NZ748101B2 - Stable liquid pharmaceutical preparation - Google Patents

Stable liquid pharmaceutical preparation Download PDF

Info

Publication number
NZ748101B2
NZ748101B2 NZ748101A NZ74810117A NZ748101B2 NZ 748101 B2 NZ748101 B2 NZ 748101B2 NZ 748101 A NZ748101 A NZ 748101A NZ 74810117 A NZ74810117 A NZ 74810117A NZ 748101 B2 NZ748101 B2 NZ 748101B2
Authority
NZ
New Zealand
Prior art keywords
ser
val
thr
leu
gly
Prior art date
Application number
NZ748101A
Other versions
NZ748101A (en
Inventor
Won Yong Han
Su Hyeon Hong
So Young Kim
Su Jung Kim
Joon Won Lee
Jun Seok Oh
Yeon Kyeong Shin
Original Assignee
Celltrion Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celltrion Inc filed Critical Celltrion Inc
Priority claimed from PCT/KR2017/006855 external-priority patent/WO2018004260A1/en
Publication of NZ748101A publication Critical patent/NZ748101A/en
Publication of NZ748101B2 publication Critical patent/NZ748101B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • 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
    • 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
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)

Abstract

stable liquid pharmaceutical preparation according to the present invention contains: the antibody infliximab; a surfactant; a sugar or a derivative thereof; and a buffer comprising acetate or histidine. The stable liquid pharmaceutical preparation according to the present invention has low viscosity while containing a high amount of the antibody, has excellent long-term storage stability on the basis of excellent stability under accelerated conditions and extreme conditions, and can be administered subcutaneously. ity while containing a high amount of the antibody, has excellent long-term storage stability on the basis of excellent stability under accelerated conditions and extreme conditions, and can be administered subcutaneously.

Description

STABLE LIQUID PHARMACEUTICAL FORMULATION Field of the Invention The present invention relates to a stable liquid pharmaceutical formulation.
Description of the Related Art Tumor necrosis factor-α (TNF-α) is a cell signaling protein (cytokine) that is involved in systemic inflammation and is a cytokine that mediates acute-phase responses. TNF-α is related to various diseases and disorders, including septicemia, infection, autoimmune diseases, and graft rejection. TNF-α stimulates immune responses and causes many clinical problems associated with autoimmune abnormalities such as rheumatoid arthritis, ankylosing spondylitis, ulcerative colitis, adult Crohn's disease, pediatric Crohn's disease, psoriasis, psoriatic arthritis and the like. Such abnormalities may be treated using TNF-α inhibitors.
[0003] Infliximab is a kind of chimeric monoclonal antibody that can function as a TNF-α inhibitor. Conventional formulations containing this antibody are prepared as freeze-dried powders, which are reconstituted, diluted, and injected intravenously using a dosage regimen determined according to each disease.
For example, the Remicade label discloses a freeze-dried formulation containing infliximab, sucrose, polysorbate 80 and sodium phosphate. For intravenous injection, it discloses a reconstitution step of adding injectable water to the freeze-dried formulation, and a step of diluting the reconstituted formulation with injectable saline containing sodium chloride.
However, the mode of administration of the conventional formulation as described above (freeze drying → reconstitution → dilution → intravenous administration) has problems in that it is costly, complicated, and causes patient’s discomfort due to frequent administration, rejection, and side effects, and in that a person who administers the formulation is limited to a medically trained person.
Adalimumab is also a kind of human monoclonal antibody that can function as a TNF-α inhibitor. A liquid formulation containing adalimumab is disclosed in, for example, the Humira label. Furthermore, Korean Patent Application Publication No. 100134689 discloses a liquid formulation containing adalimumab, sodium phosphate, sodium citrate, citric acid, mannitol, sodium chloride, and polysorbate 80 (Example 1), and an improved liquid formulation containing adalimumab, sodium phosphate, sodium citrate, citric acid, mannitol, arginine, sodium chloride, and polysorbate 80 (Example 2).
However, in the case of the above-described liquid pharmaceutical formulations containing NaCl or KCl as an isotonic agent, problems such as precipitation and gelatinization may arise, and when the antibody concentration is as low as about 50 mg/ml, the administration frequency and the administration cycle may be limited.
Accordingly, there is a need for a stable liquid pharmaceutical formulation that can overcome the problems of the above-described conventional liquid pharmaceutical formulations and that contains an antibody, particularly infliximab, as a TNF-α inhibitor.
DISCLOSURE Technical Problem It is an object of the present invention to provide a stable liquid pharmaceutical formulation having low viscosity while containing a high content of an antibody.
Another object of the present invention is to provide a liquid pharmaceutical formulation having excellent long-term storage stability based on excellent stability under accelerated conditions and severe conditions.
Still another object of the present invention is to provide a stable liquid pharmaceutical formulation that may be administered subcutaneously.
Technical Solution A stable liquid pharmaceutical formulation according to one embodiment of the present invention contains: (A) an antibody or its antigen-binding fragment; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer.
In one embodiment of the present invention, the antibody (A) may comprise an antibody that binds to TNF-α.
In one embodiment of the present invention, the antibody (A) may comprise infliximab, adalimumab, certolizumab pegol, golimumab, or a mixture thereof.
In one embodiment of the present invention, the antibody (A) may comprise a chimeric human-mouse IgG monoclonal antibody.
In one embodiment of the present invention, the antibody or its antigen- binding fragment (A) may comprise: a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6.
In one embodiment of the present invention, the antibody or its antigen- binding fragment (A) may comprise: a light-chain variable region having an amino acid sequence of SEQ ID NO: 7; and a heavy-chain variable region having an amino acid sequence of SEQ ID NO: 8.
In one embodiment of the present invention, the antibody (A) may comprise: a light chain having an amino acid sequence of SEQ ID NO: 9; and a heavy chain having an amino acid sequence of SEQ ID NO: 10.
In one embodiment of the present invention, the antibody or its antigen- binding fragment (A) may be contained at a concentration of 10 to 200 mg/ml.
In one embodiment of the present invention, the surfactant (B) may comprise polysorbate, poloxamer, or a mixture thereof.
[0021] In one embodiment of the present invention, the surfactant (B) may comprise polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, or a mixture of two or more thereof.
In one embodiment of the present invention, the surfactant (B) may comprise polysorbate 80.
[0023] In one embodiment of the present invention, the surfactant (B) may be contained at a concentration of 0.02 to 0.1% (w/v).
In one embodiment of the present invention, the sugar (C) may comprise a monosacchride, a disaccharide, an oligosaccharide, a polysaccharide, or a mixture of two or more thereof, and the sugar derivative (C) may comprise sugar alcohol, sugar acid, or a mixture thereof.
In one embodiment of the present invention, the sugar or its derivative (C) may comprise sorbitol, mannitol, trehalose, sucrose, or a mixture of two or more thereof.
In one embodiment of the present invention, the sugar or its derivative (C) may be contained at a concentration of 1 to 10% (w/v).
In one embodiment of the present invention, the buffer (D) may comprise acetate or histidine.
In one embodiment of the present invention, the buffer (D) may have a concentration of 1 to 50 mM.
[0029] In one embodiment of the present invention, the formulation may have a pH of 4.0 to 5.5.
In one embodiment of the present invention, the formulation may be free of aspartic acid, lysine, arginine, or mixtures thereof.
In one embodiment of the present invention, the formulation may be free of NaCl, KCl, NaF, KBr, NaBr, Na2SO4, NaSCN, K2SO4, or mixtures thereof.
In one embodiment of the present invention, the formulation may be free of a chelating agent.
In one embodiment of the present invention, the formulation may have a viscosity of 0.5 cp to 10 cp as measured after 1 month of storage at 40°C ± 2°C, or a viscosity of 0.5 cp to 5 cp as measured after 6 months of storage at 5°C ± 3 °C.
A stable liquid pharmaceutical formulation according to one embodiment of the present invention may contain: (A) an antibody or its antigen- binding fragment, which comprises a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer comprising acetate or histidine.
A stable liquid pharmaceutical formulation according to one embodiment of the present invention may contain: (A) 90 to 145 mg/ml of an antibody or its antigen-binding fragment, which comprises a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6; (B) 0.02 to 0.1% (w/v) of a surfactant; (C) 1 to 10% (w/v) of a sugar or its derivative; and (D) 1 to 50 mM of a buffer comprising acetate or histidine.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be for subcutaneous administration.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may not be subjected to a reconstitution step, a dilution step, or both, before use.
A pre-filled syringe according to one embodiment of the present invention is filled with the stable liquid pharmaceutical formulation.
An auto-injector according to one embodiment of the present invention includes the pre-filled syringe therein.
Advantages Effects The stable liquid pharmaceutical formulation according to the present invention has low viscosity while containing a high content of an antibody, has excellent long-term storage stability based on excellent stability under accelerated conditions and severe conditions, and may be administered subcutaneously.
DESCRIPTION OF specific embodiments Stable Liquid Pharmaceutical Formulation
[0042] A stable liquid pharmaceutical formulation according to the present invention contains: (A) an antibody or its antigen-binding fragment; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer.
As used herein, the term “free of” means that the formulation is completely free of the corresponding component. In addition, the term means that the formulation is substantially free of the corresponding component, that is, contains the corresponding component in an amount that does not affect the activity of the antibody and the stability and viscosity of the liquid pharmaceutical formulation. For example, the term means that the formulation contains the corresponding component in an amount of 0 to 1% (w/v), 0 to 1 ppm (w/v), or 0 to 1 ppb (w/v), based on the total weight of the liquid pharmaceutical formulation.
