US20130136733A1 - Stable Multi-Dose Compositions Comprising an Antibody and a Preservative - Google Patents

Stable Multi-Dose Compositions Comprising an Antibody and a Preservative Download PDF

Info

Publication number
US20130136733A1
US20130136733A1 US13/699,847 US201113699847A US2013136733A1 US 20130136733 A1 US20130136733 A1 US 20130136733A1 US 201113699847 A US201113699847 A US 201113699847A US 2013136733 A1 US2013136733 A1 US 2013136733A1
Authority
US
United States
Prior art keywords
composition
composition according
cresol
phenol
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/699,847
Inventor
Henrik Parshad
Dorthe Kot Engelund
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.)
Novo Nordisk AS
Original Assignee
Novo Nordisk AS
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 Novo Nordisk AS filed Critical Novo Nordisk AS
Priority to US13/699,847 priority Critical patent/US20130136733A1/en
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ENGELUND, DORTE KOT, PARSHAD, HENRIK
Publication of US20130136733A1 publication Critical patent/US20130136733A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies

Definitions

  • the invention relates to stable, multi-dose liquid compositions containing proteins, in particular, but not exclusively stable antibodies, and to the use of said compositions in therapy, in particular for the subcutaneous delivery of said stable protein.
  • Immunoglobulins, monoclonal antibodies (mAbs) and humanized antibodies have been in development as pharmaceutical products for a number of years.
  • multi-dose formulations of mAbs must contain antimicrobial agents to protect them from microbial contamination during multiple dosage withdrawal.
  • Multi-dose formulations containing preservatives offer several advantages over single dose containers. For example, product wastage is minimized because different sized doses may be obtained from the same container. Additionally, doses may be obtained from the same container over a period of time without the concern for microbial growth.
  • packaging is minimized because multiple doses are supplied in a single vial.
  • Aggregate formation by a polypeptide during storage of a liquid pharmaceutical composition can adversely affect biological activity of that polypeptide, resulting in loss of therapeutic efficacy of the pharmaceutical composition. Furthermore, aggregate formation may cause other problems such as blockage of tubing, membranes, or pumps when the polypeptide-containing pharmaceutical composition is administered using an infusion system.
  • US 2004/0009168 (Kaisheva et al) describes a multidose IgG formulation comprising a preservative.
  • WO 2008/071394 (F. Hoffmann-La Roche) describes a stable pharmaceutical parenteral formulation containing an amyloid-beta peptide antibody.
  • US 2007/0053871 (Li et al) describes a stable pharmaceutical formulation comprising a protein or antibody, a destabilizing concentration of preservative and a stabilizing concentration of osmolyte.
  • WO 00/15224 (Eli Lilly and Company) describes a stable, soluble formulation comprising a protein, hydrophobic preservative and nicotinamide.
  • WO 2008/121615 (MedImmune, Inc) describes a high concentration liquid formulation of an antibody.
  • WO 2009/070642 MedImmune, Inc describes stable lyophilised formulations of bispecific antibodies.
  • US 2004/0197324 (Genentech, Inc) describes high concentration antibody and protein formulations with reduced viscosity, that are stable, relatively isotonic and are of low turbidity.
  • US 2008/0112953 (Amgen, Inc) describes a stable formulation comprising an EGFR antibody and a glutamic acid buffer.
  • U.S. Pat. No. 6,875,432 (Genentech, Inc) describes a concentrated protein formulation with reduced viscosity.
  • a stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • a stable, multi-dose liquid composition as defined herein for use in therapy is provided.
  • a stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • Benzyl alcohol has been demonstrated to bind to and accelerate aggregation of partially unfolded proteins.
  • a concentration of 1% benzyl alcohol resulted in cloudiness and the formation of soluble aggregates.
  • Concentrations of benzyl alcohol greater than 2% in the same monoclonal antibody formulation resulted in precipitation of the protein.
  • stable composition refers to a composition with satisfactory physical stability, satisfactory chemical stability or satisfactory physical and chemical stability.
  • the term “physical stability” of the protein composition as used herein refers to the tendency of the protein to form biologically inactive and/or insoluble aggregates of the protein as a result of exposure of the protein to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces.
  • Physical stability of the aqueous protein compositions is evaluated by means of visual inspection and/or turbidity measurements after exposing the composition filled in suitable containers (e.g. cartridges or vials) It is an inherent quality of highly concentrated formulations of mabs to exhibit opalescence due to Raleigh scattering. Thus, a composition cannot be classified as physically unstable with respect to protein aggregation, when it shows visual turbidity in daylight. However, when there are precipitates or phase separation visible in day light the formulation is classified as physically unstable.
  • chemical stability of the protein composition refers to chemical covalent changes in the protein structure leading to formation of chemical degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native protein structure.
  • chemical degradation products can be formed depending on the type and nature of the native protein and the environment to which the protein is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the protein composition is well-known by the person skilled in the art.
  • Most proteins are prone to deamidation, a process in which the side chain amide group in glutaminyl or asparaginyl residues is hydrolysed to form a free carboxylic acid.
  • Oxidation (of for instance methionine residues) can be mentioned as another variant of chemical degradation.
  • the chemical stability of the protein composition can be evaluated by measuring the amount of the chemical degradation products at various time-points after exposure to different environmental conditions (the formation of degradation products can often be accelerated by for instance increasing temperature).
  • the amount of each individual degradation product is often determined by separation of the degradation products depending on molecule size and/or charge using various chromatography techniques (e.g. SEC-HPLC and/or RP-HPLC).
  • SEC-HPLC is in particular used for quantification of protein aggregates.
  • the samples may for instance be analysed using a TSK G3000 SWXL column, isocratic elution and subsequent UV detection at 214 nm.
  • This method is used to determine monomeric IgG content and % High Molecular Weight Proteins (HMWP) consisting of dimeric species or larger which are separated according to size by the gel resin.
  • the monomeric content and % HMWP are determined relative to the total protein content detected by the method.
  • a stable composition refers to a composition with satisfactory physical stability, satisfactory chemical stability or satisfactory physical and chemical stability.
  • a satisfactory stability of a formulation may be measured by the increase of % High Molecular Weight Proteins ( ⁇ % HMWP).
  • ⁇ % HMWP % High Molecular Weight Proteins
  • a satisfactory stability of a formulation may be one wherein the increase is less than 10% and preferably less than 5% of the protein found as an aggregate ( ⁇ % HMWP) in the formulation over the testing period.
  • a composition must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • protein means a compound composed of at least five constituent amino acids connected by peptide bonds.
  • the constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may be natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids.
  • Natural amino acids which are not encoded by the genetic code are e.g. hydroxyproline, y-carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine.
  • Synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e.
  • D-isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib (a-aminoisobutyric acid), Abu ( ⁇ -aminobutyric acid), Tle (tert-butylglycine), ⁇ -alanine, 3-aminomethyl benzoic acid and anthranilic acid.
  • preservative refers to pharmaceutically acceptable excipients which prevent the growth of micro-organisms within the composition. More particularly, the invention provides a preservative containing multi-dose liquid composition which protects the composition against microbial contamination.
  • the preservative is present within the composition in an amount of between 0.001 to 2% (w/v). In one embodiment, the preservative is present within the composition in an amount of between 0.002 to 1% (w/v).
  • the one or more preservative is selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof.
  • the one or more preservative is selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • the composition comprises a single preservative. In one embodiment, the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • phenol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.1 to 0.5% (w/v), such as 0.15 or 0.5% (w/v), in particular, 0.25 to 0.5% (w/v).
  • m-cresol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.1 to 0.5% (w/v), such as 0.15 or 0.35% (w/v), in particular, approximately 0.3% (w/v).
  • benzyl alcohol is typically present within the composition in an amount from 0.1 to 2% (w/v), such as 0.1 to 1.5% (w/v), such as 0.5 or 1.1% (w/v), in particular, approximately 1% (w/v).
  • chlorobutanol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.25 to 0.75% (w/v), such as 0.3 or 0.6% (w/v), in particular, approximately 0.3 to 0.5% (w/v).
  • methyl paraben is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as approximately 0.2% (w/v).
  • propyl paraben is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as approximately 0.2% (w/v).
  • phenoxyethanol is typically present within the composition in an amount from 0.1 to 2% (w/v), such as approximately 1% (w/v).
  • composition comprises thiomersal as a single preservative
  • thiomersal is typically present within the composition in an amount from 0.002 to 0.01% (w/v).
  • the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • the composition comprises two or more preservatives. In one embodiment, the composition comprises two preservatives. Data is presented herein which surprisingly demonstrates that a lower amount of aggregates were observed for compositions comprising two preservatives when compared with the aggregation observed for the individual preservatives. In a yet further embodiment, the composition comprises two preservatives selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • the composition comprises two preservatives selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • preservatives selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • examples of such compositions comprising two preservatives include: phenol and m-cresol; and benzyl alcohol and chlorobutanol.
  • the concentration of each individual preservative will typically be lower than when a single preservative is used.
  • the composition comprises phenol and m-cresol as a two preservative containing composition
  • phenol is typically present within the composition in an amount from 0.1 to 0.75% (w/v), such as 0.1 to 0.5% (w/v), e.g. 0.15 or 0.5% (w/v)
  • m-cresol is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as 0.15 to 0.4% (w/v), e.g. 0.18 or 0.35% (w/v).
  • composition comprises benzyl alcohol and chlorobutanol as a two preservative containing composition
  • benzyl alcohol is typically present within the composition in an amount from 0.25 to 1% (w/v), such as 0.4 to 0.9% (w/v), e.g. 0.5 or 0.8% (w/v)
  • chlorobutanol is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as 0.1 to 0.4% (w/v), e.g. 0.11 or 0.3% (w/v).
  • the composition additionally comprises a salt.
  • the salt can have a buffering capacity at the relevant pH.
  • the salt is an inorganic salt, or an organic salt or a combination of one or more of these.
  • the salt is selected from the group consisting of sodium chloride, magnesium chloride, sodium thiocyanate, ammonium thiocyanate, ammonium sulfate, ammonium chloride, calcium chloride, arginine hydrochloride, zinc chloride, sodium acetate, amino acids or a combination thereof.
  • the salt is sodium chloride or magnesium chloride, optionally in combination with other salts.
  • the salt is arginine hydrochloride.
  • the salt is a combination of an inorganic salt and arginine hydrochloride.
  • the salt is an amino acid. In one embodiment the L-stereoisomer of the amino acid is used. In one embodiment, the salt is selected from arginine, glycine, lysine, aspartic acid, or glutamic acid, or a combination thereof. In one embodiment, the amino acid is arginine or glycine. In one embodiment, the amino acid is arginine, such as L-arginine. The amino acid can be added to the composition in its salt form or in its free form, whatever is suitable.
  • the composition additionally comprises a buffer.
  • the buffer is a suitable pharmaceutically acceptable buffer, which comprises both a pharmaceutically acceptable base and a pharmaceutically acceptable acid.
  • the buffer has a pKa of between 4 and 8, such as 5 to 7.
  • the buffer may be a salt.
  • Examples of pharmaceutically acceptable acid and bases may include inorganic as well as organic non-toxic acid/bases such as is well-known in the art. Examples are dosodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, maleate, succinate, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-amino methane, or mixtures thereof. Each one of these specific buffers constitutes an alternative embodiment of the invention.
  • the pharmaceutically acceptable buffer comprises histidine, maleate, succinate, phosphate, or tris(hydroxymethyl)-amino methane.
  • the pharmaceutically acceptable buffer comprises histidine.
  • the buffer has a pKa value ⁇ 1 pH unit from the target pH of the composition.
  • the composition is buffered to a pH of between 5 and 7, such as a pH of 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0, or to a pH as defined by any ranges there between.
  • the composition is buffered to a pH of between 6.0 and 7.0, such as 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0, or to a pH as defined by any ranges there between.
  • the composition is buffered to a pH of between 6.0 and 6.5.
  • the composition is buffered to a pH of 6.0 or 6.5, such as 6.5.
  • the composition additionally comprises a surfactant.
  • the surfactant is selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypropylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic® F68, poloxamer 188 and 407, Triton X-100), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (Polysorbates, e.g.
  • Tween-20, Tween-40, Tween-80 and Brij-35 monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lectins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmitoyl phosphatidic acid) and lysophospholipids (eg.
  • phospholipids eg. dipalmitoyl phosphatidic acid
  • lysophospholipids eg.
  • ceramides e.g. sodium tauro-dihydrofusidate etc.
  • C6-C12 e.g.
  • acylcarnitines and derivatives N ⁇ -acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, N ⁇ -acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, N ⁇ -acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, DSS (docusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49-4]), docusate potassium, CAS registry no [7491-09-0]), SDS (sodium dodecyl sulphate or sodium lauryl sulphate), sodium caprylate, cholic acid or derivatives thereof, bile acids and salts thereof and glycine or taurine conjugates
  • DSS docusate sodium, CAS
  • N-alkyl-N,N-dimethylammonio-1-propanesulfonates 3-cholamido-1-propyldimethylammonio-1-propanesulfonate
  • cationic surfactants quaternary ammonium bases
  • cetyl-trimethylammonium bromide cetylpyridinium chloride
  • non-ionic surfactants eg. Dodecyl ⁇ -D-glucopyranoside
  • poloxamines eg.
  • Tetronic's which are tetrafunctional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention.
  • the surfactant is Tween 80 (i.e. polysorbate 80).
  • the surfactant is present within the composition in an amount of below 0.01%. In one embodiment, the surfactant is present within the composition in an amount of below 0.0075%, i.e between 0.001% and 0.005%, such as 0.001%. In one embodiment, no surfactant is present.
  • the composition additionally comprises a tonicity modifying agent.
  • suitable tonicity modifying agents include salts (e.g sodium chloride), polyhydric alcohols (e.g propyleneglycol, glycerol, xyllitol. mannitol or D-sorbitol), monosaccarides (glucose or maltose), di saccarides (e.g sucrose), amino acids (L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophane, threonine), polyethylen glycols (e.g PEG 400) or mixtures thereof.
  • salts e.g sodium chloride
  • polyhydric alcohols e.g propyleneglycol, glycerol, xyllitol. mannitol or D-sorbitol
  • monosaccarides glucose or maltose
  • di saccarides e.
  • the tonicity modifying agent is sucrose, mannitol or propylene glycol. In one embodiment, the tonicity modifying agent is sucrose.
  • the buffer and/or salt of the composition also acts as a tonicity modifier or the tonicity modifier will act as a buffer and/or salt (and the concentration of the tonicity modifier will therefore in such cases be calculated as such).
  • the tonicity modifying agent is present within the composition in an amount of between 50 and 250 mM, such as between 100 and 200 mM, for example any one of 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 or 200 or any ranges there between. In one embodiment, the tonicity modifying agent is present within the composition in an amount of 150 mM.
  • the composition is isotonic.
  • a stable, multi-dose liquid composition comprising a protein and one or more preservatives.
  • the protein is an immunoglobulin. In one embodiment, the protein is an antibody. In one embodiment, the protein is a monoclonal antibody (mAb). In one embodiment, the protein is an IgG4 antibody.
  • antibody covers monoclonal antibodies (including full length antibodies which have an immunoglobulin Fc region), antibody compositions with polyepitopic specificity, bispecific antibodies, diabodies, and single-chain molecules, as well as antibody fragments (e.g., Fab, F(ab′) 2 , and Fv).
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • Suitable antibodies which may be formulated in a stable composition of the invention include: 3F8, Abagovomab, Abciximab, ACZ885 (canakinumab), Adalimumab, Adecatumumab, Afelimomab, Afutuzumab, Alacizumab pegol, Alemtuzumab, Altumomab pentetate, Anatumomab mafenatox, Anrukinzumab (IMA-638), Apolizumab, Arcitumomab, Aselizumab, Atlizumab (tocilizumab), Atorolimumab, Bapineuzumab, Basiliximab, Bavituximab, Bectumomab, Belimumab, Bertilimumab, Besilesomab, Bevacizumab, Biciromab, Bivatuzumab mertansine, Blinatum
  • the antibody is selected from an Anti-IL-20, Anti-TFPI, Anti-IL-21, Anti-C5Ar, Anti-NKGDA or Anti-NKG2a antibody.
  • the antibody is a monoclonal Anti-IL-20 antibody. In one embodiment, the antibody is an Anti-IL-20 antibody as described in WO 2010/000721. In one embodiment, the Anti-IL-20 monoclonal antibody is 15D2 or 5B7 as described in WO 2010/000721.
  • the antibody is a monoclonal Anti-TFPI antibody. In one embodiment, the antibody is an Anti-TFPI antibody as described in PCT/EP2009/067598. In one embodiment, the Anti-TFPI monoclonal antibody is HzTFPI4F36 as described in PCT/EP2009/067598.
  • the protein is present within the composition in high concentrations.
  • the protein is present in a concentration of 50 mg/ml or more, such as 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300 mg/ml or more.
  • the protein is present within the composition in an amount of between 50 mg/ml and 300 mg/ml, for instance between 50 mg/ml and 250 mg/ml, such as between 50 mg/ml and 200 mg/ml, for instance between 50 mg/ml and 150 mg/ml.
  • the protein is present in a concentration of between 75 mg/ml and 300 mg/ml, for instance between 75 mg/ml and 250 mg/ml, such as between 75 mg/ml and 200 mg/ml, for instance between 75 mg/ml and 150 mg/ml. In one embodiment, the protein is present in a concentration of between 100 mg/ml and 300 mg/ml, for instance between 100 mg/ml and 250 mg/ml, such as between 100 mg/ml and 200 mg/ml, for instance between 100 mg/ml and 150 mg/ml.
  • a protein composition of the invention comprises:
  • a protein composition of the invention comprises:
  • a protein composition of the invention comprises:
  • a protein composition of the invention comprises:
  • a protein composition of the invention comprises:
  • compositions of the invention have surprisingly demonstrated stability towards formation of high molecular weight proteins (HMWP) at 40° C. for 4 weeks and at 5° C. and 40° C. for 3 months.
  • HMWP high molecular weight proteins
