US20180110856A1 - Pharmaceutical Formulations and Methods of Making the Same - Google Patents

Pharmaceutical Formulations and Methods of Making the Same Download PDF

Info

Publication number
US20180110856A1
US20180110856A1 US15/788,762 US201715788762A US2018110856A1 US 20180110856 A1 US20180110856 A1 US 20180110856A1 US 201715788762 A US201715788762 A US 201715788762A US 2018110856 A1 US2018110856 A1 US 2018110856A1
Authority
US
United States
Prior art keywords
pharmaceutical composition
etanercept
formulation
buffering agent
arginine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/788,762
Other languages
English (en)
Inventor
Monica GOSS
Nicole BALL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to US15/788,762 priority Critical patent/US20180110856A1/en
Assigned to AMGEN INC. reassignment AMGEN INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BALL, Nicole, GOSS, Monica
Priority to US15/958,261 priority patent/US10307483B2/en
Publication of US20180110856A1 publication Critical patent/US20180110856A1/en
Priority to US16/144,120 priority patent/US11491223B2/en
Priority to US17/933,055 priority patent/US20230080571A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the invention relates to the formulation of pharmaceutical compositions of etanercept.
  • the invention also relates to methods of removing buffer and of formulating pharmaceutical compositions of etanercept.
  • Formulation of a protein drug can present many challenges for the pharmaceutical scientist.
  • a formulation must be found that stabilizes the protein drug and makes it resistant to degradation by proteolysis, aggregation, misfolding, etc.
  • finding appropriate stability conditions can be challenging.
  • Etanercept is a dimeric fusion protein consisting of the extracellular ligand-binding portion of the human 75 kilodalton (p75) tumor necrosis factor receptor (TNFR) linked to the Fc portion of human IgG1.
  • the Fc component of etanercept contains the CH2 domain, the CH3 domain and hinge region, but not the CH1 domain of IgG1.
  • TNFR tumor necrosis factor receptor
  • TNFR tumor necrosis factor receptor
  • Etanercept is commercially available as ENBREL® (Amgen Inc., Thousand Oaks, Calif.) and is approved to treat moderately to severely active rheumatoid arthritis, moderately to severely active polyarticularjuvenile idiopathic arthritis (JIA) in patients ages two and older, chronic moderate to severe plaque psoriasis (PsO) in adults, psoriatic arthritis (PsA) in adults, and active ankylosing spondylitis (AS). Etanercept was first available in a lyophilized formulation to be reconstituted immediately before injection.
  • ENBREL® Amgen Inc., Thousand Oaks, Calif.
  • the formulation Upon reconstitution of the lyophilized product with water for injection, the formulation is 10 mM Tris HCl, 4% mannitol, 1% sucrose, pH 7.4 at about 25 mg/mL. However, this formulation is not stable for storage. It was discovered that a liquid formulation of etanercept could be achieved using arginine to stabilize the protein (see U.S. Pat. No. 7,648,702).
  • An exemplary liquid formulation consists of 50 mg/mL etanercept, 25 mM phosphate buffer, 25 mM L-arginine hydrochloride, 100 mM NaCl, 1% Sucrose at pH 6.3 in water.
  • the invention provides pharmaceutical compositions containing etanercept that are stable and can be conveniently stored as a liquid at controlled room temperature (CRT) for extended periods of time, even in the absence of an additional buffering agent.
  • CRT controlled room temperature
  • the pharmaceutical compositions of the invention when injected into subjects, they also demonstrate significantly reduced injection pain as compared to the commercially available prior art formulation. Thus these pharmaceutical compositions are more convenient and advantageous for patients.
  • Another aspect of the invention provides methods of formulating pharmaceutical preparations of etanercept at a desired pH but in the absence of additional buffering agent in the final formulation.
  • the invention provides a pharmaceutical composition comprising etanercept, NaCl, arginine, and sucrose, wherein the pharmaceutical composition has essentially no additional buffering agent, and the pH of the composition is between 6.1 and 6.5.
  • the pharmaceutical composition is capable of maintaining the pH between 6.1 and 6.5 when stored at controlled room temperature (CRT) for 2 weeks.
  • the etanercept concentration is between 40 mg/mL and 100 mg/mL.
  • the pharmaceutical composition is isotonic.
  • the pharmaceutical composition contains: between 20 mM and 150 mM NaCl; between 5 mM and 100 mM arginine; and between 0.5% and 2% (w/v) sucrose.
  • the pharmaceutical composition comprises a surfactant.
  • the surfactant is polysorbate 20, polysorbate 80, or poloxamer 188.
  • the surfactant is polysorbate 20 at a concentration (w/v) of between 0.001% and 0.1%.
  • the surfactant is polysorbate 80 at a concentration (w/v) of between 0.001% and 0.1%.
  • the surfactant is poloxamer 188 at a concentration (w/v) of between 0.01% to 0.3%.
  • the pharmaceutical composition maintains a pH of between 5.8 and 6.7 for at least two weeks when stored at approximately 25° C., and wherein less than 6% of the total etanercept is aggregated in a high molecular weight form as assessed using size exclusion chromatography. In another embodiment, the pharmaceutical composition maintains a pH of between about 6.1 and about 6.5. In another embodiment, less than 28% of the total amount of etanercept is in a misfolded form as assessed using hydrophobic interaction chromatography. In another embodiment, the pharmaceutical composition consists essentially of about 40-100 mg/mL etanercept, about 120 mM NaCl, about 25 mM arginine, about 1% sucrose, and water. In another embodiment, the pharmaceutical composition consists essentially of about 40-100 mg/mL etanercept, about 120 mM NaCl, about 25 mM arginine, about 1% sucrose, about 0.01% polysorbate 20, and water.
  • the present invention provides a method of formulating a pharmaceutical composition of etanercept to remove an additional buffering agent and maintain pH from 6.1 to 6.5, comprising formulating the etanercept formulation in a formulation comprising an additional buffering agent at between pH 6.1 and 6.5, and exchanging the formulation comprising an additional buffering agent against a formulation that does not comprise an additional buffering agent and is between pH 5.6 and 6.5, and collecting the resulting pharmaceutical formulation.
  • the exchange step uses diafiltration.
  • the formulation that does not comprise an additional buffering agent is isotonic.
  • the formulation that does not comprise an additional buffering agent contains sucrose, arginine, and NaCl.
  • the formulation that does not comprise an additional buffering agent contains between 20 mM and 150 mM NaCl; between 5 mM and 100 mM arginine; and between 0.5% and 2% (w/v) sucrose.
  • the formulation that does not comprise an additional buffering agent consists essentially of about 120 mM NaCl, about 25 mM arginine, about 1% sucrose, and water.
  • the method of formulating a pharmaceutical composition of etanercept further comprises adding polysorbate.
  • the polysorbate is polysorbate 20 at a concentration (w/v) of between 0.001% and 0.1%.
  • the method of formulating a pharmaceutical composition of etanercept further comprises filtering the pharmaceutical composition. In another embodiment, the method of formulating a pharmaceutical composition of etanercept further comprises aliquoting the pharmaceutical composition into a drug product form.
  • the present invention provides a kit comprising a pharmaceutical composition of etanercept as described above in a drug product form and instructions for storage and use.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising etanercept, NaCl, arginine, sucrose, a phosphate buffer and benzyl alcohol, wherein the pH of the composition is between 6.1 and 6.5.
  • the benzyl alcohol is at a concentration (v/v) of between 0.1% and 5.0%.
  • the concentration of benzyl alcohol is about 0.9%.
  • the pharmaceutical composition comprising etanercept further comprises polysorbate 20 at a concentration (w/v) of between 0.001% and 0.1%.
  • the concentration of polysorbate 20 is about 0.004%.
  • the pharmaceutical composition comprising etanercept consists essentially of etanercept at about 40-100 mg/mL, arginine at about 25 mM, sodium chloride at about 100 mM, sucrose at a concentration (w/v) of about 1%, phosphate buffer at about 25 mM, and benzyl alcohol at a concentration (v/v) of about 0.9%.
  • the pharmaceutical composition comprising etanercept consists essentially of etanercept at about 40-100 mg/mL, arginine at about 25 mM, sodium chloride at about 100 mM, sucrose at a concentration (w/v) of about 1%, phosphate buffer at about 25 mM, benzyl alcohol at a concentration (v/v) of about 0.9%, and polysorbate 20 at a concentration (w/v) of about 0.004%.
  • the present invention provides a single-dose container containing a pharmaceutical composition comprising etanercept as described above.
  • the pharmaceutical composition consists essentially of etanercept at about 40-100 mg/mL, arginine at about 25 mM, sodium chloride at about 100 mM, sucrose at a concentration (w/v) of about 1%, phosphate buffer at about 25 mM, and benzyl alcohol at a concentration (v/v) of about 0.9%.
  • the pharmaceutical composition consists essentially of etanercept at about 40-100 mg/mL, arginine at about 25 mM, sodium chloride at about 100 mM, sucrose at a concentration (w/v) of about 1%, phosphate buffer at about 25 mM, benzyl alcohol at a concentration (v/v) of about 0.9%, and polysorbate 20 at a concentration (w/v) of about 0.004%.
  • the single dose container is a vial, a syringe, or an autoinjector.
  • the single-dose container contains an aqueous formulation consisting of etanercept at 50.0 mg/mL, sodium chloride at 120 mM, L-arginine at 25 mM, sucrose at 1.0% (w/v).
  • the present invention provides a method of preparing a single-dose container containing a pharmaceutical composition comprising etanercept as described above, comprising filling the single-dose container with about a single dose of the pharmaceutical composition under sterile conditions.
  • FIG. 1 shows the percent HMW (peak B) as detected by SEC for the etanercept stability assay of Example 3.
  • FIG. 2 shows the percent LMW as detected by dSEC for the etanercept stability assay of Example 3.
  • FIG. 3 shows the percent Peak 3 as detected by HIC for the etanercept stability assay of Example 3.
  • FIG. 4 shows the percent Peak 3 as detected by HIC for the stainless steel cryo-vessel storage etanercept stability assay of Example 4.
  • FIG. 5 shows the percent LMW as detected by dSEC for the stainless steel cryo-vessel storage etanercept stability assay of Example 4.
  • FIG. 6 shows the percent Peak B as detected by SEC for the stainless steel cryo-vessel storage etanercept stability assay of Example 4.
  • FIG. 7 shows the percent Peak B as detected by SEC for the freeze-thaw etanercept stability assay of Example 4.
  • FIG. 8 shows the pH stability of the conditioned AEX intermediate pool at controlled room temperature (CRT) in the assay of Example 6.
  • FIG. 9 shows the UF/DF pool pH (A) and conductivity (B) stability at CRT in the assay of Example 6.
  • FIG. 10 shows the pH (A) and conductivity (B) stability of etanercept formulated in SAS solution in the assay of Example 6.
  • the invention provides improved pharmaceutical compositions of etanercept.
  • pharmaceutical composition is understood to refer to a formulation of a polypeptide suitable for injection and/or administration into a patient in need thereof. More particularly, a pharmaceutical composition is substantially sterile and does not contain any agents that are unduly toxic or infectious to the recipient.
  • Etanercept is a soluble form of the p75 TNF receptor fused to an Fc domain of a human IgG1 (TNFR:Fc).
  • a commercially available etanercept is known as ENBREL® (Immunex Inc., Thousand Oaks, Calif.).
  • Etanercept is produced by recombinant DNA technology in a Chinese hamster ovary (CHO) mammalian cell expression system. It consists of 934 amino acids and has an apparent molecular weight of approximately 150 kilodaltons (Physicians Desk Reference, 2002, Medical Economics Company Inc.). The full sequence expressed in CHO cells is shown below. However, it is to be understood that minor modifications and deletions of this sequence (up to 10%) may be possible and can be used within the scope of the invention.
  • CHO Chinese hamster ovary
  • the invention provides a pharmaceutical composition comprising etanercept but containing essentially no additional buffering agent.
  • additional buffering agent refers to a component of an etanercept composition or formulation, other than etanercept itself, that contributes significantly to the buffering capacity of the composition or formulation.
  • Etanercept itself has been shown herein to provide all the needed buffering to maintain the pH between 6.1 and 6.5, and in particular at about 6.2-6.3, under the conditions described below. As demonstrated below in Example 1, this pH range has been shown to be effective to maintain the desired stability characteristics of an etanercept formulation (less than 6% high molecule weight aggregates and less than 28% misfolded and clipped species).