(A) Antibody or Its Antigen-Binding Fragment The term “antibody” refers to immunoglobulin molecules comprised of four polypeptide chains, two heavy chains and two light chains inter-connected by disulfide bonds. Other naturally occurring antibodies having an altered structure, for example, camelid antibodies, are also included in this definition. Each heavy chain is comprised of a heavy-chain variable region and a heavy-chain constant region. The heavy-chain constant region is comprised of three domains (CH1, CH2 and CH3).
Each light chain is comprised of a light-chain variable region and a light-chain constant region. The light-chain constant region is comprised of one domain (CL).
The heavy-chain variable region and the light-chain variable region can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each of the heavy-chain variable region and the light-chain variable region is composed of three CDRs and four FRs, which are arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
In one embodiment of the present invention, the pharmaceutical formulation may contain, as the antibody, a polyclonal antibody, a monoclonal antibody, a recombinant antibody, a single-chain antibody, a hybrid antibody, a chimeric antibody, a humanized antibody, or a fragment thereof. The term “chimeric antibody” refers to an antibody comprising heavy-chain and light-chain variable region sequences from one species and constant region sequences from another species. In one embodiment of the present invention, the pharmaceutical formulation may contain, as the antibody, a chimeric human-mouse IgG monoclonal antibody. The chimeric human-mouse IgG monoclonal antibody is comprised of mouse heavy-chain and light- chain variable regions and human heavy-chain and light-chain constant regions bound thereto. The chimeric human-mouse IgG monoclonal antibody may be produced according to a method known in the art. For example, infliximab may be produced according to a method described in US Patent No. 6,284,471.
[0047] In one embodiment of the present invention, the pharmaceutical formulation may contain, as the antibody, an antibody that binds to TNF-α or the epitope of TNF-α. The antibody that binds to TNF-α or the epitope of TNF-α may comprise infliximab, adalimumab, certolizumab pegol, golimumab, or a mixture thereof. In one embodiment of the present invention, the antibody may comprise infliximab.
In one embodiment of the present invention, the antibody or its antigen- binding fragment (A) may comprise: a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6.
In one embodiment of the present invention, the antibody or its antigen binding fragment (A) may comprise: a light-chain variable region having an amino acid sequence of SEQ ID NO: 7; and a heavy-chain variable region having an amino acid sequence of SEQ ID NO: 8.
In one embodiment of the present invention, the antibody or its antigen binding fragment (A) may comprise: a light chain having an amino acid sequence of SEQ ID NO: 9; and a heavy chain having an amino acid sequence of SEQ ID NO: 10.
The concentration of the antibody or its antigen-binding fragment may be freely controlled within a range that does not substantially adversely affect the stability and viscosity of the stable liquid pharmaceutical formulation according to the present invention. In one embodiment of the present invention, the concentration of the antibody or its antigen-binding fragment may be 10 to 200 mg/ml. In another embodiment of the present invention, the concentration of the antibody or its antigen- binding fragment may be 50 to 200 mg/ml. In still another embodiment of the present invention, the concentration of the antibody or its antigen-binding fragment may be 80 to 150 mg/ml. In still another embodiment of the present invention, the concentration of the antibody or its antigen-binding fragment may be 90 to 145 mg/ml. In yet another embodiment of the present invention, the concentration of the antibody or its antigen-binding fragment may be 110 to 130 mg/ml. If the concentration of the antibody or its antigen-binding fragment is within the above-described range, the high content of the antibody or its antigen-binding fragment makes it possible to increase the degree of freedom of dose and administration cycle, and the pharmaceutical formulation may exhibit excellent long-term stability and low viscosity.
(B) Surfactant Examples of the surfactant include, but are not limited to, polyoxyethylene sorbitan fatty acid ester (e.g., polysorbate), polyoxyethylene alkyl ether (e.g., Brij), alkylphenyl polyoxyethylene ether (e.g., Triton-X), polyoxyethylene- polyoxypropylene copolymers (e.g., Poloxamer, Pluronic), sodium dodecyl sulfate (SDS), and the like.
In one embodiment of the present invention, the surfactant may comprise polyoxyethylene sorbitan fatty acid ester (polysorbate). The polysorbate may comprise polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, or a mixture of two or more thereof. In one embodiment of the present invention, the polysorbate may comprise polysorbate 20, polysorbate 80, or a mixture thereof. In another embodiment of the present invention, the polysorbate may comprise polysorbate 80.
[0055] In one embodiment of the present invention, the concentration of the surfactant may be freely controlled within a range that does not substantially adversely affect the stability and viscosity of the stable liquid pharmaceutical formulation according to the present invention. For example, the concentration of the surfactant may be 0.001 to 5% (w/v), 0.01 to 1% (w/v), or 0.02 to 0.1% (w/v). If the concentration of the surfactant is within the above-described range, the pharmaceutical composition may exhibit excellent long-term stability and low viscosity.
(C) Sugar or Its Derivative The sugar may comprise a monosacchride, a disaccharide, an oligosaccharide, a polysaccharide, or a mixture of two or more thereof. Examples of the monosacchride include, but are not limited to, glucose, fructose, galactose, and the like. Examples of the disaccharide include, but are not limited to, sucrose, lactose, maltose, trehalose, and the like. Examples of the oligosaccharide include, but are not limited to, fructooligosaccaharides, galactooligosaccaharides, mannanoligosaccaharides, and the like. Examples of the polysaccharide include, but are not limited to, starch, glycogen, cellulose, chitin, pectin, and the like.
The sugar derivative may comprise sugar alcohol, sugar acid, or a mixture thereof. Examples of the sugar alcohol include, but are not limited to, glycerol, erythritol, threitol, arabitol, xylitol, ribitol, mannitol, sorbitol, galactitol, fucitol, iditol, inositol, volemitol, isomalt, maltitol, lactitol, maltotriitol, maltotetraitol, polyglycitol, and the like. Examples of the sugar acid include, but are not limited to, aldonic acid (glyceric acid, etc.), ulosonic acid (neuraminic acid, etc.), uronic acid (glucuronic acid, etc.), aldaric acid (tartaric acid, etc.), and the like.
In one embodiment of the present invention, the sugar or its derivative (C) may comprise sorbitol, mannitol, trehalose, sucrose, or a mixture of two or more thereof.
In one embodiment of the present invention, the concentration of the sugar or its derivative may be freely controlled within a range that does not substantially adversely affect the stability and viscosity of the stable liquid pharmaceutical formulation according to the present invention. For example, the concentration of the sugar or its derivative may be 0.1 to 30% (w/v), 1 to 20% (w/v), or 1 to 10% (w/v). If the concentration of the sugar or its derivative may be within this range, the pharmaceutical composition may exhibit excellent long-term stability and low viscosity.
(D) Buffer
[0062] The buffer that is used in the present invention is a neutralizing substance that minimizes the change in pH caused by acid or alkali. Examples of the buffer include phosphate, acetate, succinate, gluconate, glutamate, citrate, histidine, and the like. In one embodiment of the present invention, the buffer may comprise acetate or histidine. If the buffer comprises both acetate and histidine, the stability of the pharmaceutical formulation may be reduced.
In one embodiment of the present invention, the buffer may comprise acetate. Examples of the acetate include, but are not limited to, sodium acetate, zinc acetate, aluminum acetate, ammonium acetate, potassium acetate, and the like. For pH adjustment, the buffer may further comprise an acid, for example, acetic acid.
When the buffer comprises acetate, it may be most preferable in terms of pH adjustment and stability.
In one embodiment of the present invention, the buffer may comprise histidine. When the buffer comprises histidine, it may comprise a histidine salt, for example, histidine chloride, histidine acetate, histidine phosphate, histidine sulfate, or the like. For pH adjustment, the buffer may comprise an acid, for example, hydrochloric acid, acetic acid, phosphoric acid, sulfuric acid, or the like.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be free of citrate, phosphate, or a mixture thereof.
In one embodiment of the present invention, the concentration of the buffer (or the anion of the buffer) may be freely controlled within a range that does not substantially adversely affect the stability and viscosity of the stable liquid pharmaceutical formulation according to the present invention. For example, the concentration of the buffer or its anion may be 1 to 50 mM, 5 to 30 mM, or 10 to 25 mM. If the concentration of the buffer or its anion is within this range, the pharmaceutical composition may exhibit excellent long-term stability and low viscosity.
(E) pH In one embodiment of the present invention, the pH of the stable liquid pharmaceutical composition may be 4.0 to 5.5, or 4.7 to 5.3. If the pH is within this range, the pharmaceutical composition may exhibit excellent long-term stability and low viscosity. The pH of the pharmaceutical formulation may be adjusted using the buffer. In other words, if the pharmaceutical formulation contains a certain content of the buffer, it may exhibit the pH in the above-described range without having to use a separate pH-adjusting agent. If citrate, phosphate or a mixture thereof is used as the buffer, it may be difficult to show the pH in the above-described range. If the pharmaceutical formulation further contains an acid (e.g., hydrochloric acid) or a base (e.g., sodium hydroxide) as a separate pH-adjusting agent, the stability of the antibody may be reduced.
