iii) the temperature is reduced at a rate of 0.3°C per minute. In an embodiment, in step (iii) the temperature is reduced at a rate of 0.2°C per minute. In an embodiment, in step (iii) the temperature is reduced at a rate of 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C, 1.8°C, 1.9°C or 2.0°C per minute.
In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for 2.1 to 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for 2.5 to 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5 or 6 hours. In an embodiment, in step (iii) the temperature is held at the initial freezing temperature for more than 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5 or 5.5 hours. In an embodiment, in step (iv) the temperature is increased at a rate of 0.8°C per minute. In an embodiment, in step (iv) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C, 1.8°C, 1.9°C or 2.0°C per minute. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 2.1 to 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 3 to 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 5 to 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for 1, 1.5, 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 hours. In an embodiment, in step (iv) the temperature is held at the annealing temperature for more than 1, 1.5, 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9 or 9.5 hours .
In an embodiment, in step (iv) the temperature is held at the annealing temperature for 5 hours.
In an embodiment, in step (v) the temperature is reduced at a rate of 0.3°C per minute.
In an embodiment, in step (v) the temperature is held at the refreezing temperature for 1.1 to 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for 2 to 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5, 5.5 or 6 hours. In an embodiment, in step (v) the temperature is held at the refreezing temperature for more than 2, 2.1, 2.5, 3, 3.5, 4, 4.5, 5 or 5.5 hours. In an embodiment, in step (vi) the temperature is held at the refreezing temperature for 1 hour.
In an embodiment, in step (vii) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C, 0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C,
1.8°C, 1.9°C or 2.0°C per minute.
In an embodiment, in step (vii) the temperature is held at the primary drying temperature for 36 hours or more.
In an embodiment, in step (vii) the temperature is held at the primary drying temperature for 36 hours.
In an embodiment, in step (vii) the temperature is held at the primary drying temperature for 10 to 29 hours.
In an embodiment, in step (vii) the temperature is held at the primary drying temperature for 29 to 42 hours. In an embodiment, in step (vii) the primary drying temperature is -30°C to -5°C.
In an embodiment, the process further comprises measuring the temperature of the frozen solution within one or more of the containers during step (vii) , wherein in step (vii) the temperature is held at the primary drying temperature for three hours beyond the time at which the temperature of each measured container is equal to or greater than the primary drying temperature.
In an embodiment, in step (viii) the temperature is increased at a rate of 0.2°C, 0.3°C, 0.4°C, 0.5°C, 0.6°C, 0.7°C, 0.8°C,
0.9°C, 1.0°C, 1.1°C, 1.2°C, 1.3°C, 1.4°C, 1.5°C, 1.6°C, 1.7°C, 1.8°C, 1.9°C or 2.0°C per minute..
In an embodiment, in step (viii) the temperature is held at the secondary drying temperature for 4.1, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 or more hours. In an embodiment, in step (viii) the temperature is held at the secondary drying temperature for 15 hours.
In an embodiment, in step (ix) the partial atmospheric pressure is 810 mBar. In an embodiment, in step (ix) the partial atmospheric pressure is 600 T.
In an embodiment, in step (ix) the restoring to partial atmospheric pressure is adding sterile filtered nitrogen to the chamber. In an embodiment, the process further comprises the step:
(x) sealing the containers.
In an embodiment, the sealing comprises inserting a stopper.
In an embodiment, the initial freezing temperature is -49 °C to -25°C. In an embodiment, the initial freezing temperature is -47°C to -40°C. In an embodiment, the initial freezing temperature is -45°C to -35°C.
In an embodiment, the initial freezing temperature is -45°C.
In an embodiment, the annealing temperature is -19 to -10 °C.
In an embodiment, the annealing temperature is -30 to -20 °C. In an embodiment, the annealing temperature is -25 to -15°C.
In an embodiment, the annealing temperature is -19 to -15°C.
In an embodiment, the annealing temperature is -15 to -10 °C.
In an embodiment, the annealing temperature is -19°C, -18 °C, -17°C, -16°C, -15°C, -14°C, -13°C, -12°C, -11°C or -10°C. In an embodiment, the refreezing temperature is -49 to -25°C.
In an embodiment, the refreezing temperature is -45 °C. In an embodiment, the refreezing temperature is the same as the initial freezing temperature. In an embodiment, the primary drying temperature is -19 °C. to 0°C. In an embodiment, the primary drying temperature is -19°C, -18°C, -17°C, -16°C, -15°C, -14°C, -13°C, -12°C, -11°C, -10°C, -9°C, -8°C, -7°C, -6°C, -5°C, -4°C, -3°C, -2°C, -1°C or 0°C.
In an embodiment, the primary drying temperature is -10 °C.
In an embodiment, the secondary drying temperature is 5 to 30 °C. In an embodiment, the secondary drying temperature is 20°C to 30°C. In an embodiment, the secondary drying temperature is 25 °C.
In an embodiment, in step (vi) the pressure is reduced to 100 mT .
In an embodiment, the solution comprising a protein has a protein concentration from 2 to 250 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration greater than 65 mg/ml.
In an embodiment, the solution comprising a protein has a protein concentration from 65 to 250 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration from 80 to 120 mg/ml. In an embodiment, the solution comprising a protein has a protein concentration of 100, 150, 200, or 250 mg/ml.
In an embodiment, the solution comprising a protein has a protein concentration from 100 to 110 mg/ml. In an embodiment, in step (i) each of the one or more containers contains from 0.5 to 2.0 ml of the solution.
In an embodiment, each of the one or more containers contains 1.0 to 1.2 ml of the solution. In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection in 15 minutes or less.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection in 6 minutes or less.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection after one month of storage at recommended conditions in 15 minutes or less.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which reconstitutes in water for injection after one month of storage at recommended conditions in 6 minutes or less. In an embodiment, the solution comprising the protein further comprises 40 to 60 mM phosphate.
In an embodiment, the solution comprising the protein further comprises 100 to 150 mM mannitol, 20 to 40 mM trehalose, or 0.02 to 0.05 percent polysorbate 80. In an embodiment, the solution comprising the protein further comprises one or more of 100 to 150 mM mannitol, 20 to 40 mM trehalose, or 0.02 to 0.05 percent polysorbate 80. In an embodiment, the solution comprising the protein comprises 50 mM sodium phosphate, 115 mM mannitol, 35 mM trehalose, and 0.03 percent polysorbate 80. In an embodiment, the solution comprising the protein comprises 60 mM sodium phosphate, 100 mM mannitol, 30 mM trehalose, and 0.03 percent polysorbate 80. In an embodiment, the sodium phosphate comprises 16 mM sodium phosphate monobasic and 34 mM sodium phosphate dibasic.
In an embodiment, the solution comprising the protein further comprises mannitol and trehalose in a molar ratio of about 3.3 to 1. In an embodiment, the process produces a lyophiliz pharmaceutical composition containing a protein, which has residual moisture of 3.0 weight percent or less.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which has a residual moisture of 0.3 weight percent or less.
In an embodiment, the residual moisture is 3 percent or less.
In an embodiment, the residual moisture is 0.1, 0.3, 0.4, 0.5, 1 or 2 percent or less. In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which is stable under recommended storage conditions for at least six months .
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which is stable under recommended storage conditions for at least 18 months .
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 99.0% or more after storage for six months at 2-8°C.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 96.0% or more after storage for six months at 25°C.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which has a purity of 89.0% or more after storage for six months at 40 °C.
In an embodiment, the process produces a lyophilized pharmaceutical composition containing a protein, which has a 9.6% or less loss in purity after storage for six months. The present invention further provides a product produced by the process. In an embodiment, the product reconstitutes in water for injection within 15 minutes.
