WO2008029908A1 - Stable lyophilized pharmaceutical preparation comprising antibody - Google Patents

Stable lyophilized pharmaceutical preparation comprising antibody Download PDF

Info

Publication number
WO2008029908A1
WO2008029908A1 PCT/JP2007/067481 JP2007067481W WO2008029908A1 WO 2008029908 A1 WO2008029908 A1 WO 2008029908A1 JP 2007067481 W JP2007067481 W JP 2007067481W WO 2008029908 A1 WO2008029908 A1 WO 2008029908A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
mass
alanine
freeze
polyol
Prior art date
Application number
PCT/JP2007/067481
Other languages
French (fr)
Japanese (ja)
Inventor
Takahiko Ito
Tomoyoshi Ishikawa
Chigusa Suzuki
Original Assignee
Kyowa Hakko Kirin Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kyowa Hakko Kirin Co., Ltd. filed Critical Kyowa Hakko Kirin Co., Ltd.
Priority to JP2008533208A priority Critical patent/JPWO2008029908A1/en
Publication of WO2008029908A1 publication Critical patent/WO2008029908A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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

Definitions

  • the present invention relates to the field of antibody pharmaceutical formulations. Specifically, the present invention relates to a stable lyophilized pharmaceutical preparation containing an antibody.
  • Antibodies are one of the proteins useful for medical use.
  • monoclonal antibodies and polyclonal antibodies have been used for human clinical trials, aiming at treating various diseases such as antitumor, autoimmune diseases and infectious diseases. It is attracting attention in the pharmaceutical field!
  • purified monoclonal antibodies and polyclonal antibodies are recognized as having the property of easily forming aggregates and granular insoluble particles in solution. It has become a problem. Stabilized antibody formulations that do not have such undesirable properties are desired.
  • a therapeutically effective antibody that has not been previously lyophilized and an acetate buffer maintained at PH 5.0. It describes an aqueous pharmaceutical formulation containing an agent, a surfactant and a polyol. Solution formulations are easy and most economical to handle for manufacturers, and are the easiest to administer to patients. In general, however, the effects of chemical degradation (deamidation or oxidation) and physical degradation (aggregation and precipitation) in solution formulations Therefore, precise control of storage conditions is necessary to maintain stability, and low stability! Can be a drawback for antibodies for commercial applications.
  • lyophilization of a product that is relatively unstable in solution may result in a stabilized product and thus has a longer shelf life and stability than a solution formulation.
  • a technique known as lyophilization is often used for injectable pharmaceuticals that exhibit poor stability in aqueous solutions. This process requires Freeze-drying, which causes the ice to sublime from the frozen solution, leaving a solid, dried component of the original liquid.
  • the process involves processing an aqueous solution in a liquid state and filling a dosing container, drying at a low temperature, thereby eliminating harmful thermal effects, and storing it in a dried state where it can be more stable. It has a number of advantages.
  • the lyophilized product can usually be rapidly dissolved and easily reconstituted prior to administration to a patient.
  • Patent Document 1 discloses a freeze-dried preparation containing an antibody, a sugar or amino sugar, an amino acid, and a surfactant. However, it is suggested that a freeze-dried preparation consisting of a combination of alanine and polyol is suitable.
  • Patent Document 2 discloses a freeze-dried preparation consisting only of protein, buffer, alanine and mannitol! However, it is suggested that a freeze-dried preparation consisting of a combination of alanine and amorphous polyol is suitable.
  • Patent Document 3 discloses a stable lyophilized preparation using 1 to 20 mg of D-mannitol per 1 mg of human monoclonal antibody. It is also preferable to use a lyophilized preparation composed of a combination of alanine and amorphous polyol. Also present a suitable mixing ratio.
  • freeze-dried preparation disclosed in the existing patents causes generation of foreign substances, an increase in the polymer, an increase in the oxidant, and the like, and a stable preparation could not be found.
  • freeze-dried preparations containing maltose which is a reducing sugar, showed an increase in oxidant, particularly during the storage period.
  • lyophilized preparations containing mannitol showed an increase in polymer especially during the storage period.
  • Patent Document 1 International Publication No. W098 / 22136A2 Pamphlet
  • Patent Document 2 Pamphlet of International Publication No. W097 / 17064
  • Patent Document 3 Japanese Patent Laid-Open No. 5-25058
  • Patent Document 4 Japanese Translation of Japanese Patent Publication 3-504605
  • Patent Document 5 International Publication W089 / 11297 Pamphlet
  • Patent Document 1 “Remington's Pnarmaceutical Sciences” 15 / 3 ⁇ 43 ⁇ 4, McKnouno, Mack Publishing Co., Easton, Philadelphia, pp 1483-1485 Disclosure of Invention
  • An object of the present invention is to provide a stable lyophilized pharmaceutical preparation containing an antibody. Specifically, an object of the present invention is to provide a stable lyophilized pharmaceutical preparation containing a therapeutically effective amount of an antibody and containing a crystalline substance alanine and a non-reducing sugar and an amorphous polyol. Means for solving the problem
  • the present inventors have conducted extensive research to obtain a stable lyophilized preparation containing a therapeutically effective amount of antibody.
  • the result is a therapeutically effective amount of antibody plus non-reduced crystalline material. It has been found that a lyophilized preparation can be obtained by containing a polyol that is a sugar and an amorphous substance, and has an effect of suppressing the formation of a polymer generated during a storage period that is not found in a conventionally known lyophilized preparation.
  • the present invention has been completed.
  • the present invention is as follows.
  • a method for producing a stable freeze-dried preparation comprising adding alanine and polyol to an antibody and freeze-drying.
  • Mass force S of alanine and polyol for antibody mass 1 is 5 / 6-50 [15]-[
  • the preparation containing the antibody of the present invention has a polymer, a degradation product, a deamide, an antibody polymer during the storage period even when stored at 25 ° C or 40 ° C for 3 months. It was confirmed that the antibody was stable without increasing its oxidant, and the biological activity of the antibody was retained.
  • the preparation according to the present invention is stable for at least 1 year at low temperature (2 ° C to 8 ° C), and stable for at least 25 months at 3 months and / or at 40 ° C for 1 month.
  • FIG. 1 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C.
  • the amount of polymer was measured by size exclusion HPLC.
  • Alanine alone shows remarkable polymer formation, but shows that a formulation combining alanine with trehalose or sucrose is stable.
  • Fig. 2 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C.
  • the amount of polymer was measured by size exclusion HPLC.
  • the ratio of sucrose in combination with alanine indicates that a formulation with R of 0.25 to 2.5 is stable.
  • Fig. 3 shows changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C.
  • FIG. The amount of polymer was measured by size exclusion HPLC. This indicates that a preparation with a Q (ratio of antibody mass to alanine mass) of 0.05 to 3 is stable.
  • Fig. 4 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C. The amount of polymer was measured by size exclusion HPLC. It shows that lyophilized preparations with anti-HLA-DR antibody and anti-CD40 antibody using alanine and sucrose as excipients are stable.
  • a “stable formulation” does not contain ingredients that are toxic to the patient to whom the formulation is administered, and by adding additives other than active ingredients that do not disrupt the patient's biostasis as much as possible. These active ingredients retain chemical and / or physical and / or biological stability during storage. ⁇ Additives other than active ingredients that do not disrupt the patient's homeostasis as much as possible '' are those whose safety has been sufficiently confirmed by past treatment results, or past administration results have not been! O! / Even if there is toxicity evaluation for cells and animals! /, Means that safety is sufficiently predicted by other methods. “Preserving patient homeostasis” means that additives other than the active ingredient have no biological activity unacceptable to the patient and / or isotonic (essentially with human blood). Have the same osmotic pressure).
  • Lyophilization is a lyophilization often used in the preparation of pharmaceuticals!
  • a liquid composition is prepared and then lyophilized to form a dried cake-like product.
  • This method generally dries an already frozen sample under vacuum to remove the ice, leaving the non-aqueous component in the form of a powder or cake-like material.
  • the lyophilized product can be stored for extended periods of time and at elevated temperatures without loss of biological activity and can be easily reconstituted into a particulate-free solution by adding an appropriate diluent.
  • a suitable diluent is any biologically acceptable liquid in which the lyophilized powder becomes completely soluble. Water, particularly water for injection, is a suitable diluent because it does not contain salts or other substances that affect antibody stability.
  • the advantage of freeze-drying is the moisture content to a level where various molecular events are significantly reduced (this is The product becomes unstable during long-term storage).
  • the lyophilized formulation can also easily withstand the physical stress of transport.
  • the reconstituted product is free of particulate matter and can therefore be administered parenterally, preferably intravenously or intramuscularly or subcutaneously, to a subject without prior filtration.
  • An important feature of a lyophilized formulation is the product reconstitution time or the time required to rehydrate.
  • a cake with a highly porous structure is important in order to enable very fast complete rehydration.
  • Cake structure is a function of many parameters, including protein concentration, excipient type and concentration, and process parameters of the lyophilization cycle.
  • reconstitution time increases with increasing protein concentration, so short reconstitution time is an important goal in the development of high concentration lyophilized antibody formulations. Longer reversion times degrade product quality because the protein is exposed to longer time in a more concentrated solution.
  • Protein stability is measured by various analytical methods. For example, the new 'protein refinement theory and practice', as outlined by RK Scopes, Springer's “Fairark Tokyo Publishing”. Protein stability includes chemical stability, physical stability and biological stability. “Chemical stability” can be assayed by detecting the chemically altered state of the protein. Chemical changes can be assessed by size modification such as clipping, which can be assessed by size exclusion chromatography or SDS-PAGE, charge state changes that can be assessed by ion exchange chromatography (eg, as a result of deamidation), and hydrophobic chromatography. Includes possible changes in the hydrophilic / hydrophobic state (eg resulting from oxidation).
  • “Physical stability” includes the absence of insoluble particulates and / or turbidity and / or agglomeration that can be assessed during visual inspection of color and / or transparency and / or by size exclusion chromatography. “Biological stability” can be evaluated by assaying the binding activity to an antigen that can be evaluated by, for example, size exclusion chromatography or ELISA.
  • Proteins useful for therapeutic applications include antibodies. Antibodies are expressed on the cell surface. Attempts have been made to use antibodies that bind to proteins and have the effect of inducing cell death or damage to cells for the treatment of cancer and the like. Currently, it is used as a target disease for monoclonal antibodies such as chimeric antibodies (Rituximab) targeting CD20, a receptor on the cell membrane, humanized antibodies targeting Her2 / neu, and malignant tumors. The therapeutic effect is recognized. In addition, as disclosed in the pamphlet of International Publication No. WO2003 / 033538!
  • a monoclonal antibody (anti-HLA-DR antibody) against an HLA-DR antibody which is a type of class II major histocompatibility complex (MHC) molecule Is also considered useful.
  • Antibodies are particularly useful as anti-tumor agents because of their high specificity for antigens with long blood half-lives. For example, in the case of an antibody that targets a tumor-specific antigen, the administered antibody is presumed to accumulate in the tumor, so that it depends on the capture-dependent cytotoxic activity (CDC) and antibody-dependent cytotoxicity. Attacks against cancer cells of the immune system can be expected by activity (ADCC).
  • ADCC activity
  • the bound drug can be efficiently delivered to the tumor site. Reducing side effects can be expected by reducing the amount of drug delivered. If a tumor-specific antigen has an activity that induces cell death, administration of an antibody having agonistic activity can be performed. Also, tumor-specific antibody accumulation and tumor growth due to antibody activity Stop or retraction is expected.
  • the antibody is considered to be suitable for application as an antitumor agent due to its characteristics as described above.
  • the “lyophilized preparation” included in this patent is a form in which an active ingredient which is an antibody having a medicinal effect is lyophilized, and the form in which the active ingredient is clearly effective in a solid, It does not contain any additional ingredients that may disrupt biostasis to the patient to whom the product is administered.
  • the active ingredient shall be clearly effective means that the active ingredient contained therein maintains its activity and does not lose its pharmaceutical effect.
  • An additive other than the active ingredient refers to those whose safety has been sufficiently confirmed by past treatment results, or no past administration results! It means substances whose safety can be sufficiently predicted by toxicity evaluation for cells, animals or other methods.