  • a pharmaceutical formulation comprising the antibody and the excipients (including one or more preservatives) is prepared.
  • the pharmaceutical compositions of the invention are stable for more than 6 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical compositions of the invention are stable for more than 4 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical compositions of the invention are stable for more than 4 weeks of usage and for more than 2 years of storage.
  • the pharmaceutical compositions of the invention are stable for more than 2 weeks of usage and for more than 2 years of storage.
  • the pharmaceutical compositions of the invention are stable for more than 1 week of usage and for more than 6 months of storage.
  • a stable, multi-dose liquid composition as defined herein for use in therapy is provided.
  • treatment means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder.
  • the term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relieve the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications.
  • the patient to be treated is preferably a mammal; in particular a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
  • the stable compositions of Anti-IL-20 antibodies of the present invention may be used in the treatment of an inflammatory disease, in particular autoinflammatory diseases, such as psoriasis, systemic lupus erythomatosus, rheumatoid arthritis, Crohn's disease and psoriatic arthritis or otherwise as described in WO 2010/000721.
  • autoinflammatory diseases such as psoriasis, systemic lupus erythomatosus, rheumatoid arthritis, Crohn's disease and psoriatic arthritis or otherwise as described in WO 2010/000721.
  • the invention provides a method of treating such an inflammatory disease which comprises administering to a patient a therapeutically effective amount of a stable composition of an Anti-IL-20 antibody of the present invention.
  • the invention also provides a stable composition of an Anti-IL-20 antibody of the present invention for use in the treatment of such an inflammatory disease.
  • the invention also provides the use of a stable composition of an Anti-IL-20 antibody of the present invention in the manufacture of a medicament for the treatment of such an inflammatory disease.
  • the invention also provides a stable pharmaceutical composition comprising a stable composition of an Anti-IL-20 antibody of the present invention for use in the treatment of such an inflammatory disease.
  • compositions of Anti-TFPI antibodies of the present invention may be used in the treatment of a coagulopathy (bleeding disorder), such as haemophilia A, with or without inhibitors, and haemophilia B, with or without inhibitors or otherwise as described in PCT/EP2009/067598.
  • a coagulopathy bleeding disorder
  • haemophilia A with or without inhibitors
  • haemophilia B with or without inhibitors or otherwise as described in PCT/EP2009/067598.
  • the invention provides a method of treating a coagulopathy which comprises administering to a patient a therapeutically effective amount of a stable composition of an Anti-TFPI antibody of the present invention.
  • the invention also provides a stable composition of an Anti-TFPI antibody of the present invention for use in the treatment of a coagulopathy.
  • the invention also provides the use of a stable composition of an Anti-TFPI antibody of the present invention in the manufacture of a medicament for the treatment of a coagulopathy.
  • the invention also provides a pharmaceutical composition comprising a stable composition of an Anti-TFPI antibody of the present invention for use in the treatment of a coagulopathy.
  • parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe.
  • parenteral administration can be performed by means of an infusion pump.
  • the formulations were prepared (see Table 1 below). The formulations were prepared from a stock solution containing ca. 150 mg/ml of the Anti-IL-20 antibody and 10 mM histidine buffer, pH 6.5. This stock solution was prepared by conventional UF/DF/UF. Stock solution of the excipients were prepared and mixed in the correct proportion. The final formulations were filled in 3 ml Penfill® cartridges, type 1 glass. The formulations were stored at 40° C. for 4 weeks and then analysed chemically, pharmaceutically and biophysically. The increase in the formation of protein aggregates (% HMWP) was measured by SEC-HPLC (as described above).
  • compositions containing 2 preservatives generally resulted in lower aggregate formation than the results achieved with the individual preservatives alone.
  • the composition containing 5 mg/ml phenol alone resulted in an increase of 2.5% HMWP
  • the composition containing 3.5 mg/ml m-cresol alone resulted in an increase of 3.1% HMWP
  • a composition containing a combination of these two preservatives at identical concentrations resulted in a lowering of aggregate formation (an increase of 1.2% HMWP).
  • An increase of 1.1% is seen for reference without preservative.
  • composition containing 3.5 mg/ml m-cresol resulted in the smallest increase in HMWP. After 3 months storage the increase in HMWP for this formulation is lower compared to reference without preservative.
  • composition containing phenol and m-cresol, respectively showed no increase in % HMWP over a period of 12 months at 5° C.
  • a preservative efficacy screening test was performed.
  • the efficacy of the preservative was measured using a modified USP/Ph Eur preservative efficacy test.
  • formulations were tested against Staphylococcus aureus . After inoculation, samples were stored for 6 and 24 hours at room temperature and the total bacterial counts were measured using a colony counter. The log reduction values were calculated as log (initial count/final count).
  • Embodiment 1 A stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • Embodiment 2 A composition according to embodiment 1, wherein the preservative is present within the composition in an amount of between 0.001 to 2% (w/v).
  • Embodiment 3 A composition according to embodiment 1 or 2, wherein the preservative is present within the composition in an amount of between 0.002 to 1% (w/v).
  • Embodiment 4 A composition according to any of embodiments 1 to 3, wherein the one or more preservative is selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof.
  • the one or more preservative is selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof.
  • Embodiment 5 A composition according to any of embodiments 1 to 4, wherein the one or more preservative is selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 6 A composition according to any of embodiments 1 to 5, wherein the composition comprises a single preservative.
  • Embodiment 7 A composition according to any of embodiments 1 to 6, wherein the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • a single preservative selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • Embodiment 8 A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 9 A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 10 A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount of 0.15 or 0.5% (w/v).
  • Embodiment 11 A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.25 to 0.5% (w/v).
  • Embodiment 12 A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 13 A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 14 A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount of 0.15 or 0.35% (w/v).
  • Embodiment 15 A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount of approximately 0.3% (w/v).
  • Embodiment 16 A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount from 0.1 to 2% (w/v).
  • Embodiment 17 A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount from 0.1 to 1.5% (w/v).
  • Embodiment 18 A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount of 0.5 or 1.1% (w/v).
  • Embodiment 19 A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount of approximately 1% (w/v).
  • Embodiment 20 A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 21 A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.25 to 0.75% (w/v).
  • Embodiment 22 A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount of 0.3 or 0.6% (w/v).
  • Embodiment 23 A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.3 to 0.5% (w/v).
  • Embodiment 24 A composition according to any of embodiments 1 to 7, which comprises methyl paraben as a single preservative, wherein said methyl paraben is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 25 A composition according to any of embodiments 1 to 7, which comprises methyl paraben as a single preservative, wherein said methyl paraben is present within the composition in an amount of approximately 0.2% (w/v).
  • Embodiment 26 A composition according to any of embodiments 1 to 7, which comprises propyl paraben as a single preservative, wherein said propyl paraben is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 27 A composition according to any of embodiments 1 to 7, which comprises propyl paraben as a single preservative, wherein said propyl paraben is present within the composition in an amount of approximately 0.2% (w/v).
  • Embodiment 28 A composition according to any of embodiments 1 to 7, which comprises phenoxyethanol as a single preservative, wherein said phenoxyethanol is present within the composition in an amount from 0.1 to 2% (w/v).
  • Embodiment 29 A composition according to any of embodiments 1 to 7, which comprises phenoxyethanol as a single preservative, wherein said phenoxyethanol is present within the composition in an amount of approximately 1% (w/v).
  • Embodiment 30 A composition according to any of embodiments 1 to 7, which comprises thiomersal as a single preservative, wherein said thiomersal is present within the composition in an amount from 0.002 to 0.01% (w/v).
  • Embodiment 31 A composition according to any of embodiments 1 to 7, which comprises a single preservative selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 32 A composition according to any of embodiments 1 to 5, which comprises two or more preservatives.
  • Embodiment 33 A composition according to embodiment 32, which comprises two preservatives.
  • Embodiment 34 A composition according to embodiment 32 or 33, which comprises two preservatives selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • Embodiment 35 A composition according to embodiment 34, which comprises two preservatives selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 36 A composition according to any of embodiments 32 to 35, wherein said two preservatives are phenol and m-cresol.
  • Embodiment 37 A composition according to embodiment 36, wherein phenol is present within the composition in an amount from 0.1 to 0.75% (w/v).
  • Embodiment 38 A composition according to embodiment 36 or 37, wherein phenol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 39 A composition according to any of embodiments 36 to 38, wherein phenol is present within the composition in an amount of 0.15 or 0.5% (w/v).
  • Embodiment 40 A composition according to any of embodiments 36 to 39, wherein m-cresol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 41 A composition according to any of embodiments 36 to 40, wherein m-cresol is present within the composition in an amount from 0.15 to 0.4% (w/v).
  • Embodiment 42 A composition according to any of embodiments 36 to 41, wherein m-cresol is present within the composition in an amount of 0.18 or 0.35% (w/v).
  • Embodiment 43 A composition according to any of embodiments 32 to 35, wherein said two preservatives are benzyl alcohol and chlorobutanol.
  • Embodiment 44 A composition according to embodiment 43, wherein benzyl alcohol is present within the composition in an amount from 0.25 to 1% (w/v).
  • Embodiment 45 A composition according to embodiment 43 or 44, wherein benzyl alcohol is present within the composition in an amount from 0.4 to 0.9% (w/v).
  • Embodiment 46 A composition according to any of embodiments 43 to 45, wherein benzyl alcohol is present within the composition in an amount of 0.5 or 0.8% (w/v).
  • Embodiment 47 A composition according to any of embodiments 43 to 46, wherein chlorobutanol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 48 A composition according to any of embodiments 43 to 47, wherein chlorobutanol is present within the composition in an amount from 0.1 to 0.4% (w/v).
  • Embodiment 49 A composition according to any of embodiments 43 to 48, wherein chlorobutanol is present within the composition in an amount of 0.11 or 0.3% (w/v).
  • Embodiment 50 A composition according to any embodiments 1 to 49, wherein a buffer is present, and the buffer has a pKa between 4 to 8.
  • Embodiment 51 A composition according to embodiment 50, wherein the buffer has a pKa between 5 to 7.
  • Embodiment 52 A composition according to embodiment 50 or 51, wherein a buffer is present, and the buffer is dosodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, maleate, succinate, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, or tris(hydroxymethyl)-amino methane, or mixtures thereof.
  • a buffer is present, and the buffer is dosodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, maleate, succinate, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, or tris(hydroxymethyl)-amino methane, or mixtures thereof.
  • Embodiment 53 A composition according to embodiment 52, wherein the buffer is histidine, maleate, succinate, phosphate, or tris(hydroxymethyl)-amino methane.
  • Embodiment 54 A composition according to embodiment 53, wherein the buffer is histidine.
  • Embodiment 55 A composition according to embodiment 54, wherein the buffer has a pKa value ⁇ 1 pH unit from the target pH of the composition.
  • Embodiment 56 A composition according to any of embodiments 1 to 55, wherein a salt is present and the salt is selected from the group consisting of sodium chloride, magnesium chloride, sodium thiocyanate, ammonium thiocyanate, ammonium sulfate, ammonium chloride, calcium chloride, arginine hydrochloride, zinc chloride and sodium acetate or any combination thereof.
  • Embodiment 57 A composition according to embodiment 56, wherein the salt is sodium chloride or magnesium chloride.
  • Embodiment 58 A composition according to embodiment 57, wherein the salt is sodium chloride.
  • Embodiment 59 A composition according to embodiment 56, wherein the salt is arginine-HCl.
  • Embodiment 60 A composition according to any of embodiments 1 to 59, which has a pH of between 5.0 and 7.0.
  • Embodiment 61 A composition according to embodiment 60, which has a pH of between 6.0 and 7.0.
  • Embodiment 62 A composition according to embodiment 61, which has a pH of 6.0 or 6.5.
  • Embodiment 63 A composition according to embodiment 62, which has a pH of 6.5.
  • Embodiment 64 A composition according to any of embodiments 1 to 63 which additionally comprises a surfactant.
  • Embodiment 65 A composition according to embodiment 64, wherein the surfactant is Tween 80 (i.e. polysorbate 80).
  • Embodiment 66 A composition according to embodiment 64 or 65, wherein the surfactant is present within the composition in an amount of below 0.01%.
  • Embodiment 67 A composition according to any of embodiments 64 to 66, wherein the surfactant is present within the composition in an amount of below 0.0075%.
  • Embodiment 68 A composition according to any of embodiments 64 to 67, wherein the surfactant is present within the composition in an amount between 0.001% and 0.005%.
  • Embodiment 69 A composition according to any of embodiments 64 to 68, wherein the surfactant is present within the composition in an amount of 0.001%.
  • Embodiment 70 A composition according to any of embodiments 1 to 63 wherein no surfactant is present.
  • Embodiment 71 A composition according to any of embodiments 1 to 70, which additionally comprises a tonicity modifying agent.
  • Embodiment 72 A composition according to embodiment 71, wherein the tonicity modifying agent is sucrose or propylene glycol.
  • Embodiment 73 A composition according to embodiment 72, wherein the tonicity modifying agent is sucrose.
  • Embodiment 74 A composition according to embodiment 72, wherein the tonicity modifying agent is propylene glycol.
  • Embodiment 75 A composition according to any of embodiments 71 to 74, wherein the tonicity modifying agent is present within the composition in an amount of between 50 and 250 mM.
  • Embodiment 76 A composition according to any of embodiments 71 to 75, wherein the tonicity modifying agent is present within the composition in an amount of between 100 and 200 mM.
  • Embodiment 77 A composition according to any of embodiments 71 to 76, wherein the tonicity modifying agent is present in an amount of 100 mM.
  • Embodiment 78 A composition according to any of embodiments 1 to 77, wherein the composition is pharmaceutically acceptable.
  • Embodiment 79 A stable composition according to any of embodiments 1 to 78,
  • the antibody is present within the composition in a concentration of between 50 mg/ml and 300 mg/ml.
  • Embodiment 80 A stable composition according to embodiment 79, wherein the antibody is present within the composition in a concentration of between 75 mg/ml and 300 mg/ml.
  • Embodiment 81 A stable composition according to embodiment 80, wherein the antibody is present within the composition in a concentration of between 100 mg/ml and 300 mg/ml.
  • Embodiment 82 A stable composition according to embodiment 81, wherein the antibody is present within the composition in a concentration of between 50 mg/ml and 200 mg/ml.
  • Embodiment 83 A stable composition according to embodiment 82, wherein the antibody is present within the composition in a concentration of 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, or 300 mg/ml.
  • Embodiment 84 A composition according to embodiment 1 which comprises:
  • Embodiment 85 A composition according to embodiment 1 which comprises:
  • Embodiment 86 A composition according to embodiment 1 which comprises:
  • Embodiment 87 A composition according to embodiment 1 which comprises:
  • Embodiment 88 A composition according to embodiment 1 which comprises:
  • Embodiment 89 A composition according to embodiment 1 which comprises:
  • Embodiment 90 A composition according to embodiment 1 which comprises:
  • Embodiment 91 A composition according to embodiment 1 which comprises:
  • Embodiment 92 A composition according to embodiment 1 which comprises:
  • Embodiment 93 A composition according to embodiment 1 which comprises:
  • Embodiment 94 A composition according to embodiment 1 which comprises:
  • Embodiment 95 A composition according any of embodiments 1-94, wherein the antibody is of the IgG4 subtype.
  • Embodiment 96 A composition according to any of embodiments 1 to 95, wherein the antibody is a monoclonal antibody.
  • Embodiment 97 A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 monoclonal antibody.
  • Embodiment 98 A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 monoclonal antibody as described in WO 2010/000721.
  • Embodiment 99 A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 antibody 15D2 or 5B7 as described in WO 2010/000721.
  • Embodiment 100 A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-TFPI monoclonal antibody.
  • Embodiment 101 A composition according to embodiment 96, wherein the monoclonal antibody is the Anti-TFPI monoclonal antibody HzTFPI4F36 as described in PCT/EP2009/067598.
  • Embodiment 102 A stable, multi-dose liquid composition according to any of embodiments 1 to 101 for use in therapy.
  • Embodiment 103 A method of treating an inflammatory disease which comprises administering to a patient a therapeutically effective amount of a composition according to any of embodiments 97-99.
  • Embodiment 104 A composition according to any of embodiments 97-99 for use in the treatment of an inflammatory disease.
  • Embodiment 105 Use of a composition according to any of embodiments 97-99 in the manufacture of a medicament for the treatment of an inflammatory disease.
  • Embodiment 106 A pharmaceutical composition comprising an Anti-IL-20 composition according to any of embodiments 97-99 for use in the treatment of an inflammatory disease.
  • Embodiment 107 A method of treating a coagulopathy which comprises administering to a patient a therapeutically effective amount of a composition according to any of embodiments 100-101.
  • Embodiment 108 A composition according to any of embodiments 100-101 for use in the treatment of a coagulopathy.
  • Embodiment 109 Use of a composition according to any of embodiments 100-101 in the manufacture of a medicament for the treatment of a coagulopathy.
  • Embodiment 110 A pharmaceutical composition comprising an Anti-TFPI composition according to any of embodiments 100-101 for use in the treatment of a coagulopathy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Dermatology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to stable, multi-dose liquid compositions containing proteins, in particular, but not exclusively stable antibodies, and to the use of said compositions in therapy, in particular for the subcutaneous delivery of said stable protein.