  • total additional buffering agent refers collectively to all of the components of an etanercept composition or formulation, except for etanercept itself, that contribute significantly to the buffering capacity of the composition or formulation.
  • compositions according to the invention comprise less than 2.0 mM total additional buffering agent, less than 1.5 mM total additional buffering agent, less than 1.0 mM total additional buffering agent, less than 0.5 mM total additional buffering agent, less than 0.25 mM total additional buffering agent, less than 0.1 mM total additional buffering agent, or less than 0.05 mM of total additional buffering agent.
  • additional buffering agents are used to maintain the pH in a desired range, often at concentrations of 5.0 mM or higher.
  • additional buffering agents are histidine, potassium phosphate, sodium or potassium citrate, maleic acid, ammonium acetate, tris-(hydroxymethyl)-aminomethane (tris), various forms of acetate and diethanolamine.
  • One common buffering agent is sodium phosphate as its buffering capacity is at or near pH 6.2.
  • Sodium phosphate is the buffering agent used in the current commercial liquid formulation of etanercept since its desired pH is 6.3. In the invention described herein, essentially no sodium phosphate is present in the pharmaceutical formulation of etanercept.
  • the pH of the inventive pharmaceutical composition is maintained between 6.1 and 6.5, even after extended storage.
  • the pharmaceutical composition with essentially no additional buffering agent results in significantly less pain than the current buffered commercial formulation.
  • phosphate is often chosen as a buffer for pharmaceutical compositions because of the close to neutral pH buffering capacity and belief that it is one of the less painful buffer components (as compared to, for example, citrate buffer), the instant inventors have determined that phosphate buffer at around pH 6.3 does contribute to pain upon injection.
  • a “formulation solution” or “formulation buffer” is a solution or buffer that does not itself contain etanercept but is used to make a formulation comprising etanercept.
  • the etanercept concentration in the pharmaceutical compositions of the invention is between about 40 mg/mL and about 200 mg/mL in an aqueous formulation (e.g., water as the solvent). More preferably, the etanercept concentration is between about 40 mg/mL and about 100 mg/mL, yet more preferably between about 40 mg/mL and about 75 mg/mL, and optionally about 50 mg/mL.
  • the pharmaceutical compositions of the invention also contain arginine.
  • Arginine has been shown to make a substantial contribution to stabilizing etanercept in a liquid formulation (see U.S. Pat. No. 7,648,702, incorporated herein by reference).
  • Pharmaceutically appropriate forms of arginine are commercially available.
  • L-arginine e.g., L-arginine HCl or L-arginine base
  • L-arginine is the arginine used for pharmaceutical formulations. It is understood that within the pH range of 6.0 and 6.6, and in particular at a pH of about 6.2-6.3, arginine does not contribute meaningfully to the buffering capacity of a formulation.
  • the concentration of arginine in the compositions of the invention are preferably from about 1 mM to about 1 M, more preferably from about 10 mM to about 200 mM, or alternatively from about 5 mM to about 100 mM, more preferably from about 10 mM to about 100 mM, even more preferably from about 15 mM to about 75 mM, and yet more preferably at about 25 mM.
  • the pharmaceutical composition comprises about 50 mg/mL to 75 mg/mL etanercept and about 25 mM arginine, wherein the pharmaceutical composition has essentially no additional buffering agent, and the pH of the composition is between 6.0 and 6.6.
  • the term “about” is understood to mean that there can be variation in the concentration of a component of the described formulation that can be up to and including 10% of the given value. For example, if a formulation has about 10 mg/mL of a polypeptide, this is understood to mean that a formulation can have between 9 to 11 mg/mL of the stated polypeptide.
  • the pharmaceutical compositions may contain additional excipients, as long as those excipients are not additional buffering agents, and in particular are not phosphate buffering agents.
  • additional excipients include but are not limited to sugars/polyols such as: sucrose, lactose, glycerol, xylitol, sorbitol, mannitol, maltose, inositol, trehalose, glucose; polymers such as: serum albumin (bovine serum albumin (BSA), human SA or recombinant HA), dextran, PVA, hydroxypropyl methylcellulose (HPMC), polyethyleneimine, gelatin, polyvinylpyrrolidone (PVP), hydroxyethylcellulose (HEC); non-aqueous solvents such as: polyhydric alcohols, (e.g., PEG, ethylene glycol and glycerol) dimethysulfoxide (DMSO) and dimethylformamide (DM).
  • the excipients include NaCl and/or sucrose.
  • NaCl may be present in the pharmaceutical composition at a concentration of from about 5 mM to about 200 mM, more preferably between about 20 mM to about 150 mM, even more preferably between about 80 mM to about 140 mM.
  • Sucrose may be added to a concentration of between about 0.5% to about 2% (w/v) sucrose, more preferably between about 0.8% to about 1.2% (w/v) sucrose, even more preferably at about 1% (w/v) sucrose.
  • the osmolality of a pharmaceutical composition is preferably regulated in order to maximize the active ingredient's stability and also to minimize discomfort to the patient upon administration. It is generally preferred that a pharmaceutical composition be isotonic with serum, i.e., having the same or similar osmolality, which is achieved by addition of a tonicity modifier. Serum is approximately 300+/ ⁇ 50 milliosmolals per kilogram, thus it is contemplated that the osmolality of an isotonic pharmaceutical composition will be from about 180 to about 420 milliosmolals. In some embodiments, the range will be from about 250 to about 350 milliosmolals.
  • a tonicity modifier is understood to be a molecule that contributes to the osmolality of a solution.
  • tonicity modifiers suitable for modifying osmolality include, but are not limited to amino acids (e.g., arginine, cysteine, histidine and glycine), salts (e.g., sodium chloride, potassium chloride and sodium citrate) and/or saccharides (e.g., sucrose, glucose and mannitol).
  • the concentration of the tonicity modifier in the formulation is preferably between about 1 mM to 1 M, more preferably about 10 mM to about 200 mM. In some embodiments, the concentrations of NaCl and sucrose are adjusted to generate a pharmaceutical composition that is isotonic.
  • the pharmaceutical composition contains about 40-100 mg/mL etanercept, about 120 mM NaCl, about 25 mM arginine, about 1% sucrose, and water.
  • the pharmaceutical composition can consist essentially of about 50-100 mg/mL etanercept, about 120 mM NaCl, about 25 mM arginine, about 1% sucrose, about 0.01% polysorbate 20, and water.
  • the pharmaceutical compositions of the invention may include a surfactant.
  • Surfactants are agents that reduce solution/surface induced stress.
  • examples of surfactants are polysorbates, such as polysorbate 20, polysorbate 40, polysorbate 60, and polysorbate 80 (e.g., TWEEN-20® (Sigma-Aldrich, St. Louis, Mo.) or TWEEN-80® (Sigma-Aldrich, St. Louis, Mo.)), sodium dodecyl sulfate (SDS), polyoxyethylene copolymer, poloxamers, such as poloxomer 188 (e.g., PLURONIC® F-68 (Sigma-Aldrich, St.
  • polysorbate 20 can be included in the pharmaceutical compositions at a concentration (w/v) of between about 0.001% and about 0.03%. In particular embodiments illustrated below by example, polysorbate 20 can be included in the pharmaceutical formulations at a concentration (w/v) of 0.01% or at about 0.004%.
  • the examples below illustrate how one of skill in art can determine whether a formulation is capable of maintaining the pH in a desired range.
  • the pharmaceutical composition is formulated and stored in the test containers (which may be glass vials, glass syringes, plastic syringes, stainless steel vessels, or any manner of sterile device suitable for pharmaceutical compositions) and the pH is assessed at time 0, and then at indicated times as appropriate.
  • the testing conditions will anticipate needs for storage of the pharmaceutical composition, and will stress those conditions.
  • the formulations of the invention are able to maintain the desired pH under controlled room temperature (CRT) for at least 2 weeks, at least 4 weeks, at least 8 weeks, at least 12 weeks, and at least 24 weeks.
  • CRT controlled room temperature
  • CRT is defined by the USP, and has a temperature maintained thermostatically that encompasses the usual and customary working environment of 20° C. to 25° C. (68° F. to 77° F.); that results in a mean kinetic temperature calculated to be not more than 25° C.; and that allows for excursions between 15° C. and 30° C. (59° F. and 86° F.) that are experienced in pharmacies, hospitals, and warehouses.
  • the pharmaceutical compositions of the invention exhibit particular quality attributes.
  • the testing of these quality attributes is also described below by way of example.
  • the pharmaceutical compositions of the invention contain less than 6% of the total etanercept aggregated in a high molecular weight form as assessed using size exclusion chromatography.
  • the pharmaceutical compositions of the invention contain less than 28% of the total amount of etanercept is in a misfolded form as assessed using hydrophobic interaction chromatography.
  • the pharmaceutical compositions of the invention are capable of remaining stable by maintaining pH and/or other noted quality attributes (minimum high molecular weight forms and minimum misfolded forms) for the following temperatures and extended time periods (1) at ⁇ 30° C. (frozen) for at least 4 weeks, at least 3 months, at least 6 months, at least 12 months, and at least 36 months; (2) for up to 1 freeze/thaw cycle, up to 2 freeze/thaw cycles, up to 3 freeze/thaw cycles, and up to 5 freeze/thaw cycles; (3) at 4° C. (refrigerated temperature) for at least 2 weeks, at least 4 weeks, at least 8 weeks, at least 12 weeks, at least at least 24 weeks, and at least 52 weeks; (4) at 25° C. (room temperature) for at least 2 weeks, at least 4 weeks, at least 8 weeks, at least 12 weeks, at least 24 weeks; and (5) at 40° C. (accelerated stability testing) for at least 2 weeks.
  • pH and/or other noted quality attributes minimum high molecular weight forms and minimum mis
  • the pharmaceutical compositions of the invention also exhibit the surprising result of reduced pain upon injection into a subject.
  • This property can be assessed using the Visual Analog Scale (VAS) that has been validated by Gallagher et al, 2002, Am. J. Em. Med. v20; i4: 287-290. Trained health professionals administer the drug via injection, and within 30 seconds after each injection, subjects assessed their level of injection pain using a 100 mm Visual Analog Scale (VAS). A difference of 13 to 16 mm on the VAS is considered to be clinically meaningful. Using this technique, it was demonstrated that a placebo formulation without phosphate induced significantly less pain than a placebo formulation containing phosphate at pH 6.3 and the current commercial formulation containing both etanercept and phosphate at pH 6.3.
  • VAS Visual Analog Scale
  • etanercept is expressed recombinantly in CHO cells and secreted into the medium.
  • the medium is collected, filtered, and purified using, for example, various chromatography techniques.
  • protein A can be used to purify Fc domain containing polypeptides such as etanercept, and is advantageous as a first processing step.
  • the invention also provides a method of formulating a pharmaceutical composition of etanercept to remove buffer and maintain pH at a target range, comprising formulating the etanercept in a buffered formulation in the target range, and exchanging the buffered formulation against an unbuffered formulation that is within in or just below that target range, and collecting the resulting pharmaceutical formulation of the etanercept.
  • the method provides formulating a pharmaceutical composition of etanercept to remove buffer and maintain pH from 6.0 to 6.6, comprising formulating the etanercept formulation in a buffered formulation at between pH 6.0 to 6.6, and exchanging the buffered formulation against an unbuffered formulation that is between pH 5.6 and 6.5, and collecting the resulting pharmaceutical formulation.
  • a pharmaceutical composition of etanercept to remove buffer and maintain pH from 6.0 to 6.6
  • formulating the etanercept formulation in a buffered formulation at between pH 6.0 to 6.6 comprising formulating the etanercept formulation in a buffered formulation at between pH 6.0 to 6.6, and exchanging the buffered formulation against an unbuffered formulation that is between pH 5.6 and 6.5, and collecting the resulting pharmaceutical formulation.
  • the starting buffered etanercept formulation is at pH 7.2, it will be adjusted with a strong acid, such as HCl, to within the range 6.1 to 6.5.