  • the lyophilized pharmaceutical preparation of the present invention may contain an active ingredient which is an antibody having a pharmaceutical effect and other pharmaceutically acceptable additives.
  • “maintaining patient homeostasis” means that additives other than the active ingredient are not acceptable to the patient! /, Have no biological activity, and / or are isotonic if possible (human blood And have the same osmotic pressure! /).
  • a “stable formulation” is one in which the active ingredient therein retains chemical and / or physical and / or biological stability upon storage. Preferably stable for at least 1 year at low temperatures (2 ° C to 8 ° C) and preferably stable for at least 3 months at 25 ° C and / or at least 1 month at 40 ° C. Stable to freeze-thaw, light irradiation and vibration. Protein stability is measured by various analytical methods. “Chemical stability” can be assayed by detecting and quantifying the chemically altered state of the protein.
  • Chemical changes include, for example, size modifications such as clipping that can be evaluated by size exclusion chromatography or SDS-PAGE, charge changes that can be evaluated by ion exchange chromatography (eg, as a result of deamide), and hydrophilicity / Changes in the hydrophobic state (eg resulting from oxidation) and the like.
  • Physical stability includes the absence of insoluble foreign matter and / or aggregates, and can be evaluated by visual inspection of color and / or transparency and / or size exclusion chromatography.
  • Biological stability can be evaluated by assaying the binding activity to an antigen that can be evaluated by, for example, size exclusion chromatography or ELISA. An antibody retains biological stability when it has not undergone chemical or physical changes.
  • the preparation retains chemical stability and physical stability, it can be said that the preparation is stable.
  • whether the preparation is stable can be determined by measuring the presence or absence of changes in the chemical and physical properties of the contained antibody.
  • Stable preparations do not have the ability to increase the amount of antibody polymer, degradation products, deamides, oxidants, etc. during storage so as to reduce the pharmaceutical effect of the preparation, and no insoluble foreign matter or turbidity is observed.
  • the stable formulation of the present invention is Antibodies can be stored for at least 1 year at low temperatures (2 ° C to 8 ° C), or stored for at least 3 months at 25 ° C, or stored at 40 ° C for 1 month
  • the polymers, degradation products, deamides, and oxidants do not increase so as to reduce the pharmaceutical effect of the preparation. Also, even if the product is freeze-thawed or subjected to vibration, the antibody polymer, degradation product, deamide, and oxidant do not increase so as to reduce the pharmaceutical effect of the preparation.
  • the antibody polymer in the stable preparation of the present invention does not increase during storage, for example, when it is measured by size exclusion chromatography after being stored at 25 ° C or 40 ° C for one month, A small proportion of the polymer is desirable.
  • the degradation product of the antibody in the stable preparation of the present invention does not increase greatly during storage, for example, the degradation product for the antibody in the formulation even if stored for 1 month at 25 ° C or 40 ° C. It is desirable that the ratio of is small.
  • the amount of deamidated antibody in the stable preparation of the present invention does not increase greatly during storage, for example, even if stored at 25 ° C or 40 ° C for 1 month, It is desirable that the ratio of the deamide is small.
  • the oxidized form of the antibody in the stable preparation of the present invention does not increase greatly during storage, for example, even if it is stored at 25 ° C or 40 ° C for 1 month, A small percentage is desirable.
  • the freeze-dried preparation of the present invention is characterized in that the amount of the polymer due to the antibody does not increase during storage.
  • the polymer content and the degradation product content can be measured by, for example, size exclusion chromatography, the deamide content can be measured by, for example, ion exchange chromatography, and the oxidant content can be measured, for example, by hydrophobic HPLC.
  • the "therapeutically effective amount” of the antibody in the present invention refers to an amount effective for preventing or treating a disease for which the antibody is effective for treatment.
  • Disease is any condition that would benefit from treatment with an antibody. This includes chronic and acute diseases or illnesses, including pathological conditions that predispose to mammalian diseases.
  • the therapeutically effective amount of antibody present in the formulation is determined, for example, taking into account the desired dose and mode of administration.
  • About 1 mg / mL to about 200 mg / mL, preferably about 5 mg / mL to about 50 mg / mL, most preferably about 10 mg / mL and / or about 20 mg / mL are exemplary antibody concentrations in the formulation.
  • antibody included in this patent is used in the broadest sense, and in particular as long as it retains a monoclonal antibody, a polyclonal antibody, a multispecific antibody, or a desired biological activity. Includes antibody fragments.
  • the antibodies included in this patent are not particularly limited as long as they bind to a desired antigen.
  • Mouse antibodies, rat antibodies, rabbit antibodies, Hedge antibodies, camel antibodies, chicken antibodies, chimeric antibodies, humanized antibodies, human antibodies, etc. Can be used as appropriate.
  • the antibody may be a polyclonal antibody or a monoclonal antibody.
  • the present invention relates to a stable lyophilized pharmaceutical preparation containing an antibody.
  • the antibodies in the formulation are prepared using techniques well known to those skilled in the art that are available in the relevant field to produce antibodies.
  • Exemplary monoclonal antibody preparation methods include: (1) purification of biopolymers used as immunogens and / or creation of cells overexpressing antigenic proteins on the cell surface; (2) antigens in animals After immunization by injection into cells, blood is collected and the antibody titer is assayed to determine the timing of removal of the spleen and the like, and then antibody-producing cells are prepared.
  • Myeloma cells ( (4) cell fusion between antibody-producing cells and myeloma, (5) selection of hyperidoma groups producing the desired antibody, (6) splitting into single cell clones (cloning), (7 ) In some cases, culture of high-pridoma to produce a large amount of monoclonal antibody, or breeding of animals transplanted with high-pridoma. (8) In some cases, human monoclonal antibody is obtained from antibody-producing cells such as hyperidoma.
  • the gene to be coded is cloned, incorporated into an appropriate vector, and introduced into a host (eg, mammalian cell line, E. coli, yeast cell, insect cell, plant cell, etc.), and gene recombination technology is used.
  • a host eg, mammalian cell line, E. coli, yeast cell, insect cell, plant cell, etc.
  • polyclonal antibodies can also be prepared by methods well known to those skilled in the art.
  • the antibody included in this patent has a heavy chain having an amino acid sequence in which one or several amino acids are deleted, substituted or added in the amino acid sequences of the heavy chain and / or light chain of the antibody. And / or an antibody consisting of a light chain is also included.
  • Partial modification (deletion, substitution, insertion, addition) of the amino acid as described above into the amino acid sequence of the antibody of the present invention involves partial modification of the base sequence encoding the amino acid sequence IJ. Can be introduced. This partial modification of the base sequence is a known site-specific mutagenesis method. Can be introduced by a conventional method. (Proc Natl Acad Sci USA., 1984 Vol81: 56 62).
  • the antibodies included in this patent include antibodies having any immunoglobulin class and isotype.
  • the antibody includes a modified antibody in which a subclass is recombined, or a modified antibody in which a heavy chain constant region is further modified.
  • bacterial toxins such as Pseudomonas aeruginosa toxin, diphtheria toxin, ricin, and methotrexate, such as iodine, yttrium, indium, and technetium.
  • the antibodies included in this patent also include functional fragments of antibodies.
  • the term “functional fragment” means a part (partial fragment) of an antibody that retains at least one action of the antibody on the antigen. Specifically, F (ab ′), Fab ′, Fab, Fv, disulfide binding Fv, single chain Fv (scFv), diapodi, and polymers thereof (DJKing. Applications an a Engineering of Monoclonal Antibodies., 1998 TJ International Ltd) 0
  • the antibody included in this patent is a polyclonal antibody or a monoclonal antibody, and is preferably a human antibody, a humanized antibody or a chimeric antibody.
  • the antibody is preferably IgG, and is preferably any one of IgG subclass IgG1, IgG2, and IgG4. Further, IgG may be an IgG in which amino acid deletion and / or substitution and / or insertion has been performed on a part of the amino acid sequence of the constant region by genetic modification.
  • the hybridomas producing HD4, HD6, HD8 and HD10 are FERM BP-7771, FERM BP_7772, FERM BP-7773, and FERM BP-7774, respectively.
  • Research Institute Patent Biological Deposit Center (Ibaraki, Japan) It is deposited internationally at Tsukuba Sakai Higashi 1-chome 1 1 Central 6).
  • hybridomas producing HD3 and HD7 have been deposited internationally as FERM BP-08534 and FERM BP-08536 on October 31, 2003, respectively. Furthermore, this patent also includes a human monoclonal antibody against CD40, preferably KM281-1-10, M281-2-10, which are anti-CD40 antagonist antibodies described in International Publication No. WO2002 / 088186 pamphlet.
  • Hypridoma clones KM302_1, KM281_1_10, and M281-2-10-1-2 were dated May 9, 2001 as clones KM341-1-19 as FERM BP_7578, FERM BP-75 79, and FERM BP-7580, respectively.
  • An international deposit has been made at Tsukuba Sakai Higashi 1-Chome 1-Chome 1-Chuo 6) under the Budapest Treaty.
  • plasmids with variable regions of F2-103, F5-77 and F5-157 of heavy chain and light chain were ATCC PTA_3302 (F2_103 heavy chain) and ATCC PTA-3 303 (F2) as of April 19, 2001, respectively.
  • the lyophilized preparation according to the present invention can be prepared by preparing a solution preparation containing the above-described antibody and various additives as active ingredients, and then lyophilizing the solution.
  • the lyophilized preparation contains, in addition to the antibody, a polyol, a crystalline substance, and further contains a buffer and a surfactant.
  • "Freeze-dried”, “lyophilized” and “Freeze-dried” means that the substance to be dried is first frozen and then the ice or freezing solvent is placed in a vacuum environment. It means the process of sublimation and removal.
  • an excipient may be contained in the preparation prior to lyophilization. The freeze-drying process is performed by a known method with a force S.
  • the "additive" included in the present invention includes all components included in the lyophilized preparation other than the active component, such as a buffer, a pH adjuster, an isotonic agent, an excipient, and a stabilizer. , Surfactants, preservatives and the like. In addition, one type of additive component that exhibits two or more types of effects is also included.
  • Buffer refers to an additive that moderates changes in pH by the action of an acid-base conjugate component.
  • the buffer concentration is about 1 mmol / L to about 50 mmo / L, preferably 5 mmol / L to 20 mmol / L, and most preferably about 10 mmol / L, depending on its buffer capacity and / or the desired osmotic pressure.
  • Preferred buffering agents include glutamate (eg, sodium glutamate), citrate and other organic acid buffers. Most preferred is glutamate.
  • Polyol is a substance having a plurality of hydroxyl groups, and is also used as an excipient in lyophilized preparations. Contains sugars (reducing and non-reducing sugars), sugar alcohols and sugar acids. Suitable polyols herein have a molecular weight that is less than about 600 KD (eg, in the range of about 120 KD to about 400 KD). “Reducing sugar” includes a force capable of reducing metal ions, or a hemiacetal group capable of reacting covalently with other amino groups and lysine in protein. “Non-reducing sugar” It does not have these properties of sugar.
  • reducing sugars are funolectose, mannose, manoleose, ratatose, arabinose, xylose, ribose, rhamnose, galactose and glucose.
  • non-reducing sugars include sucrose, trenosose, sorbose, meletitose, and raffinose.
  • sugar alcohols include mannitol, xylitol, erythritol, thritol, sorbitol, and daricerose.
  • Sugar acids include L-darconic acid and its metal salts. The use of reducing sugars in lyophilized formulations is not preferred because it promotes the formation of oxidants.
  • Non-freezing Illustrative examples of polyols having a crystalline structure include sucrose, trehalose, sorbitol and the like.
  • the polyol of the present invention is preferably a non-reducing sugar and amorphous polyol, or a polyol such as a non-reducing sugar and amorphous sugar or sugar alcohol. More preferred are non-reducing sugar and amorphous polyols such as sucrose, trehalose, sorbose, meretitol and raffinose, and most preferred are sucrose and trehalose.
  • the freeze-dried preparation of the present invention may contain a plurality of polyols.