Description

    FIELD OF THE INVENTION
  • The invention relates to stable, multi-dose liquid compositions containing proteins, in particular, but not exclusively stable antibodies, and to the use of said compositions in therapy, in particular for the subcutaneous delivery of said stable protein.
  • BACKGROUND OF THE INVENTION
  • Immunoglobulins, monoclonal antibodies (mAbs) and humanized antibodies have been in development as pharmaceutical products for a number of years. There is a clear incentive for developing multi-dose formulations of mAbs due to the potential for repeated dosage which results in higher convenience for the patient. However, multi-dose formulations must contain antimicrobial agents to protect them from microbial contamination during multiple dosage withdrawal. Multi-dose formulations containing preservatives offer several advantages over single dose containers. For example, product wastage is minimized because different sized doses may be obtained from the same container. Additionally, doses may be obtained from the same container over a period of time without the concern for microbial growth. Furthermore, packaging is minimized because multiple doses are supplied in a single vial.
  • There is therefore a great need for a stable, multi-dose liquid pharmaceutical antibody composition.
  • However, the effect of the preservative on protein stability is a major concern. Anti-microbial preservatives are known to interact with proteins and cause stability problems such as aggregation. Thus, identifying formulation-compatible preservatives at concentrations that also provide the desired antimicrobial efficacy is a major challenge during drug product development. There is a general consensus that development of high-concentration formulations of mAbs poses serious challenges with respect to the physical and chemical stability of the mAb, such as increased formation of soluble as well as insoluble aggregates which enhance the probability of an immunogenic response as well as give rise to low bioactivity.
  • Aggregate formation by a polypeptide during storage of a liquid pharmaceutical composition can adversely affect biological activity of that polypeptide, resulting in loss of therapeutic efficacy of the pharmaceutical composition. Furthermore, aggregate formation may cause other problems such as blockage of tubing, membranes, or pumps when the polypeptide-containing pharmaceutical composition is administered using an infusion system.
  • US 2004/0009168 (Kaisheva et al) describes a multidose IgG formulation comprising a preservative. WO 2008/071394 (F. Hoffmann-La Roche) describes a stable pharmaceutical parenteral formulation containing an amyloid-beta peptide antibody. US 2007/0053871 (Li et al) describes a stable pharmaceutical formulation comprising a protein or antibody, a destabilizing concentration of preservative and a stabilizing concentration of osmolyte. WO 00/15224 (Eli Lilly and Company) describes a stable, soluble formulation comprising a protein, hydrophobic preservative and nicotinamide. WO 2008/121615 (MedImmune, Inc) describes a high concentration liquid formulation of an antibody. WO 2009/070642 (MedImmune, Inc) describes stable lyophilised formulations of bispecific antibodies. US 2004/0197324 (Genentech, Inc) describes high concentration antibody and protein formulations with reduced viscosity, that are stable, relatively isotonic and are of low turbidity. US 2008/0112953 (Amgen, Inc) describes a stable formulation comprising an EGFR antibody and a glutamic acid buffer. U.S. Pat. No. 6,875,432 (Genentech, Inc) describes a concentrated protein formulation with reduced viscosity. U.S. Pat. No. 6,685,940 (Genentech, Inc) describes a stable lyophilised protein formulation suitable for subcutaneous administration. US 2008/0071063 (MedImmune, Inc) describes a stable formulation comprising a variant Fc region that improves stability by reducing the propensity to rapidly aggregate.
  • SUMMARY OF THE INVENTION
  • According to a first aspect of the invention, there is provided a stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • According to a second aspect of the invention, there is provided a stable, multi-dose liquid composition as defined herein for use in therapy.
  • DETAILED DESCRIPTION OF THE INVENTION
  • According to a first aspect of the invention, there is provided a stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • It has been reported that monoclonal antibody formulations containing preservatives, such as phenol, result in soluble and insoluble protein aggregates. Although phenol is used in many peptide and protein-based drugs, there are several reports of interactions between this preservative and protein formulations. It has furthermore been reported that benzyl alcohol causes aggregation of recombinant human interferon-γ (rhIFN-γ), recombinant human granulocyte colony stimulating factor (rhGCSF), recombinant human interleukin-1 receptor antagonist (rhIL-1ra), and monoclonal antibody formulations. It is common knowledge that most protein aggregation is associated with protein conformational denaturation or instability. Benzyl alcohol has been demonstrated to bind to and accelerate aggregation of partially unfolded proteins. In studies with a monoclonal antibody formulation, a concentration of 1% benzyl alcohol resulted in cloudiness and the formation of soluble aggregates. Concentrations of benzyl alcohol greater than 2% in the same monoclonal antibody formulation resulted in precipitation of the protein.
  • Data is presented herein which surprisingly shows that formulations containing an antibody in combination with a variety of differing preservatives, resulted in formulations with a low content of aggregates during 4 weeks of storage at 40° C. and storage at 5° C. and 40° C. for 3 months.
  • The term “stable composition” refers to a composition with satisfactory physical stability, satisfactory chemical stability or satisfactory physical and chemical stability.
  • The term “physical stability” of the protein composition as used herein refers to the tendency of the protein to form biologically inactive and/or insoluble aggregates of the protein as a result of exposure of the protein to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces. Physical stability of the aqueous protein compositions is evaluated by means of visual inspection and/or turbidity measurements after exposing the composition filled in suitable containers (e.g. cartridges or vials) It is an inherent quality of highly concentrated formulations of mabs to exhibit opalescence due to Raleigh scattering. Thus, a composition cannot be classified as physically unstable with respect to protein aggregation, when it shows visual turbidity in daylight. However, when there are precipitates or phase separation visible in day light the formulation is classified as physically unstable.
  • The term “chemical stability” of the protein composition as used herein refers to chemical covalent changes in the protein structure leading to formation of chemical degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native protein structure. Various chemical degradation products can be formed depending on the type and nature of the native protein and the environment to which the protein is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the protein composition is well-known by the person skilled in the art. Most proteins are prone to deamidation, a process in which the side chain amide group in glutaminyl or asparaginyl residues is hydrolysed to form a free carboxylic acid. Other degradation pathways involve formation of high molecular weight transformation products where two or more protein molecules are covalently bound to each other through transamidation and/or disulfide interactions leading to formation of covalently bound dimer, oligomer and polymer degradation products (Stability of Protein Pharmaceuticals, Ahern. T. J. & Manning M. C., Plenum Press, New York 1992).
  • Oxidation (of for instance methionine residues) can be mentioned as another variant of chemical degradation. The chemical stability of the protein composition can be evaluated by measuring the amount of the chemical degradation products at various time-points after exposure to different environmental conditions (the formation of degradation products can often be accelerated by for instance increasing temperature). The amount of each individual degradation product is often determined by separation of the degradation products depending on molecule size and/or charge using various chromatography techniques (e.g. SEC-HPLC and/or RP-HPLC).
  • SEC-HPLC is in particular used for quantification of protein aggregates. The samples may for instance be analysed using a TSK G3000 SWXL column, isocratic elution and subsequent UV detection at 214 nm. This method is used to determine monomeric IgG content and % High Molecular Weight Proteins (HMWP) consisting of dimeric species or larger which are separated according to size by the gel resin. The monomeric content and % HMWP are determined relative to the total protein content detected by the method.
  • Hence, as outlined above, a stable composition refers to a composition with satisfactory physical stability, satisfactory chemical stability or satisfactory physical and chemical stability. A satisfactory stability of a formulation may be measured by the increase of % High Molecular Weight Proteins (Δ% HMWP). A satisfactory stability of a formulation may be one wherein the increase is less than 10% and preferably less than 5% of the protein found as an aggregate (Δ% HMWP) in the formulation over the testing period. In general, a composition must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • The term “protein”, “polypeptide” and “peptide” as used herein means a compound composed of at least five constituent amino acids connected by peptide bonds. The constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may be natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids. Natural amino acids which are not encoded by the genetic code are e.g. hydroxyproline, y-carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine. Synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e. D-isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib (a-aminoisobutyric acid), Abu (α-aminobutyric acid), Tle (tert-butylglycine), β-alanine, 3-aminomethyl benzoic acid and anthranilic acid.
  • The term “preservative” refers to pharmaceutically acceptable excipients which prevent the growth of micro-organisms within the composition. More particularly, the invention provides a preservative containing multi-dose liquid composition which protects the composition against microbial contamination.
  • In one embodiment, the preservative is present within the composition in an amount of between 0.001 to 2% (w/v). In one embodiment, the preservative is present within the composition in an amount of between 0.002 to 1% (w/v).
  • In one embodiment, the one or more preservative is selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof. In one embodiment, the one or more preservative is selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • In one embodiment, the composition comprises a single preservative. In one embodiment, the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • When the composition comprises phenol as a single preservative, phenol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.1 to 0.5% (w/v), such as 0.15 or 0.5% (w/v), in particular, 0.25 to 0.5% (w/v).
  • When the composition comprises m-cresol as a single preservative, m-cresol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.1 to 0.5% (w/v), such as 0.15 or 0.35% (w/v), in particular, approximately 0.3% (w/v).
  • When the composition comprises benzyl alcohol as a single preservative, benzyl alcohol is typically present within the composition in an amount from 0.1 to 2% (w/v), such as 0.1 to 1.5% (w/v), such as 0.5 or 1.1% (w/v), in particular, approximately 1% (w/v).
  • When the composition comprises chlorobutanol as a single preservative, chlorobutanol is typically present within the composition in an amount from 0.1 to 1% (w/v), such as 0.25 to 0.75% (w/v), such as 0.3 or 0.6% (w/v), in particular, approximately 0.3 to 0.5% (w/v).
  • When the composition comprises methyl paraben as a single preservative, methyl paraben is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as approximately 0.2% (w/v).
  • When the composition comprises propyl paraben as a single preservative, propyl paraben is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as approximately 0.2% (w/v).
  • When the composition comprises phenoxyethanol as a single preservative, phenoxyethanol is typically present within the composition in an amount from 0.1 to 2% (w/v), such as approximately 1% (w/v).
  • When the composition comprises thiomersal as a single preservative, thiomersal is typically present within the composition in an amount from 0.002 to 0.01% (w/v).
  • In a yet further embodiment, the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • In one embodiment, the composition comprises two or more preservatives. In one embodiment, the composition comprises two preservatives. Data is presented herein which surprisingly demonstrates that a lower amount of aggregates were observed for compositions comprising two preservatives when compared with the aggregation observed for the individual preservatives. In a yet further embodiment, the composition comprises two preservatives selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal. In a still yet further embodiment, the composition comprises two preservatives selected from phenol, m-cresol, benzyl alcohol and chlorobutanol. Examples of such compositions comprising two preservatives include: phenol and m-cresol; and benzyl alcohol and chlorobutanol.
  • It will be appreciated that when two or more preservatives are present within the composition, the concentration of each individual preservative will typically be lower than when a single preservative is used.
  • For example, when the composition comprises phenol and m-cresol as a two preservative containing composition, phenol is typically present within the composition in an amount from 0.1 to 0.75% (w/v), such as 0.1 to 0.5% (w/v), e.g. 0.15 or 0.5% (w/v) and m-cresol is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as 0.15 to 0.4% (w/v), e.g. 0.18 or 0.35% (w/v).
  • Furthermore, when the composition comprises benzyl alcohol and chlorobutanol as a two preservative containing composition, benzyl alcohol is typically present within the composition in an amount from 0.25 to 1% (w/v), such as 0.4 to 0.9% (w/v), e.g. 0.5 or 0.8% (w/v) and chlorobutanol is typically present within the composition in an amount from 0.1 to 0.5% (w/v), such as 0.1 to 0.4% (w/v), e.g. 0.11 or 0.3% (w/v).
  • In one embodiment, the composition additionally comprises a salt. In some embodiments, the salt can have a buffering capacity at the relevant pH. In one embodiment, the salt is an inorganic salt, or an organic salt or a combination of one or more of these. In one embodiment, the salt is selected from the group consisting of sodium chloride, magnesium chloride, sodium thiocyanate, ammonium thiocyanate, ammonium sulfate, ammonium chloride, calcium chloride, arginine hydrochloride, zinc chloride, sodium acetate, amino acids or a combination thereof.
  • In one embodiment, the salt is sodium chloride or magnesium chloride, optionally in combination with other salts. In one embodiment, the salt is arginine hydrochloride. In one embodiment, the salt is a combination of an inorganic salt and arginine hydrochloride.
  • In one embodiment, the salt is an amino acid. In one embodiment the L-stereoisomer of the amino acid is used. In one embodiment, the salt is selected from arginine, glycine, lysine, aspartic acid, or glutamic acid, or a combination thereof. In one embodiment, the amino acid is arginine or glycine. In one embodiment, the amino acid is arginine, such as L-arginine. The amino acid can be added to the composition in its salt form or in its free form, whatever is suitable.
  • In one embodiment, the composition additionally comprises a buffer. In one embodiment, the buffer is a suitable pharmaceutically acceptable buffer, which comprises both a pharmaceutically acceptable base and a pharmaceutically acceptable acid. In one embodiment, the buffer has a pKa of between 4 and 8, such as 5 to 7. In some embodiments, the buffer may be a salt.
  • Examples of pharmaceutically acceptable acid and bases may include inorganic as well as organic non-toxic acid/bases such as is well-known in the art. Examples are dosodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, maleate, succinate, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-amino methane, or mixtures thereof. Each one of these specific buffers constitutes an alternative embodiment of the invention. In one embodiment, the pharmaceutically acceptable buffer comprises histidine, maleate, succinate, phosphate, or tris(hydroxymethyl)-amino methane. In one embodiment, the pharmaceutically acceptable buffer comprises histidine.
  • In one embodiment, the buffer has a pKa value ±1 pH unit from the target pH of the composition.
  • In one embodiment, the composition is buffered to a pH of between 5 and 7, such as a pH of 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0, or to a pH as defined by any ranges there between. In one embodiment, the composition is buffered to a pH of between 6.0 and 7.0, such as 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9 or 7.0, or to a pH as defined by any ranges there between. In one embodiment, the composition is buffered to a pH of between 6.0 and 6.5. In one embodiment, the composition is buffered to a pH of 6.0 or 6.5, such as 6.5.
  • In one embodiment, the composition additionally comprises a surfactant. In one embodiment of the invention the surfactant is selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypropylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic® F68, poloxamer 188 and 407, Triton X-100), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (Polysorbates, e.g. Tween-20, Tween-40, Tween-80 and Brij-35), monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lectins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmitoyl phosphatidic acid) and lysophospholipids (eg. palmitoyl lysophosphatidyl-L-serine and 1-acyl-sn-glycero-3-phosphate esters of ethanolamine, choline, serine or threonine) and alkyl, alkoxyl (alkyl ester), alkoxy (alkyl ether)-derivatives of lysophosphatidyl and phosphatidylcholines, e.g. lauroyl and myristoyl derivatives of lysophosphatidylcholine, dipalmitoylphosphatidylcholine, and modifications of the polar head group, that is cholines, ethanolamines, phosphatidic acid, serines, threonines, glycerol, inositol, and the positively charged DODAC, DOTMA, DCP, BISHOP, lysophosphatidylserine and lysophosphatidylthreonine, and glycerophospholipids (eg. cephalins), glyceroglycolipids (eg. galactopyransoide), sphingoglycolipids (eg. ceramides, gangliosides), dodecylphosphocholine, hen egg lysolecithin, fusidic acid derivatives-(e.g. sodium tauro-dihydrofusidate etc.), long-chain fatty acids and salts thereof C6-C12 (eg. oleic acid and caprylic acid), acylcarnitines and derivatives, Nα-acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, Nα-acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, Nα-acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, DSS (docusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49-4]), docusate potassium, CAS registry no [7491-09-0]), SDS (sodium dodecyl sulphate or sodium lauryl sulphate), sodium caprylate, cholic acid or derivatives thereof, bile acids and salts thereof and glycine or taurine conjugates, ursodeoxycholic acid, sodium cholate, sodium deoxycholate, sodium taurocholate, sodium glycocholate, N-Hexadecyl-N,N-dimethyl-3-ammonio-1-propanesulfonate, anionic (alkyl-aryl-sulphonates) monovalent surfactants, zwitterionic surfactants (e.g. N-alkyl-N,N-dimethylammonio-1-propanesulfonates, 3-cholamido-1-propyldimethylammonio-1-propanesulfonate, cationic surfactants (quaternary ammonium bases) (e.g. cetyl-trimethylammonium bromide, cetylpyridinium chloride), non-ionic surfactants (eg. Dodecyl β-D-glucopyranoside), poloxamines (eg. Tetronic's), which are tetrafunctional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention. In one embodiment, the surfactant is Tween 80 (i.e. polysorbate 80).