  • the unbuffered formulation that is used for exchange should be titrated to between pH 5.6 and 6.5. Because the unbuffered formulation used for exchange has no buffering agent, care should be used during titration.
  • Dialysis makes use of selective diffusion through a semi-permeable membrane to remove unwanted smaller molecules from a larger protein formulation.
  • equilibrations are done serially until a desired fold reduction in the concentration of an unwanted molecule is achieved. For example, three serial equilibrations, each at or greater than 100-fold dilution, can be used to achieve a concentration reduction of 1,000,000-fold or greater.
  • Ultrafiltration and diafiltration are similar to dialysis in that they use a semi-permeable membrane. But unlike the passive diffusion of dialysis, ultrafiltration and diafiltration involves forcing solutions through the membrane using various techniques.
  • buffer exchange can be performed using gel filtration or size exclusion chromatography.
  • chromatographic techniques that also can be used to achieve buffer exchange that are well within the skill of those in the art such as ion exchange chromatography, hydrophobic interaction chromatography, and mixed mode chromatography.
  • the methods of the invention include collecting the resulting pharmaceutical formulation. At this point, essentially all buffer has been removed, but the pH is still maintained at the desired levels. For a pharmaceutical composition containing etanercept, the pH is maintained at between 6.0 and 6.6.
  • the pharmaceutical formulation may be further treated as necessary.
  • a surfactant can be added.
  • the pharmaceutical composition can be filtered.
  • the methods of the invention also include aliquoting the pharmaceutical compositions into a drug product form. Such drug product forms are distributed for final use by patients or health care providers.
  • the pharmaceutical compositions of this invention are particularly useful for parenteral administration, i.e., subcutaneously, intramuscularly, intravenously, intraperitoneal, intracerebrospinal, intra-articular, intrasynovial, and/or intrathecal. Parenteral administration can be by bolus injection or continuous infusion.
  • compositions for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers.
  • the pharmaceutical compositions may, if desired, be presented in a vial, pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the dispenser device can comprise a syringe having a single dose of the liquid formulation ready for injection.
  • the pharmaceutical composition is aliquoted into a cassette component for use with a reusable autoinjector.
  • the pharmaceutical compositions can be provided packaged in or with an on-body injector device.
  • the pharmaceutical compositions can be aliquoted into a drug product form suitable for a needleless injection device.
  • the pharmaceutical composition can also be aliquoted into a format suitable as a depot preparation.
  • Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the formulations may be modified with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • the present invention is directed to a kit or container, which contains the pharmaceutical composition of the invention.
  • the kit can also be accompanied by instructions for the storage and use of the pharmaceutical compositions.
  • the container can be, for example, a single-use container, i.e., a container that holds one dose formulation of the present invention. It is understood that a single-use container might contain a single dose plus enough extra to ensure that a full single dose can be administered to a patient from the container, but not so much extra that the container could be used to administer a second dose.
  • Examples of containers suitable for use in certain aspects of the present invention include vials, syringes, and auto-injectors.
  • suitable auto-injectors include those found in U.S. Pat. Nos. 8,177,749, 8,052,645, and 8,920,374, in U.S. patent application Ser. Nos. 12/993,163, 13/269,750, 13/454,531, 14/112,479, 14/777,255, and 14/777,259, and in PCT Publications WO 2014/0089393, WO 2016/033496, and WO 2016/033507, each of which is incorporated herein by reference in its entirety.
  • etanercept-containing compositions and formulations of the present invention can be used in the treatment of patients with conditions that respond to treatment with etanercept.
  • conditions that respond to treatment with etanercept include rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and psoriasis.
  • Methods of treating patients with etanercept are described in, for example, U.S. Pat. Nos. 7,915,225, 8,119,605, 8,410,060, 8,722,631, and 8,119,604, each of which is incorporated herein by reference in its entirety.
  • This example demonstrates the effects of pH and buffer on etanercept at 50 mg/mL, and assesses the stability of a high concentration (100 mg/mL) solution without added phosphate buffer.
  • the following formulations were tested.
  • Enbrel drug substance in PASS 25 mM phosphate buffer, 25 mM L-arginine, 100 mM NaCl, 1% sucrose
  • PASS 25 mM phosphate buffer, 25 mM L-arginine, 100 mM NaCl, 1% sucrose
  • the material was dialyzed into the new formulations (without polysorbate) and concentrated to 50 mg/mL using 10,000 MWCO centripreps.
  • a sample of 50_SAS_100NaCl was also concentrated to 100 mg/mL (100_SAS_100NaCl).
  • Benzyl alcohol was spiked into the current formulation to a final concentration of 0.9%.
  • a 1% stock solution of polysorbate 20 was prepared fresh and spiked into all formulation to a final concentration of 0.004%. All formulations were manually filled into 1 mL long BD glass syringes to a volume of 0.5 mL and then stoppered using an ASPU vacuuming stoppering unit.
  • the pH was measured using a Mettler Toledo SevenEasy pH meter combined with a Mettler Inlab MicroProbe. Samples were warmed to room temperature prior to measurements. Osmolality was measured using The Advanced Osmometer Model 3900. Each measurement was performed using 250 ⁇ L of sample and 290 osmolality standards were tested to ensure the system was operating properly. Size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software. Denatured size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software.
  • the lower pH formulations During long term storage at 25° C. and 40° C., the lower pH formulations, A45SuT, A52SuT, and A58SuT, exhibited undesirable levels of low molecular weight degraded or clipped species when analyzed using denatured size exclusion chromatography.
  • the high concentration formulation 100_SAST_100NaCl stored at 25° C. and 40° C. began to show an increase in high molecular weight aggregates when analyzed using size exclusion chromatography, but performed similarly to the current commercial formulation at 4° C.
  • the PASST+BeOH (which is the current commercial formulation modified by the addition of 0.004% polysorbate 20 and 0.9% benzyl alcohol) performed similarly to the current commercial formulation at both 4° C. and 25° C.
  • This study was a single-center, randomized, single-blind, crossover design in which 48 healthy men and women received single SC injections of 6 solutions.
  • Test formulations (detailed below in Table 9) were administered by a trained healthcare professional in 6 unique sequences with 8 subjects randomized to each sequence. Injections were administered in each quadrant of the anterior abdominal wall and administered approximately 1 hour apart. Within 30 seconds after each injection, subjects assessed their level of injection pain using a 100 mm Visual Analog Scale (VAS). Adverse events were collected from the beginning of the first injection through 30 days after the first injection. Safety follow-up phone calls were conducted on day 2 (24 hours after the sixth injection) and day 31 (+2 days).
  • VAS Visual Analog Scale
  • Test formulation 1.0 100 mM sodium chloride, 25 mM without sodium L-arginine, 1.0% (w/v) sucrose, phosphate 0.01% (w/v) polysorbate 20 E.
  • Test formulation 0.51 100 mM sodium chloride, 25 mM without sodium L-arginine, 1.0% (w/v) sucrose, phosphate 0.01% (w/v) polysorbate 20 F.
  • VAS scores were calculated for VAS scores by solution. VAS scores were analyzed using an analysis of variance (ANOVA) model, which included sequence, solution, and period as independent variables, and subject within sequence as a random effect. No adjustment was made for multiple comparisons.
  • ANOVA analysis of variance
  • Solution C non-product specific placebo with benzyl alcohol
  • Solution D non-product specific placebo without sodium phosphate
  • No significant differences in mean VAS scores were found between Solutions C and D, between Solution B (etanercept placebo) and Solution F (active etanercept), or between different injection volumes (0.51 and 1.0 mL).
  • Solution A negative pain control
  • Enbrel drug substance in PASS 25 mM phosphate buffer, 25 mM L-arginine, 100 mM NaCl, 1% sucrose was used for this study.
  • the material was diafiltered into PASS and SAS_100NaCl (25 mM L-arginine, 100 mM NaCl, 1% sucrose) at 50 mg/mL and then ultrafiltered to ⁇ 75 mg/mL.
  • the 50 mg/mL formulations were prepared by diluting the post-UF/DF PASS and SAS material with the corresponding solution.
  • the SAST_120NaCl was prepared by diluting the 75 mg/mL SAS material using a concentrated NaCl stock solution to achieve a final concentration of 120 mM NaCl.
  • a 1% stock solution of polysorbate 20 was prepared fresh and spiked into all formulation to a final concentration of 0.010%. All formulations were manually filled into 1 mL long BD glass syringes to a volume of 1 mL and then stoppered using an ASPU vacuuming stoppering unit.
  • the pH was measured using a Mettler Toledo SevenEasy pH meter combined with a Mettler Inlab MicroProbe. Samples were warmed to room temperature prior to measurements. Protein concentration measurements using absorbance at 280 nM for all samples were performed at room temperature using the DropSense96 UV/Vis Lab Chip DS system. Each sample was measured neat with at least three replicates (3 ⁇ L each), including a formulation solution blank. Osmolality was measured using The Advanced Osmometer Model 3900. Each measurement was performed using 250 ⁇ L of sample and 290 mOsm osmolality standards were tested to ensure the system was operating properly. Size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software.
  • Hydrophobic interaction HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software at an absorbance of 215 nm.
  • Denatured size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software.
  • Sub-visible particle analysis was performed using a HACH HIAC/Royco particle counter system equipped with an HRLD-150 laser and Pharm Spec software. All samples were diluted with PASS formulation buffer to 25 mg/mL. Samples were thoroughly mixed, uncapped and degassed for 2 hours at 75 torr prior to analysis. Four (4) sips of 1.0 mL each (no tare volume) were performed, with the first sip discarded and the remaining three sips averaged. Data for particle sizes 2, 5, 10, and m was collected at all time points. The results account for the dilution and are reported as cumulative counts per milliliter.
  • the protein concentration of all formulations was tested at time zero.
  • the protein concentration results for all samples can be found in Table 13. All samples met the acceptance criteria.
  • Osmolality was tested at time zero only. The osmolality results for all samples can be found in Table 14. All formulations were at their target osmolality. Due to differences in buffer and excipient levels, the osmolality was not expected to be the same across the various formulations.
  • Peak B is the amount of high molecular weight species (aggregate) that forms. Results showed no differences in Peak B between the PASST control and the buffer-less formulations at 4° C. and 25° C., with minor differences being observed after twelve weeks at 40° C. ( FIG. 1 ). Peak B represents the total aggregate detected by SE-HPLC for these formulations. All samples remained acceptable (Peak B ⁇ 6%,) after 52 weeks of storage at 4° C., 24 weeks of storage at 25° C., and after twelve weeks of storage at 40° C.
  • Sub-visible particles were monitored by light obscuration particle counting (HIAC). Results were in line with historical PFS data and were similar between the formulations across all temperatures after twelve weeks. No trends could be established from this data set, as a single vial containing three pooled syringes was used at each time point and there is a high level of syringe-to-syringe variability in the contribution to silicone oil droplets.
  • HIAC light obscuration particle counting
  • Osmolality pH Sample (mg/mL) (mOsm) 0 F/T 3 F/T 5 F/T PASS 47.9 310 6.30 6.29 6.27 SAST_100NaCl 48.8 259 6.19 6.18 6.16 SAST_120NaCl 48.1 294 6.17 6.19 6.18 SAS_120NaCl 48.6 300 6.17 6.17 6.18
  • Etanercept 25 mg/mL, in TMS (10 mM Tris HCl, 4% mannitol, 1% sucrose, pH 7.4); SAS_100NaCl solution (100 mM NaCl, 25 mM L-arginine HCl, 1% sucrose, pH 6.3) for dialysis; PASS buffer (25 mM Phosphate, 100 mM NaCl, 25 mM L-arginine HCl, 1% sucrose, pH 6.3); 10,000 MWCO centripreps; 3-12 mL Slide-A-Lyzer dialysis cassettes, 10,000 MWCO; a Mettler Toledo MP220 pH meter and Mettler Toledo InLab MicroProbe.
  • TMS 10 mM Tris HCl, 4% mannitol, 1% sucrose, pH 7.4