  • Crystalstalline substance includes crystalline amino acids such as alanine and glycine as long as they take a crystal structure upon freezing. Particularly preferred is alanine, which is contained in an amount of 5 to 40 mg / mL, preferably 10 to 30 mg / mL, more preferably 15 to 25 mg / mL, and particularly preferably 20 mg / mL.
  • Polyols and crystalline substances can act as excipients and tonicity agents.
  • the “excipient” refers to a substance having a function of maintaining the cake shape of a freeze-dried preparation and / or a function of protecting a protein against stress associated with freezing and drying.
  • a common example used for excipients is lactose.
  • An excipient provides useful properties in that it enhances protein stability during long-term storage.
  • These substances can also act as isotonic agents.
  • An “isotonic agent” refers to an agent that adjusts the osmotic pressure so that the subject formulation has essentially the same osmotic pressure as human blood.
  • the isotonic preparation has an osmotic pressure of about 250 to 350 mOsm / kg, and the osmotic pressure ratio is about 1, with the osmotic pressure of physiological saline being 1.
  • the alanine used as an excipient in the present invention maintains the cake shape, and the polyol protects the protein against stress associated with freezing and drying.
  • a polyol such as sucrose also acts as a stabilizer against antibody aggregation, and plays an important role in reducing the time required for reconstitution of a freeze-dried preparation in a solution without any particulate matter.
  • the polyol is added to the formulation in an amount that varies depending on the desired osmotic pressure of the formulation.
  • the reconstituted lyophilized formulation is isotonic but may be hypertonic or hypotonic.
  • “Surfactants” include nonionic surfactants such as polysorbates (eg, polysorbate 20, 80, etc.) or poloxamers (eg, poloxamer 188). Added The amount of detergent is such that it reduces aggregation of the formulated antibody and / or minimizes the formation of particles in the formulation and / or reduces protein adsorption to the container.
  • the surfactant preferably includes polysorbate, and most preferably includes polysorbate 80.
  • the surfactant may be present in the formulation, preferably in an amount of 0.02 mg / mL to O. lmg / mL, most preferably about 0.05 mg / mL.
  • pre-lyophilized compositions include about 1 mg / mL to 60 mg / mL IgG antibody, about 10 mmol / L to 20 mmol / L sodium glutamate ( ⁇ 5 ⁇ 5), about 0.05 mg / mL polysorbate 80 And 20 mg / mL alanine and 5 mg / mL to 50 mg / mL sucrose as excipients.
  • the pre-lyophilized formulation is lyophilized to a dry, stable powder that can be easily reconstituted into a particulate-free solution suitable for human administration.
  • a crystalline substance most preferably alanine
  • a non-reducing sugar and an amorphous polyol most preferably sucrose and trehalose
  • an antibody as excipients and freeze-dried.
  • a stable lyophilized formulation is provided.
  • the cake structure is maintained in a crystalline phase composed of a crystalline substance, and frozen and dried by an amorphous phase composed of antibodies and non-reducing sugars and / or amorphous polyols (most preferably sucrose and trehalose).
  • a stable lyophilized formulation can be derived.
  • the blending ratio and blending amount of the alanine and sucrose excipients used in the present invention are appropriately adjusted according to the antibody concentration which is the active ingredient.
  • R is the mass ratio of sucrose to alanine (sucrose mass / alanine mass) or concentration ratio (sucrose concentration / alanine concentration) present in the lyophilized preparation
  • stability is good
  • R is 0.25 or more Preferably 0.5 to 2.5, more preferably 1 to 2. Most preferably 1.5.
  • Q is the mass ratio of antibody to alanine (antibody mass / alanine mass) or concentration ratio (antibody concentration / alanine concentration) present in the lyophilized preparation
  • sucrose / Good stability with freeze-dried formulation containing alanine Q between 0.05 and 3, preferably 0
  • freeze-dried preparation of the present invention is used after being restored.
  • Restoration is the time required for rehydration of a lyophilized preparation with a solution to obtain a particle-free / clarified solution.
  • An important feature of lyophilized formulations is the product reconstitution time or the time required to rehydrate, requiring a short time.
  • a cake with a highly porous structure is important in order to allow very fast complete rehydration.
  • Cake structure is a function of many parameters, including protein concentration, excipient type and concentration, and process parameters of the lyophilization cycle.
  • the recovery time increases with increasing protein concentration. Therefore, longer reversion times degrade product quality because proteins are exposed to more concentrated solutions for longer periods of time.
  • the user cannot administer until the product is completely rehydrated. This is to ensure complete rehydration to ensure that the product is free of particulates, is administered at the correct dose, and its sterility is not affected. That is, rapid rehydration is advantageous for patients and physicians.
  • Solution formulations can be lyophilized using appropriate drying parameters.
  • the lyophilized formulation retains the stability of the biological activity of the antibody and protects the antibody intended for administration to human subjects from physical and chemical degradation in the final product.
  • the lyophilized formulation is rehydrated at the time of use with a diluent (eg, water for injection) to give a solution free of particulate matter.
  • a diluent eg, water for injection
  • the reconstituted antibody solution does not contain particulate matter after long-term storage of the lyophilized cake at ambient temperature.
  • the reconstituted solution is administered parenterally, preferably intravenously, intramuscularly or subcutaneously to the subject.
  • the composition of the present invention minimizes the formation of protein aggregates and particulates in an immunoglobulin-containing reagent and ensures that the antibody in solution maintains its immune activity over time.
  • the composition is a sterile, pharmaceutically acceptable lyophilized solution prepared from an aqueous pre-lyophilized formulation containing antibodies, alanine, and polyol and surfactant in a buffer having a neutral or acidic pH. Contains formulation.
  • the composition of the present invention further comprises alanine and And excipients other than polyols and / or tonicity agents.
  • the formulation is administered to mammals, preferably humans, in need of treatment with antibodies, in a known manner, for example, intravenously administered as a bolus or by continuous infusion over time, intramuscular, intraperitoneal, intracerebral, subcutaneous. Administered by intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • the formulation is administered to the mammal by intravenous administration.
  • the formulation is infused, for example, using a syringe or via an intravenous infusion line.
  • a freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in International Publication No. WO2003 / 033538 as an antibody.
  • HLA-DR anti-HLA-DR antibody, IgGl
  • the reagent used in this study was an anti-HLA-DR antibody (approximately 20 mg / mL, Kirin Brewery's Pharmaceutical Production Technology Laboratory (July 2007 1) according to the method described in International Publication No. W02003 / 00 33538 pamphlet. After that, prepared by Kirin Pharma Co., Ltd.
  • Each preparation specimen was aseptically filtered using a 0.22 m filter (Millipore) in a clean bench, and filled in 5 mL glass vials (conforming to the Japanese Pharmacopoeia). After filling, half-plugging was performed with a rubber plug! / ⁇ Freeze-drying was performed using a freeze dryer manufactured by Nippon Vacuum Technology Co., Ltd. Aseptic dilution of V and solution (placebo) containing anti-HLA-DR antibody for dilution of analysis specimen and analysis blank was performed using 0.22 ⁇ m bottle top filter (Nalgen) in a clean bench.
  • each preparation specimen was stressed according to the following conditions.
  • Thermal stability test Stored in an incubator (manufactured by TABAI ESPEC) controlled at 40 ° C or 25 ° C for 1 month and 3 months. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after heat stress.
  • insoluble foreign matter and turbidity The presence of insoluble foreign matter and turbidity was examined with the naked eye at a position of about 5,000 lux directly under a white light source.
  • Size exclusion HPLC assay The polymer content and degradation product content were calculated by the size exclusion high-speed liquid chromatography method. Samples were diluted to lmg / mL as necessary and 20 L injections were performed at ambient temperature. For separation, use TSKgel G3000 SWXL 30cm X 7.8mm (manufactured by Tosohichi Co., Ltd.) column, and use 20mmol / L sodium phosphate and 500mmol / L sodium chloride pH 7.0 as mobile phase and analyze at 0.5mL / min flow rate. The time was 30 minutes and detection was performed at 215 nm. The main P The polymer eluted before the catalyst was defined as the polymer, and the polymer eluted after the product was defined as the decomposition product.
  • Cation exchange HPLC assay Deamide content was calculated by the cation exchange high performance liquid chromatographic method. Sixty specimens diluted appropriately to 5 mg / mL were injected. Detection was performed at 280 nm using TSKgel BIOAssist S (manufactured by Tosoh Corporation) as a separation column. As mobile phase, 20 mM tartaric acid pH 4.5 for A and 20 mM tartaric acid, 1 M sodium chloride pH 4.5 for B were used, and the analysis was performed using the optimum gradient conditions. Degraded substances that eluted ahead of the main peak were defined as deamides.
  • Hydrophobic HPLC assay The content of oxidant was calculated by hydrophobic chromatography. Twenty samples, each appropriately diluted to 1 mg / mL, were injected using a sample processing solution (20 mmol / L sodium phosphate, 0.984 mol / L ammonium sulfate, 7.5 mmol / L methanoline). Analysis and detection were performed at 215 nm using TS gel Buty NPR (manufactured by Tosoh Corporation) as a separation column.
  • TS gel Buty NPR manufactured by Tosoh Corporation
  • the mobile phase is 20 mmol / L sodium phosphate, ⁇ 6 ⁇ 5 as A, and 20 mmol / L sodium phosphate, 1.2 mol / L ammonium sulfate, ⁇ 6.5 as ⁇ , and the analysis is performed using the optimal gradient conditions. I went. In addition, the deteriorated substance eluted ahead of the main peak was defined as an oxidant.
  • Reduction SDS-PAGE assay The sample solution was diluted to 400 g / mL as necessary. 3 ⁇ 4DS Sample Buffer (Invitrogen), sample Reducing Agent (Invitrogen3 ⁇ 43 ⁇ 4), and H 2 O were added to the sample solution and heated at 95 ° C for 5 minutes to obtain a reduced sample solution.
  • the electrophoresis basket was filled with Tris-Glycine SDS Running buffer (manufactured by Invitrogen) for electrophoresis. Apply 5 L of the sample solution to 4-20% Tris_Glycine Gel (Invitrogen) and perform electrophoresis at a constant voltage of about 125 V until the blue color of bromophenol blue contained in the sample solution moves near the bottom of the gel.
  • molecular weight markers including 97 KDa, 66 KDa, 45 KDa, 30 KDa, 20 KDa, and 14 Da proteins.
  • Non-reducing SDS-PAGE assay The sample solution was diluted to 400 ⁇ g / mL as necessary. LDS Sample Buffer (Invitrogen) and H0 were added to the sample solution to obtain a non-reduced sample solution. The electrophoresis gel was filled with Tris-Acetate SDS Running buffer (Invitrogen) for electrophoresis. Apply 5 L of the sample solution to 3-8% Tris_Acetate Gel (Invitrogen), perform electrophoresis at a constant voltage of about 125 V, and the blue color of bromophenol blue in the sample solution is near the bottom of the gel. Went to move on. The gel after electrophoresis was detected by silver staining.
  • Tris-Acetate SDS Running buffer Invitrogen
  • molecular weight markers including 220 KDa, 116 KDa, 97.4 Da, 63.3 KDa, 54.4 Da, and 36.5 KDa proteins.
  • Osmotic pressure ratio V was measured using an automatic osmotic pressure measuring apparatus (OSMO STATION OM-6050 manufactured by ARKRAY, Inc.), and the ratio of the measured physiological saline to the osmotic pressure was calculated.
  • pH pH was measured using an automatic pH measuring device (Metler 'MP-230 manufactured by Toledo Co., Ltd.). Measurement was performed after calibration using standard solutions of pH 4, pH 7, and pH 9 at the start of measurement.
  • Moisture measurement The moisture content was measured using a trace moisture analyzer (AQ-2000, Hiranuma Sangyo Co., Ltd.) connected to an automatic moisture vaporizer (LE-20SA, Hiranuma Sangyo Co., Ltd.), and the moisture content was calculated.
  • AQ-2000 trace moisture analyzer
  • LE-20SA automatic moisture vaporizer
  • Figure 1 shows the change in polymer when each lyophilized formulation was stored at 25 ° C and 40 ° C.
  • the amount of polymer was measured by size exclusion chromatography.
