WO2020060183A1 - Formation de liquide pour stabiliser un anticorps de trastuzumab - Google Patents

Formation de liquide pour stabiliser un anticorps de trastuzumab Download PDF

Info

Publication number
WO2020060183A1
WO2020060183A1 PCT/KR2019/012056 KR2019012056W WO2020060183A1 WO 2020060183 A1 WO2020060183 A1 WO 2020060183A1 KR 2019012056 W KR2019012056 W KR 2019012056W WO 2020060183 A1 WO2020060183 A1 WO 2020060183A1
Authority
WO
WIPO (PCT)
Prior art keywords
liquid formulation
trastuzumab
stabilizer
acid
antigen
Prior art date
Application number
PCT/KR2019/012056
Other languages
English (en)
Korean (ko)
Inventor
김미경
유원정
이승하
이재민
이헌주
김용국
Original Assignee
삼성바이오에피스 주식회사
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 삼성바이오에피스 주식회사 filed Critical 삼성바이오에피스 주식회사
Publication of WO2020060183A1 publication Critical patent/WO2020060183A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions

Definitions

  • the present invention relates to a stable liquid formulation comprising trastuzumab or an antigen-binding fragment thereof, and more specifically, a liquid formulation capable of stabilizing trastuzumab or an antigen-binding fragment thereof that does not contain a surfactant, and its preparation It's about how.
  • Antibody pharmaceuticals may be exposed to various environments during manufacture, transportation, storage and administration or undergo phase changes such as air-liquid, solid-liquid, or liquid-liquid. In particular, when the antibody contacts the hydrophobic interface, problems such as protein aggregation or particle formation may occur.
  • surfactants such as polysorbate 20 or polysorbate 80 are mainly used.
  • Antibody formulations comprising polysorbate 20 are known to reduce protein uptake or aggregation.
  • polysorbates are known to degrade through oxidation and hydrolysis, which in turn produces reactive oxygen species (ROS), resulting in oxidative damage to antibody proteins.
  • ROS reactive oxygen species
  • One aspect of the present invention is to provide a liquid formulation capable of maintaining stability without inducing aggregation of the antibody without including a surfactant capable of decomposing by oxidation and hydrolysis to generate ROS.
  • Another aspect of the present invention adding a stabilizer and a buffering agent to the solvent to prepare a mixed solution; And adding trastuzumab to the mixed solution.
  • One aspect of the invention (a) Trastuzumab or an antigen-binding fragment thereof; (b) stabilizer; And (c) a buffering agent and no surfactant.
  • Another aspect of the present invention adding a stabilizer and a buffering agent to the solvent to prepare a mixed solution; And adding trastuzumab to the mixed solution.
  • liquid formulation according to one aspect of the present invention can maintain the stability of the antibody without including polysorbate, which may cause ROS under stress conditions.
  • Example 1 is a change in the percent high molecular weight material (percent high molecular weight,% HMW) of Example 1 and Comparative Example 1 under temperature stress conditions by size exclusion-high performance liquid chromatography (Size-Exclusion HPLC, SE-HPLC) It is a graph showing one result.
  • Example 1 The formulations of Example 1 and Comparative Example 1 were prepared.
  • trastuzumab (5 mM histidine buffer (pH 6.0), 40 mg / mL, Samsung Bioepis) and a buffering agent and a stabilizer, except for the surfactant, are made of polycarbonate material of 5L baotainer (biotainer) ) 5L buffer solution was prepared by adding to sterile distilled water in a container.
  • trastuzumab solution in 5 mM histidine buffer at pH 6.0 was placed in a dialysis cassette (MWCO 10 kDa) (Slide-A-Lyzer cassette, Thermo Fisher Scientific), and then a cassette containing this trastuzumab solution was placed in a 2L beaker containing 1,600 mL of the buffer solution to make dialysis, and the 5 mM histidine buffer at pH 6.0 was exchanged with the buffer solution. Thereafter, trastuzumab was concentrated to 120 mg / mL or more.
  • MWCO 10 kDa Slide-A-Lyzer cassette, Thermo Fisher Scientific
  • Example 1 Concentration was achieved by passing the dialysis trastuzumab containing solution through an Amicon Filter with a cutoff molecular weight of 30 kDa. Finally, to the concentrated trastuzumab solution, a surfactant prepared at a concentration of 20x, that is, 0.8 w / v%, in the buffer solution prepared above was added to a concentration of 1x.
  • the composition of Comparative Example 1 is also listed in Table 1. The formulation of Example 1 had a pH of about 5.5.
  • Comparative Example 1 was prepared in the same manner as in Example 1, except that polysorbate 20 was added as a surfactant.
  • the pH of the formulation obtained after adding the surfactant in Comparative Example 1 was about 5.5.
  • Table 1 shows the composition of the formulations of Example 1 and Comparative Example 1.
  • Example 1 Comparative Example 1 Trastuzumab (mg / mL) 120 120 Buffering agent Histidine (mM) 20 20 Stabilizer Methionine (mM) 10 10 Trehalose (w / v%) 8 8 Surfactants Polysorbate 20 (w / v%) - 0.04