  • The use of a surfactant in pharmaceutical compositions is well-known to the skilled person. For convenience, reference is made to Remington: The Science and Practice of Pharmacy, 20th edition, 2000.
  • In one embodiment, the surfactant is present within the composition in an amount of below 0.01%. In one embodiment, the surfactant is present within the composition in an amount of below 0.0075%, i.e between 0.001% and 0.005%, such as 0.001%. In one embodiment, no surfactant is present.
  • In one embodiment, the composition additionally comprises a tonicity modifying agent. Examples of suitable tonicity modifying agents include salts (e.g sodium chloride), polyhydric alcohols (e.g propyleneglycol, glycerol, xyllitol. mannitol or D-sorbitol), monosaccarides (glucose or maltose), di saccarides (e.g sucrose), amino acids (L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophane, threonine), polyethylen glycols (e.g PEG 400) or mixtures thereof. In one embodiment, the tonicity modifying agent is sucrose, mannitol or propylene glycol. In one embodiment, the tonicity modifying agent is sucrose. In some embodiments, the buffer and/or salt of the composition (as described above) also acts as a tonicity modifier or the tonicity modifier will act as a buffer and/or salt (and the concentration of the tonicity modifier will therefore in such cases be calculated as such).
  • In one embodiment, the tonicity modifying agent is present within the composition in an amount of between 50 and 250 mM, such as between 100 and 200 mM, for example any one of 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 or 200 or any ranges there between. In one embodiment, the tonicity modifying agent is present within the composition in an amount of 150 mM.
  • In one embodiment, the composition is isotonic.
  • According to a further aspect of the invention, there is provided a stable, multi-dose liquid composition comprising a protein and one or more preservatives.
  • In one embodiment, the protein is an immunoglobulin. In one embodiment, the protein is an antibody. In one embodiment, the protein is a monoclonal antibody (mAb). In one embodiment, the protein is an IgG4 antibody.
  • The term “antibody” covers monoclonal antibodies (including full length antibodies which have an immunoglobulin Fc region), antibody compositions with polyepitopic specificity, bispecific antibodies, diabodies, and single-chain molecules, as well as antibody fragments (e.g., Fab, F(ab′)2, and Fv).
  • The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • Examples of suitable antibodies, which may be formulated in a stable composition of the invention include: 3F8, Abagovomab, Abciximab, ACZ885 (canakinumab), Adalimumab, Adecatumumab, Afelimomab, Afutuzumab, Alacizumab pegol, Alemtuzumab, Altumomab pentetate, Anatumomab mafenatox, Anrukinzumab (IMA-638), Apolizumab, Arcitumomab, Aselizumab, Atlizumab (tocilizumab), Atorolimumab, Bapineuzumab, Basiliximab, Bavituximab, Bectumomab, Belimumab, Bertilimumab, Besilesomab, Bevacizumab, Biciromab, Bivatuzumab mertansine, Blinatumomab, Brentuximab vedotin, Briakinumab, Canakinumab, Cantuzumab mertansine, Capromab pendetide, Catumaxomab, Cedelizumab, Certolizumab pegol, Cetuximab, Citatuzumab bogatox, Cixutumumab, Clenoliximab, Clivatuzumab tetraxetan, CNTO 148 (golimumab), CNTO 1275 (ustekinumab), Conatumumab, Dacetuzumab, Daclizumab, Denosumab, Detumomab, Dorlimomab aritox, Dorlixizumab, Ecromeximab, Eculizumab, Edobacomab, Edrecolomab, Efalizumab, Efungumab, Elsilimomab, Enlimomab pegol, Epitumomab cituxetan, Epratuzumab, Erlizumab, Ertumaxomab, Etaracizumab, Exbivirumab, Fanolesomab, Faralimomab, Felvizumab, Fezakinumab, Figitumumab, Fontolizumab, Foravirumab, Fresolimumab, Galiximab, Gantenerumab, Gavilimomab, Gemtuzumab ozogamicin, Golimumab, Gomiliximab, Ibalizumab, Ibritumomab tiuxetan, Igovomab, lmciromab, Infliximab, Intetumumab, Inolimomab, Inotuzumab ozogamicin, Ipilimumab, Iratumumab, Keliximab, Labetuzumab, Lebrikizumab, Lemalesomab, Lerdelimumab, Lexatumumab, Libivirumab, Lintuzumab, Lucatumumab, Lumiliximab, Mapatumumab, Maslimomab, Matuzumab, Mepolizumab, Metelimumab, Milatuzumab, Minretumomab, Mitumomab, Morolimumab, Motavizumab, Muromonab-CD3, MYO-029 (stamulumab), Nacolomab tafenatox, Naptumomab estafenatox, Natalizumab, Nebacumab, Necitumumab, Nerelimomab, Nimotuzumab, Nofetumomab merpentan, Ocrelizumab, Odulimomab, Ofatumumab, Omalizumab, Oportuzumab monatox, Oregovomab, Otelixizumab, Pagibaximab, Palivizumab, Panitumumab, Panobacumab, Pascolizumab, Pemtumomab, Pertuzumab, Pexelizumab, Pintumomab, Priliximab, Pritumumab, PRO 140, Rafivirumab, Ramucirumab, Ranibizumab, Raxibacumab, Regavirumab, Reslizumab, Rilotumumab, Rituximab, Robatumumab, Rontalizumab, Rovelizumab, Ruplizumab, Satumomab, Sevirumab, Sibrotuzumab, Sifalimumab, Siltuximab, Siplizumab, Solanezumab, Sonepcizumab, Sontuzumab, Stamulumab, Sulesomab, Tacatuzumab tetraxetan, Tadocizumab, Talizumab, Tanezumab, Taplitumomab paptox, Tefibazumab, Telimomab aritox, Tenatumomab, Teneliximab, Teplizumab, TGN1412, Ticilimumab (tremelimumab), Tigatuzumab, TNX-355 (ibalizumab), TNX-650, TNX-901 (talizumab), Tocilizumab (atlizumab), Toralizumab, Tositumomab, Trastuzumab, Tremelimumab, Tucotuzumab celmoleukin, Tuvirumab, Urtoxazumab, Ustekinumab, Vapaliximab, Vedolizumab, Veltuzumab, Vepalimomab, Visilizumab, Volociximab, Votumumab, Zalutumumab, Zanolimumab, Ziralimumab, Zolimomab aritox and the like.
  • In one embodiment, the antibody is selected from an Anti-IL-20, Anti-TFPI, Anti-IL-21, Anti-C5Ar, Anti-NKGDA or Anti-NKG2a antibody.
  • In one embodiment, the antibody is a monoclonal Anti-IL-20 antibody. In one embodiment, the antibody is an Anti-IL-20 antibody as described in WO 2010/000721. In one embodiment, the Anti-IL-20 monoclonal antibody is 15D2 or 5B7 as described in WO 2010/000721.
  • In one embodiment, the antibody is a monoclonal Anti-TFPI antibody. In one embodiment, the antibody is an Anti-TFPI antibody as described in PCT/EP2009/067598. In one embodiment, the Anti-TFPI monoclonal antibody is HzTFPI4F36 as described in PCT/EP2009/067598.
  • It will be appreciated that the invention finds particular utility where the protein is present within the composition in high concentrations. Thus, in one embodiment, the protein is present in a concentration of 50 mg/ml or more, such as 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300 mg/ml or more. In one embodiment, the protein is present within the composition in an amount of between 50 mg/ml and 300 mg/ml, for instance between 50 mg/ml and 250 mg/ml, such as between 50 mg/ml and 200 mg/ml, for instance between 50 mg/ml and 150 mg/ml. In one embodiment, the protein is present in a concentration of between 75 mg/ml and 300 mg/ml, for instance between 75 mg/ml and 250 mg/ml, such as between 75 mg/ml and 200 mg/ml, for instance between 75 mg/ml and 150 mg/ml. In one embodiment, the protein is present in a concentration of between 100 mg/ml and 300 mg/ml, for instance between 100 mg/ml and 250 mg/ml, such as between 100 mg/ml and 200 mg/ml, for instance between 100 mg/ml and 150 mg/ml.
  • In one embodiment, a protein composition of the invention comprises:
      • (a) ≧50 mg/ml antibody;
      • (b) 30 mM or lower of an inorganic salt, such as sodium chloride or magnesium chloride;
      • (c) 0-25 mM of an amino acid, such as arginine or glycine;
      • (d) 50 mM or lower of a buffer such as histidine buffer;
      • (e) 0.001-0.005% of a non-ionic surfactant;
      • (f) 0.001-2% (w/v) of one or more preservatives;
        buffered to a pH of between 5 and 7.
  • In one embodiment, a protein composition of the invention comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) 0.001% polysorbate 80;
      • (f) 0.001-2% (w/v) of one or more preservatives;
        buffered to a pH of between 6 and 7.
  • In one embodiment, a protein composition of the invention comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) 0.001% polysorbate 80;
      • (f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol;
        buffered to a pH of 6.5.
  • In one embodiment, a protein composition of the invention comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) between 2 and 6 mg/ml m-cresol;
      • (f) between 85 and 130 mM sucrose
        buffered to a pH of 6.5.
  • In one embodiment, a protein composition of the invention comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) 4 mg/ml m-cresol;
      • (f) 105 mM propylene glycol
        buffered to a pH of 6.5.
  • Compositions of the invention have surprisingly demonstrated stability towards formation of high molecular weight proteins (HMWP) at 40° C. for 4 weeks and at 5° C. and 40° C. for 3 months.
  • A pharmaceutical formulation comprising the antibody and the excipients (including one or more preservatives) is prepared.
  • In one embodiment, the pharmaceutical compositions of the invention are stable for more than 6 weeks of usage and for more than 3 years of storage.
  • In one embodiment, the pharmaceutical compositions of the invention are stable for more than 4 weeks of usage and for more than 3 years of storage.
  • In one embodiment, the pharmaceutical compositions of the invention are stable for more than 4 weeks of usage and for more than 2 years of storage.
  • In one embodiment, the pharmaceutical compositions of the invention are stable for more than 2 weeks of usage and for more than 2 years of storage.
  • In one embodiment, the pharmaceutical compositions of the invention are stable for more than 1 week of usage and for more than 6 months of storage.
  • According to a second aspect of the invention, there is provided a stable, multi-dose liquid composition as defined herein for use in therapy.
  • The term “treatment” and “treating” as used herein means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relieve the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications.
  • The patient to be treated is preferably a mammal; in particular a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
  • For example, the stable compositions of Anti-IL-20 antibodies of the present invention may be used in the treatment of an inflammatory disease, in particular autoinflammatory diseases, such as psoriasis, systemic lupus erythomatosus, rheumatoid arthritis, Crohn's disease and psoriatic arthritis or otherwise as described in WO 2010/000721.
  • Thus according to a further aspect, the invention provides a method of treating such an inflammatory disease which comprises administering to a patient a therapeutically effective amount of a stable composition of an Anti-IL-20 antibody of the present invention.
  • The invention also provides a stable composition of an Anti-IL-20 antibody of the present invention for use in the treatment of such an inflammatory disease.
  • The invention also provides the use of a stable composition of an Anti-IL-20 antibody of the present invention in the manufacture of a medicament for the treatment of such an inflammatory disease.
  • The invention also provides a stable pharmaceutical composition comprising a stable composition of an Anti-IL-20 antibody of the present invention for use in the treatment of such an inflammatory disease.
  • Furthermore, the stable compositions of Anti-TFPI antibodies of the present invention may be used in the treatment of a coagulopathy (bleeding disorder), such as haemophilia A, with or without inhibitors, and haemophilia B, with or without inhibitors or otherwise as described in PCT/EP2009/067598.
  • Thus according to a further aspect, the invention provides a method of treating a coagulopathy which comprises administering to a patient a therapeutically effective amount of a stable composition of an Anti-TFPI antibody of the present invention.
  • The invention also provides a stable composition of an Anti-TFPI antibody of the present invention for use in the treatment of a coagulopathy.
  • The invention also provides the use of a stable composition of an Anti-TFPI antibody of the present invention in the manufacture of a medicament for the treatment of a coagulopathy.
  • The invention also provides a pharmaceutical composition comprising a stable composition of an Anti-TFPI antibody of the present invention for use in the treatment of a coagulopathy.
  • It is to be understood, that therapeutic and prophylactic (preventive) regimes represent separate aspects of the present invention.
  • The pharmaceutical formulations of the invention are generally suitable for parenteral administration. Parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe. Alternatively, parenteral administration can be performed by means of an infusion pump.
  • The invention is further described with reference to the following non-limiting examples.
  • EXAMPLES Example 1 4 Week Stability Analysis at 40° C. for Anti-IL-20
  • 12 formulations were prepared (see Table 1 below). The formulations were prepared from a stock solution containing ca. 150 mg/ml of the Anti-IL-20 antibody and 10 mM histidine buffer, pH 6.5. This stock solution was prepared by conventional UF/DF/UF. Stock solution of the excipients were prepared and mixed in the correct proportion. The final formulations were filled in 3 ml Penfill® cartridges, type 1 glass. The formulations were stored at 40° C. for 4 weeks and then analysed chemically, pharmaceutically and biophysically. The increase in the formation of protein aggregates (% HMWP) was measured by SEC-HPLC (as described above).
  • TABLE 1
    Composition of tested formulations
    Anti-IL-20 100 mg/ml
    Histidine 33 mM
    Arginine 25 mM
    NaCl 25 mM
    Polysorbate 80 0.01 mg/ml
    Preservative(s) Concentration
    varied
    pH 6.5
  • The results of the stability analysis are shown in Table 2 and demonstrates that all tested formulations resulted in a low content of aggregates following 4 weeks storage at 40° C.
  • TABLE 2
    SEC-HPLC Analysis the increase of % HMWP for Anti-IL-20
    formed during 4 weeks study (Δ % HMWP).
    Storage
    time (weeks)
    Δ % HMWP at 40° C.
    Formulation 1 2 4
    Phenol 5 mg/ml 1.0 1.8 2.5
    Phenol 1.5 mg/ml 0.4 0.8 1.4
    m-cresol 3.5 mg/ml 1.0 1.8 3.1
    m-cresol 1.5 mg/ml 0.4 0.9 1.9
    Benzyl alcohol 11 mg/ml 1.1 2.0 3.6
    Benzyl alcohol 5 mg/ml 0.5 1.0 2.0
    Chlorobutanol 6 mg/ml 0.0 0.0 0.6
    Chlorobutanol 3 mg/ml 0.5 0.8 1.9
    Phenol/m-cresol 1.5/1.8 mg/ml 0.4 0.8 1.7
    Phenol/m-cresol 5/3.5 mg/ml 0.3 0.7 1.2
    Benzyl alcohol/chlorobutanol 8/1.1 mg/ml 0.9 1.6 2.8
    Benzyl alcohol/chlorobutanol 5/3 mg/ml 0.8 1.4 2.6
    Reference (without preservative) 0.2 0.6 1.1
  • In particular, it should be noted that the compositions containing 2 preservatives (phenol and m-cresol) generally resulted in lower aggregate formation than the results achieved with the individual preservatives alone. Most surprisingly, it can be seen that the composition containing 5 mg/ml phenol alone resulted in an increase of 2.5% HMWP and the composition containing 3.5 mg/ml m-cresol alone resulted in an increase of 3.1% HMWP, however, a composition containing a combination of these two preservatives at identical concentrations resulted in a lowering of aggregate formation (an increase of 1.2% HMWP). An increase of 1.1% is seen for reference without preservative.
  • Potency was investigated after 4 weeks storage at 40° C. and the results are shown in Table 3. These results demonstrate that full potency was obtained for all formulations.
  • TABLE 3
    Potency Analysis of Anti-IL-20 following 4 week study
    Sample % Potency
    Phenol 5 mg/ml 106
    Phenol 1.5 mg/ml 110
    m-cresol 3.5 mg/ml 121
    m-cresol 1.5 mg/ml 113
    Benzyl alcohol 11 mg/ml 126
    Benzyl alcohol 5 mg/ml 113
    Chlorobutanol 6 mg/ml 108
    Chlorobutanol 3 mg/ml 112
    Phenol/m-cresol 1.5/1.8 mg/ml 115
    Phenol/m-cresol 5/3.5 mg/ml 115
    Benzyl alcohol/chlorobutanol 8/1.1 mg/ml 103
    Benzyl alcohol/chlorobutanol 5/3 mg/ml 96
    Reference (without preservative) 108
  • Example 2 3 Month Stability Analysis at 5° C. and 40° C. for Anti-IL-20
  • This study was conducted in an analogous manner to that described in Example 1 with the exception that the study was conducted for 3 months at 5° C. and 40° C. and the formulations contained sucrose as described in Table 4.
  • TABLE 4
    Composition of tested formulations
    Anti-IL-20 100 mg/ml
    Histidine 33 mM
    Arginine 25 mM
    NaCl 25 mM
    Polysorbate 80 0.01 mg/ml
    Sucrose To tonicity
    Preservative(s) Concentration
    varied
    pH 6.5
  • The results of the stability analysis at 40° C. are shown in Table 5 and demonstrates that all tested formulations resulted in a low content of aggregates following 1 month storage at 40° C.
  • TABLE 5
    SEC-HPLC Analysis of increase of % HMWP for Anti-IL-20
    formed during 3 month study at 40° C. (Δ % HMWP)
    Months of storage
    Δ % HMWP 0.5 1 3
    Reference 0.0 0.5 2.4
    Phenol 5 mg/ml 1.2 2.6 7.6
    m-cresol 3.5 mg/ml 0.2 0.9 0.0
    Phenol/m-cresol 1.5/1.8 mg/ml 2.9* 4.6* 8.9*
    *The absolute value of % HMWP is given as time zero value was not available
  • In particular, it should be noted that the composition containing 3.5 mg/ml m-cresol resulted in the smallest increase in HMWP. After 3 months storage the increase in HMWP for this formulation is lower compared to reference without preservative.
  • The results of the stability analysis at 5° C. are shown in Table 6 and demonstrates that all tested formulations resulted in a low content of aggregates following 12 months storage at 5° C.
  • TABLE 6
    SEC-HPLC Analysis of Δ % HMWP of Anti-IL-20 formed
    during 12 month study at 5° C.
    Months of
    storage
    Δ % HMWP 3 12
    Reference 0.0 0.0
    Phenol 5 mg/ml 0.0 0.0
    m-cresol 3.5 mg/ml 3.6 0.0
    Phenol/m-cresol 1.5/1.8 mg/ml 2.3* 2.3*
    *The absolute value of % HMWP is given as time zero value was not available
  • In particular, it should be noted that the composition containing phenol and m-cresol, respectively, showed no increase in % HMWP over a period of 12 months at 5° C.
  • Potency was investigated after 3 months storage at 40° C. and the results are shown in Table 7. These results demonstrate that substantial potency was obtained for all formulations.
  • TABLE 7
    Potency Analysis of Anti-IL-20 Following 3 month study at 40° C.
    Months of
    storage
    Potency (%) 0 3
    Reference 113 92
    Phenol 5 mg/ml 156 105
    m-cresol 3.5 mg/ml 116 89
    Phenol/m-cresol 1.5/1.8 mg/ml Not analysed 79
  • The robustness of the formulations was also assessed with respect to cycles of freeze-thaw (−80° C. to room temperature) and thermal and mechanical stress (4 hours daily rotation during 2 weeks storage at 37° C.). Data are given in below Table 8, and these indicate that the formulations are robust against physical stress.
  • TABLE 8
    Increase of Δ % HMWP compared to time zero of formulations
    of mAbs exposed to i) freeze-thaw stress (10 cycles from −80°
    to ambient temperature) and ii) combined rotation and thermal
    stress at 37° C. for 2 weeks
    Rotation and
    Δ % HMWP Freeze-thaw cycles thermal stress
    Reference 0.0 0.0
    Phenol 5 mg/ml 0.0 0.6
    m-cresol 3.5 mg/ml 0.0 0.1
    Phenol/m-cresol 1.5/1.8 mg/ml 2.3* 2.6*-
    *As time zero value of % HMWP was not available the absolute value of % HMWP after the exposed physical stress was used for this formulation