  • a polymer increase of about 5% at 40 ° C for 1 month and about 9% at 40 ° C for 3 months was confirmed.
  • foreign matter and turbidity were confirmed by visual inspection with the naked eye after redissolving, and the increase in polymer was also largely unstable.
  • the formulation combined with trehalose and sucrose was very stable with no increase in polymer even after storage at 40 ° C for 3 months.
  • a combination of alanine and polyol was suitable as an excipient of a lyophilized preparation for stabilizing an antibody. It is desirable to use an amorphous polyol, more preferably a non-reducing and amorphous polyol, and most preferably, trehalose or sucrose as the polyol.
  • Example 1 it was found that trehalose or sucrose is most preferable as the polyol combined with alanine.
  • sucrose was used as the polyol, the concentration of alanine was kept constant, the concentration of sucrose mixed with alanine was changed, and the optimum concentration ratio R for polymer suppression was examined.
  • R represents the mass of sucrose / alanine present in the lyophilized product.
  • a freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in International Publication No. WO2003 / 033538 as an antibody.
  • HLA-DR anti-HLA-DR antibody, IgGl
  • Example 2 Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TABA I ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each specimen was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after thermal stress loading.
  • Figure 2 shows the change in polymer when each lyophilized formulation was stored at 25 ° C and 40 ° C.
  • a beautiful cake was formed at 5 or less and showed a shape that could withstand long-term storage.
  • the other analysis items foreign matter, turbidity, insoluble fine particles, deamide, and oxidant) were stable with almost no change observed before and after thermal stress loading.
  • sucrose mass ratio R sucrose mass / alanine mass
  • R having good stability was 0.25 or more, preferably 0.5 to 2.5, more preferably 1 to 2, and still more preferably 0.75 to 2. Most preferably it was 1.5
  • a freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in WO2003 / 033538 pamphlet as an antibody.
  • HLA-DR anti-HLA-DR antibody, IgGl
  • Example 2 Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TAB AI ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after the heat stress.
  • the sum of alanine mass and sucrose mass relative to antibody mass (antibody mass is 1), it is 1: 5 / 6-50, preferably 1:;!-20, more preferably 1:;! To 10, particularly preferably 1: 5.
  • Anti-CD40 antibody 80 [0115] (1) Materials and methods
  • Example 2 Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TAB AI ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after the heat stress.
  • Figure 4 shows the change in polymer when each lyophilized preparation was stored at 40 ° C (Initial ratio: value divided by the amount of polymer before lyophilization, with the value before lyophilization being 1). It was. The amount of polymer was measured by size exclusion HPLC. Also in the anti-CD40 antibody, in the preparation using alanine and sucrose as excipients, suppression of polymer formation during the storage period was confirmed, and the antibody in this preparation was stably maintained regardless of temperature and time. In addition, the other analysis items (foreign matter, turbidity, degradation products, deamides, and oxidants) were stable with almost no change observed before and after thermal stress loading.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)

Abstract

Disclosed is a stable, lyophilized pharmaceutical preparation comprising an antibody. Specifically disclosed is a lyophilized preparation comprising an antibody and further comprising a crystalline substance and a polyol as excipients, wherein the crystalline substance may be alanine and the polyol may be sucrose or trehalose.

Description

明 細 書  Specification
抗体を含有する安定な凍結乾燥医薬製剤  Stable lyophilized pharmaceutical formulation containing antibody
技術分野  Technical field
[0001] 本発明は、抗体の医薬製剤の分野に関する。詳しくは、本発明は抗体を含有する 安定な凍結乾燥医薬製剤に関するものである。  [0001] The present invention relates to the field of antibody pharmaceutical formulations. Specifically, the present invention relates to a stable lyophilized pharmaceutical preparation containing an antibody.
背景技術  Background art
[0002] 近年の生物工学の進展に伴って、組換え DNA技術を駆使し医療用途のタンパク質 を大量に純度良く造りだすことが可能となった。し力もながら、タンパク質は従来の化 学合成分子とは異なり、分子量が大きくその三次元構造も複雑であるため、そのよう なタンパク質を安定に保ったまま安定に保存するには、その物性を考慮した特別な 技術が必要となる。タンパク質を安定に保った製剤は、タンパク質に含まれる多数の 異なった官能基を保護し、また活性に関与している高次構造を保つ必要があり、タン ノ ク質医薬品の分野において、タンパク質の安定性と活性の両方を維持する安定な 製剤の開発が大きな課題となっている。  [0002] With recent progress in biotechnology, it has become possible to produce a large amount of protein for medical use with high purity using recombinant DNA technology. However, unlike conventional chemically synthesized molecules, proteins have large molecular weights and complex three-dimensional structures. Therefore, in order to preserve such proteins stably, the physical properties must be considered. Special technology is required. Formulations that keep proteins stable need to protect many different functional groups in proteins and maintain higher-order structures that are involved in activity. In the field of protein pharmaceuticals, The development of stable formulations that maintain both stability and activity is a major challenge.
[0003] 医療用として有用なタンパク質の一つとして抗体があり、近年、モノクローナル抗体 やポリクローナル抗体がヒト臨床試験に供されており、抗腫瘍や自己免疫疾患、感染 症など様々な疾病治療を目的とした医薬品分野で注目されて!/、る。しかし、精製した モノクロ一ナル抗体やポリクローナル抗体は、溶液中で凝集物及び粒状不溶性微粒 子を形成しやすレ、性質を有するものと認識されており、抗体製剤の開発にお!/、て問 題となっている。そのような望ましくない性質をもたない安定化された抗体製剤が望ま れる。 [0003] Antibodies are one of the proteins useful for medical use. In recent years, monoclonal antibodies and polyclonal antibodies have been used for human clinical trials, aiming at treating various diseases such as antitumor, autoimmune diseases and infectious diseases. It is attracting attention in the pharmaceutical field! However, purified monoclonal antibodies and polyclonal antibodies are recognized as having the property of easily forming aggregates and granular insoluble particles in solution. It has become a problem. Stabilized antibody formulations that do not have such undesirable properties are desired.
[0004] 抗体を安定化する方法の一つとして、例えば国際公開第 W098/56418号パンフレ ットにあるように前もって凍結乾燥を受けていない治療上有効な抗体と PH5.0に維持 する酢酸緩衝剤と界面活性剤とポリオールを含有する水性医薬製剤に関して記述し ている。溶液製剤は製造者にとって取り扱うのが容易かつ最も経済的であり、また、 患者に投与するのが最も容易である。し力もながら一般的に、溶液製剤では化学的 分解 (脱アミド化反応もしくは酸化反応)ならびに物理的劣化 (凝集及び沈殿)の影響 を受けるため、貯蔵条件の正確な制御が安定性を保持するために必要であり、安定 性の低!/、抗体にとっては商業的応用に対する欠点となりうる。 [0004] As one of the methods for stabilizing an antibody, for example, as disclosed in WO098 / 56418 pamphlet, a therapeutically effective antibody that has not been previously lyophilized and an acetate buffer maintained at PH 5.0. It describes an aqueous pharmaceutical formulation containing an agent, a surfactant and a polyol. Solution formulations are easy and most economical to handle for manufacturers, and are the easiest to administer to patients. In general, however, the effects of chemical degradation (deamidation or oxidation) and physical degradation (aggregation and precipitation) in solution formulations Therefore, precise control of storage conditions is necessary to maintain stability, and low stability! Can be a drawback for antibodies for commercial applications.
[0005] これに対し、溶液中で比較的不安定である生成物を凍結乾燥することは安定化さ れる生成物をもたらす可能性があり、従って溶液製剤よりも長い貯蔵期間、安定性を 有することが当該技術分野で既知である。 (非特許文献 1を参照)。乾燥された固体 中では分解反応は回避される力、もしくは数年の間安定のまま留まる。従って凍結乾 燥として既知の技術が、水溶溶液中で乏しい安定性を表す注入可能な医薬品にし ばしば使用される。この工程は凍結乾燥すること(Freeze-drying)を必要とし、それに より氷が凍結された溶液から昇華して元の液体の固体の乾燥された成分を残す。該 工程は、水性溶液を液体状態で加工しかつ投薬容器に充填し、低温で乾燥してそ れにより有害な熱的影響を排除し、そしてそれがより安定でありうる乾燥された状態で 保存し得るために多数の利点を有する。加えて、凍結乾燥された生成物は通常迅速 に溶解可能であり、そして患者への投与前に容易に再構成される。 [0005] In contrast, lyophilization of a product that is relatively unstable in solution may result in a stabilized product and thus has a longer shelf life and stability than a solution formulation. Is known in the art. (See Non-Patent Document 1). In the dried solid, the decomposition reaction is a force that can be avoided or remains stable for several years. Therefore, a technique known as lyophilization is often used for injectable pharmaceuticals that exhibit poor stability in aqueous solutions. This process requires Freeze-drying, which causes the ice to sublime from the frozen solution, leaving a solid, dried component of the original liquid. The process involves processing an aqueous solution in a liquid state and filling a dosing container, drying at a low temperature, thereby eliminating harmful thermal effects, and storing it in a dried state where it can be more stable. It has a number of advantages. In addition, the lyophilized product can usually be rapidly dissolved and easily reconstituted prior to administration to a patient.
[0006] しかしながら、所望の特徴を有する凍結乾燥された生成物を達成するために、凍結 乾燥工程での使用のための条件及び賦形剤を選ぶのに適切な注意が払わなけれ ばならない。適切な賦形剤なしでは、大部分のタンパク質製剤は、凍結乾燥の間に 遭遇される凍結及び乾燥、脱水により少なくとも部分的に変性される。加えて、乾燥さ れた固体での長期安定性を保証する賦形剤を選ぶべきである。  [0006] However, in order to achieve a lyophilized product with the desired characteristics, appropriate care must be taken in choosing the conditions and excipients for use in the lyophilization process. Without suitable excipients, most protein formulations are at least partially denatured by freezing and drying, dehydration encountered during lyophilization. In addition, excipients should be selected that ensure long-term stability with the dried solid.
[0007] 抗体を含有する凍結乾燥製剤に関して以下の文献がある。  [0007] The following documents are related to lyophilized preparations containing antibodies.
[0008] 特許文献 1は、抗体、糖又はアミノ糖、アミノ酸及び界面活性剤を含む凍結乾燥製 剤を開示してレ、る。しかしァラニンとポリオールの組み合わせからなる凍結乾燥製剤 が好適であることは提示してレ、なレ、。  [0008] Patent Document 1 discloses a freeze-dried preparation containing an antibody, a sugar or amino sugar, an amino acid, and a surfactant. However, it is suggested that a freeze-dried preparation consisting of a combination of alanine and polyol is suitable.
[0009] 特許文献 2は、タンパク、緩衝液、ァラニン及びマンニトールのみからなる凍結乾燥 製剤を開示して!/、る。しかしァラニンと非晶質のポリオールの組み合わせからなる凍 結乾燥製剤が好適であることは提示してレ、なレ、。  [0009] Patent Document 2 discloses a freeze-dried preparation consisting only of protein, buffer, alanine and mannitol! However, it is suggested that a freeze-dried preparation consisting of a combination of alanine and amorphous polyol is suitable.
[0010] 特許文献 3は、ヒトモノクローナル抗体 lmgあたり l〜20mgの D-マンニトールを用い た安定な凍結乾燥製剤を開示してレ、る。し力、しァラニンと非晶質のポリオールの組み 合わせからなる凍結乾燥製剤が好適であることも、また抗体、ァラニン、スクロースの 好適な混合比率も提示してレ、なレ、。 [0010] Patent Document 3 discloses a stable lyophilized preparation using 1 to 20 mg of D-mannitol per 1 mg of human monoclonal antibody. It is also preferable to use a lyophilized preparation composed of a combination of alanine and amorphous polyol. Also present a suitable mixing ratio.