  • Test Example 1 Check the stability of the surfactant-free formulation
  • Example 1 Although the preparation of Example 1 prepared does not contain a surfactant, it was confirmed by SE-HPLC whether it can maintain stability under temperature stress and freeze-thaw stress.
  • Example 1 and Comparative Example 1 For the temperature stress, 1 mL of each of the formulations of Example 1 and Comparative Example 1 was placed in a 1.5 mL microtube made of polypropylene (Axygen), and the tube was heated to 40 ° C. in a stability thermostat (JEIO TECH). They were exposed to temperature stress conditions for 4 weeks. Specifically, the tube was placed in the stability thermostat and placed for 4 weeks under conditions of temperature 40 ⁇ 2 ° C. and relative humidity 75 ⁇ 5%. % HMW,% monomer and% LMW were measured using SE-HPLC for formulations stored for 4 weeks.
  • a stability thermostat JEIO TECH
  • HMW high molecular weight
  • LMW low molecular weight
  • Example 1 1 mL of each of the formulations of Example 1 and Comparative Example 1 was placed in a 1.5 mL microtube (Axygen) made of polypropylene, and the tube was maintained at -70 ⁇ 10 ° C. After placing it in a deep freezer (Thermo scientific) and maintaining it for 18 hours in a frozen and frozen state, the tube was taken out and left at room temperature for 1 hour to keep it in a thawed and thawed state. This process was repeated 5 times. If the antibody in the formulation loses stability, aggregation will be observed to increase the proportion of high molecular weight (HMW) material.
  • HMW high molecular weight
  • Example 1 showed better stability than Comparative Example 1 in freeze-thaw stress. Specifically, the formulation of Example 1 reduced ⁇ % HMW by 33% compared to the formulation of Comparative Example 1 in freeze-thaw stress.
  • the surfactant-free formulations identified by this example were more stable than the surfactant-containing formulations, which is a remarkable remarkable effect by those skilled in the art.
  • One aspect of the invention (a) Trastuzumab or an antigen-binding fragment thereof; (b) stabilizer; And (c) a buffering agent and no surfactant.
  • antigen binding fragment is a fragment of a portion capable of binding to an antigen in a Trastuzumab antibody, including, but not limited to, Fab, F (ab ′) 2 and Fv, for example.
  • the antigen can be a HER2 receptor.
  • Trastuzumab is a monoclonal antibody used to treat cancer, including breast cancer, and is marketed under the name Herceptin TM under one of several trade names. Trastuzumab is used for breast cancer that is positive for HER2 receptors. In addition, trastuzumab can be used alone or in combination with other chemotherapeutic agents. Trastuzumab is administered by intravenous injection and subcutaneous injection. Trastuzumab works by binding to the HER2 receptor and slows cell replication. Trastuzumab is a recombinant IgG1 kappa humanized from mice that binds to the extracellular domain of human epidermal growth factor receptor protein (HER2), a whole antibody.
  • HER2 human epidermal growth factor receptor protein
  • the concentration of trastuzumab or an antigen-binding fragment thereof in the liquid formulation may be 2 to 300 mg / mL.
  • the concentration of the antibody can be freely adjusted within a range that does not substantially affect the stability, viscosity and desired pH of the liquid formulation of the present invention.
  • the concentration of the trastuzumab or antigen binding fragment thereof is, for example, 10 to 300 mg / mL, 20 to 300 mg / mL, 30 to 300 mg / mL, 50 to 300 mg / mL, 80 To 300 mg / mL, 100 to 300 mg / mL, 80 to 250 mg / mL, 80 to 200 mg / mL, 80 to 180 mg / mL, 80 to 150 mg / mL, 100 to 150 mg / mL, 100 to 140 mg / mL, 105-135 mg / mL, 110-130 mg / mL, 115-125 mg / mL, 100-250 mg / mL, 100-200 mg / mL, 100-180 mg / mL, 100-150 mg / mL, 100-140 mg / mL, 150-300 mg / mL, 200-300 mg / mL, 220-300 mg / m
  • the liquid formulation of the present invention does not contain a surfactant.
  • the liquid formulations of the present invention may or may not contain surfactant at all. When substantially free of surfactant, it can mean, for example, that the surfactant comprises less than 0.04 w / v%, less than 0.02 w / v%, or less than 0.01 w / v%.
  • surfactants not included in the liquid formulation of the present invention include polyoxyethylene sorbitan fatty acid ester (Tween), polyethylene-polypropylene glycol, polyoxyethylene stearate, polyoxyethylene alkyl ether, polyoxyethylene- Polyoxypropylene copolymer (poloxamer), or sodium dodecyl sulfate.
  • the polyoxyethylene alkyl ether may be, for example, polyoxyethylene monolauryl ether, alkylphenylpolyoxyethylene 30 ether (Triton-X).
  • the polysorbate is a polyoxyethylene sorbitan fatty acid ester or a derivative thereof. Polysorbates are known to degrade through oxidation and hydrolysis, which in turn produces ROS, which is known to cause oxidative damage to antibody proteins.