  • SAS_100NaCl solution 100 mM NaCl, 25 mM L-arginine HCl, 1% sucrose, pH 6.3
  • the formulation solution that was chosen following this study was termed SAS (120 mM sodium chloride, 25 mM L-arginine, 1% sucrose, pH 6.3), without added phosphate buffer. Since the previous example demonstrated that it was difficult to attain the target pH of 6.3 when either dialyzing or using UF/DF when starting from etanercept in a sample at pH 7.56, a different method of exchange into the SAS formulation was needed. Two methods were evaluated that utilized separate final UF/DF starting material: 1) column 3 (AEX) intermediate pool as the starting material, and 2) Enbrel drug substance in PASS formulation buffer (PASS DS intermediate pool) as the starting material. Each method is described below and summarizes the development of a final UF/DF unit operation step to produce 50 g/L SAS formulated etanercept, including preparation of SAS formulation solution, final UF/DF load conditioning and processing.
  • SAS 120 mM sodium chloride, 25 mM L-arginine, 1% sucrose, pH 6.3
  • the SAS formulation solution is composed of 120 mM sodium chloride, 25 mM L-arginine, 1% sucrose, pH 6.3.
  • An SAS formulation solution was titrated to pH 6.3 using 10 N NaOH.
  • the volume of titrant required to reach the specific pH range was 4.4 L/L SAS formulation solution.
  • the pH of the permeate remained close to the pH of WFI rather than the pH of the SAS formulation solution. Without being bound to a particular theory, this is believed to be due to the low buffering capacity of the SAS formulation solution.
  • the expected range for conductivity of the permeate following membrane equilibration using the range of the SAS formulation solution preparation is 12-16 mS/cm.
  • a higher post equilibration pH than that of the SAS formulation solution is expected and should not raise concern or indicate that the membranes are not equilibrated.
  • AEX Intermediate Pool Starting Material Prior to transferring the AEX intermediate pool into the retentate tank of an UF/DF tank, the pool was conditioned using 2 M HCl to a target pH of 6.3 (acceptable range 6.2-6.4). The volume of titrant required to reach the specific pH range was approximately 2.8 mL/L AEX intermediate pool.
  • the conditioned AEX intermediate pool can be held for up to 52.6 hours at controlled room temperature (CRT).
  • CRT controlled room temperature
  • the pH of the pool during the hold is shown in FIG. 8 .
  • the final UF/DF SAS pool generated using AEX intermediate pool as the starting material, can be held for up to 96.3 hours at CRT.
  • the pH and conductivity during the hold are shown in FIGS. 9 A and B. Over the 96.3 hour hold, the pH and conductivity remain within acceptable limits.
  • PASS DS Intermediate Pool Starting Material No conditioning is required prior to transferring the PASS DS intermediate pool into the UF/DF retentate tank because the PASS DS intermediate pool is already within the acceptable pH range. In addition, since the starting material is 50 mg/mL PASS formulated Enbrel DS, the pool does not need to be concentrated to 50 g/L because it is already at the correct concentration to perform diafiltration.
  • the product quality results for the final SAS UF/DF pool, generated using PASS DS intermediate pool as the starting material, are shown in Table 21.
  • the step yield for Run 1 was outside of the acceptance criteria; however, it was most likely an artifact of bench-scale processing and considered not significant to the conclusions of the study.
  • the final SAS UF/DF pool also met acceptance criteria for product quality using SEC and HIC analysis, as described above.
  • the PASS pool does not require conditioning prior to UF/DF processing with SAS solution because this intermediate pool is already at the target pH (6.3).
  • a pool hold study was not performed for this intermediate pool because the conditions of the pool were unchanged from Enbrel PASS DS.
  • the pool can be held for up to 96 hours at 25° C.
  • the final UF/DF SAS pool generated using PASS DS intermediate pool as the starting material, can be held for up to 96.3 hours at CRT.
  • the pH and conductivity during the hold are shown in FIGS. 9 A and B. Over the 96.3 hour hold, the pH and conductivity remain within acceptable limits.
  • the SAS formulation solution can be held for up to 28 days at CRT.
  • the pH and conductivity are shown in FIGS. 10 A and B. Over the 42 day hold in small scale stainless steel stability chambers with very small headspace, the SAS formulation solution is demonstrated to maintain a pH within 5.6 to 6.5. There was precipitation observed at the 35 day and 42 day time points. The 21 day time point measurement of 5.09 appears to be an outlier due to the fact that the subsequent time points are within the proposed acceptance criteria.
  • the final UF/DF unit operation can produce 50 g/L SAS formulation product and achieve consistent product quality compared to the current commercial PASS formulation product under the following process recommendations: 1) utilizing either AEX intermediate pool, or PASS DS intermediate pool, as the starting material 2) the SAS solution can be held for at least 28 days at CRT and maintain a pH of 5.6 to 6.5, 3) the conditioned AEX intermediate pool can be held at CRT for at least 52.6 hours and maintain a pH of 6.3 ⁇ 0.1, and 4) the SAS formulated UF/DF pool can be held at CRT for at least 96.3 hours and maintain a pH of 6.1 to 6.5 and a conductivity of 10 to 14 mS/cm.
  • the goal of this example was to determine the effect on aggregation of increased levels of arginine, sucrose or sodium chloride on etanercept stability at 75 mg/mL at 40° C.
  • the levels of these excipients were each increased to maintain an isotonic formulation without added phosphate buffer. Additionally, histidine was evaluated as a buffer to replace phosphate.
  • Table 22 The formulations tested are summarized in Table 22.
  • Enbrel drug substance in PASS 25 mM phosphate buffer, 25 mM L-arginine, 100 mM NaCl, 1% sucrose
  • the material was dialyzed into the new formulations (without polysorbate) and concentrated to using 75 mg/mL using 30,000 MWCO centripreps.
  • a 1% stock solution of polysorbate 20 was prepared fresh and spiked into all formulation to a final concentration of 0.01%. All formulations were manually filled into 1 mL long BD glass syringes to a volume of 1.0 mL and then stoppered using an ASPU vacuuming stoppering unit.
  • the pH was measured using a Mettler Toledo pH meter combined with a Mettler MicroProbe. Samples were warmed to room temperature prior to measurements. Osmolality was measured using The Advanced Osmometer Model 3900. Each measurement was performed using 250 ⁇ L of sample and 290 osmolality standards were tested to ensure the system was operating properly. Size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software.
  • Example 8 Stability of Formulations with Various Levels of Polysorbate 20
  • the results of the study showed that the 50 mg/mL formulations tested remained similar to the current commercial formulation after 24 weeks at recommended storage of 2-8° C., as well as at accelerated temperatures of 25° C. and 40° C.
  • the 100 mg/mL SAST formulation performed comparably to the 50 mg/mL formulations in terms of pH and subvisible particles; differences in aggregate levels by SEC were attributed to protein concentration.
  • Enbrel drug substance in TMS (10 mM tris buffer, 4% mannitol, 1% sucrose) at 25 mg/mL was used for this study.
  • the bulk used for the SAS formulation was titrated to pH 6.3.
  • the material was ultrafiltered to ⁇ 50 mg/mL etanercept, then diafiltered into PASS (25 mM phosphate, 25 mM L-arginine, 120 mM NaCl, 1% sucrose) or SAS (25 mM L-arginine, 120 mM NaCl, 1% sucrose) at 50 mg/mL etanercept.
  • the material for the high concentration arm was then ultrafiltered to 100 mg/mL etanercept.
  • a 1% stock solution of polysorbate 20 was prepared fresh and spiked into the formulations to the final concentrations as listed in Table 25. All formulations were manually filled into 1 mL long BD glass syringes to a volume of 1 mL and then stoppered using an ASPU vacuuming stoppering unit.
  • the pH of all formulations was measured at time zero and after twelve weeks at 40° C. and 24 weeks at 4° C. and 25° C. No trends were observed as a function of time or storage temperature.
  • the measured pH values for all samples can be found in Table 26. No drift in pH was observed after twelve weeks of storage at 40° C. or 24 weeks at 4° C. and 25° C. and all samples met the acceptance criteria of +/ ⁇ 0.2 pH units from the target pH of 6.3.
  • the protein concentration and osmolality of all formulations was tested at time zero. The protein concentration and osmolality results for all samples can be found in Table 26.
  • Peak B is the amount of high molecular weight species (aggregate) that forms. Results showed no differences in Peak B between the PASS control and the bufferless formulations at all temperatures at their respective protein concentrations (Table 27-29). Peak B represents the total aggregate detected by SE-HPLC for these formulations. All 50 mg/mL samples remained acceptable (Peak B ⁇ 6%,) after 24 weeks of storage at 4° C. and 25° C., and after 2 weeks of storage at 40° C.
  • Etanercept drug substance in TMS (10 mM tris buffer, 4% mannitol, 1% sucrose) at 25 mg/mL etanercept was used for this study.
  • the bulk used for the SAS formulation was titrated to pH 6.3. The material was ultrafiltered to ⁇ 50 mg/mL etanercept, then diafiltered into PASS (25 mM phosphate, 25 mM L-arginine, 120 mM NaCl, 1% sucrose) or SAS (25 mM L-arginine, 120 mM NaCl, 1% sucrose) at 50 mg/mL etanercept.
  • the pH was measured using a Mettler Toledo SevenEasy pH meter combined with a Mettler Inlab MicroProbe. Samples were warmed to room temperature prior to measurements. Protein concentration measurements using absorbance at 280 nM for all samples were performed at room temperature using the Nano Drop system. Size exclusion HPLC was run on an Agilent 1100 HPLC with Chromeleon 7.2 software. Sub-visible particle analysis was performed using a HACH HIAC/Royco particle counter system equipped with an HRLD-150 laser and Pharm Spec software. All samples were diluted with PASS formulation buffer to 25 mg/mL. Samples were thoroughly mixed, uncapped and degassed for 2 hours at 75 torr prior to analysis.
  • Stability in the plastic silicone oil free syringes is similar to stability in glass siliconized syringes.
  • the protein concentration of all formulations was tested at time zero.
  • the pH of all formulations was measured at time zero and after twelve weeks at 40° C. and 24 weeks at 4° C. and 25° C. No trends were observed as a function of time or storage temperature and all samples met the pH acceptance criteria of +/ ⁇ 0.2 pH units from the target pH of 6.3.
  • the protein concentration and measured pH values for all samples can be found in Table 31.
  • Peak B is the amount of high molecular weight species (aggregate) that forms. Results showed no differences in Peak B between the glass syringes and the COP plastic silicone oil free syringes (Table 32-34). Peak B represents the total aggregate detected by SE-HPLC for these formulations. All samples remained acceptable (Peak B ⁇ 6%,) after 24 weeks of storage at 4° C. and 25° C.
  • the long-term stability of the SAS formulation at 50 mg/mL etanercept and the current commercial etanercept formulation stored in glass siliconized syringes and COP silicone oil free syringes was assessed at 4° C., 25° C. and 40° C. No significant differences were observed between the formulations as a function of syringe type after 24 weeks by SE-HPLC. No drift in pH was observed and all formulations remained within acceptable ranges. Sub-visible particles were reduced in the COP silicone oil free plastic syringes and were consistent with historical PFS results when stored in the glass syringes. The results of the study showed that the SAS formulation at 50 mg/mL etanercept was stable and similar to the current commercial formulation after 24 weeks at the recommended storage temperature of 2° C. to 8° C. in various syringe types.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Inorganic Chemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US15/788,762 2016-10-21 2017-10-19 Pharmaceutical Formulations and Methods of Making the Same Abandoned US20180110856A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US15/788,762 US20180110856A1 (en) 2016-10-21 2017-10-19 Pharmaceutical Formulations and Methods of Making the Same
US15/958,261 US10307483B2 (en) 2016-10-21 2018-04-20 Pharmaceutical formulations and methods of making the same
US16/144,120 US11491223B2 (en) 2016-10-21 2018-09-27 Pharmaceutical formulations and methods of making the same
US17/933,055 US20230080571A1 (en) 2016-10-21 2022-09-16 Pharmaceutical formulations and methods of making the same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662411458P 2016-10-21 2016-10-21
US15/788,762 US20180110856A1 (en) 2016-10-21 2017-10-19 Pharmaceutical Formulations and Methods of Making the Same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/958,261 Continuation US10307483B2 (en) 2016-10-21 2018-04-20 Pharmaceutical formulations and methods of making the same