[0011] またタンパク質の凍結乾燥医薬製剤において、タンパク質の安定化への賦形剤の 効果に関する多くの科学文献が存在する。しかし、凍結乾燥生成物の構造とその安 定性との間の関係についての理論は、一般的に受け入られていない。同様に、ポリ オール及びアミノ酸単独で又は組み合わせての役割は、一組の一般化される性質に 従って開示されず、研究されるタンパク質及び使用される賦形剤量に従って、相反す る結果が見られた。 [0011] There are also many scientific literatures on the effects of excipients on protein stabilization in protein lyophilized pharmaceutical formulations. However, the theory of the relationship between the structure of a lyophilized product and its stability is not generally accepted. Similarly, the role of polyols and amino acids alone or in combination is not disclosed according to a set of generalized properties, and conflicting results will be seen depending on the protein studied and the amount of excipient used. It was.
[0012] また既存特許で開示されている凍結乾燥製剤では異物発生、重合体の増加、酸化 体の増加などを生じ、安定な製剤を見出す事ができなかった。例えば特許文献 4や 特許文献 5に開示されているように、還元糖であるマルトースを含有する凍結乾燥製 剤では特に保存期間に酸化体の増加が見られた。特許文献 2や特許文献 3に開示 されているようにマンニトールを含有する凍結乾燥製剤では特に保存期間に重合体 の増加が見られた。  [0012] Furthermore, the freeze-dried preparation disclosed in the existing patents causes generation of foreign substances, an increase in the polymer, an increase in the oxidant, and the like, and a stable preparation could not be found. For example, as disclosed in Patent Documents 4 and 5, freeze-dried preparations containing maltose, which is a reducing sugar, showed an increase in oxidant, particularly during the storage period. As disclosed in Patent Document 2 and Patent Document 3, lyophilized preparations containing mannitol showed an increase in polymer especially during the storage period.
特許文献 1:国際公開第 W098/22136A2号パンフレット  Patent Document 1: International Publication No. W098 / 22136A2 Pamphlet
特許文献 2:国際公開第 W097/17064号パンフレット  Patent Document 2: Pamphlet of International Publication No. W097 / 17064
特許文献 3:特開平 5-25058号公報  Patent Document 3: Japanese Patent Laid-Open No. 5-25058
特許文献 4:特表平 3-504605  Patent Document 4: Japanese Translation of Japanese Patent Publication 3-504605
特許文献 5:国際公開 W089/11297号パンフレット  Patent Document 5: International Publication W089 / 11297 Pamphlet
特許文献 1:『Remington s Pnarmaceutical Sciences』 15/¾¾、マック ノヽノ、、リツシン グ カンパニー (Mack Publishing Co.)、フィラデルフィア州イーストン、 pp 1483-1485 発明の開示  Patent Document 1: “Remington's Pnarmaceutical Sciences” 15 / ¾¾, McKnouno, Mack Publishing Co., Easton, Philadelphia, pp 1483-1485 Disclosure of Invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0013] 本発明は、抗体を含有する安定な凍結乾燥医薬製剤の提供を目的とする。具体的 には治療上有効な量の抗体を含み、結晶性物質であるァラニン及び非還元糖であり 非晶質であるポリオールを含有する安定な凍結乾燥医薬製剤の提供を目的とする。 課題を解決するための手段  [0013] An object of the present invention is to provide a stable lyophilized pharmaceutical preparation containing an antibody. Specifically, an object of the present invention is to provide a stable lyophilized pharmaceutical preparation containing a therapeutically effective amount of an antibody and containing a crystalline substance alanine and a non-reducing sugar and an amorphous polyol. Means for solving the problem
[0014] 本発明者らは、治療上有効な量の抗体を含む安定な凍結乾燥製剤を得るべく鋭意 研究を行なった。その結果、治療上有効な量の抗体に、さらに結晶性物質と非還元 糖でありかつ非晶質であるポリオールを含有させることにより従来知られている凍結 乾燥製剤にはない保存期間に発生する重合物の生成を抑制する効果を有する凍結 乾燥製剤が得られることを見出し、本発明を完成するに至った。 [0014] The present inventors have conducted extensive research to obtain a stable lyophilized preparation containing a therapeutically effective amount of antibody. The result is a therapeutically effective amount of antibody plus non-reduced crystalline material. It has been found that a lyophilized preparation can be obtained by containing a polyol that is a sugar and an amorphous substance, and has an effect of suppressing the formation of a polymer generated during a storage period that is not found in a conventionally known lyophilized preparation. The present invention has been completed.
[0015] すなわち本発明は以下の通りである。 That is, the present invention is as follows.
[0016] [1] 抗体を含み、さらにァラニン及びポリオールを賦形剤として含む凍結乾燥製剤。  [0016] [1] A freeze-dried preparation containing an antibody and further containing alanine and polyol as excipients.
[0017] [2] ポリオールが非還元糖であり、かつ非晶質のポリオールである [1]の凍結乾燥製 剤。 [0017] [2] The freeze-dried preparation according to [1], wherein the polyol is a non-reducing sugar and is an amorphous polyol.
[0018] [3] ポリオールが非還元糖であり、かつ非晶質の糖である [1]の凍結乾燥製剤。  [0018] [3] The lyophilized preparation according to [1], wherein the polyol is a non-reducing sugar and is an amorphous sugar.
[0019] [4] ポリオールがスクロース又はトレハロースである [3]の凍結乾燥製剤。  [4] The freeze-dried preparation according to [3], wherein the polyol is sucrose or trehalose.
[0020] [5] 抗体質量 1に対するァラニン及びポリオールの質量が、 5/6〜50であることを特 徴とする [1]〜[4]のいずれかの凍結乾燥製剤。  [5] The lyophilized preparation according to any one of [1] to [4], wherein the mass of alanine and polyol with respect to antibody mass 1 is 5/6 to 50.
[0021] [6] ポリオール質量/ァラニン質量比が、 0.5〜2.5である [5]の凍結乾燥製剤。 [0021] [6] The freeze-dried preparation according to [5], wherein the polyol mass / alanine mass ratio is 0.5 to 2.5.
[0022] [7] 抗体質量/ァラニン質量比が、 0.05〜3である [5]又は [6]の凍結乾燥製剤。 [0022] [7] The freeze-dried preparation according to [5] or [6], wherein the antibody mass / alanine mass ratio is 0.05 to 3.
[0023] [8] 抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0.3〜20: 0.5[0023] [8] The ratio of the mass of the three components of antibody mass, alanine mass, and sucrose mass is 1: 0.3 to 20: 0.5
〜30である [1]〜[7]のいずれかの凍結乾燥製剤。 The freeze-dried preparation according to any one of [1] to [7].
[0024] [9] さらに、緩衝液を含む [1]〜[8]のいずれかの凍結乾燥製剤。 [0024] [9] The lyophilized preparation according to any one of [1] to [8], further comprising a buffer.
[0025] [10] 緩衝液がグルタミン酸緩衝液である、 [9]の凍結乾燥製剤。 [10] The lyophilized preparation according to [9], wherein the buffer is a glutamate buffer.
[0026] [11] 緩衝液の濃度が 10mMである [9]又は [10]の凍結乾燥製剤。 [0026] [11] The freeze-dried preparation according to [9] or [10], wherein the buffer solution has a concentration of 10 mM.
[0027] [12] さらに、界面活性剤を含む [1]〜[; 11]のいずれかの凍結乾燥製剤。 [0027] [12] The lyophilized preparation according to any one of [1] to [; 11], further comprising a surfactant.
[0028] [13] 界面活性剤がポリソルベートである、 [12]の凍結乾燥製剤。 [13] The lyophilized preparation according to [12], wherein the surfactant is polysorbate.
[0029] [14] 抗体を有効成分として含む医薬製剤である、 [1]〜[; 13]のいずれかの凍結乾 燥製剤。 [14] The freeze-dried preparation according to any one of [1] to [; 13], which is a pharmaceutical preparation containing an antibody as an active ingredient.
[0030] [15] 抗体にァラニン及びポリオールを添加して凍結乾燥することを含む、安定な凍 結乾燥製剤を製造する方法。  [0030] [15] A method for producing a stable freeze-dried preparation comprising adding alanine and polyol to an antibody and freeze-drying.
[0031] [16] ポリオールが非還元糖であり、かつ非晶質のポリオールである [15]の凍結乾燥 製剤を製造する方法。 [16] The method for producing a freeze-dried preparation according to [15], wherein the polyol is a non-reducing sugar and is an amorphous polyol.
[0032] [17] ポリオールが非還元糖であり、かつ非晶質の糖である [15]の凍結乾燥製剤を 製造する方法。 [0033] [18] ポリオールがスクロース又はトレハロースである [16]の凍結乾燥製剤を製造す る方法。 [0032] [17] The method for producing a freeze-dried preparation according to [15], wherein the polyol is a non-reducing sugar and is an amorphous sugar. [0033] [18] The method for producing a freeze-dried preparation according to [16], wherein the polyol is sucrose or trehalose.
[0034] [19] 抗体質量 1に対するァラニン及びポリオールの質量力 S、 5/6〜50である [15]〜[ [0034] [19] Mass force S of alanine and polyol for antibody mass 1 is 5 / 6-50 [15]-[
18]のいずれかの凍結乾燥製剤を製造する方法。 [18] The method for producing a freeze-dried preparation according to any one of [18].
[0035] [20] ポリオール質量/ァラニン質量比が、 0·5〜2·5である [19]の凍結乾燥製剤を 製造する方法。 [0035] [20] The method for producing a freeze-dried preparation according to [19], wherein the polyol mass / alanine mass ratio is from 0.5 to 2.5.
[0036] [21] 抗体質量/ァラニン質量比力 S、0.05〜3である [19]又は [20]の凍結乾燥製剤 を製造する方法。  [0036] [21] A method for producing a freeze-dried preparation according to [19] or [20], wherein the antibody mass / alanine mass specific force S is 0.05 to 3.
[0037] [22] 抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0.3〜20: 0.  [0037] [22] The ratio of the mass of the three components of antibody mass, alanine mass, and sucrose mass is 1: 0.3 to 20: 0.
5〜30である [15]〜[21]のいずれかの凍結乾燥製剤を製造する方法。  A method for producing a freeze-dried preparation according to any one of [15] to [21], which is 5 to 30.
発明の効果  The invention's effect
[0038] 実施例に示したように、本願発明の抗体を含む製剤は、 25°C又は 40°Cにおいて 3 箇月保存しても、保存期間中に抗体の重合体、分解物、デアミド体、及び酸化体が 増加することなぐ安定であり、また抗体の生物学的活性も保持されていることが確認 された。本願発明に係る製剤は少なくとも低温 (2°Cから 8°C)において 1年以上安定 であり、また少なくとも 25°Cにおいて 3箇月間及び/又は 40°Cにおいて 1箇月間安定 である。  [0038] As shown in the Examples, the preparation containing the antibody of the present invention has a polymer, a degradation product, a deamide, an antibody polymer during the storage period even when stored at 25 ° C or 40 ° C for 3 months. It was confirmed that the antibody was stable without increasing its oxidant, and the biological activity of the antibody was retained. The preparation according to the present invention is stable for at least 1 year at low temperature (2 ° C to 8 ° C), and stable for at least 25 months at 3 months and / or at 40 ° C for 1 month.
[0039] 本明細書は本願の優先権の基礎である日本国特許出願 2006-243144号の明細書 および/または図面に記載される内容を包含する。  [0039] This specification includes the contents described in the specification and / or drawings of Japanese Patent Application No. 2006-243144, which is the basis of the priority of the present application.
図面の簡単な説明  Brief Description of Drawings
[0040] [図 1]図 1は各凍結乾燥製剤を 25°Cあるいは 40°Cで保存した時の重合体量の変化を 示す図である。重合体量はサイズ排除 HPLCにより測定した。ァラニン単独では重合 体形成が顕著だがァラニンとトレハロース又は及びスクロースと組み合わせた製剤が 安定であることを示す。  [0040] FIG. 1 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C. The amount of polymer was measured by size exclusion HPLC. Alanine alone shows remarkable polymer formation, but shows that a formulation combining alanine with trehalose or sucrose is stable.
[図 2]図 2は各凍結乾燥製剤を 25°Cあるいは 40°Cで保存した時の重合体量の変化を 示す図である。重合体量はサイズ排除 HPLCにより測定した。ァラニンと組み合わせ るスクロースの比として Rが 0.25から 2.5の製剤が安定であることを示す。  [Fig. 2] Fig. 2 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C. The amount of polymer was measured by size exclusion HPLC. The ratio of sucrose in combination with alanine indicates that a formulation with R of 0.25 to 2.5 is stable.