  • the liquid formulation of the present invention can maintain the stability of the antibody and antigen-binding fragment thereof without containing a surfactant such as polysorbate. Further, in one embodiment, the surfactant that is not included in the liquid formulation of the present invention may be polysorbate.
  • the polysorbate may mean polysorbate 20 or polysorbate 80.
  • the liquid formulation of the present invention may be one that does not include polysorbate 20 and polysorbate 80.
  • the stabilizer may be one or more selected from the group consisting of sugar, sugar alcohol, sugar acid, polyol, metal salt and amino acid.
  • the sugar may be selected from monosaccharides, disaccharides, oligosaccharides, polysaccharides, or derivatives thereof.
  • the sugar derivative may mean sugar alcohol or sugar acid.
  • the sugar, sugar alcohol or sugar acid is glucose, fructose, galactose, sucrose, lactose, maltose, trehalose, fructooligosaccharide, galactoligosaccharide, mango-oligosaccharide, starch, glycogen, cellulose, chitin, pectin , Glycerol, erythritol, thritol, arabitol, xylitol, ribitol, mannitol, sorbitol, galactitol, fusitol, iditol, inositol, boletitol, isomalt, maltitol, lactitol, maltotriitol, maltotetraitol ,
  • the concentration of the sugar, sugar alcohol or sugar acid may be 1 to 20w / v%. In another embodiment, the concentration of the sugar, sugar alcohol or sugar acid is 2 to 20w / v%, 4 to 20w / v%, 6 to 20w / v%, 6 to 12w / v%, 4 to 14w / v%, respectively.
  • the concentration of sugar, sugar alcohol or sugar acid can be freely adjusted within a desired range to maintain the stability of the trastuzumab or antigen-binding fragment thereof in the liquid formulation of the present invention and the viscosity of the liquid formulation, and each specific sugar, sugar alcohol or It can vary individually depending on the sugar acid.
  • polyol can mean an organic compound having two or more hydroxyl groups (-OH) in its molecule.
  • the polyol is any one selected from the group consisting of propanediol, glycerin, butylene glycol, propylene glycol, dipropylene glycol, pentylene glycol, hexylene glycol, polyethylene glycol, and sorbitol You can.
  • the metal salt may be NaCl, KCl, NaF, KBr, NaBr, Na 2 SO 4 , NaSCN, or K 2 SO 4 , but is not limited thereto. In one embodiment, the metal salt may be NaCl. The concentration of the metal salt may be 0.1 to 10w / v%.
  • the concentration of the metal salt is 0.1 to 8w / v%, 0.1 to 6w / v%, 0.1 to 5w / v%, 0.1 to 3w / v%, 0.1 to 1w / v%, 0.3 to 10w / v %, 0.3 to 8 w / v%, 0.3 to 6 w / v%, 0.3 to 5 w / v%, 0.3 to 3 w / v% or 0.3 to 1 w / v%, 0.5 to 10 w / v%, 0.5 to 8 w / v% , 0.5 to 6w / v%, 0.5 to 5w / v%, 0.5 to 3w / v% or 0.5 to 1w / v%.
  • the concentration of the metal salt can be freely adjusted within a range in which the stability of trastuzumab or an antigen-binding fragment thereof in the liquid formulation of the present invention is not precipitated, and
  • the amino acid is glycine, alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, histidine, glutamine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine It may be one or more selected from the group consisting of.
  • the concentration of amino acids can be freely adjusted within a range that does not affect the desired pH of the liquid formulation of the present invention, and does not affect the stability of trastuzumab or antigen-binding fragments thereof, and can be individually adjusted for each specific amino acid. It may vary.
  • the amino acid may be methionine.
  • the stabilizer may include two or more stabilizers.
  • the stabilizer may be one or more selected from the group consisting of trehalose and methionine.
  • the stabilizer may be one or more selected from the group consisting of 1 to 15w / v% trehalose and 1 to 50 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 2 to 14w / v% trehalose and 1 to 30 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 3 to 13w / v% trehalose and 1 to 20 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 4 to 12w / v% trehalose and 1 to 20 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 5 to 11w / v% trehalose and 2 to 18 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 6 to 10w / v% trehalose and 2 to 18 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 6 to 10w / v% trehalose and 4 to 16 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 6 to 10w / v% trehalose and 6 to 14 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 7 to 9w / v% trehalose and 8 to 12 mM methionine.
  • the stabilizer may be at least one selected from the group consisting of 2 to 8w / v% trehalose and 1 to 20 mM methionine.
  • the buffering agent may be to provide a pH of 4.0 to 7.0.
  • the liquid formulation may have a pH of 4.0 to 7.0.
  • the pH of the liquid formulation may be 4.5 to 6.5, 5.0 to 6.0 or 5.5 ⁇ 0.2.
  • the pH of the liquid formulation can be freely adjusted within a range suitable for preventing aggregation of the antibody and maintaining activity.
  • the liquid formulation of the present invention may contain a buffering agent to maintain the desired pH for stabilizing trastuzumab or an antigen-binding fragment thereof.