(F) Other Components
[0070] In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be free of aspartic acid, lysine, arginine, or mixtures thereof. If the stable liquid pharmaceutical formulation contains these amino acids, it may become solid. In one embodiment of the present invention, the stable liquid pharmaceutical formulation may contain one or more amino acids, excluding the above-described three amino acids. In this case, the stable liquid pharmaceutical formulation may contain the one or more amino acid in an amount of 5% (w/v) or less, for example, 0.001 to 5% (w/v), 0.001 to 1% (w/v), 0.01 to 5% (w/v), 0.01 to 1% (w/v), 0.1 to 5% (w/v), or 0.1 to 1% (w/v).
In another embodiment of the present invention, the stable liquid pharmaceutical formulation may contain taurine. In this case, the taurine may be contained in an amount of 5% (w/v) or less, for example, 0.001 to 5% (w/v), 0.001 to 1% (w/v), 0.01 to 5% (w/v), 0.01 to 1% (w/v), 0.1 to 5% (w/v), or 0.1 to 1% (w/v).
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be free of NaCl, KCl, NaF, KBr, NaBr, Na2SO4, NaSCN, K2SO4 or the like as a metal salt. If the stable liquid pharmaceutical formulation contains these metal salts, precipitation in the formulation may occur, and the formulation may be gelatinized and may have poor stability.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be free of a chelating agent (e.g., EDTA). If the pharmaceutical formulation contains a chelating agent, the oxidation rate thereof may be increased.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation may be free of a preservative. Examples of the preservative include octadecyl dimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl alcohol, benzyl alcohol, alkyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, m-cresol, and the like. If the pharmaceutical formulation contains the preservative, the preservative may not help improve the stability of the pharmaceutical formulation.
In one embodiment of the present invention, the stable liquid pharmaceutical formulation of the present invention may further contain an additive known in the art, which does not substantially adversely affect the activity of the antibody and the stability and low viscosity of the formulation. For example, the pharmaceutical formulation may further contain an aqueous carrier, an antioxidant, or a mixture of two or more thereof. The aqueous carrier is a carrier that is pharmaceutically acceptable (safe and non-toxic when administered to humans) and is useful for preparation of liquid pharmaceutical formulations. Examples of the aqueous carrier include, but are not limited to, sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), sterile saline solution, Ringer’s solution, dextrose, and the like. Examples of the antioxidant include, but are not limited to, ascorbic acid and the like.
(G) “Stable” Liquid Pharmaceutical Formulation The term “stable” in the "stable" liquid pharmaceutical formulation of the present invention means that the antibody according to the present invention essentially retains its physical stability and/or chemical stability and/or biological activity during production and/or upon storage. Various analytical techniques for measuring protein stability are readily available in the art.
Physical stability may be assessed by methods known in the art, which include measurement of a sample's apparent attenuation of light (absorbance, or optical density). Such a measurement of light attenuation is related to the turbidity of a formulation. In addition, for physical stability, the contents of high-molecular-weight components, the contents of low-molecular-weight components, the amounts of intact proteins, the number of sub-visible particles, and the like, may be measured.
Chemical stability can be assessed by, for example, detecting and quantifying chemically altered forms of the antibody. Chemical stability includes charge alteration (for example, occurring as a result of deamidation or oxidation) which can be evaluated by, for example, ion-exchange chromatography. For chemical stability, charge variants (acidic or basic peaks) may be measured.
Biological activity may be assessed by methods known in the art. For example, antigen binding affinity may be measured by ELISA.
[0081] In one embodiment of the present invention, the liquid pharmaceutical formulation may be stable for a long period of time.
In one embodiment of the present invention, the term “stable” liquid pharmaceutical formulation means a liquid pharmaceutical formulation satisfying one or more of the following criteria.
[0083] Turbidity - a liquid pharmaceutical formulation having an absorbance A600 of 0 to 0.0300, or 0 to 0.0700, as measured by a spectrophotometer after 4 weeks of storage at a temperature of 40°C ± 2°C; - a liquid pharmaceutical formulation having an absorbance A600 of 0 to 0.0300, or 0 to 0.0700, as measured by a spectrophotometer after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition; Content of Main Component (main peak) - a liquid pharmaceutical formulation in which the content of a main component content after 4 weeks of storage at a temperature of 40°C ± 2°C is 98% to 100% as measured by SE-HPLC; - a liquid pharmaceutical formulation in which the content of a main component content after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 98% to 100% as measured by SE-HPLC; Content of High-Molecular-Weight Components (a peak whose retention time is earlier than that of the main peak (intact IgG)) - a liquid pharmaceutical formulation in which the content of high- molecular-weight components after 12 months of storage at a temperature of 5°C ± 3°C is 0 to 1.00% as measured by SE-HPLC; - a liquid pharmaceutical formulation in which the content of high- molecular-weight components after 12 months of storage at a temperature of 5°C ± 3°C under a closed condition is 0 to 1.00% as measured by SE-HPLC; Content of Low-Molecular-Weight Components (a peak whose retention time is later than that of the main peak (intact IgG) - a liquid pharmaceutical formulation in which the content of low- molecular-weight components after 12 months of storage at a temperature of 5°C ± 3°C is 0 to 0.40% as measured by SE-HPLC; - a liquid pharmaceutical formulation in which the content of low- molecular-weight components after 12 months of storage at a temperature of 5°C ± 3°C under a closed condition is 0 to 0.40% as measured by SE-HPLC; Content of Intact Immunoglobulin G - a liquid pharmaceutical formulation in which the content of intact immunoglobulin G (intact IgG %) after 12 months of storage at a temperature of 5°C ± 3°C is 94.0% to 100% as measured by non-reduced CE-SDS; - a liquid pharmaceutical formulation in which the content of intact immunoglobulin G (intact IgG %) after 12 months of storage at a temperature of 5°C ± 3°C under a closed condition is 94.0% to 100% as measured by non-reduced CE-SDS; - a liquid pharmaceutical formulation in which the content of intact immunoglobulin G (intact IgG %) after 4 weeks of storage at a temperature of 40°C ± 2°C is 94.0% to 100% as measured by non-reduced CE-SDS; - a liquid pharmaceutical formulation in which the content of intact immunoglobulin G content (intact IgG %) after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 94.0% to 100% as measured by non-reduced CE-SDS; Content of Intact Heavy Chain and Light Chain - a liquid pharmaceutical formulation in which the content of intact heavy chain and light chain (intact HC+LC %) after 12 months of storage at a temperature of 5°C ± 3°C is 99.0% to 100% as measured by reduced CE-SDS;
[00102] - a liquid pharmaceutical formulation in which the content of intact heavy chain and light chain (intact HC+LC %) after 12 months of storage at a temperature of 5°C ± 3°C under a closed condition is 99.0% to 100% as measured by reduced CE-SDS; - a liquid pharmaceutical formulation in which the content of intact heavy chain and light chain (intact HC+LC %) after 4 weeks of storage at a temperature of 40°C ± 2°C is 98.0% to 100% as measured by reduced CE-SDS; - a liquid pharmaceutical formulation in which the content of intact heavy chain and light chain content (intact HC+LC %) after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 98.0% to 100% as measured by reduced CE-SDS; Number of sub-visible particles - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <400.00 μm) after 12 months of storage at a temperature of 5°C ± 3°C is 0 to 1,000 as measured by HIAC;
[00107] - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <400.00 μm) after 12 months of storage at a temperature of 5°C ± 3°C under a closed condition is 0 to 1,000 as measured by HIAC; - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥1.00 μm, <100.00 μm) after 4 weeks of storage at a temperature of 40°C ± 2°C is 0 to 30,000 as measured by MFI; - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥1.00 μm, <100.00 μm) after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 0 to 30,000 as measured by MFI;
[00110] - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <100.00 μm) after 4 weeks of storage at a temperature of 40°C ± 2°C is 0 to 200 as measured by MFI; - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <100.00 μm) after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 0 to 200 as measured by MFI; - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <100.00 μm) after 6 weeks of storage at a temperature of 40°C ± 2°C is 0 to 500 as measured by MFI;
[00113] - a liquid pharmaceutical formulation in which the number of sub- visible particles (≥10.00 μm, <100.00 μm) after 6 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 0 to 500 as measured by MFI; Oxidation Rate
[00115] - a liquid pharmaceutical formulation in which the oxidation rate of heavy-chain Met 255 after 4 weeks of storage at a temperature of 40°C ± 2°C is 0% to 2.5% as measured by LC-MS; - a liquid pharmaceutical formulation in which the oxidation rate of heavy-chain Met 255 after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition is 0% to 2.5% as measured by LC-MS; Charge Variants - a liquid pharmaceutical formulation showing an acidic peak of 20% to % as measured by IEC-HPLC after 4 weeks of storage at a temperature of 40°C ± 2°C; - a liquid pharmaceutical formulation showing an acidic peak of 20% to % as measured by IEC-HPLC after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition; - a liquid pharmaceutical formulation showing a basic peak of 33% to 40% as measured by IEC-HPLC after 4 weeks of storage at a temperature of 40°C ± 2°C; - a liquid pharmaceutical formulation showing a basic peak of 33% to 40% as measured by IEC-HPLC after 4 weeks of storage at a temperature of 40°C ± 2°C and a relative humidity of 75 ± 5% under a closed condition;
[00122] TNF-α Binding Affinity - a liquid pharmaceutical formulation having a TNF-α binding affinity of 80% to 120% as measured by ELISA after 12 months of storage at a temperature of °C ± 3°C; and - a liquid pharmaceutical formulation having a TNF-α binding affinity of 80% to 120% as measured by ELISA after 12 months of storage at a temperature of °C ± 3°C under a closed condition.