In an embodiment, the product reconstitutes in water for injection within 7, 8, 9, 10, 11, 12, 13 or 14 minutes. In an embodiment, the product reconstitutes in water for injection within 6 minutes.
In an embodiment, the product reconstitutes to a protein concentration from 2 to 250 mg/ml.
In an embodiment, the product reconstitutes to a protein concentration from 65 to 250 mg/ml. In an embodiment, the product reconstitutes to a protein concentration from 80 to 120 mg/ml.
In an embodiment, the product reconstitutes to a protein concentration from 100 to 110 mg/ml. The present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes, thereby producing an injectable pharmaceutical composition.
The present invention further provides a method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient. The present invention further provides a process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process, and reconstituting the lyophilized pharmaceutical composition with water for injecting within 6 minutes, thereby producing an injectable pharmaceutical composition. The present invention further provides a method of treating a patient with a therapeutic protein composition, comprising a protein produced, reconstituting the lyophilized pharmaceutical composition with water for injection within 6 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
In an embodiment, the osmolality of the reconstituted solution is from 250 to 350 mOsm/kg. In an embodiment, the osmolality of the reconstituted solution is from 275 to 325 mOsm/kg. In an embodiment, the osmolality of the reconstituted solution is
300 mOsm/kg.
In an embodiment, the reconstituted solution has a pH of 6.9- 7.5. In an embodiment, the reconstituted solution has a pH of 7.1-7.3. In an embodiment, the reconstituted solution has a pH of 7.2.
The present invention further provides a sealed package comprising the lyophilized pharmaceutical composition.
In an embodiment, the sealed package comprises 80-120 mg of protein. In an embodiment, the sealed package comprises 100- 110 mg of protein.
In an embodiment, the pharmaceutical composition is stable under recommended storage conditions for at least 6-36 months. In an embodiment, the pharmaceutical composition is stable under recommended storage conditions for at least 6 months . In an embodiment, the pharmaceutical composition is stable under recommended storage conditions for at least 9, 12, 18, 24, 30, or 36 months. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 17. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 18. In a specific embodiment, the pharmaceutical composition meets or exceeds 1, 2, 3, 4, 5 or more of the stability parameters set forth in Table 19.
In an embodiment, the container is a vial.
In an embodiment, the vial is made of glass. In an embodiment, the vial is made of USP Type 1 glass. In an embodiment, the container is made of flint glass.
In an embodiment, the vial is closed by a stopper. In an embodiment, the stopper is sealed by an aluminum seal. In an embodiment, the stopper has a FLUROTEC™ coating.
In an embodiment, the volume of the vial is from 1.5 to 5 ml. In an embodiment, the volume of the vial is 3 ml.
In an embodiment, the sealing comprises inserting a stopper. In an embodiment, the stopper is elastomeric. In an embodiment, the stopper comprises rubber. In an embodiment, the stopper comprises butyl rubber. In an embodiment, the stopper is halogenated. In an embodiment, the stopper comprises chlorobutyl rubber. In an embodiment, the stopper is coated with a coating. In an embodiment, the coating is FLUROTEC™.
In an embodiment, the protein is a fusion protein. In an embodiment, the fusion protein is a fusion of human serum albumin and a therapeutic protein. In an embodiment, the therapeutic protein is one of: Interferon alpha (Interferon alfa-2b; Interferon alfa-2a; recombinant; Interferon alfa-nl; Interferon alfan3; Peginterferon alpha-2b; Ribavirin and interferon alfa-2b; Interferon alfacon-1; interferon consensus; YM 643; CIFN; interferonalpha consensus; recombinant methionyl consensus interferon; recombinant consensus interferon; CGP 35269; RO 253036; RO 258310; Intron A; Pegintron; Oif; Omniferon; Pegomniferon; Veldona; Pegrebetron; Roferon A; Wellferon; Alferon N/Ldo; Rebetron; Altemol; Viraferonpeg; Pegasys; Viraferon; Virafon; Ampligen; Infergen; Infarex; Oragen) Atrial natriuretic peptide (ANP; atrial natriuretic factor; ANF) B-type natriuretic peptide
(BNP, brain natriuretic peptide) Long-acting natriuretic peptide (LANP; proANP (31-67) ) ; Vessel Dialator (VDP; proANP- (79-98)); Kaliuretic Peptide (KUP; proANP- (99-126) ) ; C-type Natriuretic Peptide (CNP) ; Dendroaspis natriuretic peptide (DNP) ; Beta defensin-2 (beta defensin 4; SAP1; DEFB2; HBD-2;
DEFB-2; DEFB102; skin-antimicrobial peptide 1); Human chemokine HCC-1 (ckBeta-1; CKB-1; HWFBD) ; Fractalkine (neurotactin; chemokine CX3C) ; Oxyntomodulin; Killer Toxin; Killer Toxin Peptide (KP) ; TIMP-4 (Tissue Inhibitor of Metalloprotease) ; PYY (Peptide YY, including PYY 3-36 (amino acid residues 31-64 of full length PYY, amino acid residues 3- 36 of mature PYY) and also including PYY (3-36) (G9R) ; Adrenomedullin; Ghrelin; Calcitonin gene-related peptide (CGRP) ; Insulin-like growth factor-1 (Mecasermin; Somazon; IGF-1; IGF-1 complex; CEP 151; CGP 35126; FK 780; Mecar;
RHIGF-I; Somatomedin- 1 ; Somatomedin-C; Somatokine; Myotrophin; IGEF; DepoIGF-1); Neuraminidase (Influenza A virus (A/Goose/Guangdong/I / 96 (H5N1)); Hemagglutinin [Influenza A virus (A/HongKong/213/03 (HK213 : H5N1 ) ) ] ; Butyryl- cholinesterase (BchE, Serum Cholinesterase , pseudo- cholinesterase El (CHE1)); Endothelin (ET-1; GenbankAccession No. NP_001946) ; Mechano Growth Factor (MGF; IGF-IEc; Genbank Accession No. P05019) . In an embodiment, the fusion protein is a fusion of human serum albumin and butyryl-cholinesterase . In an embodiment, the fusion protein is Composition 1. In an embodiment, the fusion protein is a fusion of human serum albumin and human growth hormone. Examples of such proteins which may be used in embodiments of the invention are disclosed in U.S. Patent Application Publication Nos. US 2011/0002888 and US 2009/0029914, and U.S. Patents Nos.
7,569,384 and 7,482,013, each of which is hereby incorporated by reference. In an embodiment, the fusion protein is Composition 2. In an embodiment, the fusion protein is Composition 3. In an embodiment, the fusion protein is Composition 4. In an embodiment, the fusion protein is Composition 5. In an embodiment, the protein is a therapeutic protein. In an embodiment, the protein is an antibody. In an embodiment, the protein is not an antibody.
In an embodiment, the protein is one of: Insulin; Humulin;
Novolin; Insulin human inhalation; Exubera; Insulin aspart; Novolog (aspart) ; Insulin glulisine; Apidra (glulisine) ;
Insulin lispro; Humalog (lispro) ; Isophane insulin; NPH;
Insulin detemir; Levemir (detemir) ; Insulin glargine; Lantus
(glargine) ; Insulin zinc extended; Lente; Ultralente;
Pramlintide acetate; Symlin; Growth hormone (GH) ; somatotropin; genotropin; humatrope; norditropin;
NorlVitropin; Nutropin; Omnitrope; Protropin; Siazen;
Serostim; Valtropin; Mecasermin; Increlex; Mecasermin rinfabate; IPlex; Factor VIII; Bioclate; Helixate; Kogenate;
Recominate; ReFacto; Factor IX; Benefix; Antithromin III (AT- III); Thrombate III; Protein C concentrate; Ceprotin; β-
Glucocerebrosidase ; Cerezyme; β-Glucocerebrosidase ; Ceredase
(purified from pooled human placenta) ; Alglucosidase- ;
Myozyme; Laronidase ( -1-iduronidase ) ; Aldurazyme; Idursulphase
(Iduronate-2-sulphatase) ; Elaprase; Galsulphase; Naglazyme; Agalsidase-β (human -galactosidase A) ; Fabrazyme; -1-
Proteinase inhibitor; Aralast; Prolastin; Lactase; Lactaid;
Pancreatic enzymes (lipase, amylase, protease) ; Arco-Lase,
Cotazym, Creon, Donnazyme, Pancrease, Viokase, Zymase,
Adenosine deaminase (pegademase bovine, PEG-ADA) ; Adagen; Pooled immunoglobulins; Octagam; Human albumin; Albumarc;
Albumin; Albuminar; AlbuRx; Albutein; Flexbumin; Buminate;
Plasbumin; Erythropoietin; Epoetin- ; Epogen; Procrit;
Darbepoetin- ; Aranesp; Filgrastim (granulocyte colony stimulating factor; G-CS F) ; Neupogen; Pegfilgrastim (Peg-G- CSF) ; Neulasta; Sargramostim (granulocytemacrophage colony stimulating factor; GM-CS F) ; Leukine; Oprelvekin (interleukinll ; IL11); Neumega; Human follicle-stimulating hormone (FSH) ; Gonal-F; Follistim; Human chorionic gonadotropin (HCG) ; Ovidrel; Luveris; Type I alpha-interferon; interferon alfacon 1; consensus interferon; Infergen; Interferon- 2a (IFN 2a); Roferon-A; Peglnterferon- 2a;
Pegasys; Interferon- 2b (IFN 2b); Intron A; Peglnterferon- 2b;
Peg-Intron; Interferon- n3 (IFN n3) ; Alferon N; Interferon- pia (rIFN-β) ; Avonex; Rebif ; Interferon- ib (rIFN-β) ; Betaseron;
Interferon-ylb (IFNy) ; Actimmune; Aldesleukin (interleukin 2 (IL2); epidermal thymocyte activating factor; ETAF) ;