[図 3]図 3は各凍結乾燥製剤を 25°Cあるいは 40°Cで保存した時の重合体量の変化を 示す図である。重合体量はサイズ排除 HPLCにより測定した。 Q (ァラニン質量に対す る抗体質量の比)が 0.05から 3の製剤が安定であることを示す。 [Fig. 3] Fig. 3 shows changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C. FIG. The amount of polymer was measured by size exclusion HPLC. This indicates that a preparation with a Q (ratio of antibody mass to alanine mass) of 0.05 to 3 is stable.
[図 4]図 4は各凍結乾燥製剤を 25°Cあるいは 40°Cで保存した時の重合体量の変化を 示す図である。重合体量はサイズ排除 HPLCにより測定した。抗 HLA-DR抗体及び抗 CD40抗体でァラニン及びスクロースを賦形剤とする凍結乾燥製剤が安定であること を示す。  [Fig. 4] Fig. 4 is a graph showing changes in the amount of polymer when each lyophilized preparation is stored at 25 ° C or 40 ° C. The amount of polymer was measured by size exclusion HPLC. It shows that lyophilized preparations with anti-HLA-DR antibody and anti-CD40 antibody using alanine and sucrose as excipients are stable.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0041] 本発明に関連した開示の説明 [0041] Description of Disclosure Related to the Invention
「安定な製剤」は、その製剤を投与される患者に対して毒性がある成分を含まず、ま た患者の生体恒常性をできる限り崩さない活性成分以外の添加物を加えることにより 、その中の活性成分が保存時において化学的及び/又は物理的及び/又は生物 学的安定性を保持するものである。「患者の生体恒常性をできる限り崩さない活性成 分以外の添加物」とは過去の治療実績により十分に安全性が確認されたもの、ある いは過去の投与実績がな!/、物質にお!/、ても細胞、動物への毒性評価ある!/、はその 他の方法により安全性が十分に予測されるものを言う。また「患者の生体恒常性を保 つ」とは、活性成分以外の添加剤が患者に許容できない生物活性を有さないこと、及 び/又は可能であれば等張 (ヒトの血液と本質的に同じ浸透圧を有していること)であ ることなどが含まれる。  A “stable formulation” does not contain ingredients that are toxic to the patient to whom the formulation is administered, and by adding additives other than active ingredients that do not disrupt the patient's biostasis as much as possible. These active ingredients retain chemical and / or physical and / or biological stability during storage. `` Additives other than active ingredients that do not disrupt the patient's homeostasis as much as possible '' are those whose safety has been sufficiently confirmed by past treatment results, or past administration results have not been! O! / Even if there is toxicity evaluation for cells and animals! /, Means that safety is sufficiently predicted by other methods. “Preserving patient homeostasis” means that additives other than the active ingredient have no biological activity unacceptable to the patient and / or isotonic (essentially with human blood). Have the same osmotic pressure).
[0042] 凍結乾燥 (lyophilization)は、医薬品の調製にお!/、てしばしば使用される凍結乾燥  [0042] Lyophilization is a lyophilization often used in the preparation of pharmaceuticals!
(Freeze drying)法である。液体組成物が調製され、次に凍結乾燥して乾燥したケー キ様の製品が形成される。この方法は一般に、すでに凍結しておいた試料を真空下 で乾燥して氷を除去し、粉末又はケーキ様物質の形で非水性成分をそのまま残す。 凍結乾燥された生成物は、生物活性が喪失することなく長期間かつ高温で保存する ことができ、適切な希釈剤を加えることにより粒状物を含まない溶液に容易に復元で きる。適切な希釈剤は、生物学的に許容される任意の液体であり、その中で凍結乾 燥粉末が完全に可溶性になる液体である。水、特に注射用水が好適な希釈剤であり 、これは、抗体の安定性に影響を与える塩又は他の物質を含まないからである。凍結 乾燥の利点は、種々の分子イベントが大幅に低下するレベルまで水分含量 (これは、 長期保存で製品を不安定化する)を低下させられることである。凍結乾燥製剤はまた 、輸送の物理的ストレスに容易に耐えることができる。復元した製品は、粒状物を含ま ず、従ってあらかじめ濾過することなぐ被験体に非経口的に、好ましくは静脈内又は 筋肉内又は皮下投与を行なうことができる。 (Freeze drying) method. A liquid composition is prepared and then lyophilized to form a dried cake-like product. This method generally dries an already frozen sample under vacuum to remove the ice, leaving the non-aqueous component in the form of a powder or cake-like material. The lyophilized product can be stored for extended periods of time and at elevated temperatures without loss of biological activity and can be easily reconstituted into a particulate-free solution by adding an appropriate diluent. A suitable diluent is any biologically acceptable liquid in which the lyophilized powder becomes completely soluble. Water, particularly water for injection, is a suitable diluent because it does not contain salts or other substances that affect antibody stability. The advantage of freeze-drying is the moisture content to a level where various molecular events are significantly reduced (this is The product becomes unstable during long-term storage). The lyophilized formulation can also easily withstand the physical stress of transport. The reconstituted product is free of particulate matter and can therefore be administered parenterally, preferably intravenously or intramuscularly or subcutaneously, to a subject without prior filtration.
[0043] 凍結乾燥製剤の重要な特徴は、製品の復元時間又は再水和するのに必要な時間 である。非常に早い完全な再水和を可能にするために、高多孔性の構造を有するケ ーキが重要である。ケーキ構造はタンパク質濃度、賦形剤の種類と濃度、及び凍結 乾燥サイクルのプロセスパラメータを含む多くのパラメータの関数である。一般に復元 時間は、タンパク質濃度が上昇すると上昇し、従って、短い復元時間は、高濃度凍結 乾燥抗体製剤の開発において重要な目標である。長い復元時間は、より濃縮された 溶液にタンパク質がより長時間暴露されるため、製品の品質を劣化させる。さらに使 用者の側からは、製品が完全に再水和されるまでに投与することができない。これは 、製品が粒状物を含まず、正しい用量が投与され、その無菌性が影響を受けないこと を確保するためである。すなわち、迅速な再水和は、患者と医師にとっても好都合で ある。 [0043] An important feature of a lyophilized formulation is the product reconstitution time or the time required to rehydrate. A cake with a highly porous structure is important in order to enable very fast complete rehydration. Cake structure is a function of many parameters, including protein concentration, excipient type and concentration, and process parameters of the lyophilization cycle. In general, reconstitution time increases with increasing protein concentration, so short reconstitution time is an important goal in the development of high concentration lyophilized antibody formulations. Longer reversion times degrade product quality because the protein is exposed to longer time in a more concentrated solution. Furthermore, from the user's side, it cannot be administered until the product is completely rehydrated. This is to ensure that the product is free of particulates, the correct dose is administered and its sterility is not affected. That is, rapid rehydration is also convenient for patients and physicians.
[0044] タンパク質の安定性は、様々な分析方法により測定される。例えば新'タンパク質精 製法理論と実際、 RKスコープス著、シュプリンガー'フエアラーク東京出版に概説され ている。タンパク質の安定性は、化学的安定性、物理的安定性及び生物的安定性を 含む。 「化学的安定性」はタンパク質の化学的に変化した状態を検出することにより 検定すること力 Sできる。化学的変化は例えばサイズ排除クロマトグラフや SDS-PAGE により評価でき得るクリッピングなどのサイズ修飾や、イオン交換クロマトグラフにより 評価でき得る電荷状態の変化(例えば脱アミドの結果生じる)及び疎水クロマトグラフ により評価でき得る親水性/疎水性状態の変化(例えば酸化の結果生じる)などを含 む。 「物理的安定性」は色及び/又は透明性の目視検査時及び/又はサイズ排除クロ マトグラフにより評価でき得る、不溶性微粒子及び/又は濁り及び/又は凝集体を生じ ないことを含む。「生物的安定性」は例えばサイズ排除クロマトグラフや ELISAなどに より評価可能な抗原への結合活性を検定することにより評価可能である。  [0044] Protein stability is measured by various analytical methods. For example, the new 'protein refinement theory and practice', as outlined by RK Scopes, Springer's “Fairark Tokyo Publishing”. Protein stability includes chemical stability, physical stability and biological stability. “Chemical stability” can be assayed by detecting the chemically altered state of the protein. Chemical changes can be assessed by size modification such as clipping, which can be assessed by size exclusion chromatography or SDS-PAGE, charge state changes that can be assessed by ion exchange chromatography (eg, as a result of deamidation), and hydrophobic chromatography. Includes possible changes in the hydrophilic / hydrophobic state (eg resulting from oxidation). “Physical stability” includes the absence of insoluble particulates and / or turbidity and / or agglomeration that can be assessed during visual inspection of color and / or transparency and / or by size exclusion chromatography. “Biological stability” can be evaluated by assaying the binding activity to an antigen that can be evaluated by, for example, size exclusion chromatography or ELISA.
[0045] 治療用途に有用なタンパク質には抗体が含まれる。抗体は細胞表面に発現するタ ンパク質に結合し、細胞に対して細胞死あるいは傷害性を誘導する作用を有する抗 体を癌などの治療に用いることが試みられている。現在、細胞膜上に存在するレセプ ターである CD20を標的としたキメラ抗体(Rituximab)、 Her2/neuを標的としたヒト化抗 体などのモノクローナル抗体力 S、悪性腫瘍を対象疾患として使用されており、その治 療効果が認められている。また国際公開第 WO2003/033538号パンフレットに公開さ れて!/、るようにクラス II主要組織適合遺伝子複合体(MHC)分子の一種である HLA— DR抗体に対するモノクローナル抗体(抗 HLA-DR抗体)も有用であると考えられる。 抗体は、血中半減期が長ぐ抗原への特異性が高いという特徴を持ち、抗腫瘍剤とし て特に有用である。例えば、腫瘍特異的な抗原を標的とした抗体であれば、投与し た抗体は腫瘍に集積することが推定されるので、捕体依存性細胞傷害性活性 (CDC )や抗体依存性細胞障害性活性 (ADCC)による、免疫システムの癌細胞に対する攻 撃が期待できる。また、その抗体に放射性核種や細胞毒性物質などの薬剤を結合し ておくことにより、結合した薬剤を効率よく腫瘍部位に送達することが可能となり、同 時に、非特異的な他組織への該薬剤到達量が減少することで、副作用の軽減も見 込むことができる。腫瘍特異的抗原に細胞死を誘導するような活性がある場合はァゴ 二スティック活性を持つ抗体を投与することで、また、腫瘍特異的な抗体の集積と、抗 体の活性による腫瘍の増殖停止又は退縮が期待される。抗体は、上記のようにその 特徴から抗腫瘍剤として適用するのに適切であると考えられる。 [0045] Proteins useful for therapeutic applications include antibodies. Antibodies are expressed on the cell surface. Attempts have been made to use antibodies that bind to proteins and have the effect of inducing cell death or damage to cells for the treatment of cancer and the like. Currently, it is used as a target disease for monoclonal antibodies such as chimeric antibodies (Rituximab) targeting CD20, a receptor on the cell membrane, humanized antibodies targeting Her2 / neu, and malignant tumors. The therapeutic effect is recognized. In addition, as disclosed in the pamphlet of International Publication No. WO2003 / 033538! /, A monoclonal antibody (anti-HLA-DR antibody) against an HLA-DR antibody, which is a type of class II major histocompatibility complex (MHC) molecule Is also considered useful. Antibodies are particularly useful as anti-tumor agents because of their high specificity for antigens with long blood half-lives. For example, in the case of an antibody that targets a tumor-specific antigen, the administered antibody is presumed to accumulate in the tumor, so that it depends on the capture-dependent cytotoxic activity (CDC) and antibody-dependent cytotoxicity. Attacks against cancer cells of the immune system can be expected by activity (ADCC). In addition, by binding a drug such as a radionuclide or a cytotoxic substance to the antibody, the bound drug can be efficiently delivered to the tumor site. Reducing side effects can be expected by reducing the amount of drug delivered. If a tumor-specific antigen has an activity that induces cell death, administration of an antibody having agonistic activity can be performed. Also, tumor-specific antibody accumulation and tumor growth due to antibody activity Stop or retraction is expected. The antibody is considered to be suitable for application as an antitumor agent due to its characteristics as described above.