    Visual appearance analysis was performed in light cabinet and architect lamp for all formulations to assess the possibility of formation of particles. At time zero all samples were found to be clear to slightly opalescent without any visible particles using both analytical methods. No difference was observed after the formulations were exposed to the above stress conditions.
  • Example 3 Results from Modified Preservative Efficacy Test for Anti-IL-20
  • To analyze the efficacy of different preservatives, a preservative efficacy screening test was performed. The efficacy of the preservative was measured using a modified USP/Ph Eur preservative efficacy test. In the modified test, formulations were tested against Staphylococcus aureus. After inoculation, samples were stored for 6 and 24 hours at room temperature and the total bacterial counts were measured using a colony counter. The log reduction values were calculated as log (initial count/final count).
  • In the unmodified USP/Ph Eur preservative efficacy tests several bacteria and fungi are tested. The USP and Ph Eur regulatory requirements are listed here below in Table 9. It should be noted that the Ph Eur requirements are more stringent than those of the USP, and that the Ph Eur requirements offer a minimal level that must be achieved (B criteria) and a suggested level that is recommended (A criteria). The preservative efficacy test was modified in this study to reduce the sample requirements and cost per analysis. Staphylococcus aureus was chosen in the modified preservative test because it was the most resistant pathogen for the chosen formulations.
  • TABLE 9
    USP and Ph Eur requirements for preservative efficacy testing
    Ph Eur requirements
    (6th edition)
    USP (24) Suggested (A
    Time point requirements criteria) Minimum (B criteria)
    Requirements for
    Bacterial log reduction
     6 hours Not required 2 Not required
    24 hours Not required 3 1
     7 days 1 Not required 3
    14 days 3 Not required Not required
    28 days No increase No recovery No increase
    Requirements for
    fungal log reduction
     7 days No increase 2 Not required
    14 days No increase Not required 1
    28 days No increase No increase No increase
  • 11 formulations were prepared (see below Table 10). The formulations were tested in the modified preservative efficacy test as described above and results are given in Table 11.
  • TABLE 10
    Composition of tested formulations of Anti-IL-20
    Formulation Anti-IL- Phenol m-cresol His NaCl Arg Sucrose Propylene Tween 80
    number 20 mg/ml mg/ml mg/ml mM mM mM mM glycol mM mg/ml pH
    1 0 5 33 25 25 99 0.01 6.5
    2 100 0 33 25 25 150 0.01 6.5
    3 100 3 33 25 25 119 0.01 6.5
    4 100 5 33 25 25 99 0.01 6.5
    5 100 7 33 25 25 78 0.01 6.5
    6 100 5 33 25 25 95 0.01 6.5
    7 0 4 33 25 25 109 6.5
    8 100 2 33 25 25 129 6.5
    9 100 4 33 25 25 109 6.5
    10 100 6 33 25 25 88 6.5
    11 100 4 33 25 25 105 6.5
  • TABLE 11
    Log reduction of Staphylococcus aureus of formulations of Anti-IL-20
    in the modified preservative efficacy test
    Log reduction Log reduction
    Anti-IL-20 (mg/ml) Phenol (mg/ml) 6 hours 24 hours
     0 5 2.71 5.57
    100 0 0 0
    100 3 0 1.4
    100 5 1.64 5.57
    100 7 4.67 5.57
     100* 5 1.28 5.57
    Log reduction Log reduction
    Anti-IL-20 mg/ml m-Cresol mg/ml 6 hours 24 hours
     0 4 5.57 5.57
    100 0 0 0
    100 2 0 0
    100 4 5.57 5.57
    100 6 5.57 5.57
     100* 4 5.57 5.57
    *Propylene glycol
  • The results of the modified preservative efficacy test show that the formulation with 3 mg/ml phenol complies with criteria B in Ph Eur and formulations with 5-7 mg/ml phenol and 4-6 mg/ml m-cresol demonstrate a complete kill of Staphylococcus aureus after 24 hours. By the performed test the formulations have been demonstrated to be suitable as multi-dose formulations.
  • The stability of above formulations used in the preservative efficacy screening study with respect to formation of % HMWP is given in below in Table 12. As can be observed, increasing amounts of preservatives leads to increasing amounts of % HMWP, however, several of above formulations are stable and have satisfactory preservative effect.
  • TABLE 12
    Formation of % HMWP (Δ % HMWP) over 3 months at 40° C.
    Months of storage
    at 40° C.
    Δ % HMWP 1 3
    Mg/ml preservative
    0 mg/ml phenol 0.7 2.5
    3 mg/ml phenol 1.6 5.8
    5 mg/ml phenol 3.6 11.6
    7 mg/ml phenol 9.1 23.4
    5 mg/ml phenol* 3.7 12.1
    2 mg/ml m-cresol 1.2 5.1
    4 mg/ml m-cresol 4.1 12.5
    6 mg/ml m-cresol 13.9 13.7
    4 mg/ml m-cresol* 4.6 23.1
    *With propylene glycol
  • Example 4 3 Month Stability Analysis at 5° C. and 40° C. for Anti-TFPI
  • This study was conducted in an analogous manner to that described in Example 2 with the exception that the study was conducted with an Anti-TFPI antibody rather than the Anti-IL-20 antibody. The formulations tested are as described in Table 13.
  • TABLE 13
    Composition of tested formulations
    Anti-TFPI 100 mg/ml
    Histidine 33 mM
    Arginine 25 mM
    NaCl 25 mM
    Polysorbate 80 0.01 mg/ml
    Sucrose To tonicity
    Preservative(s) Concentration
    varied
    pH 6.0
  • The results of the stability analysis at 40° C. are shown in Table 14 and demonstrates that all tested formulations resulted in a significantly higher content of aggregates following 3 months storage at 40° C.
  • TABLE 14
    SEC-HPLC Analysis of increase of % HMWP for Anti-IL-20
    formed during 3 month study at 40° C. (Δ % HMWP).
    Months of storage
    at 40° C.
    Δ % HMWP 0.5 1 3
    Reference 0.0 0.9 3.2
    Phenol 5 mg/ml 3.3 9.5 17.4
    Phenol/m-cresol 1.5/1.8 mg/ml 0.5 2.2 7.6
  • In particular, it can be seen that 5 mg/ml phenol resulted in a 4 fold increase in aggregate formation when compared with the reference formulation. By contrast, the same concentration of preservative in the Anti-IL-20 containing formulation resulted in only a 2.1 fold increase (see Table 5). However, the results with the combination containing phenol and m-cresol were equivalent to those obtained for Anti-IL-20 (i.e. a 1.9 fold increase in aggregate formation for Anti-TFPI c.f. a 1.8 fold increase for Anti-IL-20; see Table 5).
  • The results of the stability analysis at 5° C. are shown in Table 15 which demonstrates that all tested formulations resulted in a low content of aggregates following 3 months storage at 5° C.
  • TABLE 15
    SEC-HPLC Analysis of Δ % HMWP of Anti-IL-20 formed during
    12 month study at 5° C.12
    Months
    of storage
    % Δ HMWP 3 12
    Reference 0.0 0.0
    Phenol 5 mg/ml 0.0 0.0
    Phenol/m-cresol 1.5/1.8 mg/ml 0.0 0.0
  • The robustness of the formulations was also assessed with respect to cycles of freeze-thaw (−80° C. to room temperature) and thermal and mechanical stress (4 hours daily rotation during 2 weeks storage at 37° C.). Data are given in below Table 16, and these indicate that the formulations are robust against physical stress.
  • TABLE 16
    Increase of Δ % HMWP compared to time zero of formulations
    of mAbs exposed to i) freeze-thaw stress (10 cycles from −80°
    to ambient temperature) and ii) combined rotation and thermal
    stress at 37° C. for 2 weeks
    Rotation and
    Δ % HMWP Freeze-thaw cycles thermal stress
    Reference 0.0 0.0
    Phenol 5 mg/ml 0.0 1.0
    Phenol/m-cresol 1.5/1.8 mg/ml 0.0 0.2
  • Visual appearance analysis was performed in light cabinet and architect lamp for all formulations to assess the possibility of formation of particles. At time zero all samples were found to be clear to slightly opalescent without any visible particles using both analytical methods. No difference was observed after the formulations were exposed to the above stress conditions.
  • The following is a non-limiting list of embodiments of the present invention.
  • Embodiment 1: A stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
  • Embodiment 2: A composition according to embodiment 1, wherein the preservative is present within the composition in an amount of between 0.001 to 2% (w/v).
  • Embodiment 3: A composition according to embodiment 1 or 2, wherein the preservative is present within the composition in an amount of between 0.002 to 1% (w/v).
  • Embodiment 4: A composition according to any of embodiments 1 to 3, wherein the one or more preservative is selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof.
  • Embodiment 5: A composition according to any of embodiments 1 to 4, wherein the one or more preservative is selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 6: A composition according to any of embodiments 1 to 5, wherein the composition comprises a single preservative.
  • Embodiment 7: A composition according to any of embodiments 1 to 6, wherein the composition comprises a single preservative selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • Embodiment 8: A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 9: A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 10: A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount of 0.15 or 0.5% (w/v).
  • Embodiment 11: A composition according to any of embodiments 1 to 7, which comprises phenol as a single preservative, wherein said phenol is present within the composition in an amount from 0.25 to 0.5% (w/v).
  • Embodiment 12: A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 13: A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 14: A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount of 0.15 or 0.35% (w/v).
  • Embodiment 15: A composition according to any of embodiments 1 to 7, which comprises m-cresol as a single preservative, wherein said m-cresol is present within the composition in an amount of approximately 0.3% (w/v).
  • Embodiment 16: A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount from 0.1 to 2% (w/v).
  • Embodiment 17: A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount from 0.1 to 1.5% (w/v).
  • Embodiment 18: A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount of 0.5 or 1.1% (w/v).
  • Embodiment 19: A composition according to any of embodiments 1 to 7, which comprises benzyl alcohol as a single preservative, wherein said benzyl alcohol is present within the composition in an amount of approximately 1% (w/v).
  • Embodiment 20: A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.1 to 1% (w/v).
  • Embodiment 21: A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.25 to 0.75% (w/v).
  • Embodiment 22: A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount of 0.3 or 0.6% (w/v).
  • Embodiment 23: A composition according to any of embodiments 1 to 7, which comprises chlorobutanol as a single preservative, wherein said chlorobutanol is present within the composition in an amount from 0.3 to 0.5% (w/v).
  • Embodiment 24: A composition according to any of embodiments 1 to 7, which comprises methyl paraben as a single preservative, wherein said methyl paraben is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 25: A composition according to any of embodiments 1 to 7, which comprises methyl paraben as a single preservative, wherein said methyl paraben is present within the composition in an amount of approximately 0.2% (w/v).
  • Embodiment 26: A composition according to any of embodiments 1 to 7, which comprises propyl paraben as a single preservative, wherein said propyl paraben is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 27: A composition according to any of embodiments 1 to 7, which comprises propyl paraben as a single preservative, wherein said propyl paraben is present within the composition in an amount of approximately 0.2% (w/v).
  • Embodiment 28: A composition according to any of embodiments 1 to 7, which comprises phenoxyethanol as a single preservative, wherein said phenoxyethanol is present within the composition in an amount from 0.1 to 2% (w/v).
  • Embodiment 29: A composition according to any of embodiments 1 to 7, which comprises phenoxyethanol as a single preservative, wherein said phenoxyethanol is present within the composition in an amount of approximately 1% (w/v).
  • Embodiment 30: A composition according to any of embodiments 1 to 7, which comprises thiomersal as a single preservative, wherein said thiomersal is present within the composition in an amount from 0.002 to 0.01% (w/v).
  • Embodiment 31: A composition according to any of embodiments 1 to 7, which comprises a single preservative selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 32: A composition according to any of embodiments 1 to 5, which comprises two or more preservatives.
  • Embodiment 33: A composition according to embodiment 32, which comprises two preservatives.
  • Embodiment 34: A composition according to embodiment 32 or 33, which comprises two preservatives selected from phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal.
  • Embodiment 35: A composition according to embodiment 34, which comprises two preservatives selected from phenol, m-cresol, benzyl alcohol and chlorobutanol.
  • Embodiment 36: A composition according to any of embodiments 32 to 35, wherein said two preservatives are phenol and m-cresol.
  • Embodiment 37: A composition according to embodiment 36, wherein phenol is present within the composition in an amount from 0.1 to 0.75% (w/v).