  • buffer refers to a neutralizing material that minimizes the change in pH by acid or alkali, and in one embodiment, the buffering agent is histidine, phosphoric acid, citric acid, maleic acid, tartaric acid, succinic acid, citrate, acetate , Carbonate and salts thereof, but are not limited thereto.
  • the buffering agent is histidine. In one embodiment, the buffering agent is 1 to 100 mM histidine. The concentration of histidine is, for example, 1 to 80 mM, 1 to 70 mM, 1 to 60 mM, 1 to 50 mM, 1 to 40 mM, 1 to 30 mM, 2 to 80 mM, 2 to 70 mM, 2 To 60 mM, 2 to 50 mM, 2 to 40 mM, 2 to 30 mM, 2 to 25 mM, 3 to 80 mM, 3 to 70 mM, 3 to 60 mM, 3 to 50 mM, 3 to 40 mM, 3 To 30 mM, 3 to 25 mM, 4 to 80 mM, 4 to 70 mM, 4 to 60 mM, 4 to 50 mM, 4 to 40 mM, 4 to 30 mM, 4 to 25 mM, 5 to 80 mM, 5 To 70 mM, 5 to 60 mM, 5 to 60 m
  • the histidine may be in the form of histidine and its conjugate acid.
  • the conjugate acid may be histidine HCl.
  • the ratio of histidine and its conjugate acid may vary depending on the pH of the selected agent. For example, when the pH of the formulation is 5.5 ⁇ 0.2, the concentration ratio of histidine and histidine HCl monohydrate may be about 1: 6.
  • the stabilizer, buffering agent, and surfactant may be different.
  • the trastuzumab or antigen-binding fragment thereof may be stabilized.
  • the liquid formulation may be a stable liquid pharmaceutical formulation.
  • stabilization means that trastuzumab or an antigen-binding fragment thereof substantially retains its physical stability, chemical stability and / or biological activity before and after administration, during further manufacturing processes, storage or storage. Physical stability, chemical stability and / or biological activity can be assessed by commonly known methods.
  • Stability can be measured for a selected period of time at a selected temperature.
  • a stable formulation is one in which no significant change is observed at 2-8 ° C. for 12 months or longer.
  • a stable formulation is one that does not exhibit significant changes at 2-8 ° C. for 18 months or longer.
  • a stable formulation is one where no significant change is observed for 3 months or longer at 23-27 ° C.
  • a stable formulation is one where no significant changes are observed for 6 months or longer at 23-27 ° C.
  • a stable formulation is one where no significant change is observed at 23-27 ° C. for 12 months or longer.
  • a stable formulation is one where no significant change is observed at 23-27 ° C.
  • Stability criteria for antibody formulations are as follows.
  • the antibody monomer is 10% or less, for example, 5% or less, or 2.5% or less relative to the initial monomer amount.
  • the antibody potency is within 60 to 140%, or 80 to 120%, of the control or standard antibody.
  • LMW low molecular weight
  • the change in the amount of the low molecular weight species is less than 10%, less than 5%, or less than 2.5% relative to its initial amount. It may have.
  • the change in the amount of the high molecular weight species is 10% or less, 5% or less, or 2.5% or less relative to its initial amount. It may have.
  • the concentration of the formulation, and the pH may have a change of 0% or less, ⁇ 5% or less, or ⁇ 2.5% or less relative to the initial value.
  • the initial pH is 10% or less, 5% or less, or 2.5% or less
  • the pH changes within the range.
  • the formulation is placed in a freezer maintained at -70 ⁇ 10 ° C and maintained for 18 hours to maintain a frozen state, and then the process of removing the tube and leaving it at room temperature for 1 hour to maintain it in a thawed state is repeated 5 times.
  • the pH changes within the range of 10% or less, 5% or less, or 2.5% or less of the initial pH.
  • the preparation is when 1 mL is placed in a 1.5 mL microtube (Axygen) made of polypropylene, and the tube is mounted on a stirrer (Heidolph) and stirred at 400 rpm for 72 hours at room temperature.
  • PH is changed within the range of 10% or less, 5% or less, or 2.5% or less with respect to the initial pH.
  • the trastuzumab or antigen-binding fragment thereof retains chemical stability within the pharmaceutical agent when it is chemically stable at a predetermined time that appears to still retain biological activity.
  • Chemical stability can be assessed by detecting and quantifying a chemically modified form of the antibody.
  • Chemical modification includes size modification or charge change.
  • the charge change can be, for example, a charge change that occurs as a result of deamidation.
  • the size modification is size exclusion chromatography (SE-HPLC), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), capillary electrophoresis-sodium dodecyl sulfate, CE-SDS ) Analysis, and substrate-assisted laser desorption / ionization / time-of-flight mass spectrometry (MALDI / TOF MS).
  • charge changes can be evaluated by ion exchange chromatography (IEC), and imaged capillary isoelectric focusing (icIEF).