Proleukin; Alteplase (tissue plasminogen activator; tPA) ;
Activase; Reteplase (deletion mutein of tPA) ; Retavase;
Tenecteplase ; TNKase; Urokinase; Abbokinase; Factor Vila;
NovoSeven; Drotrecogin- (activated protein C) ; Xigris; Salmon calcitonin; Fortical; Miacalcin; Teriparatide (human parathyroid hormone residues 1-34); Forteo; Exenatide; Byetta;
Octreotide; Sandostatin; Dibotermin- (recombinant human bone morphogenic protein 2; rhBMP2 ) ; Infuse; Recombinant human bone morphogenic protein 7 (rhBMP7); Osteogenic protein 1; Histrelin acetate (gonadotropin releasing hormone; GnRH) ;
Supprelin LA; Vantas; Palifermin ( keratinocyte growth factor
KGF) ; kepivance; Becaplermin (platelet-derived growth factor;
PDGF) ; Regranex; Trypsin; Granulex; Nesiritide; Natrecor;
Botulinum toxin type A; Botox; Botulinum toxin type B; Myoblock; Collagenase; Santyl; Human deoxy-ribonuclease I; dornase- ; pulmozyme; Hyaluronidase (bovine, ovine); Amphadase (bovine) ; hydase (bovine) ; Vitrase (ovine) ; Hyaluronidase (recombinant human) ; hylenex; Papain; accuzyme; panafil; L- asparaginase ; ELSPAR; Peg-asparaginase ; Oncaspar; Rasburicase; Elitek; Lepirudin; Refludan; Bivalirudin; Angiomax;
Streptokinase; Streptase; Anistreplase (anisoylated plasminogen streptokinase activator complex; APSAC) ; Eminase;
Bevacizumab; Avastin; Cetuximab; Erbitux; Panitumumab;
Vectibix; Alemtuzumab; Campath; Rituximab; Rituxan; Trastuzumab; Herceptin; Abatacept; Orencia; Anakinra; Antril;
Kineret; Abalimumab; Humira; Etanercept; Enbrel; Infliximab;
Remicade; Alefacept; Amevive; Natalizumab; Tysabri; Eculizumab; Soliris; Antithymocyte globulin (rabbit); Thymoglobulin; Basiliximab; Simulect; Daclizumab; Zenapax; Muromonab-CD3 ; Orthoclone; OKT3; Omalizumab; Xolair; Palivizumab; Synagis; Enfuvirtide; Fuzeon; Abciximab; ReoPro; Pegvisomant; Somavert; Crotalidae polyvalent immune Fab
(ovine) ; Crofab; Digoxin immune serum Fab (ovine) ; Digifab Ranibizumab; Lucentis; Denileukin; Diftitox; Ontak Ibritumomab; Tiuxetan; Zevalin; Gemtuzumab; Ozogamicin Mylotarg; Tositumomab and I-tositumomab; Bexxar; Bexxar 1-131 Hepatitis B surface antigen (HBsAg) ; Engerix; Recombivax HB;
HPV vaccine; Gardasil; OspA; LYMErix; Anti-Rhesus (Rh) immunoglobulin G; Rhophylac; Recombinant purified protein derivative (DPPD) ; Glucagon; GlucaGen; Growth hormone releasing hormone (GHRH) ; Geref ; Secretin; ChiRhoStim (human peptide) , SecreFlo (porcine peptide) ; Thyroid stimulating hormone (TSH) ; thyrotropin; Capromab pendetide; ProstaScint; Indium-lll-octreotide; OctreoScan; Satumomab pendetide; OncoScint; Arcitumomab; CEA-scan; Nofetumomab; Verluma; Apcitide; Acutect; Imciromab pentetate; Myoscint; Technetium fanolesomab; NeutroSpec; HIV antigens; Enzyme immunoassay;
OraQuick; Uni-Gold; Hepatitis C antigens; Recombinant immunoblot assay (RI BA) . Examples of proteins which may be used in this invention are disclosed in Leader et al . 2008, which is hereby incorporated by reference. For the foregoing embodiments, each embodiment disclosed herein is contemplated as being applicable to each of the other disclosed embodiment.
All combinations and sub-combinations of each of the various elements of the methods and embodiments described herein are envisaged and are within the scope of the invention.
This invention will be better understood by reference to the Examples which follow, which are set forth to aid in an understanding of the subject matter but are not intended to, and should not be construed to, limit in any way the claims which follow thereafter. Examples
Example 1 : Experimental Determination of Novel Formulation
Composition 1, a recombinant protein composed of the mature form of recombinant human serum albumin (rHSA) fused at its amino terminus to the carboxy-terminus of a mutated human butyrylcholinesterase (BChE) , was used to develop a novel lyophilization process and a suitable formulation. U.S. Provisional Application No. 61/752,740, filed January 15, 2013, is hereby incorporated by reference into this application.
Pre-formulation Studies
Ionic Strength Effects Sodium Chloride Spiking
Ionic strength effects were evaluated with Composition 1 (50 mg/mL in PMTT (which comprises 10 mM phosphate, 200 mM mannitol, 60 mM trehalose and 0.01% PS80, at pH 7.2)) at six target sodium chloride concentrations (5 mM, 10 mM, 20 mM, 50 mM, 80 mM, 120 mM) .
Vials of each sample were incubated at 25 °C for 5 days. Samples were removed from incubation after 5 days. The samples were compared to the 0 day and 0 mM sodium chloride controls by visual inspection and SE-HPLC.
The results suggest that increased concentrations of sodium chloride reduce purity loss. At or above 6 mS/cm, there is no significant change in SE-HPLC purity (Table 1) . All tested samples were clear, pale yellow, and essentially free from foreign particulate matter. Table 1 : Ionic Strength Effects Measured by Sodium Chloride Spiking .
Figure imgf000023_0001
Buffer controls containing 5 mM, 10 mM, 20 mM, 50 mM, 80 mM, and 120 mM sodium chloride were measured for conductivity. Buffer controls containing 10 mM, 20 mM, 30 mM, 40 mM, 50 mM, and 60 mM phosphate were measured for conductivity.
When the concentration of phosphate is ≥50 mM, the conductivity of the solution is ≥6 mS/cm (Table 2) . Therefore, phosphate can be used to replace NaCI while maintaining the ionic strength.
Table 2 : Sodium Chloride and Sodium Phosphate Buffer Conductivity Comparison.
Figure imgf000023_0002
Phosphate Spiking
Ionic strength effects were evaluated with Composition 1 (100 mg/mL in 200 mM mannitol, 60 mM trehalose, 0.03% PS80, pH 7.2) at six target phosphate concentrations (10 mM, 20 mM, 30 mM, 40 mM, 50 mM, 60 mM) .
Vials of each sample were incubated at 25 °C for 5 days. Samples were removed from incubation after 3 and 5 days. The samples were compared to the 0 day controls by visual inspection and SE-HPLC. Buffer controls were measured for conductivity.
The results show that increasing buffer conductivity decreases SE-HPLC purity loss. At a conductivity of approximately 4.5 mS/cm or higher (~≥30 mM sodium phosphate), there is no significant SE-HPLC purity loss after 5 days at 25°C (, Table 3) . All tested samples were clear, pale yellow, and essentially free from foreign particulate matter. Therefore, increasing ionic strength could prevent the protein from forming aggregates .
Table 3: Sodium Phosphate Spiking Data.
Figure imgf000024_0001
Polysorbate 80 Effects
The effects of PS80 were evaluated with Composition 1 (100 mg/mL in 10 mM phosphate, 200 mM mannitol, 60 mM trehalose, pH 7.2) at four target PS80 concentrations (0.01%, 0.05%, 0.1%, and 0.2%) . The samples were incubated at 2-8°C and 25°C for 1,
2 and 3 days . Samples were compared to the 0 point and the PS80-free controls by visual inspection and SE-HPLC. Osmolality was measured for the 0 points .
There was no change in purity for samples incubated at 2-8 °C (Table 4) . Samples at 100 mg/ml in PMTT incubated at 25°C showed 5-6% purity loss, but with no significant differences across the PS80 concentrations (Table 4) . There was no change in appearance across all PS80 concentrations, temperatures and time points, with the reconstituted solution always a clear pale yellow liquid essentially free from foreign particulate matter. There was no change in osmolality (Table 5) . Since there was no significant difference, 0.03% PS80, considered an acceptable middle point, was selected. This data also demonstrated that PMTT was not a suitable formulation for a higher dose of concentrated product.
Table 4: PS80 Spiking Purity Data
SEC Purity (%) SEC Purity Loss (%)
PS80 concentration (% ) PS80 concentration (°/ '>)
Temperature Day 0 0.01 0.05 0.1 0.2 0 0.01 0.05 0.1 0.2
0 99.7 99.7 99.8 99.8 99.7 NA NA NA NA NA
1 99.7 99.7 99.7 99.7 99.7 0.0 0.0 -0.1 0.0 0.0
2-8°C
2 99.8 99.7 99.7 99.7 99.7 0.0 0.0 -0.1 0.0 0.0
3 99.7 99.7 99.7 99.7 99.7 0.0 0.0 0.0 -0.1 0.0
0 99.7 99.7 99.8 99.8 99.7 NA NA NA NA NA
1 97.5 97.5 97.6 97.6 97.6 -2.2 -2.2 -2.2 -2.1 -2.1
25°C
2 95.5 95.5 95.6 95.8 95.8 -4.2 -4.3 -4.1 -4.0 -3.9
3 94.0 94.1 94.1 94.3 94.3 -5.7 -5.6 -5.7 -5.5 -5.5 Table 5: PS80 Spiking Osmolality Data
Figure imgf000026_0001
Formulation buffers containing varying concentrations of phosphate (40 mM, 50 mM and 60mM) , mannitol (60-200 mM) , trehalose (18-60 mM) and 0.03% PS80 were made by combining varying amounts of 500 mM phosphate (pH 7.2) stock solution, 500 mM mannitol stock solution and 200 mM trehalose stock solution, while keeping the ratio of trehalose to mannitol the same as PMTT. The osmolality of each buffer was tested and compared to the osmolality of PMTT (Table 6) .
Table 6: Phosphate Buffer Combinations
Figure imgf000026_0002
0.03 50 80 24 285
0.03 60 200 60 484
0.03 60 134 40 406
0.03 60 130 39 384
0.03 60 126 38 382
0.03 60 120 36 372
0.03 60 114 34 363
0.03 60 110 33 361
0.03 60 80 24 318
0.03 60 74 22 313
0.03 60 70 21 308
0.03 60 66 20 303
0.03 60 60 18 297