  • Embodiment 38: A composition according to embodiment 36 or 37, wherein phenol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 39: A composition according to any of embodiments 36 to 38, wherein phenol is present within the composition in an amount of 0.15 or 0.5% (w/v).
  • Embodiment 40: A composition according to any of embodiments 36 to 39, wherein m-cresol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 41: A composition according to any of embodiments 36 to 40, wherein m-cresol is present within the composition in an amount from 0.15 to 0.4% (w/v).
  • Embodiment 42: A composition according to any of embodiments 36 to 41, wherein m-cresol is present within the composition in an amount of 0.18 or 0.35% (w/v).
  • Embodiment 43: A composition according to any of embodiments 32 to 35, wherein said two preservatives are benzyl alcohol and chlorobutanol.
  • Embodiment 44: A composition according to embodiment 43, wherein benzyl alcohol is present within the composition in an amount from 0.25 to 1% (w/v).
  • Embodiment 45: A composition according to embodiment 43 or 44, wherein benzyl alcohol is present within the composition in an amount from 0.4 to 0.9% (w/v).
  • Embodiment 46: A composition according to any of embodiments 43 to 45, wherein benzyl alcohol is present within the composition in an amount of 0.5 or 0.8% (w/v).
  • Embodiment 47: A composition according to any of embodiments 43 to 46, wherein chlorobutanol is present within the composition in an amount from 0.1 to 0.5% (w/v).
  • Embodiment 48: A composition according to any of embodiments 43 to 47, wherein chlorobutanol is present within the composition in an amount from 0.1 to 0.4% (w/v).
  • Embodiment 49: A composition according to any of embodiments 43 to 48, wherein chlorobutanol is present within the composition in an amount of 0.11 or 0.3% (w/v).
  • Embodiment 50: A composition according to any embodiments 1 to 49, wherein a buffer is present, and the buffer has a pKa between 4 to 8.
  • Embodiment 51: A composition according to embodiment 50, wherein the buffer has a pKa between 5 to 7.
  • Embodiment 52: A composition according to embodiment 50 or 51, wherein a buffer is present, and the buffer is dosodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, maleate, succinate, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, or tris(hydroxymethyl)-amino methane, or mixtures thereof.
  • Embodiment 53: A composition according to embodiment 52, wherein the buffer is histidine, maleate, succinate, phosphate, or tris(hydroxymethyl)-amino methane.
  • Embodiment 54: A composition according to embodiment 53, wherein the buffer is histidine.
  • Embodiment 55: A composition according to embodiment 54, wherein the buffer has a pKa value ±1 pH unit from the target pH of the composition.
  • Embodiment 56: A composition according to any of embodiments 1 to 55, wherein a salt is present and the salt is selected from the group consisting of sodium chloride, magnesium chloride, sodium thiocyanate, ammonium thiocyanate, ammonium sulfate, ammonium chloride, calcium chloride, arginine hydrochloride, zinc chloride and sodium acetate or any combination thereof.
  • Embodiment 57: A composition according to embodiment 56, wherein the salt is sodium chloride or magnesium chloride.
  • Embodiment 58: A composition according to embodiment 57, wherein the salt is sodium chloride.
  • Embodiment 59: A composition according to embodiment 56, wherein the salt is arginine-HCl.
  • Embodiment 60: A composition according to any of embodiments 1 to 59, which has a pH of between 5.0 and 7.0.
  • Embodiment 61: A composition according to embodiment 60, which has a pH of between 6.0 and 7.0.
  • Embodiment 62: A composition according to embodiment 61, which has a pH of 6.0 or 6.5.
  • Embodiment 63: A composition according to embodiment 62, which has a pH of 6.5.
  • Embodiment 64: A composition according to any of embodiments 1 to 63 which additionally comprises a surfactant.
  • Embodiment 65: A composition according to embodiment 64, wherein the surfactant is Tween 80 (i.e. polysorbate 80).
  • Embodiment 66: A composition according to embodiment 64 or 65, wherein the surfactant is present within the composition in an amount of below 0.01%.
  • Embodiment 67: A composition according to any of embodiments 64 to 66, wherein the surfactant is present within the composition in an amount of below 0.0075%.
  • Embodiment 68: A composition according to any of embodiments 64 to 67, wherein the surfactant is present within the composition in an amount between 0.001% and 0.005%.
  • Embodiment 69: A composition according to any of embodiments 64 to 68, wherein the surfactant is present within the composition in an amount of 0.001%.
  • Embodiment 70: A composition according to any of embodiments 1 to 63 wherein no surfactant is present.
  • Embodiment 71: A composition according to any of embodiments 1 to 70, which additionally comprises a tonicity modifying agent.
  • Embodiment 72: A composition according to embodiment 71, wherein the tonicity modifying agent is sucrose or propylene glycol.
  • Embodiment 73: A composition according to embodiment 72, wherein the tonicity modifying agent is sucrose.
  • Embodiment 74: A composition according to embodiment 72, wherein the tonicity modifying agent is propylene glycol.
  • Embodiment 75: A composition according to any of embodiments 71 to 74, wherein the tonicity modifying agent is present within the composition in an amount of between 50 and 250 mM.
  • Embodiment 76: A composition according to any of embodiments 71 to 75, wherein the tonicity modifying agent is present within the composition in an amount of between 100 and 200 mM.
  • Embodiment 77: A composition according to any of embodiments 71 to 76, wherein the tonicity modifying agent is present in an amount of 100 mM.
  • Embodiment 78: A composition according to any of embodiments 1 to 77, wherein the composition is pharmaceutically acceptable.
  • Embodiment 79: A stable composition according to any of embodiments 1 to 78,
  • wherein the antibody is present within the composition in a concentration of between 50 mg/ml and 300 mg/ml.
  • Embodiment 80: A stable composition according to embodiment 79, wherein the antibody is present within the composition in a concentration of between 75 mg/ml and 300 mg/ml.
  • Embodiment 81: A stable composition according to embodiment 80, wherein the antibody is present within the composition in a concentration of between 100 mg/ml and 300 mg/ml.
  • Embodiment 82: A stable composition according to embodiment 81, wherein the antibody is present within the composition in a concentration of between 50 mg/ml and 200 mg/ml.
  • Embodiment 83: A stable composition according to embodiment 82, wherein the antibody is present within the composition in a concentration of 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, or 300 mg/ml.
  • Embodiment 84: A composition according to embodiment 1 which comprises:
      • (a) ≧50 mg/ml antibody;
      • (b) 30 mM or lower of a in-organic salt, such as sodium chloride or magnesium chloride;
      • (c) 0-25 mM of an amino acid, such as arginine or glycine;
      • (d) 50 mM or lower of a buffer such as histidine buffer;
      • (e) 0.001-0.005% of a non-ionic surfactant;
      • (f) 0.001-2% (w/v) of one or more preservatives;
      • (g) 100 mM sucrose;
        buffered to a pH of between 5 and 7.
  • Embodiment 85: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) 0.001% polysorbate 80;
      • (f) 0.001-2% (w/v) of one or more preservatives;
      • (g) 100 mM sucrose;
        buffered to a pH of between 6 and 7.
  • Embodiment 86: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histidine buffer;
      • (d) 25 mM arginine;
      • (e) 0.001% polysorbate 80;
      • (f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol;
      • (g) 120 mM sucrose;
        buffered to a pH of 6.5.
  • Embodiment 87: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 0.01 mg/ml poysorbate 80;
      • (f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol;
      • (g) 120 mM sucrose.
        Buffered to a pH between 5 and
  • Embodiment 88: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-TFPI;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 0.01 mg/ml poysorbate 80;
      • (f) 5 mg/ml phenol;
      • (g) 100 mM sucrose.
  • Embodiment 89: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml antibody;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 0.01 mg/ml poysorbate 80;
      • (f) 5 mg/ml phenol;
      • (g) 95 mM propylene glycol.
  • Embodiment 90: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-IL-20;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 0.01 mg/ml poysorbate 80;
      • (f) 4 mg/ml m-cresol;
      • (g) 105 mM propylene glycol.
  • Embodiment 91: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-IL-20;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 2 mg/ml m-cresol;
      • (f) 129 mM sucrose.
  • Embodiment 92: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-IL-20;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 4 mg/ml m-cresol;
      • (f) 109 mM sucrose.
  • Embodiment 93: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-IL-20;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 6 mg/ml m-cresol;
      • (f) 88 mM sucrose.
  • Embodiment 94: A composition according to embodiment 1 which comprises:
      • (a) 100 mg/ml Anti-IL-20;
      • (b) 25 mM sodium chloride;
      • (c) 33 mM histdine buffer;
      • (d) 25 mM arginine;
      • (e) 4 mg/ml m-cresol;
      • (f) 105 mM propylene glycol.
  • Embodiment 95: A composition according any of embodiments 1-94, wherein the antibody is of the IgG4 subtype.
  • Embodiment 96: A composition according to any of embodiments 1 to 95, wherein the antibody is a monoclonal antibody.
  • Embodiment 97: A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 monoclonal antibody.
  • Embodiment 98: A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 monoclonal antibody as described in WO 2010/000721.
  • Embodiment 99: A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-IL-20 antibody 15D2 or 5B7 as described in WO 2010/000721.
  • Embodiment 100: A composition according to embodiment 96, wherein the monoclonal antibody is an Anti-TFPI monoclonal antibody.
  • Embodiment 101: A composition according to embodiment 96, wherein the monoclonal antibody is the Anti-TFPI monoclonal antibody HzTFPI4F36 as described in PCT/EP2009/067598.
  • Embodiment 102: A stable, multi-dose liquid composition according to any of embodiments 1 to 101 for use in therapy.
  • Embodiment 103: A method of treating an inflammatory disease which comprises administering to a patient a therapeutically effective amount of a composition according to any of embodiments 97-99.
  • Embodiment 104: A composition according to any of embodiments 97-99 for use in the treatment of an inflammatory disease.
  • Embodiment 105: Use of a composition according to any of embodiments 97-99 in the manufacture of a medicament for the treatment of an inflammatory disease.
  • Embodiment 106: A pharmaceutical composition comprising an Anti-IL-20 composition according to any of embodiments 97-99 for use in the treatment of an inflammatory disease.
  • Embodiment 107: A method of treating a coagulopathy which comprises administering to a patient a therapeutically effective amount of a composition according to any of embodiments 100-101.
  • Embodiment 108: A composition according to any of embodiments 100-101 for use in the treatment of a coagulopathy.
  • Embodiment 109: Use of a composition according to any of embodiments 100-101 in the manufacture of a medicament for the treatment of a coagulopathy.
  • Embodiment 110: A pharmaceutical composition comprising an Anti-TFPI composition according to any of embodiments 100-101 for use in the treatment of a coagulopathy.
  • All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference in their entirety and to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein (to the maximum extent permitted by law), regardless of any separately provided incorporation of particular documents made elsewhere herein.
  • The use of the terms “a” and “an” and “the” and similar referents in the context of describing the invention are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. For example, the phrase “the compound” is to be understood as referring to various “compounds” of the invention or particular described aspect, unless otherwise indicated.
  • Unless otherwise indicated, all exact values provided herein are representative of corresponding approximate values (e.g., all exact exemplary values provided with respect to a particular factor or measurement can be considered to also provide a corresponding approximate measurement, modified by “about,” where appropriate). When a range is given, the range includes both end values, unless otherwise indicated.
  • The description herein of any aspect or aspect of the invention using terms such as “comprising”, “having,” “including,” or “containing” with reference to an element or elements is intended to provide support for a similar aspect or aspect of the invention that “consists of”, “consists essentially of”, or “substantially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition described herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context).