Publications (1)

Publication Number Publication Date
US20180110856A1 true US20180110856A1 (en) 2018-04-26

Family

ID=61971152

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/788,762 Abandoned US20180110856A1 (en) 2016-10-21 2017-10-19 Pharmaceutical Formulations and Methods of Making the Same
US15/958,261 Active US10307483B2 (en) 2016-10-21 2018-04-20 Pharmaceutical formulations and methods of making the same
US16/144,120 Active US11491223B2 (en) 2016-10-21 2018-09-27 Pharmaceutical formulations and methods of making the same
US17/933,055 Pending US20230080571A1 (en) 2016-10-21 2022-09-16 Pharmaceutical formulations and methods of making the same

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/958,261 Active US10307483B2 (en) 2016-10-21 2018-04-20 Pharmaceutical formulations and methods of making the same
US16/144,120 Active US11491223B2 (en) 2016-10-21 2018-09-27 Pharmaceutical formulations and methods of making the same
US17/933,055 Pending US20230080571A1 (en) 2016-10-21 2022-09-16 Pharmaceutical formulations and methods of making the same

Country Status (14)

Country Link
US (4) US20180110856A1 (zh)
EP (1) EP3528787A4 (zh)
JP (3) JP6884858B2 (zh)
KR (2) KR102446838B1 (zh)
CN (2) CN109982685B (zh)
AU (1) AU2017345490B2 (zh)
BR (1) BR112019007858A2 (zh)
CA (1) CA3040899A1 (zh)
CL (1) CL2019001053A1 (zh)
EA (1) EA201990998A1 (zh)
IL (1) IL266132B (zh)
MX (1) MX2019004580A (zh)
SG (1) SG11201903521XA (zh)
WO (1) WO2018075818A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10307483B2 (en) 2016-10-21 2019-06-04 Amgen Inc. Pharmaceutical formulations and methods of making the same
CN114206391A (zh) * 2019-08-09 2022-03-18 艾瑞克有限公司 包含抗体的新组合物
US11607451B2 (en) 2005-06-14 2023-03-21 Amgen Inc. Self-buffering antibody formulations

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202200615A (zh) 2020-03-12 2022-01-01 美商安進公司 用於治療和預防患者的crs之方法
US11777543B2 (en) 2020-05-12 2023-10-03 Nec Corporation Distortion compensation apparatus and distortion compensation method