In one embodiment of the present invention, the pharmaceutical formulation may have a viscosity of 0.5 cp to 10.0 cp as measured after 1 month of storage at a temperature of 40°C ± 2°C. In another embodiment of the present invention, the pharmaceutical formulation may have a viscosity of 0.5 cp to 5.0 cp as measured after 6 months of storage at a temperature of 5°C ± 3°C.
Method for Preparation of Stable Liquid Pharmaceutical Formulation The stable liquid pharmaceutical formulation of the present invention may be prepared using any known method which is not limited to a particular method.
For example, the stable liquid pharmaceutical formulation may be prepared by adding a buffer to a solution containing a surfactant and a sugar or its derivative while adjusting the pH of the solution, and then adding an antibody to the mixed solution.
Alternatively, the liquid pharmaceutical formulation may be prepared by preparing a solution containing some excipients in the final step of a purification process, and then adding the remaining component to the solution. For example, the liquid pharmaceutical formulation may be prepared by preparing a solution containing an antibody, a buffer and a sugar or its derivative, and then adding a surfactant to the solution.
In addition, the method for preparation of the formulation may comprise or not comprise a freeze-drying step.
When the preparation method does not comprise the freeze-drying step, for example, the liquid pharmaceutical formulation prepared according to the present invention may be treated by sterilization, and then immediately placed in a closed container.
[00130] When the preparation method comprises the freeze-drying step, for example, the liquid pharmaceutical formulation prepared according to the present invention may be freeze-dried or freeze-dried and stored, and then components removed or modified by freeze drying and/or storage may be supplemented or replaced, thereby preparing the liquid pharmaceutical formulation according to the present invention. Alternatively, only components of the liquid pharmaceutical formulation of the present invention, excluding components that may be removed or modified by freeze drying and/or storage, may be freeze-dried or freeze-dried and stored, and then the excluded components may be added thereto, thereby preparing the liquid pharmaceutical formulation according to the present invention.
[00131] Method of Use of Stable Liquid Pharmaceutical Formulation The stable liquid pharmaceutical formulation according to the present invention may be used for treating diseases in which the activity of TNF-α acts as a harmful factor. Examples of diseases in which the activity of TNF-α acts as a harmful factor include, but are not limited to, septicemia, autoimmune diseases, infectious diseases, graft rejection, malignant cancer, lung disorders, bowel disorders, heart disorders, and the like.
In one embodiment of the present invention, the diseases in which the activity of TNF-α acts as a harmful factor may be selected from among rheumatoid arthritis, ankylosing spondylitis, ulcerative colitis, adult Crohn's disease, pediatric Crohn's disease, psoriasis, and psoriatic arthritis.
The stable liquid pharmaceutical formulation according to the present invention may be provided as a single-dosage form, a multiple-dosage form, or a form for subcutaneous self-injection.
The concentrations of other components, including the antibody, in the liquid pharmaceutical formulation, are as described above, and the total volume of the liquid pharmaceutical formulation may be 0.2 to 2.0 mL.
The dose and timing of administration of the liquid pharmaceutical formulation may vary depending on the kind of disease, the severity and course of the disease, the patient’s health and response to treatment, and the judgment of the treating physician, and is not limited to a particular dose and timing of administration. For example, one or several products containing the liquid pharmaceutical formulation may be administered at a dose of 1 to 10 mg/kg based on the antibody concentration, and then the same or different doses may be administered at intervals of one week, two weeks, three weeks, one month, two months or three months.
[00137] In one embodiment of the present invention, the stable liquid pharmaceutical formulation may not be subjected to a reconstitution step, a dilution step, or both, before use.
Treatment Method and Stabilization Method The present invention also provide a method for treating a patient having a disease in which TNF-α activity acts as a harmful factor, the method comprising administering to the patient a stable liquid pharmaceutical formulation containing: (A) an antibody or its antigen binding fragment; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer.
The present invention also provides a method of stabilizing an antibody in a liquid pharmaceutical formulation, the method comprising preparing a stable liquid pharmaceutical containing: (A) an antibody or its antigen binding fragment; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer.
In one embodiment of the treating method or the stabilizing method, the antibody (A) may comprise an antibody that binds to TNF-α.
[00142] In one embodiment of the treating method or the stabilizing method, the antibody (A) may comprise infliximab, adalimumab, certolizumab pegol, golimumab, or a mixture thereof.
In one embodiment of the treating method or the stabilizing method, the antibody (A) may comprise a chimeric human-mouse IgG monoclonal antibody.
[00144] In one embodiment of the treating method or the stabilizing method, the antibody (A) or its the antigen binding fragment thereof may comprise: a light- chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6.
In one embodiment of the treating method or the stabilizing method, the antibody or its antigen binding fragment (A) may comprise: a light-chain variable region having an amino acid sequence of SEQ ID NO: 7; and a heavy-chain variable region having an amino acid sequence of SEQ ID NO: 8.
In one embodiment of the treating method or the stabilizing method, the antibody (A) may comprise: a light chain having an amino acid sequence of SEQ ID NO: 9; and a heavy chain having an amino acid sequence of SEQ ID NO: 10.
In one embodiment of the treating method or the stabilizing method, the antibody or its antigen binding fragment (A) may be contained at a concentration of to 200 mg/ml.
In one embodiment of the treating method or the stabilizing method, the surfactant (B) may comprise polysorbate, poloxamer, or a mixture thereof.
In one embodiment of the treating method or the stabilizing method, the surfactant (B) may comprise polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, or a mixture of two or more thereof.
In one embodiment of the treating method or the stabilizing method, the surfactant (B) may comprise polysorbate 80.
In one embodiment of the treating method or the stabilizing method, the surfactant (B) may be contained at a concentration of 0.02 to 0.1% (w/v).
In one embodiment of the treating method or the stabilizing method, the sugar (C) may comprise a monosacchride, a disaccharide, an oligosaccharide, a polysaccharide, or a mixture of two or more thereof, and the sugar derivative (C) may comprise sugar alcohol, sugar acid, or a mixture thereof.
In one embodiment of the treating method or the stabilizing method, the sugar or its derivative (C) may comprise sorbitol, mannitol, trehalose, sucrose, or a mixture of two or more thereof.
[00154] In one embodiment of the treating method or the stabilizing method, the sugar or its derivative (C) may be contained at a concentration of 1 to 10% (w/v).
In one embodiment of the treating method or the stabilizing method, the buffer (D) may comprise acetate or histidine.
In one embodiment of the treating method or the stabilizing method, the buffer (D) may have a concentration of 1 to 50 mM.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may have a pH of 4.0 to 5.5.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may be free of aspartic acid, lysine, arginine, or mixtures thereof.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may be free of NaCl, KCl, NaF, KBr, NaBr, Na2SO4, NaSCN, K2SO4, or mixtures thereof.
[00161] In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may be free of a chelating agent.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may be free of a preservative.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may further contain an aqueous carrier, an antioxidant, or a mixture of two or more thereof.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may have a viscosity of 0.5 cp to 10 cp as measured after 1 month of storage at a temperature of 40°C ± 2°C, or a viscosity of 0.5 cp to 5 cp as measured after 6 months of storage at a temperature of 5°C ± 3 °C.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may contain: (A) an antibody or its antigen binding fragment, which comprises a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6; (B) a surfactant; (C) a sugar or its derivative; and (D) a buffer comprising acetate or histidine.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may contain: (A) 90 to 145 mg/ml of an antibody or its antigen binding fragment, which comprises a light-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 1, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 2, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 3; and a heavy-chain variable region comprising a CDR1 domain comprising an amino acid sequence of SEQ ID NO: 4, a CDR2 domain comprising an amino acid sequence of SEQ ID NO: 5, and a CDR3 domain comprising an amino acid sequence of SEQ ID NO: 6; (B) 0.02 to 0.1% (w/v) of a surfactant; (C) 1 to 10% (w/v) of a sugar or its derivative; and (D) 1 to 50 mM of a buffer comprising acetate or histidine.
In one embodiment of the treating method, the stable liquid pharmaceutical formulation may be administered subcutaneously.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may not be subjected to a reconstitution step, a dilution step, or both, before use.
In one embodiment of the treating method or the stabilizing method, the stable liquid pharmaceutical formulation may be filled in a pre-filled syringe before use.
[00170] In one embodiment of the treating method or the stabilizing method, the pre-filled syringe may be included in an auto-injector before use.
Product The present invention also provides a product comprising: the stable liquid pharmaceutical formulation; and a container receiving the stable liquid pharmaceutical formulation in a closed state.
The stable liquid pharmaceutical formulation is as described above.
In one embodiment of the present invention, the container may be formed of a material such as glass, a polymer (plastic), a metal or the like, but is not limited thereto. In one embodiment of the present invention, the container is a bottle, a vial, a cartridge, a syringe (pre-filled syringe, auto-syringe), or a tube, but is not limited thereto. In one embodiment of the present invention, the container may be a glass or polymer vial, or a glass or polymer pre-filled syringe.
Specific product forms of the above-described vial, cartridge, pre-filled syringe or auto-syringe, and methods of filling the stable liquid pharmaceutical formulation into the vial, cartridge, pre-filled syringe or auto-syringe, may be readily available or implemented by any person skilled in the technical field to which the present invention pertains. For example, US Patent Nos. 4,861,335 and 6,331,174, etc., disclose the specific product form of a pre-filled syringe and a filling method.