Claims (15)

1. A stable, multi-dose liquid composition comprising an antibody and one or more preservatives.
2. A composition according to claim 1, wherein the preservative is present within the composition in an amount of between 0.001 to 2% (w/v), such as 0.002 to 1% (w/v).
3. A composition according to claim 1, wherein the one or more preservative is selected from the group consisting of phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride, thiomersal or any combinations thereof, such as phenol, m-cresol, benzyl alcohol and chlorobutanol.
4. A composition according to claim 1, which comprises a single preservative, selected from the group consisting of phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal, such as phenol, m-cresol, benzyl alcohol and chlorobutanol.
5. A composition according to claim 1, which comprises two or more preservatives selected from the group consisting of phenol, m-cresol, benzyl alcohol, chlorobutanol, ethanol, phenoxyethanol, p-chlor-m-cresol, methyl paraben, propyl paraben, benzalkonium chloride and thiomersal, such as phenol, m-cresol, benzyl alcohol and chlorobutanol, in particular: phenol and m-cresol; and benzyl alcohol and chlorobutanol.
6. A composition according to claim 1, which is buffered to a pH of between 5 and 7, such as a pH of between 6.0 and 7.0, such as 6.0 or 6.5, in particular 6.5.
7. A composition according to claim 1, which further comprises a tonicity modifying agent selected from the group consisting of sucrose and propylene glycol.
8. A composition according to claim 1, wherein the antibody is present within the composition in a concentration selected from the group consisting of 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, and 300 mg/ml.
9. A composition according to claim 1, wherein no surfactant is present.
10. A composition according to claim 1 comprising:
(a) 100 mg/ml antibody;
(b) 25 mM sodium chloride;
(c) 33 mM histidine buffer;
(d) 25 mM arginine;
(e) 0.001% polysorbate 80;
(f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol; and
(g) 120 mM sucrose;
buffered to a pH of 6.5.
11. A composition according to claim 1 comprising:
(a) 100 mg/ml antibody;
(b) 25 mM sodium chloride;
(c) 33 mM histidine buffer;
(d) 25 mM arginine;
(e) 0.01% polysorbate 80;
(f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol; and
(g) 120 mM sucrose;
buffered to a pH of 6.5.
12. A composition according to claim 1 comprising:
(a) 100 mg/ml antibody;
(b) 25 mM sodium chloride;
(c) 33 mM histidine buffer;
(d) 25 mM arginine;
(e) 0.01% polysorbate 80;
(f) 1.5 mg/ml phenol and 1.8 mg/ml m-cresol; and
(g) 120 mM sucrose;
buffered to a pH of 6.
13-15. (canceled)
16. A method of treating an inflammatory disease comprising administering a stable antibody composition according to claim 1.
17. A method of treating a coagulopathy comprising administering a stable antibody composition according to claim 1.
US13/699,847 2010-05-28 2011-05-26 Stable Multi-Dose Compositions Comprising an Antibody and a Preservative Abandoned US20130136733A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/699,847 US20130136733A1 (en) 2010-05-28 2011-05-26 Stable Multi-Dose Compositions Comprising an Antibody and a Preservative

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP10164298.1 2010-05-28
EP10164298 2010-05-28
US35152210P 2010-06-04 2010-06-04
US13/699,847 US20130136733A1 (en) 2010-05-28 2011-05-26 Stable Multi-Dose Compositions Comprising an Antibody and a Preservative
PCT/EP2011/058648 WO2011147921A1 (en) 2010-05-28 2011-05-26 Stable multi-dose compositions comprising an antibody and a preservative

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/058648 A-371-Of-International WO2011147921A1 (en) 2010-05-28 2011-05-26 Stable multi-dose compositions comprising an antibody and a preservative

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/799,276 Continuation US10835602B2 (en) 2010-05-28 2017-10-31 Stable multi-dose compositions comprising an antibody and a preservative

Publications (1)

Publication Number Publication Date
US20130136733A1 true US20130136733A1 (en) 2013-05-30

Family

ID=42562357

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/699,847 Abandoned US20130136733A1 (en) 2010-05-28 2011-05-26 Stable Multi-Dose Compositions Comprising an Antibody and a Preservative
US15/799,276 Active US10835602B2 (en) 2010-05-28 2017-10-31 Stable multi-dose compositions comprising an antibody and a preservative

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/799,276 Active US10835602B2 (en) 2010-05-28 2017-10-31 Stable multi-dose compositions comprising an antibody and a preservative

Country Status (11)

Country Link
US (2) US20130136733A1 (en)
EP (1) EP2575761A1 (en)
JP (3) JP6294075B2 (en)
KR (1) KR20130086144A (en)
CN (2) CN102905692B (en)
AU (1) AU2011257219B2 (en)
BR (1) BR112012030139A2 (en)
CA (1) CA2800188A1 (en)
MX (1) MX2012013586A (en)
RU (1) RU2012153786A (en)
WO (1) WO2011147921A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9795674B2 (en) 2010-02-26 2017-10-24 Novo Nordisk A/S Stable antibody containing compositions
US9849181B2 (en) 2012-08-31 2017-12-26 Bayer Healthcare Llc High concentration antibody and protein formulations
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
US10835602B2 (en) 2010-05-28 2020-11-17 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
WO2021094917A1 (en) * 2019-11-13 2021-05-20 Pfizer Inc. Stable aqueous anti-tfpi antibody formulation

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003062278A1 (en) 2002-01-25 2003-07-31 G2 Therapies Ltd MONOCLONAL ANTIBODIES AGAINST EXTRACELLULAR LOOPS OF C5aR
WO2009103113A1 (en) 2008-02-20 2009-08-27 G2 Inflammation Pty Ltd HUMANIZED ANTI-C5aR ANTIBODIES
US8287861B2 (en) 2008-06-30 2012-10-16 Novo Nordisk A/S Anti-human interleukin-20 antibodies
US8454956B2 (en) 2009-08-31 2013-06-04 National Cheng Kung University Methods for treating rheumatoid arthritis and osteoporosis with anti-IL-20 antibodies
CN103596982B (en) 2011-06-06 2016-11-02 诺沃—诺迪斯克有限公司 Therapeutic antibodies
JP6345123B2 (en) * 2012-02-16 2018-06-20 サンタラス, インコーポレイテッド Anti-VLA1 (CD49A) antibody pharmaceutical composition
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
PT2830658T (en) * 2012-03-26 2018-12-28 Sanofi Sa Stable igg4 binding agent formulations
US20140004131A1 (en) 2012-05-04 2014-01-02 Novartis Ag Antibody formulation
US9221904B2 (en) 2012-07-19 2015-12-29 National Cheng Kung University Treatment of osteoarthritis using IL-20 antagonists
US8852588B2 (en) 2012-08-07 2014-10-07 National Cheng Kung University Treating allergic airway disorders using anti-IL-20 receptor antibodies
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
TWI694836B (en) 2014-05-16 2020-06-01 英商葛蘭素史克智慧財產管理有限公司 Antibody formulation
MX2018002048A (en) 2015-08-19 2018-04-13 Pfizer Tissue factor pathway inhibitor antibodies and uses thereof.
KR20190141243A (en) * 2017-05-05 2019-12-23 암젠 인크 Pharmaceutical compositions comprising bispecific antibody constructs for improved storage and administration
US20200113912A1 (en) 2018-09-12 2020-04-16 Silverback Therapeutics, Inc. Methods and Compositions for the Treatment of Disease with Immune Stimulatory Conjugates
KR102337683B1 (en) * 2018-09-21 2021-12-13 주식회사 녹십자 Highly efficient anti-TFPI antibody composition
MX2021015533A (en) 2019-06-19 2022-02-10 Silverback Therapeutics Inc Anti-mesothelin antibodies and immunoconjugates thereof.
JP2023514727A (en) 2020-02-21 2023-04-07 シルバーバック セラピューティックス インコーポレイテッド Nectin-4 antibody conjugates and uses thereof
CA3183993A1 (en) 2020-07-01 2022-01-06 Peter R. Baum Anti-asgr1 antibody conjugates and uses thereof
CA3194881A1 (en) 2020-08-12 2022-02-17 Innate Pharma Subcutaneous anti-c5ar antagonist treatment regimen with avdoralimab
JP7278503B2 (en) * 2020-09-24 2023-05-19 ノヴォ・ノルディスク・ヘルス・ケア・アーゲー Pharmaceutical formulation of consizumab and method for producing same
CN112244025A (en) * 2020-11-27 2021-01-22 上海创宏生物科技有限公司 Sterile treatment agent for macromolecular protein and/or nucleic acid and preparation method and application thereof
WO2024090488A1 (en) * 2022-10-26 2024-05-02 日本メジフィジックス株式会社 Radioactive pharmaceutical composition