  • Activity can be determined through HER2 receptor binding assay.
  • the HER2 receptor binding assay can be performed by Enzyme-linked immuonsorbent assay (ELISA).
  • ELISA analysis measures the ratio (%) of trastuzumab binding to the HER2 receptor.
  • ELISA can be performed using the Her2 ELISA kit (R & D system). After reacting with the trastuzumab sample and the HER2 receptor on the ELISA substrate, the binding degree can be determined by measuring the absorbance at 450 nm to obtain a relative binding rate (%).
  • the trastuzumab or antigen-binding fragment thereof retains biological activity in the pharmaceutical agent when the trastuzumab or antigen-binding fragment thereof is biologically active for its intended purpose.
  • the biological activity is, for example, about 30%, about 20%, within about 10%, about 5% of the biological activity at which the biological activity of the trastuzumab or antigen-binding fragment thereof in the pharmaceutical agent is indicated at the time of manufacture of the pharmaceutical agent.
  • Biological activity is retained within or within analytical error.
  • the biological activity can be determined, for example, in antigen binding assays.
  • Stabilization is temperature stress, e.g., 1 to 4 weeks at 40 ° C, freeze-thaw stress, e.g., 5 repetitions of cycles at -70 ° C freeze and thaw at room temperature, or stirring stress, e.g. , Can be evaluated by measuring% HMW,% monomer and / or% LMW using SE-HPLC, by applying 400 rpm rotational force for 72 hours in a stirrer.
  • the liquid formulations of the present invention may have less ⁇ % HMW, ⁇ % monomer and / or ⁇ % LMW values compared to Herceptin®.
  • the liquid formulation provided herein has a change in% HMW measured when stored at 40 ° C for 4 weeks at 40 ° C when the antibody content is 120 mg / ml (pH 5.5), that is,% HMW at week 4 of storage- Week 0% HMW value is about 5% or less, about 3% or less, about 2.5% or less, about 2.3% or less, about 2.0% or less, about 0.5 to about 5%, about 0.8 to about 3%, about 0.1 to About 2.25%, about 1.00 to about 2.23%, about 1.15% ⁇ 0.08%, and also about 1.15% ⁇ 0.04%.
  • the "week 0" indicates the initial time of storage.
  • the liquid formulation provided herein is thawed by maintaining the liquid composition at -70 ⁇ 10 ° C for 18 hours, and then standing at room temperature for 1 hour when the antibody content is 120 mg / ml (pH 5.5).
  • the change amount of% HMW measured by conventional SEC is about 2% or less, about 1.5% or less, about 1.0% or less, about 0.5% or less, about 0.1% or less, about 0.05% or less, or about 0.01% or less, about 0.001% or less, about 0.1 to about 1%, about -0.05 to about 0.05%, about -0.03 to about 0.03%, about -0.02% ⁇ 0.02%, or about -0.02% ⁇ 0.01% have.
  • the liquid formulation may be for subcutaneous or intravenous injection.
  • the liquid formulation may further include an aqueous carrier suitable for injection.
  • the aqueous carrier may be a safe and non-toxic pharmaceutically acceptable drug when administered to humans.
  • the aqueous carrier includes, but is not limited to, sterile alcohol, for example, sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), sterile saline solution, Ringer's solution, and dextrose.
  • the liquid formulation of the present invention may have an appropriate range of osmolality upon subcutaneous or intravenous injection.
  • Osmolarity may be, for example, 200 to 400 mOsm / kg, 250 to 380 mOsm / kg, or 270 to 360 mOsm / kg.
  • the osmolarity concentration can be appropriately adjusted to minimize pain that may occur during administration.
  • the liquid formulation of the present invention may have an appropriate range of viscosity when subcutaneously or intravenously injected.
  • the viscosity for example, 0.5 to 50 cp, can be appropriately adjusted to minimize pain that may occur upon administration.
  • the liquid formulation of the present invention may include a hyaluronidase enzyme.
  • the amount of the hyaluronidase enzyme may vary depending on the hyaluronidase enzyme used.
  • the amount of hyaluronidase enzyme can be an amount sufficient to increase the dispersion and absorption of an anti-HER2 antibody, e.g., trastuzumab or an antigen binding fragment thereof, administered together.
  • the amount of hyaluronidase enzyme can be 150 U / ml or more.
  • An effective amount of hyaluronidase enzyme is, for example, about 1,000 to 16,000 U / ml, where the amount corresponds to about 0.01 mg to 0.16 mg based on 100,000 U / mg of estimated specific activity.
  • the concentration of the hyaluronidase enzyme may be about 1,500 to 12,000 U / ml, or about 2,000 to 12,000 U / ml. This amount corresponds to the amount of hyaluronidase enzyme added to the formulation.
  • the amount of hyaluronidase enzyme measured in the final formulation can vary within a specific range.
  • the ratio (w / w) of hyaluronidase enzyme to anti-HER2 antibody is 1: 5,00 to 1: 100,000, 1: 1,000 to 1: 50,000, 1: 1,000 to 1: 30,000, 1: 1,000 to 1: 20,000, 1: 1,000 to 1: 15,000, 1: 1,000 to 1: 13,000, 1: 1,000 to 1: 12,000, 1: 1,000 to 1: 10,000, 1: 1,000 to 1: 8,000, 1: 4,000 to 1: 5,000, Or about 1: 6,000.