For example, US Patent Nos. 5,085,642 and 5,681,291, etc., disclose the specific product form of an auto-syringe and an assembly method. The above-described vial, cartridge, pre-filled syringe or auto-syringe that is used in the present invention may be a commercially available product, or a product separately manufactured considering the physical properties of the stable liquid pharmaceutical formulation, an area to which the formulation is to be administered, the dose of the formulation, and the like.
[00176] In one embodiment of the present invention, the inside of the container may not be coated with silicone oil. If it is coated with silicone oil, the stability of the formulation may be reduced. The container may be a single-dose or multiple-dose container.
In one embodiment of the present invention, the product may further comprise instructions providing a method of using the stable liquid pharmaceutical formulation, a method of storing the formulation, or both. The method of using the formulation includes a method for treating a disease in which TNF-α activity acts as a harmful factor, and may include the route of administration, the dose of the formulation, and the timing of administration.
[00178] In one embodiment of the present invention, the product may comprise other required utensils (e.g., a needle, a syringe, etc.) in a commercial viewpoint and a user viewpoint.
Hereinafter, the present invention will be described with reference to examples. It is to be understood, however, that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention.
Examples The antibody used in the following experimental examples was infliximab purified from commercially available Remsima (manufactured by Celltrion).
[00182] The physical stability, chemical stability and biological activity of liquid pharmaceutical formulations used in the following experimental examples were measured using the following methods.
- Turbidity The absorbance at 600 nm was measured using a UV-Vis spectrophotometer.
- Content of main component The main component content (main peak %) was measured using size exclusion high-performance liquid chromatography (HPLC).
- Content of high-molecular-weight components
[00188] The content of high-molecular-weight components (pre-peak %) was measured using size exclusion high-performance liquid chromatography (HPLC).
- Content of low-molecular-weight components The content of low-molecular-weight components (post-peak %) was measured using size exclusion high-performance liquid chromatography (HPLC).
[00191] - Content of intact immunoglobulin G (intact IgG %) The content of intact immunoglobulin G (%) was measured using Non- Reduced Capillary Electrophoresis-Sodium Dodecyl Sulfate (NR CE-SDS).
- Content of intact heavy chain and light chain (intact HC+LC %) The content of intact heavy chain and light chain (%) was measured using Reduced Capillary Electrophoresis-Sodium Dodecyl Sulfate (R CE-SDS).
- Number of sub-visible particles Experimental Examples 1 to 4: the number of sub-visible particles was measured using Micro Flow Imaging (MFI).
Experimental Example 5: the number of sub-visible particles was measured using a light-shielding liquid particle counter (model: HIAC 9703).
- Oxidation The oxidation (%) of heavy chain Met 255 was measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS).
- Change variants
[00201] Acidic and basic peaks (%) were measured by Ion Exchange Chromatography-High Performance Liquid Chromatography (IEC-HPLC).
- TNF-α binding affinity TNF-α binding affinity (%) was measured by Enzyme-Linked ImmunoSorbent Assay (ELISA).
[00204] - Viscosity 3 5 -1 Using a micro-capillary flow system (apparent shear rate: 10 to 10 s ) equipped with a flow cell (B05 sensor type; 50 μm cell depth), viscosity was measured in a 500 μL syringe at 25°C ± 0.1°C.
Experimental Example 1: Comparison of Sugar Alcohol with NaCl; Comparison of Acetate/Histidine Buffer with Citrate/Phosphate Buffer; Comparison of pH 4-5.5 with pH 6-7 For preparation of liquid pharmaceutical formulations to be used in Experimental Example 1, each buffer was prepared so as to have a desired pH, and sorbitol or NaCl was added thereto. Then, an antibody was added thereto and a surfactant was added, thereby preparing the samples shown in Table 1 below. The specific content of each component is shown in Table 1 below. The concentration of the buffer means the molecular/anion concentration of the corresponding compound.
The total volume was 1 ml.
Table 1 Antibody Surfactant Sugar alcohol or NaCl Buffer pH content (mg/ml) Example 1 100 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 4.0 0.05% (w/v) 10 mM Example 2 100 Polysorbate 80 Sorbitol 5 %(w/v) Histidine 5.5 0.05% (w/v) 10 mM Example 3 100 Polysorbate 20 Sorbitol 5 %(w/v) Histidine 5.5 0.05 %(w/v) 10 mM Comparative 100 Polysorbate 80 NaCl 140 mM Sodium acetate 4.0 Example 1 0.05 %(w/v) 10 mM Comparative 100 Polysorbate 80 NaCl 140 mM Sodium citrate 10 5.0 Example 2 0.05 %(w/v) mM Comparative 100 Polysorbate 80 Sorbitol 5 %(w/v) Sodium citrate 10 5.0 Example 3 0.05 %(w/v) mM Comparative 100 Polysorbate 80 NaCl 140 mM Histidine 5.5 Example 4 0.05 %(w/v) 10 mM Comparative 100 Polysorbate 80 NaCl 140 mM Sodium 6.0 Example 5 0.05 %(w/v) phosphate mM Comparative 100 Polysorbate 80 Sorbitol 5 %(w/v) Sodium 6.0 Example 6 0.05 %(w/v) phosphate mM Comparative 100 Polysorbate 80 NaCl 140 mM Sodium 7.0 Example 7 0.05 %(w/v) phosphate mM Comparative 100 Polysorbate 80 Sorbitol 5 %(w/v) Sodium 7.0 Example 8 0.05 %(w/v) phosphate mM Liquid pharmaceutical formulations prepared according to Examples 1 to 3 and Comparative Examples 1 to 8 were stored for 2 weeks at a temperature of 40 ± 2°C and a relative humidity of 75 ± 5%. As a result, the formulations containing NaCl (Comparative Examples 1, 2, 4, 5 and 7) all showed precipitation and a form like gelatin. In addition, Comparative Example 3, containing sorbitol, but containing sodium citrate, and Comparative Example 8 containing sorbitol, but containing sodium phosphate, also showed a form like gelatin.
Among the formulations containing sorbitol, only the formulations of Examples 1, 2 and 3 and Comparative Example 6 did not show a gelatin form. The formulations were measured for their stability after 0, 2 and 4 weeks of storage at a temperature of 5 ± 3°C and their stability after 2 and 4 weeks of storage at a temperature of 40 ± 2°C and a relative humidity of 75 ± 5%. The results of the measurement are shown in Tables 2 to 9 below.
Turbidity Table 2 After 0 week After 2 weeks at After 4 weeks at After 2 weeks at After 4 weeks at at 5±3°C 5±3°C 5±3°C 40±2°C 40±2°C Example 1 0.0082 0.0060 0.0087 0.0364 0.0263 Example 2 0.0099 0.1550 0.0082 0.0291 0.0562 Example 3 0.0112 0.0059 0.0082 0.0358 0.0643 Comparative 0.0120 0.0228 0.0138 0.1127 0.3113 Example 6 As can be seen in Table 2 above, the formulation of Example 1, having a pH of 4 and containing acetate as the buffer, was the best in terms of turbidity, and particularly, showed an absorbance of 0.0300 or lower after 4 weeks of storage at 40°C. Furthermore, it can be seen that the formulations of Example 2 and 3, having a pH of 5.5 and containing histidine as the buffer, also showed an absorbance of 0.0700 or lower after 4 weeks of storage at 40°C.
However, it can be seen that the formulation of Comparative Example 6, having a pH of 6 and containing phosphate as the buffer, showed significantly increased turbidity after 2 and 4 weeks of storage at 40°C.
Content of high-molecular-weight components Table 3 After 0 week at After 2 weeks at After 4 weeks at After 2 weeks at After 4 weeks at ±3°C 5±3°C 5±3°C 40±2°C 40±2°C Example 1 0.4 0.8 0.6 0.8 0.7 Example 2 0.6 1.1 0.9 1.6 1.4 Example 3 0.6 1.1 0.8 1.4 1.3 Comparati 0.8 1.5 1.2 2.4 2.3 ve Example As can be seen in Table 3 above, the formulation of Example 1 showed the lowest high-molecular-weight component content under all the conditions.
Particularly, the formulation of Example 1 showed a high-molecular-weight component content of 1.0% or less after 4 weeks of storage at a temperature of 40°C.
Furthermore, it can be seen that the formulations of Examples 2 and 3 showed a high- molecular-weight component content of 1.5% or less after 4 weeks of storage at a temperature of 40°C.
[00218] Content of intact immunoglobulin G (Intact IgG %) Table 4 After 0 week at After 2 weeks After 4 weeks After 2 weeks After 4 weeks ±3°C at 5±3°C at 5±3°C at 40±2°C at 40±2°C Example 1 97.7 98.8 98.0 96.9 94.5 Example 2 97.4 98.7 98.2 97.4 94.6 Example 3 97.2 98.9 97.8 97.4 94.4 Comparati 97.2 98.6 98.3 97.1 93.6 Example 6 As can be seen in Table 4 above, the contents of intact immunoglobulin in the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C were 94.0% or more, which was higher than that of Comparative Example 6.
Content of intact heavy chain and light chain (Intact HC+LC %)
[00222] Table 5 After 0 week After 2 weeks at After 4 weeks at After 2 weeks at After 4 weeks at at 5±3°C 5±3°C 5±3°C 40±2°C 40±2°C Example 1 99.5 99.6 99.5 99.2 98.3 Example 2 99.5 99.6 99.4 99.3 98.0 Example 3 99.6 99.6 99.4 99.3 98.3 Comparative 99.6 99.6 99.4 99.3 97.6 Example 6 As can be seen in Table 5 above, the contents of intact heavy chain and light in the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C were 98.0% or more, which was higher than that of Comparative Example 6.