Family Cites Families (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5567584A (en) 1988-01-22 1996-10-22 Zymogenetics, Inc. Methods of using biologically active dimerized polypeptide fusions to detect PDGF
US20020099179A1 (en) 1989-12-21 2002-07-25 Linda K. Jolliffe Cdr-grafted antibodies
DK261490D0 (en) 1990-10-31 1990-10-31 Novo Nordisk As NEW PHARMACEUTICAL COMPOUND
US6165467A (en) 1991-07-20 2000-12-26 Yoshihide Hagiwara Stabilized human monoclonal antibody preparation
US5789192A (en) 1992-12-10 1998-08-04 Schering Corporation Mammalian receptors for interleukin-10 (IL-10)
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
JP3681206B2 (en) 1995-12-26 2005-08-10 株式会社三菱化学ヤトロン Anti-factor Xa / tissue factor pathway inhibitor complex monoclonal antibody and use thereof
CN100502047C (en) 1996-06-28 2009-06-17 精工爱普生株式会社 Thin film transistor
EP2065396A1 (en) 1996-08-30 2009-06-03 Human Genome Sciences, Inc. Interleukin-19
US5985614A (en) 1996-08-30 1999-11-16 Human Genome Sciences, Inc. Polynucleotides encoding interleukin-19
US5945511A (en) 1997-02-20 1999-08-31 Zymogenetics, Inc. Class II cytokine receptor
CA2288994C (en) 1997-04-30 2011-07-05 Enzon, Inc. Polyalkylene oxide-modified single chain polypeptides
US6486301B1 (en) 1997-07-16 2002-11-26 Human Genome Sciences, Inc. Interleukin-20
JP2001510035A (en) 1997-07-16 2001-07-31 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Interleukin-20
US6020163A (en) 1997-08-06 2000-02-01 Zymogenetics, Inc. Lipocalin homolog
US6576743B1 (en) 1997-11-26 2003-06-10 Zymogenetics, Inc. Mammalian cytokine-like polypeptide-10
IL137178A0 (en) 1998-01-23 2001-07-24 Immunex Corp Il-18 receptors
CA2323236A1 (en) 1998-03-09 1999-09-16 Schering Corporation Human receptor proteins; related reagents and methods
NZ507435A (en) 1998-03-10 2003-12-19 Genentech Inc Novel polypeptides and nucleic acids with homology to cornichon
US7198789B2 (en) 1998-03-17 2007-04-03 Genetics Institute, Llc Methods and compositions for modulating interleukin-21 receptor activity
CN1233476A (en) 1998-04-24 1999-11-03 陆道培 Medicine for treating acute leukemia, and method for preparing same
AU5203199A (en) 1998-05-26 1999-12-13 Procter & Gamble Company, The Chimeric molecules comprising an extracellular ligand binding domain of a receptor and an ige fc or constant region, and their use in an assay system
AU4411699A (en) 1998-06-05 1999-12-20 Human Genome Sciences, Inc. Interferon receptor hkaef92
US7071304B2 (en) 1998-09-01 2006-07-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6440930B1 (en) * 1998-09-17 2002-08-27 Eli Lilly And Company Protein formulations
US7087215B2 (en) * 1998-12-21 2006-08-08 Generex Pharmaceuticals Incorporated Methods of administering and enhancing absorption of pharmaceutical agents
US6451286B1 (en) * 1998-12-21 2002-09-17 Generex Pharmaceuticals Incorporated Pharmaceutical compositions for buccal and pulmonary administration comprising an alkali metal alkyl sulfate and at least three micelle-forming compounds
EP1141008A1 (en) 1998-12-31 2001-10-10 Millennium Pharmaceuticals, Inc. Class ii cytokine receptor-like proteins and nucleic acids encoding them
EP1210418B1 (en) 1999-06-02 2010-08-18 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20050060101A1 (en) 1999-06-28 2005-03-17 Bevilacqua Michael P. Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
CZ20021186A3 (en) 1999-10-04 2002-11-13 Chiron Corporation Pharmaceutical preparations containing stabilized liquid polypeptide
KR100399156B1 (en) * 1999-11-19 2003-09-26 주식회사 엘지생명과학 Liquid Formulation of α-Interferon
US6610286B2 (en) 1999-12-23 2003-08-26 Zymogenetics, Inc. Method for treating inflammation using soluble receptors to interleukin-20
DK1246846T3 (en) 1999-12-23 2008-12-08 Zymogenetics Inc Soluble interleukin-20 receptor
ATE363915T1 (en) 2000-04-07 2007-06-15 Signal Coordinating Therapy In METHODS AND COMPOSITIONS FOR TREATING NEOPLASMS
US7015194B2 (en) 2000-05-10 2006-03-21 Novo Nordisk A/S Pharmaceutical composition comprising factor VIIa and anti-TFPI
EP1336410A4 (en) 2000-08-04 2005-10-12 Chugai Pharmaceutical Co Ltd Protein injection preparations
AU2001290524A1 (en) 2000-08-08 2002-02-18 Zymogenetics Inc. Soluble zcytor 11 cytokine receptors
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
CN1503677A (en) 2001-02-28 2004-06-09 Use of LP 82 to treat hematopoietic disorders
AU2002245652A1 (en) 2001-03-09 2002-09-24 Zymogenetics, Inc. Soluble heterodimeric cytokine receptor
MXPA04000747A (en) * 2001-07-25 2004-07-08 Protein Desing Labs Inc Stable lyophilized pharmaceutical formulation of igg antibodies.
ES2629395T3 (en) 2001-10-04 2017-08-09 Genetics Institute, Llc Methods and compositions to modulate the activity of interleukin-21
US7022289B1 (en) 2001-10-10 2006-04-04 The United States Of America As Represented By The Secretary Of The Army Chemical and biological sampling device and kit and method of use thereof
CN100374457C (en) * 2001-11-14 2008-03-12 森托科尔公司 Anti-IL-6 antibodies, compositions, methods and uses
AU2002357283B2 (en) 2001-12-17 2007-08-16 Zymogenetics, Inc. Method for treating cervical cancer
AU2003211990A1 (en) 2002-02-14 2003-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
US20030180287A1 (en) 2002-02-27 2003-09-25 Immunex Corporation Polypeptide formulation
JP2005530716A (en) 2002-03-27 2005-10-13 アメリカ合衆国 Methods for treating cancer in humans
US20040009168A1 (en) * 2002-04-05 2004-01-15 Elizabet Kaisheva Multidose antibody formulation
ES2381265T3 (en) 2002-06-07 2012-05-24 Zymogenetics, Inc. Use of IL-21 and monoclonal antibody to treat solid cancers
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US20040022792A1 (en) 2002-06-17 2004-02-05 Ralph Klinke Method of stabilizing proteins at low pH
EP1551875A4 (en) 2002-06-21 2006-06-28 Biogen Idec Inc Buffered formulations for concentrating antibodies and methods of use thereof
US7258221B2 (en) 2002-07-18 2007-08-21 Fabio Perini S.P.A. Storage unit for elongated products
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
WO2004055164A2 (en) 2002-12-13 2004-07-01 Abgenix, Inc. System and method for stabilizing antibodies with histidine
TR201808801T4 (en) 2003-02-10 2018-07-23 Biogen Ma Inc Immunoglobulin formulation and method of preparation thereof.
PL2335725T3 (en) 2003-04-04 2017-04-28 Genentech, Inc. High concentration antibody and protein formulations
ME02785B (en) 2003-07-15 2012-12-31 Amgen Inc Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
EP1698640B2 (en) 2003-10-01 2019-06-19 Kyowa Hakko Kirin Co., Ltd. Method of stabilizing antibody and stabilized solution-type antibody preparation
US20070184050A1 (en) 2003-12-25 2007-08-09 Kirin Beer Kabushiki Kaisha Stable water-based medicinal preparation containing antibody
EP1755677A4 (en) 2004-06-14 2009-11-25 Medimmune Vaccines Inc High pressure spray-dry of bioactive materials
AU2005299809B2 (en) 2004-10-22 2010-12-02 Zymogenetics, Inc. Anti-IL-22RA antibodies and binding partners and methods of using in inflammation
JP2008532931A (en) 2005-02-08 2008-08-21 ザイモジェネティクス, インコーポレイテッド Anti-IL-20, anti-IL-22 and anti-IL-22RA antibodies and binding partners and uses in inflammation
US7525604B2 (en) 2005-03-15 2009-04-28 Naxellent, Llc Windows with electrically controllable transmission and reflection
ATE501248T1 (en) 2005-07-02 2011-03-15 Arecor Ltd STABLE AQUEOUS SYSTEMS WITH PROTEINS
WO2007006858A2 (en) 2005-07-12 2007-01-18 Oy Jurilab Ltd Method for treatment of cardiovascular and metabolic diseases and detecting the risk of the same
CA2915270C (en) * 2005-08-05 2017-07-11 Amgen Inc. Stable aqueous protein or antibody pharmaceutical formulations and their preparation
WO2007038754A2 (en) 2005-09-27 2007-04-05 Source Mdx Gene expression profiling for identification monitoring and treatment of rheumatoid arthritis
WO2007038501A2 (en) 2005-09-27 2007-04-05 The Feinstein Institute For Medical Research Rheumatoid arthritis markers
BRPI0618850A2 (en) 2005-11-21 2011-09-13 Sanofi Pasteur Ltd stabilization formulations for recombinant viruses
EP1962907A2 (en) 2005-12-21 2008-09-03 Wyeth a Corporation of the State of Delaware Protein formulations with reduced viscosity and uses thereof
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
CA2638811A1 (en) 2006-02-03 2007-08-16 Medimmune, Llc Protein formulations
DE102006005094A1 (en) 2006-02-04 2007-08-09 Degussa Gmbh Titanium dioxide and polycarboxylate ether-containing dispersion
TW200744634A (en) 2006-02-21 2007-12-16 Wyeth Corp Methods of using antibodies against human IL-22
WO2007105133A2 (en) 2006-03-15 2007-09-20 Koninklijke Philips Electronics N.V. Remote control pointing technology with roll detection
TW200806317A (en) 2006-03-20 2008-02-01 Wyeth Corp Methods for reducing protein aggregation
EP2006380A4 (en) 2006-03-23 2010-08-11 Kyowa Hakko Kirin Co Ltd Agonistic antibody directed against human thrombopoietin receptor
AU2007229554A1 (en) * 2006-03-28 2007-10-04 F. Hoffmann-La Roche Ag Anti-IGF-1R human monoclonal antibody formulation
EP1857559A1 (en) 2006-05-16 2007-11-21 INSERM (Institut National de la Santé et de la Recherche Médicale) A method for predicting responsiveness to TNF alpha blocking agents
WO2007147001A2 (en) 2006-06-14 2007-12-21 Imclone Systems Incorporated Lyophilized formulations of anti-egfr antibodies
EP2029171A1 (en) 2006-06-19 2009-03-04 Wyeth a Corporation of the State of Delaware Methods of modulating il-22 and il-17
BRPI0712953B8 (en) 2006-06-30 2021-05-25 Novo Nordisk As anti-nkg2a antibodies, their use, and pharmaceutical composition
MX2009003635A (en) 2006-10-06 2009-04-22 Amgen Inc Stable formulations.
AU2007308145A1 (en) 2006-10-12 2008-04-17 Wyeth Modification of ionic strength in antibody-solutions to reduce opalescence/aggregates
US20100267575A1 (en) 2006-10-17 2010-10-21 Childrens Hospital Medical Center Gene array technique for predicting response in inflammatory bowel diseases
JP2010508838A (en) 2006-11-09 2010-03-25 アンスティテュ ナシオナル ドゥ ラ サントゥ エ ドゥ ラ ルシェルシェ メディカル(イーエヌエスエーエールエム) Method for predicting therapeutic response to TNF-α blocker
TW200831133A (en) 2006-12-11 2008-08-01 Hoffmann La Roche Mab Abeta lyophylized formulation
JP5419709B2 (en) 2007-01-09 2014-02-19 ワイス・エルエルシー Anti-IL-13 antibody preparation and use thereof
WO2008104608A1 (en) 2007-03-01 2008-09-04 Universite Catholique De Louvain Method for the determination and the classification of rheumatic conditions
CN101601127B (en) 2007-03-23 2011-02-16 松下电器产业株式会社 Conductive bump, method for producing the same, and electronic component mounted structure
JP5456658B2 (en) 2007-03-30 2014-04-02 メディミューン,エルエルシー Antibody preparation
GB0707938D0 (en) 2007-04-25 2007-05-30 Univ Strathclyde Precipitation stabilising compositions
WO2008132176A2 (en) 2007-04-27 2008-11-06 Universite Catholique De Louvain Method for evaluating the response of an individual to tnf blocking therapy
MX2009013410A (en) 2007-06-08 2010-03-22 Biogen Idec Inc Biomarkers for predicting anti-tnf responsiveness or non-responsiveness.
PT2170390T (en) 2007-06-14 2019-02-12 Biogen Ma Inc Natalizumab antibody formulations
US20090208492A1 (en) 2007-06-14 2009-08-20 Elan Pharmaceuticals, Inc. Lyophilized Immunoglobulin Formulations and Methods of Preparation
US20090004189A1 (en) 2007-06-18 2009-01-01 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
EP2170268A2 (en) 2007-06-25 2010-04-07 Amgen, Inc. Compositions of specific binding agents to hepatocyte growth factor
WO2009009406A1 (en) 2007-07-06 2009-01-15 Smithkline Beecham Corporation Antibody formulations
UA107557C2 (en) 2007-07-06 2015-01-26 OFATUMUMAB ANTIBODY COMPOSITION
WO2009013538A2 (en) 2007-07-20 2009-01-29 National Institute For Bioprocessing Research And Training Glycosylation markers for cancer and chronic inflammation
US20100261613A1 (en) 2007-07-26 2010-10-14 Michael Centola Methods for inflammatory disease management
US9308257B2 (en) 2007-11-28 2016-04-12 Medimmune, Llc Protein formulation
CN110698561A (en) 2007-12-14 2020-01-17 诺沃—诺迪斯克有限公司 Anti-human NKG2D antibodies and uses thereof
WO2009103113A1 (en) 2008-02-20 2009-08-27 G2 Inflammation Pty Ltd HUMANIZED ANTI-C5aR ANTIBODIES
WO2009120684A1 (en) 2008-03-25 2009-10-01 Medimmune, Llc Antibody formulation
CN103396480A (en) 2008-05-15 2013-11-20 诺沃—诺迪斯克有限公司 Antibody purification process
TWI451876B (en) * 2008-06-13 2014-09-11 Lilly Co Eli Pegylated insulin lispro compounds
US8287861B2 (en) 2008-06-30 2012-10-16 Novo Nordisk A/S Anti-human interleukin-20 antibodies
UA112050C2 (en) 2008-08-04 2016-07-25 БАЄР ХЕЛСКЕР ЛЛСі THERAPEUTIC COMPOSITION CONTAINING MONOCLONAL ANTIBODY AGAINST TISSUE FACTOR INHIBITOR (TFPI)
KR20110048536A (en) 2008-08-28 2011-05-11 와이어쓰 엘엘씨 Uses of il-22, il-17, and il-1 family cytokines in autoimmune diseases
BRPI0918419A2 (en) 2008-09-19 2015-11-24 Hoffmann La Roche new antibody formulation
EP2196476A1 (en) 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
RU2562114C2 (en) * 2008-12-22 2015-09-10 Ново Нордиск А/С Antibodies against inhibitor of tissue factor metabolic pathway
DK2414542T3 (en) 2009-03-30 2017-11-27 Tel Hashomer Medical Res Infrastructure & Services Ltd PROCEDURES FOR PREDICTING CLINICAL PROCEDURE AND TREATMENT OF MULTIPLE SCLEROSIS
MX353186B (en) 2009-09-03 2018-01-05 Genentech Inc Methods for treating, diagnosing, and monitoring rheumatoid arthritis.
US8580528B2 (en) 2009-10-16 2013-11-12 Stichting Vu-Vumc Method for prognosticating the clinical response of a patient to B-lymphocyte inhibiting or depleting therapy
WO2011047073A2 (en) 2009-10-16 2011-04-21 Mayo Foundation For Medical Education And Research Assessing rheumatoid arthritis
CN107496917B (en) 2010-02-26 2021-06-11 诺沃—诺迪斯克有限公司 Compositions comprising stable antibodies
WO2011147921A1 (en) 2010-05-28 2011-12-01 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
JP2013529089A (en) 2010-06-07 2013-07-18 エフ.ホフマン−ラ ロシュ アーゲー Gene expression markers for predicting response to drug treatment with monoclonal antibodies that inhibit interleukin-6 receptor
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9795674B2 (en) 2010-02-26 2017-10-24 Novo Nordisk A/S Stable antibody containing compositions
US10709782B2 (en) 2010-02-26 2020-07-14 Novo Nordisk A/S Stable antibody containing compositions
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
US10835602B2 (en) 2010-05-28 2020-11-17 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
US9849181B2 (en) 2012-08-31 2017-12-26 Bayer Healthcare Llc High concentration antibody and protein formulations
WO2021094917A1 (en) * 2019-11-13 2021-05-20 Pfizer Inc. Stable aqueous anti-tfpi antibody formulation

Also Published As

Publication number Publication date
JP6239657B2 (en) 2017-11-29
JP6568917B2 (en) 2019-08-28
JP2018030879A (en) 2018-03-01
CN105055306B (en) 2019-10-01
BR112012030139A2 (en) 2017-06-13
JP2013527189A (en) 2013-06-27
CN102905692B (en) 2015-09-16
KR20130086144A (en) 2013-07-31
CA2800188A1 (en) 2011-12-01
AU2011257219B2 (en) 2014-12-04
US10835602B2 (en) 2020-11-17
CN105055306A (en) 2015-11-18
WO2011147921A1 (en) 2011-12-01
US20180104335A1 (en) 2018-04-19
MX2012013586A (en) 2013-01-24
JP6294075B2 (en) 2018-03-14
JP2016084367A (en) 2016-05-19
RU2012153786A (en) 2014-07-10
EP2575761A1 (en) 2013-04-10
CN102905692A (en) 2013-01-30
AU2011257219A1 (en) 2012-11-22

Similar Documents

Publication Publication Date Title
US10835602B2 (en) Stable multi-dose compositions comprising an antibody and a preservative
US10709782B2 (en) Stable antibody containing compositions
US20200352857A1 (en) Excipients to reduce the viscosity of antibody formulations and formulation compositions
WO2016103034A1 (en) Protein compositions and use thereof
US20200147070A1 (en) Inducing phospholipidosis for enhancing therapeutic efficacy
TW202106711A (en) Antibody formulation
TW202045206A (en) Antibody formulation

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVO NORDISK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PARSHAD, HENRIK;ENGELUND, DORTE KOT;SIGNING DATES FROM 20121128 TO 20121129;REEL/FRAME:029650/0215

STCB Information on status: application discontinuation

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