Family Cites Families (156)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2837168A1 (de) 1978-08-25 1980-03-06 Blutspendedienst Dt Rote Kreuz Verfahren zur herstellung einer fuer die intravenoese anwendung geeigneten immunglobulinloesung
US4362661A (en) 1979-08-09 1982-12-07 Teijin Limited Immunoglobulin composition having a high monomer content, and process for production thereof
CA1153695A (en) 1979-08-30 1983-09-13 Syoji Ono S-sulfonated immunoglobulin composition having a high monomer content and a process for production thereof
US4374763A (en) 1979-09-17 1983-02-22 Morishita Pharmaceutical Co., Ltd. Method for producing gamma-globulin for use in intravenous administration and method for producing a pharmaceutical preparation thereof
US4396608A (en) 1981-08-24 1983-08-02 Cutter Laboratories Intravenously injectable immune serum globulin
US5702699A (en) 1982-09-23 1997-12-30 Cetus Corporation Process for the recovery of lipophilic proteins
US4681713A (en) 1984-03-15 1987-07-21 Toyo Boseki Kabushiki Kaisha Method of making a hollow fiber membrane for dialysis
US4597966A (en) 1985-01-09 1986-07-01 Ortho Diagnostic Systems, Inc. Histidine stabilized immunoglobulin and method of preparation
US6673347B1 (en) 1986-04-30 2004-01-06 Gryphon Therapeutics Polypeptide and protein derivatives and process for their preparation
GB8628104D0 (en) 1986-11-25 1986-12-31 Connaught Lab Pasteurization of immunoglobin solutions
DE3640513A1 (de) 1986-11-27 1988-06-09 Biotest Pharma Gmbh Verfahren zur herstellung eines virussicheren, lagerstabilen und intravenoes vertraeglichen immunglobulin-g-praeparates
JP2547556B2 (ja) 1987-02-06 1996-10-23 株式会社 ミドリ十字 r−グロブリンの液状製剤
WO1990005183A1 (en) 1988-10-31 1990-05-17 Immunex Corporation Interleukin-4 receptors
US5395760A (en) 1989-09-05 1995-03-07 Immunex Corporation DNA encoding tumor necrosis factor-α and -β receptors
DE59010908D1 (de) 1989-09-12 2000-08-10 Hoffmann La Roche TNF-bindende Proteine
DE3939346A1 (de) 1989-11-29 1991-06-06 Behringwerke Ag Arzneimitel zur subkutanen oder intramuskulaeren applikation enthaltend polypeptide
US5177194A (en) 1990-02-01 1993-01-05 Baxter International, Inc. Process for purifying immune serum globulins
US5945098A (en) 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
GB9017002D0 (en) 1990-08-02 1990-09-19 Health Lab Service Board Improved method for the purification of erwina l-asparaginase
JP3179538B2 (ja) 1990-12-11 2001-06-25 ノバルティス アクチエンゲゼルシャフト 安定なヒトカルシトニンの水性溶液
US5110910A (en) 1991-03-25 1992-05-05 Miles Inc. Virucidal euglobulin precipitation
US5256571A (en) 1991-05-01 1993-10-26 Cytyc Corporation Cell preservative solution
KR100240159B1 (ko) 1991-12-20 2000-01-15 킨 캐리 당단백질 gpiib/iiia와 반응성인 사람화된 면역글로불린
US6004555A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US5298410A (en) 1993-02-25 1994-03-29 Sterling Winthrop Inc. Lyophilized formulation of polyethylene oxide modified proteins with increased shelf-life
US5484892A (en) 1993-05-21 1996-01-16 Dana-Farber Cancer Institute, Inc. Monoclonal antibodies that block ligand binding to the CD22 receptor in mature B cells
DE4344824C1 (de) 1993-12-28 1995-08-31 Immuno Ag Hochkonzentriertes Immunglobulin-Präparat und Verfahren zu seiner Herstellung
GB9401448D0 (en) 1994-01-26 1994-03-23 Ciba Geigy Ag Stable dry powders
US5580856A (en) 1994-07-15 1996-12-03 Prestrelski; Steven J. Formulation of a reconstituted protein, and method and kit for the production thereof
IL114909A (en) 1994-08-12 1999-10-28 Immunomedics Inc Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
CN1300173C (zh) 1996-02-09 2007-02-14 艾博特生物技术有限公司 结合人TNFα的人抗体
TW518219B (en) 1996-04-26 2003-01-21 Chugai Pharmaceutical Co Ltd Erythropoietin solution preparation
GB9610992D0 (en) 1996-05-24 1996-07-31 Glaxo Group Ltd Concentrated antibody preparation
TW555765B (en) 1996-07-09 2003-10-01 Amgen Inc Low molecular weight soluble tumor necrosis factor type-I and type-II proteins
JP2000515857A (ja) 1996-07-18 2000-11-28 シーエスエル リミテッド 免疫グロブリン溶液の低温殺菌
TW491855B (en) 1996-08-07 2002-06-21 Csl Ltd Purification of immunoglobulins
EP0852951A1 (de) 1996-11-19 1998-07-15 Roche Diagnostics GmbH Stabile lyophilisierte pharmazeutische Zubereitungen von mono- oder polyklonalen Antikörpern
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US5886154A (en) 1997-06-20 1999-03-23 Lebing; Wytold R. Chromatographic method for high yield purification and viral inactivation of antibodies
JP3679406B2 (ja) 1998-05-20 2005-08-03 協和醗酵工業株式会社 遺伝子組換え抗体
US6436897B2 (en) 1998-06-01 2002-08-20 Celtrix Pharmaceuticals, Inc. Pharmaceutical formulations for IGF/IGFBP
ES2527915T3 (es) 1998-06-09 2015-02-02 Csl Behring Ag Producto de inmunoglobulina G (IgG) líquido
US6610206B1 (en) 1998-10-20 2003-08-26 Advanced Renal Technologies Buffered compositions for dialysis
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
US7294481B1 (en) 1999-01-05 2007-11-13 Immunex Corporation Method for producing recombinant proteins
DK2332978T3 (da) 1999-02-03 2014-06-23 Amgen Inc Nye polypeptider, der er involveret ved et immunrespons
IL145597A0 (en) 1999-04-08 2002-06-30 Genentech Inc Composition based on oppositely-charged polypeptides
JP2002541208A (ja) 1999-04-09 2002-12-03 オーソ−マクニール・フアーマシユーチカル・インコーポレーテツド エリトロポイエチンの薬剤組成物
US20010021380A1 (en) 1999-04-19 2001-09-13 Pluenneke John D. Soluble tumor necrosis factor receptor treatment of medical disorders
WO2000062790A2 (en) 1999-04-19 2000-10-26 Immunex Corporation Soluble tumor necrosis factor receptor treatment of medical disorders
US20040220103A1 (en) 1999-04-19 2004-11-04 Immunex Corporation Soluble tumor necrosis factor receptor treatment of medical disorders
CZ20021186A3 (cs) 1999-10-04 2002-11-13 Chiron Corporation Farmaceutické prostředky obsahující stabilizovaný kapalný polypeptid
ATE403437T1 (de) 1999-12-14 2008-08-15 Genentech Inc Lfa-1 antagonisten und tnf-alpha antagonisten zur behandlung von rheumatoiden arthritis
JP2004500063A (ja) 1999-12-16 2004-01-08 アムジェン インコーポレイテッド Tnfr/opg様分子およびその使用
BR0108193A (pt) 2000-02-10 2003-02-25 Wyeth Corp Processo para tratamento ou inibição de dano celular ou morte celular
CA2396793A1 (en) 2000-02-16 2001-08-23 Genentech, Inc. Uses of agonists and antagonists to modulate activity of tnf-related molecules
US20050209447A1 (en) 2000-07-25 2005-09-22 Takashi Ito Process for producing recombinant protein
EP1336410A4 (en) 2000-08-04 2005-10-12 Chugai Pharmaceutical Co Ltd PROTEIN INJECTION PREPARATIONS
EP2311492B1 (en) 2000-08-11 2017-10-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilized preparations
AU2002213441B2 (en) 2000-10-12 2006-10-26 Genentech, Inc. Reduced-viscosity concentrated protein formulations
PL363239A1 (en) 2000-12-14 2004-11-15 Fuijsawa Pharmaceutical Co, Ltd. Silensed anti-cd28 antibodies and use thereof
US6693173B2 (en) 2000-12-26 2004-02-17 Alpha Therapeutic Corporation Method to remove citrate and aluminum from proteins
EP1399483B1 (en) 2001-01-05 2010-04-14 Pfizer Inc. Antibodies to insulin-like growth factor i receptor
ES2184594B1 (es) 2001-01-17 2004-01-01 Probitas Pharma Sa Procedimiento para la produccion de gammaglobulina g humana inactivada de virus.
JP2005505494A (ja) 2001-02-23 2005-02-24 イミュネックス・コーポレーション 活性タンパク質の回収増加
GB0113179D0 (en) 2001-05-31 2001-07-25 Novartis Ag Organic compounds
US7187129B2 (en) 2001-06-25 2007-03-06 Koninklijke Philips Electronics, N.V. High pressure gas discharge lamp and method of manufacturing the same
KR20040023630A (ko) 2001-06-26 2004-03-18 아브게닉스, 인크. Opgl에 결합하는 항체
US7247304B2 (en) 2001-08-23 2007-07-24 Genmab A/S Methods of treating using anti-IL-15 antibodies
BR0212136A (pt) * 2001-08-23 2004-12-07 Genmab As Anticorpo monoclonal humano isolado, métodos para inibir a produção de tnfa em células t ou monócitos e a proliferação de célula t, hibridoma, transfectoma, animal não humano transgênico, método para produzir um anticorpo monoclonal humano, composição farmacêutica, métodos para tratar ou prevenir um distúrbio mediado pela il-15 humana, a psorìase e a artrite reumatóide, método para diagnosticar uma doença, ácido nucleico, e, vetor de expressão
AU2002305767B2 (en) 2001-09-20 2008-04-10 Cornell Research Foundation, Inc. Methods and compositions for treating and preventing skin disorders using binding agents specific for PSMA
KR100913714B1 (ko) 2001-11-08 2009-08-24 패시트 바이오테크 코포레이션 Igg 항체의 안정한 액상 약학 제형물
WO2003047510A2 (en) 2001-11-30 2003-06-12 Centocor, Inc. Anti-tnf antibodies, compositions, methods and uses