[0046] 以上のように、治療用途に好適な多くの抗体が研究、開発、実用化され、医療現場 で頻繁に使用されるようになった現状においては、より安定性の優れた抗体を含有 する凍結乾燥製剤が当該分野で必要とされている。  [0046] As described above, many antibodies suitable for therapeutic use have been researched, developed and put into practical use, and in the current situation where they are frequently used in the medical field, they contain more stable antibodies. There is a need in the art for lyophilized formulations.
[0047] 発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
本特許に含まれる「凍結乾燥製剤」とは、医薬効果を有する抗体である活性成分が 凍結乾燥された形態であり、固体中において活性成分を明確に効果的であるものと する形態であり、製剤が投与される患者に対して生体恒常性を崩す可能性がある更 なる成分を含まないものをいう。ここで「活性成分を明確に効果的であるものとする」と は、含まれる活性成分がその活性を維持しており、医薬効果を失っていないことをい つ。 [0048] 「患者の生体恒常性をできる限り崩さな!/、活性成分以外の添加剤」とは過去の治療 実績により十分に安全性が確認されたもの、あるいは過去の投与実績が無!/、物質に おレ、ても細胞、動物への毒性評価あるいはその他方法により安全性が十分に予測さ れるものを言う。すなわち、本発明の凍結乾燥医薬製剤は、医薬効果を有する抗体 である活性成分と医薬的に許容できるその他の添加剤を含み得る。また「患者の生 体恒常性を保つ」とは、活性成分以外の添加剤が患者に許容できな!/、生物活性を 有さないこと、及び/又は可能であれば等張 (ヒトの血液と本質的に同じ浸透圧を有 して!/、ること)であることなどが含まれる。 The “lyophilized preparation” included in this patent is a form in which an active ingredient which is an antibody having a medicinal effect is lyophilized, and the form in which the active ingredient is clearly effective in a solid, It does not contain any additional ingredients that may disrupt biostasis to the patient to whom the product is administered. Here, “the active ingredient shall be clearly effective” means that the active ingredient contained therein maintains its activity and does not lose its pharmaceutical effect. [0048] “Does not disrupt the patient's homeostasis as much as possible! /, An additive other than the active ingredient” refers to those whose safety has been sufficiently confirmed by past treatment results, or no past administration results! It means substances whose safety can be sufficiently predicted by toxicity evaluation for cells, animals or other methods. That is, the lyophilized pharmaceutical preparation of the present invention may contain an active ingredient which is an antibody having a pharmaceutical effect and other pharmaceutically acceptable additives. In addition, “maintaining patient homeostasis” means that additives other than the active ingredient are not acceptable to the patient! /, Have no biological activity, and / or are isotonic if possible (human blood And have the same osmotic pressure! /).
[0049] 「安定な製剤」とはその中の活性成分が保存時において化学的及び/又は物理的 及び/又は生物学的安定性を保持するものである。好ましくは少なくとも低温(2°Cか ら 8°C)において 1年以上安定であり、また好ましくは 25°Cにおいて少なくとも 3箇月及 び/又は 40°Cにおいて少なくとも 1箇月間安定であり、製剤の凍結融解、光照射及 び振動に安定であることをいう。タンパク質の安定性は、様々な分析方法により測定 される。「化学的安定性」はタンパク質の化学的に変化した状態を検出し定量するこ とにより検定すること力 Sできる。化学的変化は例えばサイズ排除クロマトグラフや SDS- PAGEにより評価できるクリッピングなどのサイズ修飾、イオン交換クロマトグラフにより 評価できる電荷の変化(例えばデアミドの結果生じる)、及び疎水クロマトグラフにより 評価できる親水性/疎水性状態の変化(例えば酸化の結果生じる)などを含む。「物 理的安定性」は不溶性異物及び/又は凝集体を生じないことなどを含み、色及び/ 又は透明性の目視検査及び/又はサイズ排除クロマトグラフにより評価できる。そし て、「生物的安定性」は例えばサイズ排除クロマトグラフや ELISAなどにより評価可能 な抗原への結合活性を検定することにより評価可能である。抗体が化学的又は物理 的な変化を受けていない場合、その抗体は生物的安定性を保持している。従って、 製剤中の抗体が化学的安定性及び物理的安定性を保持している場合、その製剤は 安定であるといえる。すなわち製剤が安定であるかどうかは、含まれる抗体の化学的 及び物理的特性の変化の有無を測定することによりわかる。安定な製剤は、抗体の 重合体、分解物、デアミド体、酸化体等が保存中に製剤の医薬効果を減じるほど増 カロすること力 Sなく、また不溶性異物や濁りも認められない。本発明の安定な製剤は、 少なくとも低温 (2°Cから 8°C)において 1年以上保存しても、あるいは少なくとも 25°Cに おいて 3箇月間保存しても、あるいは 40°Cにおいて 1箇月間保存しても、抗体の重合 体、分解物、デアミド体、酸化体が製剤の医薬効果を減じるほど増加しない。また製 剤を凍結融解しても又は振動を与えても、抗体の重合体、分解物、デアミド体、酸化 体が製剤の医薬効果を減じるほど増加しない。本発明の安定な製剤中の抗体の重 合体は、保存中に増加することなぐ例えば 25°C又は 40°Cで 1箇月保存した後にサイ ズ排除クロマトグラフで測定した場合、製剤中の抗体に対する重合体の割合は小さ いことが望ましい。また、本発明の安定な製剤中の抗体の分解物は、保存中に大きく 増加することはなぐ例えば 25°C又は 40°Cで 1箇月保存しても、製剤中の抗体に対す る分解物の割合は小さいことが望ましい。また、本発明の安定な製剤中の抗体のデ アミド体の量は、保存中に大きく増加することはなぐ例えば 25°C又は 40°Cで 1箇月保 存しても、製剤中の抗体に対するデアミド体の割合は小さいことが望ましい。さらに、 本発明の安定な製剤中の抗体の酸化体は、保存中に大きく増加することはなぐ例 えば 25°C又は 40°Cで 1箇月保存しても、製剤中の抗体に対する酸化体の割合は小さ いことが望ましい。本発明の凍結乾燥製剤は、保存中に抗体に起因する重合体量が 増加することがないことを特徴とする。重合体含量及び分解物含量は、例えばサイズ 排除クロマトグラフィーにより測定でき、デアミド体含量は、例えばイオン交換クロマト グラフィ一により測定でき、酸化体含量は、例えば、疎水 HPLCにより測定することが できる。 [0049] A "stable formulation" is one in which the active ingredient therein retains chemical and / or physical and / or biological stability upon storage. Preferably stable for at least 1 year at low temperatures (2 ° C to 8 ° C) and preferably stable for at least 3 months at 25 ° C and / or at least 1 month at 40 ° C. Stable to freeze-thaw, light irradiation and vibration. Protein stability is measured by various analytical methods. “Chemical stability” can be assayed by detecting and quantifying the chemically altered state of the protein. Chemical changes include, for example, size modifications such as clipping that can be evaluated by size exclusion chromatography or SDS-PAGE, charge changes that can be evaluated by ion exchange chromatography (eg, as a result of deamide), and hydrophilicity / Changes in the hydrophobic state (eg resulting from oxidation) and the like. “Physical stability” includes the absence of insoluble foreign matter and / or aggregates, and can be evaluated by visual inspection of color and / or transparency and / or size exclusion chromatography. “Biological stability” can be evaluated by assaying the binding activity to an antigen that can be evaluated by, for example, size exclusion chromatography or ELISA. An antibody retains biological stability when it has not undergone chemical or physical changes. Therefore, if the antibody in the preparation retains chemical stability and physical stability, it can be said that the preparation is stable. In other words, whether the preparation is stable can be determined by measuring the presence or absence of changes in the chemical and physical properties of the contained antibody. Stable preparations do not have the ability to increase the amount of antibody polymer, degradation products, deamides, oxidants, etc. during storage so as to reduce the pharmaceutical effect of the preparation, and no insoluble foreign matter or turbidity is observed. The stable formulation of the present invention is Antibodies can be stored for at least 1 year at low temperatures (2 ° C to 8 ° C), or stored for at least 3 months at 25 ° C, or stored at 40 ° C for 1 month The polymers, degradation products, deamides, and oxidants do not increase so as to reduce the pharmaceutical effect of the preparation. Also, even if the product is freeze-thawed or subjected to vibration, the antibody polymer, degradation product, deamide, and oxidant do not increase so as to reduce the pharmaceutical effect of the preparation. The antibody polymer in the stable preparation of the present invention does not increase during storage, for example, when it is measured by size exclusion chromatography after being stored at 25 ° C or 40 ° C for one month, A small proportion of the polymer is desirable. In addition, the degradation product of the antibody in the stable preparation of the present invention does not increase greatly during storage, for example, the degradation product for the antibody in the formulation even if stored for 1 month at 25 ° C or 40 ° C. It is desirable that the ratio of is small. In addition, the amount of deamidated antibody in the stable preparation of the present invention does not increase greatly during storage, for example, even if stored at 25 ° C or 40 ° C for 1 month, It is desirable that the ratio of the deamide is small. Furthermore, the oxidized form of the antibody in the stable preparation of the present invention does not increase greatly during storage, for example, even if it is stored at 25 ° C or 40 ° C for 1 month, A small percentage is desirable. The freeze-dried preparation of the present invention is characterized in that the amount of the polymer due to the antibody does not increase during storage. The polymer content and the degradation product content can be measured by, for example, size exclusion chromatography, the deamide content can be measured by, for example, ion exchange chromatography, and the oxidant content can be measured, for example, by hydrophobic HPLC.
[0050] 本発明における抗体の「治療上有効な量」とは、治療に抗体が有効である疾患の 予防又は治療に有効な量を指す。「疾患」とは抗体での治療による利益がある任意 の症状である。これは、哺乳類の疾患の素因になる病理的状態を含む慢性及び急 性疾患又は疾病を含む。製剤中に存在する抗体の治療上有効な量は、例えば望ま しい用量及び投与形態を考慮に入れて決定される。約 lmg/mLから約 200mg/mL、好 ましくは約 5mg/mLから約 50mg/mL、最も好ましくは約 10mg/mL及び/又は約 20mg/ mLが製剤中の例示的な抗体濃度である。  [0050] The "therapeutically effective amount" of the antibody in the present invention refers to an amount effective for preventing or treating a disease for which the antibody is effective for treatment. “Disease” is any condition that would benefit from treatment with an antibody. This includes chronic and acute diseases or illnesses, including pathological conditions that predispose to mammalian diseases. The therapeutically effective amount of antibody present in the formulation is determined, for example, taking into account the desired dose and mode of administration. About 1 mg / mL to about 200 mg / mL, preferably about 5 mg / mL to about 50 mg / mL, most preferably about 10 mg / mL and / or about 20 mg / mL are exemplary antibody concentrations in the formulation.
[0051] 本特許に含まれる「抗体」とは最も広い意味において使用され、特にモノクローナル 抗体、ポリクローナル抗体、多重特異性抗体、又は所望の生物活性を保持する限り は抗体断片を含む。また、本特許に含まれる抗体は、所望の抗原と結合する限り特 に制限はなぐマウス抗体、ラット抗体、ゥサギ抗体、ヒッジ抗体、ラクダ抗体、ニヮトリ 抗体、キメラ抗体、ヒト化抗体、ヒト抗体等を適宜用いることができる。抗体はポリクロ ーナル抗体であってもモノクローナル抗体であってもよい。 [0051] The term "antibody" included in this patent is used in the broadest sense, and in particular as long as it retains a monoclonal antibody, a polyclonal antibody, a multispecific antibody, or a desired biological activity. Includes antibody fragments. The antibodies included in this patent are not particularly limited as long as they bind to a desired antigen. Mouse antibodies, rat antibodies, rabbit antibodies, Hedge antibodies, camel antibodies, chicken antibodies, chimeric antibodies, humanized antibodies, human antibodies, etc. Can be used as appropriate. The antibody may be a polyclonal antibody or a monoclonal antibody.