Buffers with osmolality approximately equal to 300 mOsm/kg were made, and conductivity and osmolality were measured for the buffers and for Composition 1 (100 mg/mL in PMTT) (Table 7) . Table 7 : Proto-formulation Buffer Measurements (Bolded lines indicate P50MTT and P60MTT)
Figure imgf000027_0001
From measuring Composition 1 (100 mg/mL in PMTT) and PMTT alone, it was calculated that Composition 1 at 100 mg/mL contributes approximately 31.5 mOsm/kg to osmolality. Targeting an osmolality of 300 mOsm/kg, two formulations were selected: P50MTT (267 mOsm/kg) , and P60MTT (268 mOsm/kg) .
P50MTT comprises 50 mM sodium phosphate, 115 mM mannitol, 35 mM trehalose and 0.03% PS80, at pH 7.2, while P60MTT comprises 60 mM phosphate, 100 mM mannitol, and 30 mM trehalose and 0.03% PS80, at pH 7.2. Measurements were performed for Composition 1 (100 mg/mL) in the new P50MTT and P60MTT formulations (Table 8) .
Table 8: P50MTT and P60MTT Measurements
Figure imgf000028_0001
Pre-formulation Conclusions
The pre-formulation studies were executed to determine potential formulation candidates for the lyophilization formulation of the concentrated product. Previous studies showed that Composition 1 was affected by concentration dependent aggregation, suggesting that aggregation is a major degradation pathway. In response, the ionic strength study was conducted to determine if increasing the ionic strength of the formulation buffer would have an effect on reducing aggregation. The results of the study demonstrate that there is a significant ionic strength effect, and in the higher ionic strength formulation there was a significant reduction in dose dependent aggregation at a protein concentration of 100 mg/ml.
The results of the PS80 spiking study show no difference between PS80 concentrations. Therefore, 0.03% PS80, which is within the acceptable range, was selected for the formulations .
Mannitol and trehalose concentrations in the candidate formulations were modified to target an osmolality of 300 mOsm/kg, while maintaining the ratio between mannitol and trehalose as established during development of the previous PMTT formulation. Two proto-formulations , P50MTT and P60MTT, were selected for additional studies .
Proto-formulation Evaluation Freeze-Thaw Effects
The effects of repeated freezing and thawing were evaluated with Composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) . Samples were frozen for 2-16 hours at ≤-65°C and then thawed for 3 hours at room temperature. Samples were collected after 1, 2, 4, 6 and 10 complete cycles of freezing and thawing. Samples were compared to the 0 point by visual inspection and SE-HPLC. Select samples were also tested by SDS-PAGE and potency analysis.
The results show no change in SE-HPLC purity after 10 cycles of freeze and thaw on Composition 1 in both P50MTT and P60MTT.
The SDS-PAGE results support the results of SE-HPLC. All tested samples were clear, pale yellow, and essentially free from foreign particulate matter. There was no significant change in potency (Table 9) .
Table 9: Freeze-Thaw Effects on Composition 1
Figure imgf000030_0001
Shaking Effects
The effects of shaking-induced aggregation were evaluated with Composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) . Samples were shaken horizontally at 150 rpm. Samples were incubated at 2-8°C and 25°C from 0 to 24 hours. Samples were compared to the 0 point by visual inspection, SE-HPLC and HI-HPLC.
The results show no change in SE-HPLC purity or HI-HPLC purity for Composition 1 in both P50MTT and P60MTT. All tested samples were clear, pale yellow, and essentially free from foreign particulate matter. This suggests that Composition 1 is not sensitive to shaking induced aggregation (Table 10) .
Table 10: Shaking Effects on Composition 1.
SE-HPLC SE-HPLC HI-HPLC HI-HPLC
Purity Purity Purity Purity
Formulation Temp Hrs (%) Change (%) (%) Change (%)
0 99.5 NA 91.5 NA
1 99.5 0.0 NT NT
3 99.6 0.1 91.6 0.1
2-8°C
6 99.5 0.0 NT NT
12 99.5 0.0 91.6 0.1
24 99.5 0.0 91.6 0.1
P50MTT
0 99.5 NA 91.5 NA
1 99.4 0.0 NT NT
3 99.5 0.0 91.7 0.2
22°C
6 99.5 0.0 NT NT
12 99.4 0.0 91.6 0.1
24 99.4 -0.1 91.6 0.1
0 99.5 NA 91.7 NA
1 99.5 0.0 NT NT
3 99.5 0.0 91.7 0.0
2-8°C
6 99.5 0.0 NT NT
12 99.5 0.0 91.6 -0.1
24 99.5 0.0 91.7 0.0
P60MTT
0 99.5 NA 91.7 NA
1 99.505 0.0 NT NT
3 99.525 0.0 91.6 -0.1
22°C
6 99.505 0.0 NT NT
12 99.5 0.0 91.6 -0.1
24 99.48 0.0 91.7 0.0
Short-Term Liquid Stability
Composition 1 (101.6 mg/mL P50MTT and 100.8 mg/mL in P60MTT) was used for this study. Samples were incubated at 2-8°C and 25 °C for 6 days. Samples were removed from incubation after 1,
3 and 6 days. Samples were compared to the 0 point by visual inspection, SE-HPLC and HI-HPLC. All tested samples were clear, pale yellow, and essentially free from foreign particulate matter. Select samples were also tested by SDS- PAGE and potency analysis (Table 11) . Table 11: Short Term Liquid Stability Results
Figure imgf000032_0001
The results show that Composition 1 in both P50MTT and P60MTT had no change in SE-HPLC and HI-HPLC purity (after incubation in 2-8°C and 25°C for 6 days. This is a significant change from the prior formulation (PMTT) , which had an approximate
SE-HPLC purity loss of 4.5% and HI-HPLC purity loss of 7.2% after incubation at 25 °C after 5 days. The SDS-PAGE results support the results of SE-HPLC. There was no significant change in potency (Table 11) . Proto-formulation Conclusion
The results of the proto-formulation studies indicate that Composition 1 at 100 mg/mL is stable at 2-8°C and 25°C for up to 6 days in both P50MTT and P60MTT formulations. Composition 1 in P50MTT and in P60MTT was not sensitive to freeze-thaw or shaking effects. Overall, there was no difference between the P50MTT and P60MTT formulations. Both could support the lyophilization process and would be potential formulation candidates for an initial lyophilization evaluation. Lyophilization Formulation Evaluation
Initial Lyophilization Evaluation
An initial lyophilization cycle evaluation was carried out using Composition 1 (101.6 mg/mL in P50MTT and 100.8 mg/mL in P60MTT) . The TBU lyophilization cycle is summarized in Table 12. Post-lyophilization tests include visual inspection pre- and post-reconstitution and residual moisture content analysis. 0-12 hour post-reconstitution samples were analyzed by SE-HPLC and HI-HPLC. Selected samples were also tested by potency analysis. Table 12: TBU Lyophilization Cycle
Figure imgf000033_0001
The lyophilization products were pharmaceutically acceptable cakes (white to off-white in color and intact) .
There was no change in SE-HPLC purity, HI-HPLC purity, and potency between pre- and post-lyophilization. The residual moisture content for both cakes was 0.1%. Lyophilization Cycle Evaluation
Low Temperature Thermal Analysis
To characterize the physio-chemical behavior of Composition 1 (100 mg/mL in P50MTT) at low temperatures, low temperature thermal analysis was performed. The analysis consisted of electrical resistance measurements (using a Kaye Validator instrument) , observations of freeze drying behavior using a freeze-drying microscope (FDM) , and low temperature differential scanning calorimetry (LT-DSC) .
The results of the analysis are summarized below:
• Phase transition at -17°C
• A minimum temperature of -29 °C is required for complete solidification
• Liquid-like movement occurs at -4°C
• Recommended temperature for primary drying at or below a range of -6°C to -8°C
The TBU lyophilization cycle conditions are summarized as follows :
• Freezing and refreezing steps at -45°C
• Annealing step at -18°C
• Primary drying at -10°C
This data supports that the TBU lyophilization cycle is appropriately designed and suitable for this product.
TBU and Other Lyophilization Cycle Evaluations
Composition 1 (100 mg/mL in P50MTT) was lyophilized using the TBU cycle and used for the long term stability study.
Two randomly selected vials from the batch were analyzed by visual inspection and pre and post lyophilization analysis. The results indicate that the TBU cycle produces pharmaceutically acceptable cakes that are white to off-white in color and intact.
Additional lyophilization cycle evaluation was carried out using Composition 1 (103 mg/mL in P50MTT) . A total of 7 development lyophilization cycles, as well as the TBU cycle as a control, were completed with variations to the freezing, annealing, primary drying, and secondary drying steps. Upon completion of the lyophilization process, samples were analyzed by visual inspection, moisture content analysis, HI- HPLC and SE-HPLC.
The lyophilization cycle evaluation was carried out using Composition 1 (103 mg/mL in P50MTT) . Visual inspection, residual moisture content measurement, SE-HPLC and HI-HPLC purity analysis were performed. See Table 13 for detailed information pertaining to the various lyophilization cycle parameters .
Table 13: Lyophilization Cycle Parameters Summary
Figure imgf000036_0001