  • the hyaluronidase enzyme may be derived from an animal, bacterial, or human sample or prepared by recombinant DNA technology.
  • the hyaluronidase enzyme may be soluble hyaluronidase glycoproteins (sHASEGPs). These soluble hyaluronidase glycoproteins can be administered together with the agent or separately. The addition of the soluble hyaluronidase glycoprotein can facilitate the subcutaneous administration of the therapeutic drug.
  • soluble hyaluronidase glycoprotein By rapidly depolymerizing hyaluronan (HA) in the extracellular space, the soluble hyaluronidase glycoprotein lowers the viscosity of the interstitium, increasing hydraulic conductance, making it easier for large volumes to subcutaneous tissue. Make it administered comfortably.
  • the increased hydraulic conductivity induced by soluble hyaluronidase glycoproteins through reduced interstitial viscosity can result in better dispersion, thereby increasing the systemic bioavailability of the subcutaneously administered therapeutic drug.
  • the hyaluronidase enzyme may be licensed for use in humans in any one or more countries.
  • the hyaluronidase enzyme may be, for example, Hylase TM , Dessau, Hyalase TM , EU approved, Vitrase TM , Hydase TM , Amphadase TM , or Hylenex TM , licensed in the United States.
  • the hyaluronidase enzyme may be a human hyaluronidase enzyme such as PH20 or rHuPH20. Recombinant human PH20 (rHuPH20) is a member of the neutral and acid-active ⁇ -1,4 glycosyl hydrolase family.
  • rHuPH20 depolymerizes hyaluronan by hydrolysis of a ⁇ -1,4 bond between the C1 position of N-acetyl glucosamine and the C4 position of glucuronic acid.
  • rHuPH20 may be a truncated deletion variant without amino acid at the carboxy terminal required for lipid attachment developed by Halozyme.
  • Another aspect of the present invention provides a liquid formulation, wherein the trastuzumab or antigen-binding fragment thereof is 2 to 300 mg / mL, the stabilizer is a combination of trehalose and methionine, and the buffering agent is histidine. .
  • the stabilizer is a combination of 1 to 20 w / v% trehalose and 1 to 20 mM methionine, and the buffering agent is 10 to 50 mM histidine.
  • the concentration of the trehalose is 2 to 20w / v%, 4 to 20w / v%, 6 to 20w / v%, 1 to 18w / v%, 1 to 15w / v%, 1 to 12w / v%, 1 to 10w / v%, 2 to 18w / v%, 2 to 15w / v%, 2 to 12w / v%, 2 to 10w / v%, 4 to 18w / v%, 4 to 15w / v%, 4 to 12w / v% or 4 to 10 w / v%.
  • the concentration of the methionine is 1 to 20 mM, 2 to 20 mM, 5 to 20 mM, 8 to 20 mM, 1 to 18 mM, 2 to 18 mM, 5 to 18 mM, 1 to 15 mM, 2 to 15 mM, 5 to 15 mM, 8 to 15 mM, 1 to 12 mM, 2 to 12 mM, 5 to 12 mM, or 8 to 12 mM.
  • the histidine may be 10 to 45 mM, 10 to 40 mM, 10 to 35 mM, 10 to 30 mM, 15 to 25 mM, or 18 to 22 mM.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention (a) Trastuzumab or an antigen-binding fragment 80 to 160 mg / ml thereof; (b) 1 to 20 mM methionine and 1 to 15 (w / v)% of trehalose as stabilizers; And (c) 1-50 mM histidine as a buffering agent.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention is (a) Trastuzumab or an antigen-binding fragment 80 to 160 mg / ml thereof; (b) 1 to 20 mM methionine and 1 to 15 (w / v)% trehalose as stabilizers; And (c) 1 to 40 mM histidine as a buffering agent.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention is (a) Trastuzumab or an antigen-binding fragment 100 to 140 mg / ml thereof; (b) 3 to 18 mM of methionine and 2 to 13 (w / v)% of trehalose as stabilizers; And (c) 10-30 mM histidine as a buffering agent.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention (a) Trastuzumab or an antigen-binding fragment 110 to 130 mg / ml thereof; (b) 5 to 15 mM methionine as stabilizer and 4 to 12 (w / v)% trehalose; And (c) 15 to 25 mM histidine as a buffering agent.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention (a) Trastuzumab or an antigen-binding fragment 110 to 130 mg / ml thereof; (b) 8 to 12 mM methionine and 6 to 10 (w / v)% threonyl as stabilizers; And (c) 16 to 24 mM histidine as a buffering agent.
  • the formulation may be pH 5.5 ⁇ 0.2.
  • One embodiment of the present invention (a) Trastuzumab or an antigen-binding fragment thereof 120 mg / ml ⁇ 5%; (b) 10 mM ⁇ 5% methionine as stabilizer and 8% (w / v)% ⁇ 5% trehalose; And (c) 20 mM ⁇ 5% histidine as a buffering agent.
  • ⁇ 5% indicates that there is a variation of “ ⁇ 5%” with respect to the basic amount.
  • the liquid formulation may be pH 5.5 ⁇ 0.2.
  • the histidine may include histidine and its conjugate acid, for example, histidine HCl.
  • the buffering agent may include only histidine and its conjugate acid.
  • the formulation may be one that does not contain a surfactant, for example, a nonionic surfactant.
  • the formulation may not contain polysorbate 20 or 80.
  • Another aspect of the present invention adding a stabilizer and a buffering agent to the solvent to prepare a mixed solution; And adding trastuzumab to the mixed solution.