EP1463942B2 (en) 2001-12-21 2012-06-20 Immunex Corporation Methods for purifying protein
AU2003211990A1 (en) 2002-02-14 2003-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
AU2003215365A1 (en) 2002-02-21 2003-09-09 Duke University Treatment methods using anti-cd22 antibodies
US20030180287A1 (en) 2002-02-27 2003-09-25 Immunex Corporation Polypeptide formulation
EP1497444B1 (en) 2002-03-27 2015-11-04 Immunex Corporation Methods for increasing polypeptide production
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US20060234226A1 (en) 2002-04-26 2006-10-19 Fahner Robert L Non-affinity purification of proteins
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
GB0210121D0 (en) 2002-05-02 2002-06-12 Celltech R&D Ltd Biological products
EP1551875A4 (en) 2002-06-21 2006-06-28 Biogen Idec Inc TAMPON PREPARATIONS FOR CONCENTRATING ANTIBODIES AND METHODS FOR USE THEREOF
TWI430810B (zh) 2002-07-19 2014-03-21 Abbott Lab S A TNFα相關病症之治療
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
CA2504134C (en) 2002-11-01 2012-03-06 Bayer Healthcare Llc Process for concentration of macromolecules
US20040086532A1 (en) 2002-11-05 2004-05-06 Allergan, Inc., Botulinum toxin formulations for oral administration
MEP57508A (en) 2002-12-20 2011-05-10 Amgen Inc Binding agents which inhibit myostatin
EP1603948A1 (en) 2003-03-14 2005-12-14 Pharmacia Corporation Antibodies to igf-i receptor for the treatment of cancers
PL2335725T3 (pl) 2003-04-04 2017-04-28 Genentech, Inc. Preparaty zawierające wysokoskoncentrowane przeciwciała i białka
WO2004104021A2 (en) 2003-05-14 2004-12-02 Dow Corning Corporation Controlled release of active agents utilizing repeat sequence protein polymers
EP1631317A2 (en) 2003-06-06 2006-03-08 Regeneron Pharmaceuticals, Inc. Use of vegf inhibitors for tumor regression
ATE414106T1 (de) 2003-06-30 2008-11-15 Domantis Ltd Pegylierte single-domain-antikörper (dab)
UA93653C2 (ru) 2003-11-07 2011-03-10 Иммунекс Корпорейшн Выделенное антитело, kotopoe специфически связывается c человеческим рецептором il-4 (il-4r)
EP1532983A1 (en) 2003-11-18 2005-05-25 ZLB Bioplasma AG Immunoglobulin preparations having increased stability
CN1953768B (zh) 2004-02-12 2010-10-13 默克专利有限公司 抗-egfr抗体的高浓缩液体制剂
WO2005095454A1 (en) 2004-03-25 2005-10-13 Diadexus, Inc. Lng105 antibody composition and methods of use, and use of lng105 to assess lung cancer risk
CA2566075A1 (en) 2004-05-12 2005-12-01 Baxter Healthcare S.A. Microspheres comprising protein and showing injectability at high concentrations of said agent
WO2006003999A1 (ja) 2004-07-05 2006-01-12 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute ヒト抗ヒトb7rp-1抗体およびその抗体フラグメント
US7648742B2 (en) * 2004-07-29 2010-01-19 Merck Patent Gmbh Difluorosubstituted heterocyclic compounds and the use thereof in the form of components in liquid crystalline media
TW200621282A (en) 2004-08-13 2006-07-01 Wyeth Corp Stabilizing formulations
TWI364458B (en) 2004-08-27 2012-05-21 Wyeth Res Ireland Ltd Production of tnfr-lg
WO2006024497A1 (en) 2004-08-30 2006-03-09 Lonza Biologics Plc. Affinity- plus ion exchange- chromatography for purifying antibodies
US20060051347A1 (en) 2004-09-09 2006-03-09 Winter Charles M Process for concentration of antibodies and therapeutic products thereof
WO2006064373A2 (en) 2004-12-16 2006-06-22 Genzyme Polyclonals S.A.S. Methods of purifying immunologlobulins
JP4498939B2 (ja) 2005-02-01 2010-07-07 東京応化工業株式会社 ポジ型レジスト組成物およびレジストパターン形成方法
US8597709B2 (en) 2005-04-12 2013-12-03 Inovobiologic Inc. Dietary supplement and methods of use
AU2006259664A1 (en) 2005-06-14 2006-12-28 Amgen Inc. Self-buffering protein formulations
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070049732A1 (en) 2005-09-01 2007-03-01 Zurlo Eugene J Ultra-high yield intravenous immune globulin preparation
NZ567517A (en) 2005-09-21 2011-09-30 Burcon Nutrascience Mb Corp Preparation of canola protein isolate involving isoelectric precipitation
US8067350B2 (en) 2005-12-15 2011-11-29 Kimberly-Clark Worldwide, Inc. Color changing cleansing composition
US20070202051A1 (en) 2006-02-10 2007-08-30 Pari Gmbh Aerosols for sinunasal drug delivery
US20080003220A1 (en) 2006-04-21 2008-01-03 Amgen, Inc. Buffering agents for biopharmaceutical formulations
AU2007240507B2 (en) 2006-04-21 2013-07-18 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
JP2010503683A (ja) 2006-09-15 2010-02-04 バロフォールド, インコーポレイテッド 免疫原性低下のためのタンパク質の高圧処理
WO2008057240A2 (en) 2006-10-27 2008-05-15 Abbott Biotechnology Ltd. Crystalline anti-htnfalpha antibodies
MX2009006594A (es) 2006-12-21 2009-08-12 Amgen Inc Formulaciones de ph regulado y estables que contienen polipeptidos.
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
KR20190045414A (ko) * 2007-11-30 2019-05-02 애브비 바이오테크놀로지 리미티드 단백질 제형 및 이의 제조방법
AU2009210741A1 (en) * 2008-02-07 2009-08-13 Amgen Inc. Stabilized protein compositions
US8052645B2 (en) 2008-07-23 2011-11-08 Avant Medical Corp. System and method for an injection using a syringe needle
EP2276527B1 (en) 2008-05-20 2018-02-28 Avant Medical Corp. Autoinjector system
US8177749B2 (en) 2008-05-20 2012-05-15 Avant Medical Corp. Cassette for a hidden injection needle
WO2010032220A1 (en) * 2008-09-19 2010-03-25 Pfizer Inc. Stable liquid antibody formulation
MX345226B (es) * 2008-10-29 2017-01-20 Ablynx Nv Formulaciones de moleculas de union a antigeno de dominio sencillo.
AU2010221156A1 (en) 2009-03-06 2011-09-22 Genentech, Inc. Antibody formulation
US20110070227A1 (en) 2009-09-18 2011-03-24 Anna-Marie Novotney-Barry Treatment of Autoimmune and Inflammatory Diseases
MX344727B (es) 2010-11-11 2017-01-05 Abbvie Biotechnology Ltd Formulaciones liquidas de anticuerpos anti-tnf-alfa de alta concentracion mejoradas.
US9173999B2 (en) * 2011-01-26 2015-11-03 Kaleo, Inc. Devices and methods for delivering medicaments from a multi-chamber container
CA2833748C (en) 2011-04-20 2019-07-16 Amgen Inc. Autoinjector apparatus
EP2699265B1 (en) * 2011-04-20 2019-10-16 Sandoz AG STABLE PHARMACEUTICAL LIQUID FORMULATIONS OF THE FUSION PROTEIN TNFR:Fc
UY34105A (es) * 2011-06-03 2012-07-31 Lg Life Sciences Ltd Formulación líquida estable de etanercept
US10995130B2 (en) 2011-07-01 2021-05-04 Biogen Ma Inc. Arginine-free TNFR:Fc-fusion polypeptide compositions and methods of use
AU2012326171B2 (en) 2011-10-18 2017-03-09 Coherus Biosciences, Inc. Etanercept formulations stabilized with sodium chloride
JP2015519382A (ja) * 2012-06-12 2015-07-09 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 治療用抗体のための医薬処方物
JP6479655B2 (ja) 2012-07-05 2019-03-06 ユーエヌエル ホールディングス エルエルシーUNL Holdings LLC 注射可能カートリッジ用自動注射器及びその為の駆動制御機構
EP2869816A4 (en) * 2012-07-09 2016-04-20 Coherus Biosciences Inc FORMANTS OF ETANERCEPT HAVING A REDUCTION MARKED IN INVISIBLE PARTICLES IN THE NU
DK2895188T3 (en) * 2012-09-11 2018-02-26 Coherus Biosciences Inc CORRECTLY FOLDED ETHNECCEPT IN HIGH PURITY AND EXCELLENT YIELD
JP6431844B2 (ja) * 2012-10-26 2018-11-28 ルピン アトランティス ホールディングス エスエーLupin Atlantis Holdings Sa Tnfr:fc融合プロテインの安定な医薬組成物
EP2919812A4 (en) 2012-11-19 2016-05-18 Merck Sharp & Dohme Liquid formulations for TNFR: FC fusion proteins
JP6026002B2 (ja) * 2012-11-27 2016-11-16 アルテオジェン インコーポレイテッド タンパク質とFcドメインを融合した融合タンパク質の安定化用組成物
WO2014106116A2 (en) 2012-12-28 2014-07-03 Abbott Cardiovascular Systems Inc. Delivery of biologic therapeutics
EP2968760B1 (en) 2013-03-15 2024-01-03 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
JP6768501B2 (ja) 2013-03-15 2020-10-14 アムゲン・インコーポレーテッド 薬物カセット、自動注入機、および自動注入機システム
JP2016518386A (ja) 2013-05-02 2016-06-23 マブキシェンス エス.アー. TNFR:Fc融合ポリペプチドの代替配合物
KR102238065B1 (ko) * 2013-10-24 2021-04-07 아스트라제네카 아베 안정한 수성 항체 제제
JP6798882B2 (ja) 2013-11-29 2020-12-09 アレス トレーディング ソシエテ アノニム TNFR及びFc領域を含む融合タンパク質の液体製剤
EP2975050B1 (en) 2014-07-18 2019-05-29 Sandoz Ag Quantification of misfolded TNFR2:Fc
ES2834023T3 (es) 2014-08-28 2021-06-16 Unl Holdings Llc Sistemas de sensores para dispositivos de administración de fármacos
AU2015308659B2 (en) 2014-08-28 2020-07-02 Unitract Syringe Pty Ltd Skin sensors for drug delivery devices
CN107205925B (zh) * 2014-12-22 2020-12-11 阿雷斯贸易股份有限公司 液体药物组合物
WO2016149139A1 (en) 2015-03-13 2016-09-22 Samsung Bioepis Co., Ltd. Anti-tnf-alpha polypeptide composition and use thereof
US9763976B1 (en) 2016-08-02 2017-09-19 Zo Skin Health, Inc. Composition and method for treating skin conditions
SG11201903521XA (en) 2016-10-21 2019-05-30 Amgen Inc Pharmaceutical formulations and methods of making the same