The results of the lyophilization cycle evaluation further confirm that the TBU lyophilization cycle is more appropriate for Composition 1. The data suggests that the TBU cycle produces pharmaceutically acceptable cakes, with the lowest residual moisture (0.3%) compared to the other lyophilization cycles tested during the evaluation (Table 14) .
Table 14: Lyophilization Cycle Evaluation Results Summary
Figure imgf000037_0001
* Samples vials were reconstituted with 1 mL sWFI at ambient laboratory conditions. Samples were inverted 5X upon addition of sWFI and then incubated at ambient laboratory conditions without additional agitation until complete dissolution was observed. ® 1.0 mL fill volume. * 1.1 mL fill volume. Λ Bulk TV-1380 purity 89.8%, 99.6% as determined by HIC and SEC-
HPLC, respectively.
Pre- and Post-Lyophilization Analysis
Composition 1 (100 mg/ml in P50MTT after reconstitution with 1.1 ml of WFI ) 0 month was used for the pre- and post- lyophilization analysis. Time points were 0, 4, 8 and 12 hours. Visual inspection was performed prior to reconstitution. Reconstitution time was recorded. Post- reconstitution, samples were analyzed by visual inspection, pH, osmolality, concentration measurement, SE-HPLC, SDS-PAGE, potency analysis and free thiol content (Table 15) .
Table 15: Pre and Post Reconstitution Summary
Figure imgf000038_0001
The results indicate that the TBU cycle produces pharmaceutically acceptable cakes that are white to off-white in color and intact. Post reconstitution, samples are clear and free of particulate matter. Additionally, samples up to 12 hours post-reconstitution pass acceptance criteria (Tables 15, 16) .
Table 16: Acceptance Criteria
Figure imgf000039_0001
Conclusion of Lyophilization Evaluation
For essentially equivalent formulations, it is preferable to use the formulation containing a lower concentration of salt for the lyophilization process. Therefore, the P50MTT formulation was selected as the final concentrated product formulation and was used for the lyophilization formulation evaluation and long term stability program.
To summarize the lyophilization evaluation studies, the TBU lyophilization cycle is appropriate for the lyophilization of Composition 1. The results of the low thermal analysis study indicate that the parameters of the TBU lyophilization cycle meet the minimum temperature requirements and the pre and post lyophilization results suggest that there is no change in protein quality. Cakes produced using the TBU lyophilization cycle are white to off-white in color and are intact, which is considered to be pharmaceutically acceptable.
The results of the lyophilization evaluation also suggest that the P50MTT is an appropriate formulation for the concentrated product. Upon reconstitution, samples remain clear and free of particulate matter.
Conclusion
The formulation studies were executed to determine an appropriate formulation for the lyophilized concentrated product .
The pre-formulation studies demonstrated that increasing ionic strength results in a significant reduction in dose dependent aggregation at a protein concentration of 100 mg/ml. PS80 concentration had no significant effect and a concentration of 0.03% was selected for use in the proto-formulations .
Two proto-formulations, P50MTT and P60MTT, were selected for additional studies .
The study results indicates that Composition 1 drug substances at 100 mg/mL with these two formulations are stable at 2-8°C and 25°C for up to 6 days, and are neither sensitive to freeze-thaw nor shaking effects, which could support the lyophilization process. There is no significant impact on the product quality by post-lyophilization . Overall, the two formulations are comparable in terms of the product quality and stability.
However, the P50MTT formulation was selected as a formulation candidate for an additional lyophilization cycle evaluation and long term stability study, due to its lower ionic strength compared to P60MTT, which might negatively impact lyophilization process and lyophilization product.
The lyophilization evaluation studies support that the TBU cycle produces pharmaceutically acceptable cakes. Overall, the results of the formulation studies demonstrate that P50MTT is a suitable lyophilization formulation for the concentrated product and the TBU lyophilization program would be an appropriate lyophilization process to use for concentrated product fill. Example 2: Long Term Stability Testing of Composition 1
Methods
Composition 1 (100 mg/ml in P50MTT after reconstitution with 1.1 ml of WFI) was used for the stability program study. The lyophilized product was stored at 2-8°C, 25°C and 40°C. Results
At the end of 6 months, there is no significant change in SE- HPLC purity for Composition 1 when stored at 2-8°C (Table 17) . The quality attributes of samples stored at the recommended conditions meet all acceptance criteria to at least 6 months. When stored at elevated temperature conditions, such as 25°C and 40°C, there is a 2.5% and 9.6% loss in SE-HPLC purity after 6 months, respectively (Tables 18 and 19) . However, potency was within tolerances for all temperature conditions up to 6 months (Tables 17, 18 and 19) . Table 17: Stability Data for Composition 1 When Stored at Recommended Conditions, 2-8°C
Figure imgf000042_0001
'Result is an average of 3 vials (1 each from beginning, middle, and end) Table 18: Stability Data for Composition 1, 25°C
Figure imgf000043_0001
WC = White Cake, CYF = Clear, Yellow solution, essentially free from foreign particulate matter 'Result is an average of 3 vials (1 each from beginning, middle, and end)
Table 19: Stability Data for Composition 1, 40°C
Figure imgf000043_0002
WC = White Cake, CYF = Clear, Yellow solution, essentially free from foreign particulate matter 'Result is an average of 3 vials (1 each from beginning, middle, and end) Samples stored under recommended conditions are stable under the recommended conditions for 18 months.
Samples stored under recommended conditions are stable for 24 months . Samples stored under recommended conditions are stable for 36 months .
Example 3: Long Term Stability Testing of Composition 2 Methods
Composition 2, known as Neugranin™, is a protein derived from the direct genetic fusion of the genes for Granulocyte Colony
Stimulating Factor (GCSF) and human serum albumin. The TBU lypholization cycle applied to Composition 2 (15 mg/ml) is summarized in Table 20. The lyophilized product was stored at 2-8°C, 25°C and 40°C. Results
At the end of 6 months, there is no significant change in SEC- HPLC purity for Composition 2 when stored at 2-8°C (Table 21) . The quality attributes of samples stored at the recommended conditions meet all acceptance criteria to at least 6 months. When stored at elevated temperature conditions, such as 25°C and 40°C, there is a .4% gain and a .1% loss in SEC-HPLC purity after 6 months, respectively (Tables 22-23) . However, potency was within tolerances for all temperature conditions up to 6 months (Tables 21, 22 and 23) . Table 20: TBU Lyophilization Cycle
Figure imgf000044_0001
i Engage vacuum and adjust to 100 mT as controlled by capacitance manometer.
j Hold shelf temperature at -45°C until vacuum set point is achieved.
k Hold at -45°C for lhour.
1 Warm shelf temperature to - 10°C at 0.8°C/min.
m Hold shelf temperature at -10°C for 36 hours (primary drying).
n Ensure that all functioning product thermocouples have been at or above - 10°C for at least 3 hours before proceeding to the next step. If not, continue to hold the shelf temperature at -10°C until all functioning product thermocouples have been at or above - 10°C for at least 3 hours.
0 Warm shelf temperature to 25°C at 0.8°C/min.
P Hold at 25°C for 15 hours (secondary drying).
q At end of secondary drying, slowly restore chamber to approximately 600
T with sterile filtered nitrogen, NF/EP and seat stoppers.
Table 21: Stability Data for Composition 2 When Stored at Recommended Conditions, 2-8°C
Figure imgf000045_0001
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; FIO- For information only; NT-Not Test Table 22: Stability Data for Composition 2, 25°C
Figure imgf000046_0001
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT- Not tested Table 23: Stability Data for Composition 2, 40 ° C
Figure imgf000046_0002
(RP-HPLC) RRT
10.2 10.3 10.2 10.8 10.5 11.1
0.98
MP+
96.7 97.5 97.2 97.8 96.1 94.1
RRT .98
Purity (IEC-HPLC) NT NT NT NT NT 69.4
Potency (bioassay) 134 122 101 107 83 108
Residual Moisture 0.4 0.7 0.6 0.7 1.6 1.3
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT- Not tested
Samples stored under recommended conditions are stable under the recommended conditions for 18 months.
Samples stored under recommended conditions are stable for 24 months .
Samples stored under recommended conditions are stable for 36 months .
Example 4 : Long Term Stability Testing of Composition 3 Methods Composition 3, known as Albuferon™-Beta, is a product derived from the direct genetic fusion of the genes for human interferon-beta (IFN-beta) and human serum albumin. The TBU lypholization cycle applied to Composition 3 (2.0 mg/ml) is summarized in Table 24. The lyophilized product was stored at 2-8°C, 25°C and 40°C.