Oxidation rate (heavy-chain Met 255) Table 6 After 0 week at 40±2°C After 4 weeks at 40±2°C Example 1 2.2 2.4 Example 2 2.0 2.5 Example 3 2.1 2.5 Comparative Example 6 2.2 4.1 As can be seen in Table 6 above, the oxidation rates of heavy-chain Met 255 in the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C were 2.5% or less, which was lower than that of Comparative Example 6.
Charge variants (acidic peaks) Table 7 After 0 week After 2 weeks at After 4 weeks at After 2 weeks at After 4 weeks at at 5±3°C 5±3°C 5±3°C 40±2°C 40±2°C Example 1 20.5 20.5 20.5 27.0 33.5 Example 2 20.6 20.8 20.6 27.9 34.5 Example 3 20.3 20.9 20.8 27.5 34.4 Comparative 20.4 20.9 20.9 30.3 38.6 Example 6 As can be seen in Table 7, the acidic peaks of the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C were 35% or less, which was lower than that of Comparative Example 6. It indicates that the formulations of Examples 1 to 3 are stable formulations in which deamidation that is a major cause of increasing acidic peaks less occurs.
Charge variants (basic peaks) Table 8 After 0 week After 2 weeks at After 4 weeks at After 2 weeks at After 4 weeks at at 5±3°C 5±3°C 5±3°C 40±2°C 40±2°C Example 1 40.6 40.1 40.2 37.4 34.4 Example 2 40.5 39.8 39.8 36.3 33.1 Example 3 40.4 39.6 39.8 36.5 33.3 Comparative 40.4 39.8 40.0 35.1 30.9 Example 6 As can be seen in Table 8 above, the basic peaks of the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C were 33% or more, which was higher than that of Comparative Example 6.
Number of sub-visible particles (≥1.00 μm, <100.00 μm) Table 9 After 0 week at After 4 weeks at After 4 weeks at ±3°C 5±3°C 40±2°C Example 1 1527 7645 7005 Example 2 4405 14257 29500 Example 3 4525 1493 26923 Comparative 13282 6688 2319386 Example 6 As can be seen in Table 9, the number of sub-visible particles (≥1.00 μm, <100.00 μm) in the formulations of Examples 1 to 3 after 4 weeks of storage at a temperature of 40°C was 30,000 or less, which was smaller than that of Comparative Example 6.
Experimental Example 2: Effect of Amino Acid For preparation of liquid pharmaceutical formulations to be used in Experimental Example 2, a buffer comprising sodium acetate was prepared so as to have a desired pH, and sorbitol was added thereto. Then, an antibody was added thereto and a surfactant and amino acid/taurine were added, thereby preparing the samples shown in Table 10 below. The concentration of each component is shown in Table 10 below. The concentration of the buffer means the concentration of acetate anion. The total volume was 1 ml.
Table 10 Antibody Surfactant Sugar Buffer pH Amino content alcohol or acid/taurine (mg/ml) NaCl Example 1 100 Polysorbate 80 Sorbitol 5 Sodium 4.0 - 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-alanine Example 1 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-asparagine Example 2 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-glutamine Example 3 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-glutamic acid Example 4 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-glycine Example 5 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-isoleucine Example 6 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-leucine Example 7 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-methionine Example 8 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-phenylalanine Example 9 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-proline Example 10 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-serine Example 11 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-threonine Example 12 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-tryptophan Example 13 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-tyrosine Example 14 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 Valine Example 15 0.05 %(w/v) %(w/v) acetate mM Reference 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 Taurine Example 16 0.05 %(w/v) %(w/v) acetate mM Comparative 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-aspartic acid Example 9 0.05 %(w/v) %(w/v) acetate mM Comparative 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-histidine Example 10 0.05 %(w/v) %(w/v) acetate mM Comparative 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-lysine Example 11 0.05 %(w/v) %(w/v) acetate mM Comparative 100 Polysorbate 80 Sorbitol 4 Sodium 4.0 L-arginine Example 12 0.05 %(w/v) %(w/v) acetate mM Amino acid or taurine was added in an amount of 5% (w/v) or less.
The formulations of Comparative Examples 9, 10, 11 and 12, containing aspartic acid, histidine, lysine and arginine, respectively, became solid after 24 hours of storage at 50 ± 2°C.
[00241] For the formulations containing other amino acids or taurine, the stabilities after 24 hours of storage at 5±3°C and 50±2°C were measured, but there was no significant difference between these formulations and between the these formulations and the formulation of Example 1.
Experimental Example 3: Protein Concentration; Surfactant Concentration; and the Kind of Sugar For preparation of liquid pharmaceutical formulations to be used in Experimental Example 3, a buffer comprising sodium acetate was prepared so as to have a desired pH, and sorbitol, mannitol, trehalose or sucrose was added thereto.
Then, an antibody was added thereto and a surfactant was added, thereby preparing the samples shown in Table 11 below. The content of each component is shown in Table 11 below. The concentration of the buffer means the concentration of acetate anion.
The total volume was 1 ml.
Table 11 Antibody Surfactant Sugar Buffer pH content (mg/ml) Example 4 125 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.05 %(w/v) 10 mM Example 5 110 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.05 %(w/v) 10 mM Example 6 90 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.05 %(w/v) 10 mM Example 7 145 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.05 %(w/v) 10 mM Example 8 110 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.02 %(w/v) 10 mM Example 9 110 Polysorbate 80 Sorbitol 5 %(w/v) Sodium acetate 5.0 0.1 %(w/v) 10 mM Example 10 110 Polysorbate 80 Mannitol 5 Sodium acetate 5.0 0.05 %(w/v) %(w/v) 10 mM Example 11 110 Polysorbate 80 Trehalose 10 Sodium acetate 5.0 0.05 %(w/v) %(w/v) 10 mM Example 12 110 Polysorbate 80 Sucrose 10 Sodium acetate 5.0 0.05 %(w/v) %(w/v) 10 mM The formulations were measured for their stabilities after 0, 2 and 4 weeks of storage at a temperature of 5 ± 3°C and for their stabilities after 2 and 4 weeks of storage at a temperature of 40 ± 2°C and a relative humidity of 75 ± 5%.
The results of the measurement are shown in Tables 12 to 17 below.
[00246] Protein Concentration Content of high-molecular-weight components Table 12 Antibody After 0 week After 2 After 4 After 2 After 4 content weeks at 5°C weeks at 5°C weeks at weeks at (mg/ml) 40°C 40°C Example 6 90 1.0 1.1 1.1 0.8 0.8 Example 5 110 1.1 1.1 1.2 1.0 1.0 Example 4 125 1.1 1.2 1.2 1.2 1.2 Example 7 145 1.2 1.2 1.3 1.3 1.3 As can be seen in Table 12 above, the high-molecular-weight component content increased as the antibody concentration increased. However, at an antibody concentration ranging from 90 to 145 mg/ml, the high-molecular-weight component contents after 4 weeks of storage at 5°C and 40°C were generally low.
[00250] Surfactant Concentration Number of sub-visible particles (≥1.00 μm, <100.00 μm) Table 13 Surfactant After 0 week After 2 weeks After 4 weeks at 40°C at 40°C Example 8 Polysorbate 80 590 9235 5581 0.02 %(w/v) Example 5 Polysorbate 80 6076 3957 6458 0.05 %(w/v) Example 9 Polysorbate 80 997 2678 1672 0.1 %(w/v) As can be seen in Table 13 above, at a surfactant concentration ranging from 0.02 to 0.1% (w/v), the number of sub-visible particles (≥1.00 μm, <100.00 μm) after 4 weeks of storage at 40°C was 10,000 or less.
The Kind of Sugar Content of a main component (main peak) Table 14 Sugar After 0 week After 2 weeks at After 4 weeks at 40°C 40°C Example 5 Sorbitol 98.9 98.5 98.1 %(w/v) Example 10 Mannitol 98.9 98.6 98.2 %(w/v) Example 11 Trehalose 98.9 98.6 98.2 %(w/v) Example 12 Sucrose 98.9 98.6 98.1 %(w/v) As can be seen in Table 14 above, the formulations containing sorbitol, mannitol, trehalose or sucrose as a sugar showed a main component content of 98% or more after 4 weeks of storage at 40°C.
[00258] Charge variants (acidic peaks) Table 15 Sugar After 0 week After 2 weeks at After 4 weeks at 40°C 40°C Example 5 Sorbitol 19.6 27.2 33.9 %(w/v) Example 10 Mannitol 19.7 27.2 33.7 %(w/v) Example 11 Trehalose 19.6 27.3 34.0 %(w/v) Example 12 Sucrose 19.7 27.3 33.8 %(w/v) As can be seen in Table 15 above, the formulations containing sorbitol, mannitol, trehalose or sucrose as a sugar showed an acidic peak of 35% or less after 4 weeks of storage at 40°C.