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11607451B2 (en) 2005-06-14 2023-03-21 Amgen Inc. Self-buffering antibody formulations
US10307483B2 (en) 2016-10-21 2019-06-04 Amgen Inc. Pharmaceutical formulations and methods of making the same
US11491223B2 (en) 2016-10-21 2022-11-08 Amgen Inc. Pharmaceutical formulations and methods of making the same
CN114206391A (zh) * 2019-08-09 2022-03-18 艾瑞克有限公司 包含抗体的新组合物

Also Published As

Publication number Publication date
CA3040899A1 (en) 2018-04-26
US20190022217A1 (en) 2019-01-24
IL266132A (en) 2019-06-30
EP3528787A4 (en) 2020-05-06
SG11201903521XA (en) 2019-05-30
JP7402195B2 (ja) 2023-12-20
AU2017345490A1 (en) 2019-05-09
WO2018075818A1 (en) 2018-04-26
KR102413592B1 (ko) 2022-06-27
IL266132B (en) 2021-12-01
CN109982685B (zh) 2022-03-11
KR20210158878A (ko) 2021-12-31
EP3528787A1 (en) 2019-08-28
CN109982685A (zh) 2019-07-05
US10307483B2 (en) 2019-06-04
JP2021120393A (ja) 2021-08-19
KR102446838B1 (ko) 2022-09-26
CN114917185A (zh) 2022-08-19
BR112019007858A2 (pt) 2019-07-02
MX2019004580A (es) 2019-08-12
JP2019533679A (ja) 2019-11-21
AU2017345490B2 (en) 2022-07-07
JP2024010026A (ja) 2024-01-23
EA201990998A1 (ru) 2019-11-29
US11491223B2 (en) 2022-11-08
US20230080571A1 (en) 2023-03-16
US20180256718A1 (en) 2018-09-13
CL2019001053A1 (es) 2019-08-02
KR20190071760A (ko) 2019-06-24
JP6884858B2 (ja) 2021-06-09
CN114917185B (zh) 2023-11-14

Similar Documents

Publication Publication Date Title
US20230080571A1 (en) Pharmaceutical formulations and methods of making the same
AU2018274882B2 (en) Stable protein solution formulation containing high concentration of an anti-VEGF antibody
US11129876B2 (en) Etanercept formulations stabilized with amino acids
EP2726090B1 (en) Arginine - free tnfr : fc- fusion polypeptide compositions
US10258689B2 (en) Stable liquid formulation of etanercept
US9186401B2 (en) Concentrated human immunoglobulin composition
WO2017104778A1 (ja) 抗ヒトtslp受容体抗体含有医薬組成物
US11945859B2 (en) Protein solution formulation containing high concentration of an anti-VEGF antibody
EA041785B1 (ru) Фармацевтическая композиция и способ ее получения
KR20240053633A (ko) Vegf 수용체 융합 단백질을 위한 제제
JP2022546400A (ja) 高濃度の薬理学的に活性な抗体の新規製剤

Legal Events

Date Code Title Description
AS Assignment

Owner name: AMGEN INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GOSS, MONICA;BALL, NICOLE;REEL/FRAME:044918/0556

Effective date: 20180116

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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