Results
At the end of 6 months, there is no significant change in SEC- HPLC purity for Composition 3 when stored at 2-8°C (Table 25) . The quality attributes of samples stored at the recommended conditions meet all acceptance criteria to at least 6 months.
When stored at elevated temperature conditions, such as 25°C and 40°C, there is a 1.0% and a 3.1% loss in SEC-HPLC purity after 6 months, respectively (Tables 26-27) . However, potency was within tolerances for all temperature conditions up to 6 months (Tables 25, 26 and 27) . Table 24: TBU Lyophilization Cycle
Figure imgf000048_0001
Table 25: Stability Data for Composition 3 When Stored at Recommended Conditions, 2-8°C
Figure imgf000048_0002
Purity
Report Results (X.X %)
(RP-HPLC) (%) 92.6 90.8 92.2 92.4 91.5 92.8
Potency (bioassay) (%) Report Results (X %) 90 71.8 93 84 89.2 85.5
Residual Moisture (%) < 3.0% 1.1 1.4 1.0 1.8 1.3 2.0
Deamidation Report Results
NT 0.30 0.34 0.36 0.256 (pmol/pmol) (X.X pmol/pmol)
Bioburden (Membrane O CFU/
< lO CFU/lO mL
Filtration) 10 mL
Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
WC = White Cake, CPF = Clear, pale yellow solution, essentially free from foreign particulate matter;
FIO- For information only; NT -Not tested
Table 26: Stability Data for Composition 3, 25°C
Figure imgf000049_0001
Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT- Not tested
Table 27: Stability Data for Composition 3, 40 °C
Figure imgf000049_0002
Appearance
CPF CPF CPF CPF
(Post- reconstitution)
pH 7.2 7.1 7.2 7.2
Osmolality (Freezing
299 303 316 301
point)
Protein Concentration
2.0 1 2.1 2.1 2.0
(A280) (mg/ml)
Reduced
98 98 98 98
Purity (%)
(SDS- Non- PAGE) reduced 100 100 98 99
(%)
Purity (SE-HPLC) (%) 98.4 97.2 95.8 95.3
Purity
92.6 89.7 87.4 85.0
(RP-HPLC) (%)
Potency (bioassay) (%) 90 79 83 82
Residual Moisture (%) 1.1 1.2 1.5 1.9
Deamidation
NT 0.31 0.32 0.39
(pmol/pmol)
Result is an average of vials taken from beginning, middle, and end of the lyo cycle.
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT- Not tested
Samples stored under recommended conditions are stable under the recommended conditions for 18 months. Samples stored under recommended conditions are stable for 24 months .
Samples stored under recommended conditions are stable for 36 months .
Example 5: Long Term Stability Testing of Composition 4 Methods
Composition 4, known as Albutropin™, is a contiguous protein comprised of human serum albumin (HSA) and recombinant growth hormone (rHGH) with the mature form of HSA genetically fused at its C-terminus to the N-terminus of the mature form of rHGH. The TBU lypholi zation cycle applied to Composition 4
(25.0 mg/ml) is summarized in Table 28. The lyophilized product was stored at 2-8°C, 25°C and 40°C.
Results
At the end of 6 months, there is no significant change in SEC- HPLC purity for Composition 4 when stored at 2-8°C (Table 29) . The quality attributes of samples stored at the recommended conditions meet all acceptance criteria to at least 6 months. When stored at elevated temperature conditions, such as 25°C and 40°C, there is a .8% and a 2.6% loss in SEC-HPLC purity after 6 months, respectively (Tables 30-31) . However, potency was within tolerances for all temperature conditions up to 6 months (Tables 29, 30 and 31) .
Table 28: TBU Lyophilization Cycle
Figure imgf000051_0001
Table 29: Stability Data for Composition 4 When Stored at Recommended Conditions, 2-8°C
Figure imgf000052_0001
5 'Result is an average of 3 vials (1 each from beginning, middle, and end) Table 30: Stability Data for Composition 4, 25°C
Figure imgf000053_0001
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT 'Result is an average of 3 vials (1 each from beginning, middle, and end)
Table 31: Stability Data for Composition 4, 40°C
Figure imgf000053_0002
Main Peak 99.1 98.1 97.7 96.5
Purity (SEC- RRT < .78 0.2 0.2 0.2 0.3
HPLC) RRT .86 0.4 1.0 1.7 2.7
RRT 1.09 0.3 0.7 0.4 0.5
Main Peak 95.3 93.6 91.9 88.9
RRT .64 0.3 0.3 0.5 0.4
RRT 0.88-0.91 0.3 0.3 0.4 0.5
Purity RRT 0.96-0.99 0.2 0.4 0.6 0.9
(RP-HPLC) RRT 1.03-1.06 2.2 3.0 3.7 5.4
RRT 1.09-1.10 0.4 0.5 0.6 0.7
RRT 1.13 1.3 1.8 2.2 3.3
RRT 1.15 0.0 0.0 0.0 0.0
Potency (bioassay) 111 110 121 85
Residual Moisture1 0.21 0.7 1.0 1.5
For Information Only
Purity (IEC-HPLC) NT NT NT NT
Deamidation (pmol/pmol) NT NT NT NT
WC = White Cake, CPF = Clear, pale yellow, essentially free from foreign particulate matter; NT= Not tested 'Result is an average of 3 vials (1 each from beginning, middle, and end)
Samples stored under recommended conditions are stable under the recommended conditions for 18 months. Samples stored under recommended conditions are stable for 24 months .
Samples stored under recommended conditions are stable for 36 months .
Example 6: Long Term Stability Testing of Composition 5 Methods
Composition 5, known as Cardeva™, is a recombinant human B- type natriuretic peptide (BNP) serum albumin fusion protein. The TBU lypholization cycle applied to Composition 5 (100 mg/ml) is summarized in Table 32. The lyophilized product was stored at 2-8°C, 25°C and 40°C.
Results
At the end of 6 months, there is no significant change in SEC- HPLC purity for Composition 5 when stored at 2-8°C (Table 33) . The quality attributes of samples stored at the recommended conditions meet all acceptance criteria up to 6 months. When stored at elevated temperature conditions, such as 25 °C and 40°C, there is a .7% and a 4.0% loss in SEC-HPLC purity after 6 months, respectively (Tables 34-35) . However, potency was within tolerances for all temperature conditions up to 6 months (Tables 33, 34 and 35) . Table 32: TBU Lyophilization Cycle
Figure imgf000055_0001
Table 33: Stability Data for Composition 5 When Stored at Recommended Conditions, 2-8°C
Figure imgf000055_0002
Purity Reduced (%) > 90.0% 100 100 100 100 (SDS- Non-reduced
PAGE) > 90.0% 100 100 100 99
(%)
Purity (SEC-HPLC) (%) > 90.0% 98.7 98.9 98.9 99.0
Purity
Report Results (%) 92.7 91.7 92.8 91.7 (RP-HPLC) (%)
Purity
Report Results (%) 64.9 64.6 65.1 62.8 (IE-HPLC) (%)
Potency (bioassay)
Report Results (%) 30.5 34.9 28.7 (relative potency %)
Residual Moisture (%) < 3.0% 0.03 0.03 0.06 0.9
Free Thiol (Ellman's Report Results
NT 0.9 0.7 0.7 Reagant) (mol/mol)
WC— White cake; CPF— Clear, pale yellow, essentially free from foreign particulate matter
Table 34: Stability Data for Composition 5, 25°C
Figure imgf000056_0001
WC— White cake; CPF— Clear, pale yellow, essentially free from foreign particulate matter Table 35: Stability Data for Composition 5, 40 ° C
Figure imgf000056_0002
Appearance
< 4 < 4 < 6 < 3
(Reconstitution time)
Appearance
CPF CPF CPF CPF
(Post- reconstitution)
pH 6.0 6.1 6.0 6.1
Osmolality (Freezing
284 299 305 308 point)
Protein Concentration
102.9 103.2 114.2 99.1
(A280) (mg/ml)
Purity Reduced (%) 100 100 98 95
(SDS- Non-reduced
PAGE) 100 100 96 95
(%)
Purity (SEC-HPLC) (%) 98.7 97.3 95.8 94.7
Purity
92.7 89.6 90.0 86.1
(RP-HPLC) (%)
Purity
64.9 61.9 59.1 55.7
(IE-HPLC) (%)
Potency (bioassay)
30.5 38.9 34.3
(relative potency %)
Free Thiol (Ellman's
NT 0.9 0.7 0.8
Reagant)
WC— White cake; CPF— Clear, pale yellow, essentially free from foreign particulate matter
Discussion
A more concentrated formulation can have significant advantages, including increasing convenience (since fewer or smaller vials are required to contain a given dose) and reducing the injection bolus necessary for a given dose. However, it is not always routine and often very difficult to increase the concentration of a peptide formulation.
The appropriateness of a lyophilization process is unpredictable. Freezing rates that are either too fast or too slow can lead to protein aggregation or denaturing (Rathore and Raj an 2008; Krishnamurthy and Manning 2002) . Excessive drying can destabilize the protein (Rathore and Rajan 2008) . Even the material used for the vial and the stopper can have critical effect on lyophilized protein products (Rathore and Rajan 2008) .
As the concentration of protein in the solution used to make a lyophilized product increases, the time required to reconstitute the lyophilate increases as well (Shire et al . 2004) .
The process described herein, however, represents an approach which produces pharmaceutically acceptable lyophilized cakes with fast reconstitution times .
References
Leader, Benjamin, Quentin J. Baca, and David E. Golan.
"Protein therapeutics: a summary and pharmacological classification." Nature Reviews Drug Discovery 7.1 (2008) : 21-39.
Rathore, Nitin, and Rahul S. Rajan. "Current perspectives on stability of protein drug products during formulation, fill and finish operations." Biotechnology Progress 24.3 (2008) : 504-514.
Shire, Steven J., Zahra Shahrokh, and Jun Liu. "Challenges in the development of high protein concentration formulations." Journal of Pharmaceutical Sciences 93.6 (2004) : 1390-1402.