Abstract

La présente invention concerne une formulation liquide comprenant : un trastuzumab ou un fragment de liaison d'antigène de celui-ci ; un stabilisant ; et un agent tampon, la formulation liquide étant exempte d'un tensioactif, ainsi que son procédé de préparation.
PCT/KR2019/012056 2018-09-18 2019-09-18 Formation de liquide pour stabiliser un anticorps de trastuzumab WO2020060183A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20180111666 2018-09-18
KR10-2018-0111666 2018-09-18

Publications (1)

Publication Number Publication Date
WO2020060183A1 true WO2020060183A1 (fr) 2020-03-26

Family

ID=69888559

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/KR2019/012056 WO2020060183A1 (fr) 2018-09-18 2019-09-18 Formation de liquide pour stabiliser un anticorps de trastuzumab
PCT/KR2019/012077 WO2020060192A1 (fr) 2018-09-18 2019-09-18 Formulation liquide stabilisante de trastuzumab contenant une concentration élevée de tensioactif

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/KR2019/012077 WO2020060192A1 (fr) 2018-09-18 2019-09-18 Formulation liquide stabilisante de trastuzumab contenant une concentration élevée de tensioactif

Country Status (1)

Country Link
WO (2) WO2020060183A1 (fr)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113855798A (zh) * 2021-09-30 2021-12-31 佛山汉腾生物科技有限公司 抗cd20的抗体制剂
WO2023100095A1 (fr) * 2021-11-30 2023-06-08 Enzene Biosciences Limited Composition comprenant un anticorps her2, formulation et son procédé de préparation
WO2024022414A1 (fr) * 2022-07-29 2024-02-01 舒泰神(北京)生物制药股份有限公司 Préparation d'anticorps monoclonal à haute concentration et son utilisation