Number of sub-visible particles (≥1.00 μm, <100.00 μm)
[00262] Table 16 Sugar After 0 week After 2 weeks at After 4 weeks at 40°C 40°C Example 5 Sorbitol 6076 3957 6458 %(w/v) Example 10 Mannitol 1055 865 4595 %(w/v) Example 11 Trehalose 2803 1572 3554 %(w/v) Example 12 Sucrose 1246 2416 11230 %(w/v) Number of sub-visible particles (≥10.00 μm, <100.00 μm) Table 17 Sugar After 0 week After 2 weeks at After 4 weeks at 40°C 40°C Example 5 Sorbitol 128 11 115 %(w/v) Example 10 Mannitol 36 37 84 %(w/v) Example 11 Trehalose 42 13 56 %(w/v) Example 12 Sucrose 40 42 118 %(w/v) As can be seen in Tables 16 and 17 above, in the formulations containing sorbitol, mannitol, trehalose or sucrose as a sugar, the number of sub-visible particles (≥1.00 μm, <100.00 μm) after 4 weeks of storage at 40°C was 15,000 or less, and the number of sub-visible particles (≥10.00 μm, <100.00 μm) after 4 weeks of storage at 40°C was 200 or less.
Experimental Example 4: The Kind of Surfactant and the Effect of Chelating Agent For preparation of liquid pharmaceutical formulations to be used in Experimental Example 4, a buffer comprising sodium acetate was prepared so as to have a desired pH, and sorbitol was added thereto. Then, an antibody was added thereto and a surfactant or a mixture of a surfactant and a chelating agent was added, thereby preparing the samples shown in Table 18 below. The content of each component is shown in Table 18 below. The concentration of the buffer means the concentration of acetate anion. The total volume was 1 ml.
[00268] Table 18 Antibod Surfactant Sugar Buffer pH Chelating y content agent (mg/ml) (EDTA) Example 13 120 Polysorbate 80 Sorbitol Sodium 5.0 - 0.05 %(w/v) 5 %(w/v) acetate mM Example 14 120 Polysorbate 20 Sorbitol Sodium 5.0 - 0.05 %(w/v) 5 %(w/v) acetate mM Example 15 120 Poloxamer 188 Sorbitol Sodium 5.0 - 0.8 %(w/v) 5 %(w/v) acetate mM Comparative 120 Polysorbate 80 Sorbitol Sodium 5.0 0.05 mg/ml Example 13 0.05 %(w/v) 5 %(w/v) acetate mM Comparative 120 Polysorbate 20 Sorbitol Sodium 5.0 0.05 mg/ml Example 14 0.05 %(w/v) 5 %(w/v) acetate mM Comparative 120 Poloxamer 188 Sorbitol Sodium 5.0 0.05 mg/ml Example 15 0.8 %(w/v) 5 %(w/v) acetate mM The formulations shown in Table 18 above were measured for their stabilities after 0, 3 and 6 weeks of storage at a temperature of 5 ± 3°C, a temperature of 25 ± 2°C and a relative temperature of 60 ± 5%, and a temperature of 40 ± 2°C and a relative humidity of 75 ± 5% under a closed condition. The results of the measurement are shown in Tables 19 and 20 below.
The Kind of Surfactant Number of Sub-visible particles (≥10.00 μm, <100.00 μm) Table 19 Surfactant After 0 After After After After After After week at 3 6 3 6 3 6 °C weeks weeks weeks weeks weeks weeks at 5°C at 5°C at at at at °C 25°C 40°C 40°C Example 13 Polysorbate 50 149 46 34 182 249 55 0.05 %(w/v) Example 14 Polysorbate 20 581 309 103 54 90 185 279 0.05 %(w/v) Example 15 Poloxamer 208 67 86 172 56 344 2050 0.8 %(w/v) As can be seen in Table 19 above, in the formulation of Example 13, containing polysorbate 80 as a surfactant, the number of sub-visible particles (≥10.00 μm, <100.00 μm) after 6 weeks of storage at 40°C was 100 or less (the smallest), and in the formulation of Example 15, containing poloxamer 188 as a surfactant, the number of sub-visible particles (≥10.00 μm, <100.00 μm) after 6 weeks of storage at 40°C was 2,000 or more (the largest).
Effect of Chelating Agent (EDTA) Oxidation rate (heavy-chain Met 255) Table 20 Chelating After 0 After 3 After 6 After 3 After 6 agent week at weeks at weeks at weeks at weeks at (EDTA) 5°C 5°C 5°C 40°C 40°C Example 13 - 1.9 1.9 1.9 2.3 2.5 Example 14 - 2.0 1.9 1.9 2.2 2.4 Example 15 - 1.9 1.9 1.9 2.3 2.5 Comparative 0.05 mg/ml 1.9 1.8 1.8 2.9 3.3 Example 13 Comparative 0.05 mg/ml 2.3 1.8 2.0 2.8 3.3 Example 14 Comparative 0.05 mg/ml 1.8 1.9 1.9 2.8 3.4 Example 15 As can be seen in Table 20 above, in the formulations of Comparative Examples 13 to 15, containing a chelating agent (EDTA), the oxidation rate of heavy- chain Met 255 after 6 weeks of storage at 40°C increased compared to that in the formulations of Examples 13 to 15, containing no chelating agent (EDTA).
[00278] Experimental Example 5: Long-Term Stability For preparation of a liquid pharmaceutical formulation to be used in Experimental Example 5, a buffer comprising sodium acetate was prepared so as to have a pH of 5.0, and sorbitol was added thereto. Then, an antibody was added thereto and a surfactant was added, thereby preparing the sample shown in Table 21 below. The content of each component is shown in Table 21 below. The concentration of the buffer means the concentration of acetate anion. The total volume was 1 ml.
Table 21 Antibody Surfactant Sugar Buffer pH content (mg/ml) Example 120 Polysorbate 80 Sorbitol Sodium acetate 5.0 16 0.05 %(w/v) 5 %(w/v) 25 mM The formulation shown in Table 21 was measured for its stability after 0, 3 and 6 months of storage at a temperature of 5 ± 3°C under a closed condition.
The results of the measurement are shown in Tables 22 to 27 below.
Number of sub-visible particles (≥10.00 μm, <400.00 μm) Table 22 After 0 month After 3 After 6 After 9 After 12 at 5°C months at months at months at months at °C 5°C 5°C 5°C Example 16 35 26 48 32 43
[00284] As can be seen in Table 22 above, the number of sub-visible particles (≥10.00 μm, <400.00 μm) in the formulation of Example 16 after 12 months of storage at 5°C was as small as 100 or less.
Content of intact immunoglobulin (Intact IgG %) Table 23 After 0 month After 3 After 6 After 9 After 12 at 5°C months at months at months at months at °C 5°C 5°C 5°C Example 16 94.6 93.9 94.3 94.4 94.4
[00287] As can be seen in Table 23 above, the content of intact immunoglobulin G in the formulation of Example 16 after 12 months of storage at 5°C was as high as 94% or more.
Content of intact heavy chain and light chain (Intact HC+LC %) Table 24 After 0 After 3 After 6 After 9 After 12 month at 5°C months at months at months at months at 5°C °C 5°C 5°C Example 16 99.7 99.5 99.6 99.4 99.4 As can be seen in Table 24 above, the content of intact heavy chain and light chain in the formulation of Example 16 after 12 months of storage at 5°C was as high as 99% or more.
Content of high-molecular-weight components
[00292] Table 25 After 0 After 3 After 6 After 9 After 12 month at 5°C months at months at months at months at 5°C °C 5°C 5°C Example 16 0.5 0.9 0.9 0.8 0.7 As can be seen in Table 25 above, the content of high-molecular- weight components in the formulation of Example 16 after 12 months of storage at 5°C was as low as 1.0% or less.
Content of low-molecular-weight components
[00295] Table 26 After 0 After 3 After 6 After 9 After 12 month at 5°C months at months at months at months at 5°C °C 5°C 5°C Example 16 0.0 0.1 0.1 0.1 0.3 As can be seen in Table 26 above, the content of low-molecular-weight components in the formulation of Example 16 after 12 months of storage at 5°C was as low as 0.4% or less.
TNF-α binding affinity
[00298] Table 27 After 0 After 3 After 6 After 9 After 12 month at 5°C months at months at months at months at 5°C °C 5°C 5°C Example 16 95 98 116 101 97 As can be seen in Table 27 above, the TNF-α binding affinity of the formulation of Example 16 after 12 months of storage at 5°C was as high as 95% or more.
The formulation of Example 16 was measured for its viscosity after 0, 0.5, 1, 2 and 3 months of storage at a temperature of 40 ± 2°C under a closed condition and for its viscosity after 6 months of storage at a temperature of 5 ± 3°C under a closed condition. The results of the measurement are shown in Table 28 below.
Viscosity (cP) Table 28 After 0 month After 0.5 months After 1 month After 6 months at at 40°C at 40°C 5°C Example 16 4.1 5.6 8.0 4.0
[00303] As can be seen in Table 28 above, the viscosity of the formulation of Example 16 was maintained at a low level (8.0 cp) after 1 month of storage at a temperature of 40°C ± 2°C and maintained at a low level (4.0 cp) after 6 months of storage at a temperature of 5°C ± 3°C.

Claims (22)

WHAT IS CLAIMED IS:
1. A stable liquid pharmaceutical formulation, comprising: (A) an infliximab; (B) 0.001 to 5% (w/v) of a surfactant comprising polyoxyethylene sorbitan fatty acid ester, polyoxyethylene alkyl ether, alkylphenyl polyoxyethylene ether, polyoxyethylene- polyoxypropylene copolymers, sodium dodecyl sulfate or a mixture of two or more thereof; (C) 0.1 to 30 % (w/v) of one or more sugar and/or sugar derivative selected from the group consisting of sorbitol, mannitol, sucrose and trehalose; and (D) a buffer comprising acetate or histidine.