Claims

Claims is claimed is:
A process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of:
(i) obtaining a solution comprising the protein in one or more containers;
(ii) placing the one or more containers within a chamber of a lyophilizing unit; iii) reducing the temperature to an initial freezing temperature of -60°C to -25°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the initial freezing temperature for 1 to 6 hours to form a frozen solution;
(iv) increasing the temperature to an annealing temperature of the frozen solution of -30 °C to -10 °C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the annealing temperature for 1 to 10 hours;
(v) reducing the temperature to a refreezing temperature of -60°C to -25°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the refreezing temperature for 1 to 6 hours;
(vi) reducing the pressure of the chamber to 50 to 500 mT, and continuing to hold the temperature at the refreezing temperature for an additional 0 to 4 hours; vii) increasing the temperature to a primary drying temperature of -30°C to -5°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the primary drying temperature for 10 to 72 hours; (viii) increasing the temperature to a secondary drying temperature of 5°C to 30°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the secondary drying temperature for 2 to 25 hours; and
(ix) increasing the pressure of the chamber to partial atmospheric pressure.
2. The process of claim 1, wherein step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 °C to 10 °C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
3. The process of claim 1, wherein step (ii) placing the containers within the chamber of the lyophilizing unit comprises placing the containers on a shelf which is at an initial shelf temperature of from -40 °C to 5°C within the chamber and holding the temperature of the shelf at the initial shelf temperature for 0 to 5 hours before initiating step (iii) .
4. The process of claim 2, wherein (a) the initial shelf temperature is from -5°C to 10°C or (b) the initial shelf temperature is from -5°C to 5°C.
5. The process of any one of claims 3 to 4, wherein the initial shelf temperature is 5°C.
6. The process of any one of claims 2 to 5, wherein the shelf is held at the initial shelf temperature for (a) 1.1 to 5 hours or (b) 2 hours or more.
7. The process of any one of claims 1 to 6, wherein the temperature in steps (iii) to (viii) is the shelf temperature .
8. The process of claim 1, wherein step (ii) further comprises pre-cooling the one or more containers.
9. The process of claim 8, wherein the containers are pre- cooled to a temperature (a) from -5°C to 10°C or (b) from -5°C to 5°C.
10. The process of one of claims 8 to 9, wherein the pre- cooling is by liquid nitrogen.
11. The process of one of claims 1 to 10, wherein in step (iii) the temperature is reduced at a rate of (a) 0.3°C per minute or (b) 0.2°C per minute.
12. The process of any one of claims 1 to 11, wherein in step
(iii) the temperature is held at the initial freezing temperature for (a) 2.1 to 6 hours or (b) 3 hours.
13. The process of any one of claims 1 to 12, wherein in step
(iv) the temperature is increased at a rate of 0.8°C per minute .
14. The process of any one of claims 1 to 13, wherein in step
(iv) the temperature is held at the annealing temperature for (a) 2.1 to 10 hours or (b) 5 hours.
15. The process of any one of claims 1 to 14, wherein in step
(v) the temperature is reduced at a rate of 0.3°C per minute .
16. The process of any one of claims 1 to 15, wherein in step
(v) the temperature is held at the refreezing temperature for (a) 1.1 to 6 hours or (b) 2 hours.
17. The process of any one of claims 1 to 16, wherein in step
(vi) the temperature is held at the refreezing temperature for 1 hour.
18. The process of any one of claims 1 to 17, wherein in step
(vii) the temperature is increased at a rate of 0.6°C per minute .
19. The process of any one of claims 1 to 18, wherein in step (vii) the temperature is held at the primary drying temperature for (a) 36 hours or more, (b) 36 hours, (c) 10 to 29 hours, or (d) 29 to 42 hours.
20. The process of any one of claims 1 to 18, wherein in step (vii) the primary drying temperature is -30°C to-5°C
21. The process of any one of claims 1 to 18, further comprising measuring the temperature of the frozen solution within one or more of the containers during step
(vii) , wherein in step (vii) the temperature is held at the primary drying temperature for three hours beyond the time at which the temperature of each measured container is equal to or greater than the primary drying temperature .
22. The process of any one of claims 1 to 21, wherein in step
(viii) the temperature is increased at a rate of (a) 0.6°C per minute or (b) 0.8°C per minute.
23. The process of any one of claims 1 to 22, wherein in step
(viii) the temperature is held at the secondary drying temperature for (a) 4.1 or more hours or (b) 15 hours.
24. The process of any one of claims 1 to 23, wherein in step
(ix) the partial atmospheric pressure is (a) 810 mBar or (b) 600 Torr.
25. The process of any one of claims 1 to 24, wherein in step
(ix) the restoring to partial atmospheric pressure is adding sterile filtered nitrogen to the chamber.
26. The process of any one of claims 1 to 25, further comprising the step:
(x) sealing the containers.
27. The process of claim 26, wherein the sealing comprises inserting a stopper.
28. The process of any one of claims 1 to 27, wherein the initial freezing temperature is (a) -49°C to -25°C or (b) -45°C.
29. The process of any one of claims 1 to 28, wherein the annealing temperature is (a) -19°C to -10°C or (b) -18°C.
30. The process of any one of claims 1 to 29, wherein the refreezing temperature is (a) -49°C to -25°C or (b) 45°C.
31. The process of any one of claims 1 to 30, wherein the primary drying temperature is (a) -19°C to 0°C or (b) 10°C.
32. The process of any one of claims 1 to 31, wherein the secondary drying temperature is (a) 5°C to 30 °C or (b) 25°C.
33. The process of any one of claims 1 to 32, wherein in step (vi) the pressure is reduced to 100 mTorr.
34. The process of any one of claims 1 to 33, wherein the solution comprising a protein has a protein concentration (a) greater than 65 mg/ml, (b) from 2 to 250 mg/ml, (c) from 65 to 250 mg/ml, or (d) from 100-110 mg/ml.
35. The process of any one of claims 1 to 34, wherein in step (i) each of the one or more containers contains (a) from 0.5 to 2.0 ml of the solution or (b) 1.0 to 1.2 ml of the solution .
36. The process of any one of claims 1 to 35 for producing a lyophilized pharmaceutical composition containing a protein, which (a) reconstitutes in water for injection in 15 minutes or less or 6 minutes or less, or (b) reconstitutes in water for injection after one month of storage at recommended conditions in 15 minutes or less or 6 minutes or less.
37. The process of any one of claims 1 to 36, wherein the solution comprising the protein further comprises 40 to 60 mM phosphate.
38. The process of any one of claims 1 to 37, wherein the solution comprising the protein further comprises 100 to 150 mM mannitol, 20 to 40 mM trehalose, or 0.02 to 0.05 percent polysorbate 80.
39. The process of any one of claims 1 to 38, wherein the solution comprising the protein further comprises mannitol and trehalose in concentrations at a ratio of about 3.3 to one.
40. The process of any one of claims 1 to 39 for producing a lyophilized pharmaceutical composition containing a protein, which has a residual moisture of (a) 3.0 weight percent or less or (b) 0.3 weight percent or less.
41. The process of any one of claims 1 to 40 for producing a lyophilized pharmaceutical composition containing a protein, which (a) is stable under recommended storage conditions for at least six months, (b) is stable under recommended storage conditions for at least 18 months, (c) has a purity of 99.0% or more after storage for six months at 2-8°C, (d) has a purity of 96.0% or more after storage for six months at 25°C, (e) has a purity of 89.0% or more after storage for six months at 40 °C, (f) has a 9.6% or less loss in SE-HPLC purity after storage for six months, or any combination of (a) -(f) .
42. The process of any one of claims 1 to 41 for producing a lyophilized pharmaceutical composition containing a protein, wherein the protein is Composition 1, Composition 2, Composition 3, Composition 4 or Composition 5.
43. The process of any one of claims 1 to 41 for producing a lyophilized pharmaceutical composition containing a protein, wherein the protein is (a) a fusion protein or (b) a human serum albumin fusion protein.
44. A product produced by the process of any one of claims 1 to 43.
45. The product of claim 44, which reconstitutes in water for injection within (a) 15 minutes or (b) 6 minutes.
46. The product of claim 44, which reconstitutes to a protein concentration (a) from 2 to 250 mg/ml, (b) from 65 to 250 mg/ml, or (c) from 100 to 110 mg/ml.
47. A process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process of any one of claims 1 to 43, and reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes, thereby producing an injectable pharmaceutical composition .
48. A method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process of any one of claims 1 to 43, reconstituting the lyophilized pharmaceutical composition with water for injection within 15 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
49. A process for producing an injectable pharmaceutical composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process of any one of claims 1 to 43, and reconstituting the lyophilized pharmaceutical composition with water for injection within 6 minutes, thereby producing an injectable pharmaceutical composition .
50. A method of treating a patient with a therapeutic protein composition, comprising obtaining an amount of the lyophilized pharmaceutical composition comprising a protein produced by the process of any one of claims 1 to 43, reconstituting the lyophilized pharmaceutical composition with water for injection within 6 minutes to form a reconstituted solution, and administering the reconstituted solution to the patient, thereby treating the patient.
51. A process for producing a lyophilized pharmaceutical composition containing a protein, comprising the steps of:
(i) obtaining a solution comprising the protein in one or more containers;
(ii) placing the one or more containers within a chamber of a lyophilizing unit;
(iii) reducing the temperature to an initial freezing temperature of -60°C to -25°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the initial freezing temperature for 1 to 6 hours to form a frozen solution;
(iv) reducing the pressure of the chamber to 50 to 500 mT, and continuing to hold the temperature at the freezing temperature for an additional 0 to 4 hours;
(v) increasing the temperature to a primary drying temperature of -30°C to -5°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the primary drying temperature for 10 to 72 hours; and
(vi) increasing the temperature to a secondary drying temperature of 5°C to 30°C at a rate of 0.2°C to
2.0°C per minute, and holding the temperature at the secondary drying temperature for 2 to 25 hours.
52. The process of claim 51, further comprising after step (vi) a step (vii) comprising increasing the pressure of the chamber to partial atmospheric pressure.
53. The process of claim 51 or 52, further comprising after step (iii) and before step (iv) an annealing step comprising :
(i) increasing the temperature to an annealing temperature of the frozen solution of -30 °C to -10 °C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the annealing temperature for 1 to 10 hours; and
(ii) reducing the temperature to a refreezing temperature of -60°C to -25°C at a rate of 0.2°C to 2.0°C per minute, and holding the temperature at the refreezing temperature for 1 to 6 hours.
54. The process of claim 51 or 52 for producing a lyophilized pharmaceutical composition containing a protein, wherein the protein is Composition 4.
PCT/US2014/011399 2013-01-15 2014-01-14 Lyophilization process WO2014113358A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP14740889.2A EP2945593A4 (en) 2013-01-15 2014-01-14 Lyophilization process
MX2015008944A MX2015008944A (en) 2013-01-15 2014-01-14 Lyophilization process.
CA2897062A CA2897062A1 (en) 2013-01-15 2014-01-14 Lyophilization process
HK16105645.0A HK1217633A1 (en) 2013-01-15 2016-05-17 Lyophilization process