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004066957A2 (fr) * 2003-01-30 2004-08-12 Medimmune, Inc. Preparation d'anticorps anti-integrine $g(a)v$g(b)3 et utilisations associees
JP2006511448A (ja) * 2002-06-14 2006-04-06 メディミューン,インコーポレイテッド 安定化された抗呼吸器合胞体ウイルス(rsv)抗体製剤
US7132100B2 (en) * 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
KR20130041374A (ko) * 2009-07-31 2013-04-24 에프. 호프만-라 로슈 아게 피하용 항-에이치이알2 항체 제형
KR20180003452A (ko) * 2016-06-30 2018-01-09 (주)셀트리온 안정한 액체 약제학적 제제
KR20180085801A (ko) * 2015-12-07 2018-07-27 메르크 파텐트 게엠베하 항-pd-1 항체 아벨루맙을 포함하는 수성 약학 제형

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009006301A2 (fr) * 2007-06-29 2009-01-08 Battelle Memorial Institute Stabilisation de protéines
MX345226B (es) * 2008-10-29 2017-01-20 Ablynx Nv Formulaciones de moleculas de union a antigeno de dominio sencillo.
EP3402470A4 (fr) * 2016-01-12 2019-11-13 Dr. Reddy's Laboratories Limited Composition pharmaceutique stable

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006511448A (ja) * 2002-06-14 2006-04-06 メディミューン,インコーポレイテッド 安定化された抗呼吸器合胞体ウイルス(rsv)抗体製剤
US7132100B2 (en) * 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
WO2004066957A2 (fr) * 2003-01-30 2004-08-12 Medimmune, Inc. Preparation d'anticorps anti-integrine $g(a)v$g(b)3 et utilisations associees
KR20130041374A (ko) * 2009-07-31 2013-04-24 에프. 호프만-라 로슈 아게 피하용 항-에이치이알2 항체 제형
KR20180085801A (ko) * 2015-12-07 2018-07-27 메르크 파텐트 게엠베하 항-pd-1 항체 아벨루맙을 포함하는 수성 약학 제형
KR20180003452A (ko) * 2016-06-30 2018-01-09 (주)셀트리온 안정한 액체 약제학적 제제

Also Published As

Publication number Publication date
WO2020060192A1 (fr) 2020-03-26

Similar Documents

Publication Publication Date Title
WO2020060183A1 (fr) Formation de liquide pour stabiliser un anticorps de trastuzumab
AU2016283557B9 (en) A stable liquid formulation of fusion protein with IgG Fc domain
WO2011090305A2 (fr) Formulations liquides pour un conjugué de g-csf à action prolongée
WO2012008779A2 (fr) Formulation liquide d'un conjugué d'hormone de croissance humaine à action prolongée
WO2012165917A1 (fr) Formulation liquide stable d'étanercept
EP2328559B1 (fr) Formulation comprenant un anticorps contre p-selectin
EP2399604A1 (fr) Nouvelle formulation d'anticorps
US20110158987A1 (en) Novel antibody formulation
HU220256B (hu) Granulocita-kolónia stimuláló faktort (G-CSF) tartalmazó, stabilan eltartható, liofilizált gyógyszerkészítmények és eljárás az előállításukra
US20090068196A1 (en) Pharmaceutical formulation of an antibody against IL13Ralpha1
WO2013117969A1 (fr) Procédé pour préparer des compositions pharmaceutiques stables de composés sensibles à l'hydrolyse
US20160120946A1 (en) Compositions comprising gc-macrophage activating factor and uses thereof
WO2021118321A1 (fr) Préparation pharmaceutique d'anticorps anti-pd-1 stable
WO2013147559A1 (fr) Formulation liquide de conjugué d'hormone de croissance humaine à action prolongée hautement concentrée
EA020069B1 (ru) Жидкая лекарственная форма конъюгата
KR101699677B1 (ko) 고나도트로핀을 위한 제제
US20170112903A1 (en) Lyophilized pharmaceutical composition of fc-peptide fusion protein
WO2020060179A1 (fr) Formulation de stabilisation d'anticorps trastuzumab exempte d'agent tampon
US20210188941A1 (en) Stable fusion protein formulation
WO2021125852A1 (fr) Nouvelle formulation injectable
US10052361B2 (en) Liquid pharmaceutical composition of conjugated erythropoietin
EP0508435B1 (fr) Composition pharmaceutique de [Leu 13]-motiline-Hse stabilisée
WO2021071326A1 (fr) Composition liquide stable, procédé pour sa préparation, et formulation la comprenant
WO2021154027A1 (fr) Préparation pharmaceutique d'anticorps anti-pd-1 stable

Legal Events

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

Ref document number: 19863212

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19863212

Country of ref document: EP

Kind code of ref document: A1