2. The stable liquid pharmaceutical formulation of claim 1, wherein the infliximab (A) has a concentration of 10 to 200 mg/ml.
3. The stable liquid pharmaceutical formulation of claim 1 or claim 2, wherein the surfactant (B) comprises polysorbate, poloxamer, or a mixture thereof.
4. The stable liquid pharmaceutical formulation of any one of claims 1 to 3, wherein the surfactant (B) comprises polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, or a mixture of two or more thereof.
5. The stable liquid pharmaceutical formulation of any one of claims 1 to 3, wherein the surfactant (B) comprises polysorbate 80.
6. The stable liquid pharmaceutical formulation of any one of claims 1 to 5, wherein the surfactant (B) has a concentration of 0.02 to 0.1% (w/v).
7. The stable liquid pharmaceutical formulation of any one of claims 1 to 6, wherein the one or more sugar and/or sugar derivative selected from the group consisting of sorbitol, mannitol, sucrose and trehalose (C) has a concentration of 1 to 10% (w/v).
8. The stable liquid pharmaceutical formulation of any one of claims 1 to 7, wherein the buffer (D) comprises acetate.
9. The stable liquid pharmaceutical formulation of any one of claims 1 to 8, wherein the buffer (D) has a concentration of 1 to 50 mM.
10. The stable liquid pharmaceutical formulation of any one of claims 1 to 9, which has a pH of 4.0 to 5.5.
11. The stable liquid pharmaceutical formulation of any one of claims 1 to 10, wherein the formulation is free of aspartic acid, lysine, arginine, or mixtures thereof.
12. The stable liquid pharmaceutical formulation of any one of claims 1 to 11, wherein the formulation is free of NaCl, KCl, NaF, KBr, NaBr, Na SO , NaSCN, K SO , or mixtures 2 4 2 4 thereof.
13. The stable liquid pharmaceutical formulation of any one of claims 1 to 12, which is free of a chelating agent.
14. The stable liquid pharmaceutical formulation of any one of claims 1 to 13, which has a viscosity of 0.5 cp to 10 cp after 1 month of storage at 40°C ± 2°C, or a viscosity of 0.5 cp to 5 cp after 6 months of storage at 5°C ± 3 °C.
15. A stable liquid pharmaceutical formulation, comprising: (A) 90 to 145 mg/ml of an infliximab; (B) 0.02 to 0.1% (w/v) of a surfactant comprising polyoxyethylene sorbitan fatty acid ester, polyoxyethylene alkyl ether, alkylphenyl polyoxyethylene ether, polyoxyethylene- polyoxypropylene copolymers, sodium dodecyl sulfate or a mixture of two or more thereof; (C) 1 to 10% (w/v) of one or more sugar and/or sugar derivative selected from the group consisting of sorbitol, mannitol, sucrose and trehalose; and (D) 1 to 50 mM of a buffer comprising acetate or histidine.
16. The stable liquid pharmaceutical formulation of any one of claims 1 to 15, which is for subcutaneous administration.
17. The stable liquid pharmaceutical formulation of any one of claims 1 to 16, which is not subjected to a reconstitution step, a dilution step, or both, before use.
18. A pre-filled syringe filled with the stable liquid pharmaceutical formulation of any one of claims 1 to 17.
19. An auto-injector including the pre-filled syringe of claim 18 therein.
20. The stable liquid pharmaceutical formulation of any one of claims 1 to 17, substantially as herein described with reference to any one or more of the examples but excluding comparative examples.
21. The pre-filled syringe of claim 18, substantially as herein described with reference to any one or more of the examples but excluding comparative examples.
22. The auto-injector of claim 19, substantially as herein described with reference to any one or more of the examples but excluding comparative examples. SEQUENCE LIST SEQ ID NO. 1: Gln Phe Val Gly Ser Ser SEQ ID NO. 2: Tyr Ala Ser SEQ ID NO. 3: Gln Gln Ser His Ser Trp Pro Phe Thr 5 SEQ ID NO. 4: Gly Phe Ile Phe Ser Asn His Trp SEQ ID NO. 5: Ile Arg Ser Lys Ser Ile Asn Ser Ala Thr SEQ ID NO. 6: Ser Arg Asn Tyr Tyr Gly Ser Thr Tyr Asp Tyr SEQ ID NO. 7: Asp Ile Leu Leu Thr Gln Ser Pro Ala Ile Leu Ser Val Ser Pro Gly Glu Arg Val Ser Phe Ser Cys Arg Ala Ser Gln Phe Val Gly Ser Ser Ile His Trp Tyr Gln Gln 10 Arg Thr Asn Gly Ser Pro Arg Leu Leu Ile Lys Tyr Ala Ser Glu Ser Met Ser Gly Ile Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser Ile Asn Thr Val Glu Ser Glu Asp Ile Ala Asp Tyr Tyr Cys Gln Gln Ser His Ser Trp Pro Phe Thr Phe Gly Ser Gly Thr Asn Leu Glu Val Lys SEQ ID NO. 8: Glu Val Lys Leu Glu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly 15 Ser Met Lys Leu Ser Cys Val Ala Ser Gly Phe Ile Phe Ser Asn His Trp Met Asn Trp Val Arg Gln Ser Pro Glu Lys Gly Leu Glu Trp Val Ala Glu Ile Arg Ser Lys Ser Ile Asn Ser Ala Thr His Tyr Ala Glu Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Ser Ala Val Tyr Leu Gln Met Thr Asp Leu Arg Thr Glu Asp Thr Gly Val Tyr Tyr Cys Ser Arg Asn Tyr Tyr Gly Ser Thr Tyr Asp Tyr Trp Gly Gln Gly Thr Thr Leu 20 Thr Val Ser SEQ ID NO. 9: 1 Met Asp Phe Gln Val Gln Ile Ile Ser Phe Leu Leu Ile Ser Ala Ser Val Ile Met 21 Arg Gly Asp Ile Leu Leu Thr Gln Ser Pro Ala Ile Leu Ser Val Ser Pro Gly Glu 25 Arg Page 1 41 Val Ser Phe Ser Cys Arg Ala Ser Gln Phe Val Gly Ser Ser Ile His Trp Tyr Gln 61 Arg Thr Asn Gly Ser Pro Arg Leu Leu Ile Lys Tyr Ala Ser Glu Ser Met Ser Gly Ile 5 81 Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Ser Ile Asn Thr Val 101 Glu Ser Glu Asp Ile Ala Asp Tyr Tyr Cys Gln Gln Ser His Ser Trp Pro Phe Thr Phe 121 Gly Ser Gly Thr Asn Leu Glu Val Lys Arg Thr Val Ala Ala Pro Ser Val Phe 10 Ile Phe 141 Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn 161 Phe Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn 15 181 Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr 201 Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His 221 Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys *** 20 SEQ ID NO. 10: 1 Met Gly Trp Ser Leu Ile Leu Leu Phe Leu Val Ala Val Ala Thr Arg Val Leu Ser Glu 21 Val Lys Leu Glu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly Ser Met Lys Leu Ser Page 2 41 Cys Val Ala Ser Gly Phe Ile Phe Ser Asn His Trp Met Asn Trp Val Arg Gln Ser Pro 61 Glu Lys Gly Leu Glu Trp Val Ala Glu Ile Arg Ser Lys Ser Ile Asn Ser Ala Thr 5 81 Tyr Ala Glu Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Ser Ala Val 101 Tyr Leu Gln Met Thr Asp Leu Arg Thr Glu Asp Thr Gly Val Tyr Tyr Cys Ser Arg Asn 121 Tyr Tyr Gly Ser Thr Tyr Asp Tyr Trp Gly Gln Gly Thr Thr Leu Thr Val Ser 10 Ser Ala 141 Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly 161 Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp 15 181 Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly 201 Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr 221 Ile Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu 20 Pro Lys 241 Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro 261 Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Page 3 281 Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr 301 Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser 5 321 Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu 341 Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser 361 Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp 10 Glu Leu 381 Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala 401 Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu 15 421 Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln 441 Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln 461 Lys Ser Leu Ser Leu Ser Pro Gly Lys *** Page 4
NZ748101A 2016-06-30 2017-06-28 Stable liquid pharmaceutical preparation NZ748101B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR20160083039 2016-06-30
KR10-2016-0083039 2016-06-30
PCT/KR2017/006855 WO2018004260A1 (en) 2016-06-30 2017-06-28 Stable liquid pharmaceutical preparation

Publications (2)

Publication Number Publication Date
NZ748101A NZ748101A (en) 2020-11-27
NZ748101B2 true NZ748101B2 (en) 2021-03-02

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020201249B2 (en) Stable liquid pharmaceutical preparation
JP6265978B2 (en) Antibody preparation
US10980881B2 (en) Stable liquid formula having an anti-TNFalpha antibody, acetate buffer and glycine
EA031436B1 (en) Aqueous pharmaceutical composition, pre-filled syringe comprising same and use of the composition in treating autoimmune diseases
KR20210096640A (en) Anti-PD-L1 Antibody Formulations
US11730698B2 (en) Stable liquid pharmaceutical preparation
NZ748101B2 (en) Stable liquid pharmaceutical preparation
US20230312734A1 (en) Stable pharmaceutical formulation, vial, cartridge, pre-filled syringe and auto-injector comprising the same
KR20230032347A (en) Stable pharmaceutical formulation