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361752797P 2013-01-15 2013-01-15
US61/752,797 2013-01-15
US201361784538P 2013-03-14 2013-03-14
US61/784,538 2013-03-14

Publications (1)

Publication Number Publication Date
WO2014113358A1 true WO2014113358A1 (en) 2014-07-24

Family

ID=51165301

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/011399 WO2014113358A1 (en) 2013-01-15 2014-01-14 Lyophilization process

Country Status (6)

Country Link
US (1) US20140199286A1 (en)
EP (1) EP2945593A4 (en)
CA (1) CA2897062A1 (en)
HK (1) HK1217633A1 (en)
MX (1) MX2015008944A (en)
WO (1) WO2014113358A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2015009141A (en) 2013-01-15 2016-03-16 Teva Pharma Formulations of albu-bche, preparation and uses thereof.
WO2016108534A1 (en) * 2014-12-30 2016-07-07 주식회사 삼양바이오팜 Polymer nanoparticle freeze-dried product, and preparation method therefor
SG11202002157TA (en) 2017-09-19 2020-04-29 Advaxis Inc Compositions and methods for lyophilization of bacteria or listeria strains
EP3744319B1 (en) 2019-05-28 2022-10-19 Ilkogen Ilac Sanayi Ve Ticaret A.S. A stable lyophilized formulation for hybrid fc fused g-csf
CN113116831A (en) * 2019-12-26 2021-07-16 天士力生物医药股份有限公司 Freeze-drying method of recombinant human prourokinase for injection
JP2023550203A (en) * 2020-10-19 2023-11-30 オーク ヒル バイオ リミテッド Compositions suitable for use in neonates
CN114853835A (en) * 2022-04-13 2022-08-05 广东海赫生物医药科技有限公司 Method for drying NADH

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0367090B1 (en) * 1988-11-01 1994-01-12 Schwarz Pharma Ag Lyophilized MDM composition and method of making it
US7045318B2 (en) * 1995-12-30 2006-05-16 Delta Biotechnology Limited Recombinant fusion proteins to growth hormone and serum albumin
US20070259348A1 (en) * 2005-05-03 2007-11-08 Handylab, Inc. Lyophilized pellets
WO2012142281A1 (en) * 2011-04-12 2012-10-18 Endocyte, Inc. Solid pharmaceutical composition

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5227374A (en) * 1982-11-12 1993-07-13 Mead Johnson & Company Lyophilized cyclophosphamide
US6019968A (en) * 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
AU2005272862B2 (en) * 2004-08-12 2011-03-10 Merck Sharp & Dohme Corp. Stable pegylated interferon formulation
US7572893B2 (en) * 2004-08-17 2009-08-11 Regeneron Pharmaceuticals, Inc. IL-1 antagonist formulations
ES2390354T3 (en) * 2005-07-22 2012-11-12 Amgen, Inc Lyophilisates of concentrated proteins, procedures and uses
EP1854478A1 (en) * 2006-05-12 2007-11-14 Cytos Biotechnology AG Nicotine-carrier vaccine formulation
CN102762196B (en) * 2009-11-24 2014-05-14 基立福疗法公司 Lyophilization methods, compositions, and kits

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0367090B1 (en) * 1988-11-01 1994-01-12 Schwarz Pharma Ag Lyophilized MDM composition and method of making it
US7045318B2 (en) * 1995-12-30 2006-05-16 Delta Biotechnology Limited Recombinant fusion proteins to growth hormone and serum albumin
US20070259348A1 (en) * 2005-05-03 2007-11-08 Handylab, Inc. Lyophilized pellets
WO2012142281A1 (en) * 2011-04-12 2012-10-18 Endocyte, Inc. Solid pharmaceutical composition

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LIU, W ET AL.: "Freeze-Drying of Proteins From a Sucrose-Glycine Excipient System: Effect of Formulation Composition on the Initial Recovery of Protein Activity.", AAPS PHARMSCITECH., vol. 6, no. 2, 2005, pages E150 - E157, XP055266844 *
See also references of EP2945593A4 *
YU , J ET AL.: "Synergistic Effects of Surfactants and Sugars on Lipoplex Stability During Freeze-drying and Rehydration.", JOUMAL OF PHARMACEUTICAL SCIENCES., vol. 98, no. 9, 2009, pages 3319 - 3328, XP055266849 *

Also Published As

Publication number Publication date
MX2015008944A (en) 2016-06-21
HK1217633A1 (en) 2017-01-20
US20140199286A1 (en) 2014-07-17
EP2945593A1 (en) 2015-11-25
EP2945593A4 (en) 2016-08-31
CA2897062A1 (en) 2014-07-24

Similar Documents

Publication Publication Date Title
WO2014113358A1 (en) Lyophilization process
RU2472492C2 (en) Stable non-aqueous pharmaceutical compositions
JP6063877B2 (en) Stable formulation for parenteral injection of peptide drugs
EP2593137B1 (en) Stabilized formulations containing anti-ngf antibodies
JP2008543839A5 (en)
US11351256B2 (en) Room temperature stable lyophilized protein
RU2012147070A (en) PROTEIN COMPOSITIONS AND METHODS FOR PRODUCING THEM
US20190175701A1 (en) Pharmaceutical Compositions
CA2545034A1 (en) Propylene glycol-containing peptide formulations which are optimal for production and for use in injection devices
Gokarn et al. Excipients for protein drugs
JP2010529956A5 (en)
US20150265683A1 (en) Stable aqueous composition comprising human insulin or an analogue or derivative thereof
KR20160074562A (en) Stable formulation of insulin glulisine
CA3034972A1 (en) A pharmaceutical insulin formulation
US20150065423A1 (en) Rapid acting injectable formulations
Redwan Cumulative updating of approved biopharmaceuticals
WO2009086166A1 (en) Room temperature stable, lyophilized natriuretic peptide formulations
WO2016011281A1 (en) FORMULATIONS OF AN ALBUMIN-hGH FUSION PROTEIN
IL277731B1 (en) Medical infusion pump system for the delivery of an insulin compound
WO2011108010A2 (en) A thermostable liquid formulation of gonadotropins
AU2013368990B2 (en) Pharmaceutical composition
US20200188479A1 (en) Apparatus and methods for administration of a nauseogenic compound from a drug delivery device
Gierach et al. Protein drug production and formulation
EP3225235B1 (en) Stable peptide formulations for parenteral injection
WO2017049205A2 (en) Growth hormone formulation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14740889

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2897062

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/008944

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2014740889

Country of ref document: EP