[0052] 本発明は抗体を含有する安定な凍結乾燥医薬製剤に関するものである。製剤中の 抗体は、抗体を産生するために該当分野で利用できる当業者に周知の技術を使用 して調製される。例示的なモノクローナル抗体の調製方法として、(1)免疫原として使 用する、生体高分子の精製及び/又は抗原タンパク質を細胞表面に過剰に発現し ている細胞の作成、(2)抗原を動物細胞に注射することにより免疫した後、血液を採 取しその抗体価を検定して脾臓等の摘出の時期を決定してから、抗体産生細胞を調 製する工程、(3)骨髄腫細胞(ミエローマ)の調製、(4)抗体産生細胞とミエローマと の細胞融合、(5)目的とする抗体を産生するハイプリドーマ群の選別、(6)単一細胞 クローンへの分割(クローニング)、(7)場合によってはモノクローナル抗体を大量に 製造するためのハイプリドーマの培養、又はハイプリドーマを移植した動物の飼育、( 8)場合によってはハイプリドーマ等の抗体産生細胞からヒトモノクローナル抗体をコ ードする遺伝子をクローユングし、適当なベクターに組み込んで、これを宿主 (例えば 哺乳類細胞細胞株、大腸菌、酵母細胞、昆虫細胞、植物細胞など)に導入し、遺伝 子組換え技術を用いて産生された組換え型抗体の調製、(9)このようにして得られた 抗体の精製、(10)このようにして製造されたモノクローナル抗体の生理活性及びその 認識特異性の検討、あるいは標識薬としての特性の検定、等が挙げられる。さらにポ リクローナル抗体についても同様に当業者に周知の方法により作製することができる [0052] The present invention relates to a stable lyophilized pharmaceutical preparation containing an antibody. The antibodies in the formulation are prepared using techniques well known to those skilled in the art that are available in the relevant field to produce antibodies. Exemplary monoclonal antibody preparation methods include: (1) purification of biopolymers used as immunogens and / or creation of cells overexpressing antigenic proteins on the cell surface; (2) antigens in animals After immunization by injection into cells, blood is collected and the antibody titer is assayed to determine the timing of removal of the spleen and the like, and then antibody-producing cells are prepared. (3) Myeloma cells ( (4) cell fusion between antibody-producing cells and myeloma, (5) selection of hyperidoma groups producing the desired antibody, (6) splitting into single cell clones (cloning), (7 ) In some cases, culture of high-pridoma to produce a large amount of monoclonal antibody, or breeding of animals transplanted with high-pridoma. (8) In some cases, human monoclonal antibody is obtained from antibody-producing cells such as hyperidoma. The gene to be coded is cloned, incorporated into an appropriate vector, and introduced into a host (eg, mammalian cell line, E. coli, yeast cell, insect cell, plant cell, etc.), and gene recombination technology is used. Preparation of the produced recombinant antibody, (9) Purification of the antibody thus obtained, (10) Examination of physiological activity and recognition specificity of the monoclonal antibody thus produced, or labeling agent And the like, and the like. Furthermore, polyclonal antibodies can also be prepared by methods well known to those skilled in the art.
[0053] 本特許に含まれる抗体には抗体の重鎖及び/又は軽鎖の各々のアミノ酸配列に おいて、 1又は数個のアミノ酸が欠失、置換若しくは付加されたアミノ酸配列を有する 重鎖及び/又は軽鎖からなる抗体も包含される。本発明の抗体のアミノ酸配列中へ の、前記のようなアミノ酸の部分的改変(欠失、置換、揷入、付加)は、そのアミノ酸配 歹 IJをコードする塩基配列を部分的に改変することにより導入することができる。この塩 基配列の部分的改変は、既知の部位特異的変異導入法(site specific mutagenesis) を用いて定法により導入することができる。 (Proc Natl Acad Sci USA., 1984 Vol81 : 56 62)。本特許に含まれる抗体には、いずれのィムノグロブリンクラス及びアイソタイプを 有する抗体をも包含する。抗体には、サブクラスを組み替えた改変抗体、又はさらに 重鎖定常域を改変した改変抗体をも含む。さらに、ョード、イットリウム、インジウム、テ クネチウム等の方夂 ft性核き O.W.Goding,Monoclonal Anibodies: rinciples and practi ce,1993 Academic Press)、緑膿菌毒素、ジフテリアトキシン、リシンのような細菌毒素 、及びメトトレキセート、マイトマイシン、カリキアマイシンなどの化学療法剤(D.J.King, Applications and bngineenng of Monoclonal AntiDoaies. , 1998 .j .Intenational Ltd.; M.L.Grossbard. , Monoclonal Antibody-BaseTherapy of Cancer., 1998 Marcel Dekker Inc)、さらに、 Maytansinoid等のプロドラッグ(Chari et al., Cancer Res. , 1992 Vol.52 : 1 27 ; Liu et al., Proc.Natl.Acad.Sci.USA, 1996 VoL93 : 8681)などを結合させることによ りガンなどの疾患の治療効果をさらに増強したものも包含する。 [0053] The antibody included in this patent has a heavy chain having an amino acid sequence in which one or several amino acids are deleted, substituted or added in the amino acid sequences of the heavy chain and / or light chain of the antibody. And / or an antibody consisting of a light chain is also included. Partial modification (deletion, substitution, insertion, addition) of the amino acid as described above into the amino acid sequence of the antibody of the present invention involves partial modification of the base sequence encoding the amino acid sequence IJ. Can be introduced. This partial modification of the base sequence is a known site-specific mutagenesis method. Can be introduced by a conventional method. (Proc Natl Acad Sci USA., 1984 Vol81: 56 62). The antibodies included in this patent include antibodies having any immunoglobulin class and isotype. The antibody includes a modified antibody in which a subclass is recombined, or a modified antibody in which a heavy chain constant region is further modified. In addition, OWGoding, Monoclonal Anibodies: rinciples and practices (1993 Academic Press), bacterial toxins such as Pseudomonas aeruginosa toxin, diphtheria toxin, ricin, and methotrexate, such as iodine, yttrium, indium, and technetium. , Mitomycin, calikiamycin and other chemotherapeutic agents (DJKing, Applications and bngineenng of Monoclonal AntiDoaies., 1998 .j .Intenational Ltd .; MLGrossbard., Monoclonal Antibody-Base Therapy of Cancer., 1998 Marcel Dekker Inc), Maytansinoid And other prodrugs (Chari et al., Cancer Res., 1992 Vol. 52: 1 27; Liu et al., Proc. Natl. Acad. Sci. USA, 1996 VoL93: 8681), etc. Those that further enhance the therapeutic effect of diseases such as cancer are also included.
[0054] また本特許に含まれる抗体には抗体の機能的断片も含まれる。この「機能的断片」 とは、抗体の一部分(部分断片)であって、抗体の抗原への作用を 1つ以上保持する ものを意味し、具体的には F (ab ' ) 、 Fab '、 Fab、 Fv、ジスルフイド結合 Fv、一本鎖 Fv ( scFv)、ダイアポディ、及びこれらの重合体等が挙げられる(D.J.King. Applications an a Engineering of Monoclonal Antibodies., 1998 T.J.International Ltd) 0 [0054] The antibodies included in this patent also include functional fragments of antibodies. The term “functional fragment” means a part (partial fragment) of an antibody that retains at least one action of the antibody on the antigen. Specifically, F (ab ′), Fab ′, Fab, Fv, disulfide binding Fv, single chain Fv (scFv), diapodi, and polymers thereof (DJKing. Applications an a Engineering of Monoclonal Antibodies., 1998 TJ International Ltd) 0
[0055] 本特許に含まれる抗体は、ポリクローナル抗体又は及びモノクローナル抗体であり 、好ましくはヒト抗体、ヒト型化抗体又はキメラ抗体である。該抗体は、好ましくは IgGで あり、好ましくは IgGのサブクラス力 IgGl、 IgG2、 IgG4のいずれかである。また IgGが、 定常領域のアミノ酸配列の一部に遺伝子改変により、アミノ酸の欠失、及び/又は、 置換、及び又は、揷入が行われた IgGであっても良い。  [0055] The antibody included in this patent is a polyclonal antibody or a monoclonal antibody, and is preferably a human antibody, a humanized antibody or a chimeric antibody. The antibody is preferably IgG, and is preferably any one of IgG subclass IgG1, IgG2, and IgG4. Further, IgG may be an IgG in which amino acid deletion and / or substitution and / or insertion has been performed on a part of the amino acid sequence of the constant region by genetic modification.
[0056] また、より好ましくは国際公開第 WO2003/033538号パンフレットに記載されている 抗 HLA- DRヒトモノクローナル抗体である HD3、 HD4、 HD6、 HD7、 HD8、 HD 10、 HD4 Gl、 HD4G2Ser、 HD4G4、 HD8G1、 HD8GlSer、 HD8G2、 HD8G2Ser、 HD8G4である 。このうち、 HD4、 HD6、 HD8及び HD 10を産生するハイブリドーマは、それぞれ FERM BP-7771、 FERM BP_7772、 FERM BP- 7773、 FERM BP-7774として、 2001年 10月 11 日付で独立行政法人 産業技術総合研究所特許生物寄託センター (日本国茨城県 つくば巿東 1丁目 1番地 1 中央第 6)に国際寄託されている。また、 HD3及び HD7を 産生するハイブリドーマは、それぞれ FERM BP-08534、 FERM BP-08536として、 200 3年 10月 31日付で同センターに国際寄託されている。さらに、本特許には CD40に対 するヒトモノクローナル抗体も含まれ、好ましくは国際公開第 WO2002/088186号パン フレットに記載されている抗 CD40アンタゴニスト抗体である KM281-1-10、 M281-2- 10-1-2、 M283-5, KM225- 2- 56、 KM292- 1- 24、 KM341_6_9、 4D11、 5H10、 11E1、 5G3、 3811、 3411、 3417、 F4_465、および抗 CD40ァゴニスト抗体である KM302_1、 M341- 1- 19、 KM643- 4- 11、 2053、 2105、 3821、 3822、 285、 110、 115、 F1- 102、 F2- 1 03、 F5_77、 F5-157である。このうちハイプリドーマクローン KM302_1、 KM281_1_10、 M281-2-10-1-2,は 2001年 5月 9日付でそそれぞれ FERM BP_7578、 FERM BP-75 79、 FERM BP-7580として、クローン KM341-1-19及び 4D11は 2001年 9月 27日付で それぞれ FERMBP-7759、 FERMBP-7758として、クローン 2105は 2002年 4月 17日付で FERMBP-8024として、独立行政法人産業技術総合研究所特許生物寄託センター ( 茨城県つくば巿東 1丁目 1番地 1中央第 6)にブタペスト条約に基づき国際寄託されて いる。さらに、 F2-103、 F5-77及び F5-157の重鎖及び軽鎖の可変領域を有するプラ スミドは 2001年 4月 19日付でそれぞれ ATCC PTA_3302(F2_103重鎖)、 ATCC PTA-3 303(F2-103軽鎖)、 ATCC PTA_3304(F5_77重鎖)、 ATCC PTA_3305(F5_77軽鎖)、 A TCC PTC-3306(F5_157重鎖)、及び ATCC PTA_3307(F5_157軽鎖)として、ハイブリ ドーマクローン F1-102及び F4-465は 2001年 4月 24日付でそれぞれ ATCC PTA-3337 及び ATCC PTA-3338として、 (American Type Culuture Collection, 10801 Universit y Blvd., Manassas, Virginia, USA) (アメリカ国立菌培養収集所、アメリカ合衆国 ヴァ 一ジニァ州 20110-2209 マナサス 10801 ュニバーシティブノレバード)にブダぺス ト条約に基づき国際寄託されている。本発明の凍結乾燥製剤は上記のいずれの抗 体を用いたときにも安定である。 [0056] Further, HD3, HD4, HD6, HD7, HD8, HD10, HD4Gl, HD4G2Ser, HD4G4, which are anti-HLA-DR human monoclonal antibodies described in WO 2003/033538, more preferably, HD8G1, HD8GlSer, HD8G2, HD8G2Ser, HD8G4. Of these, the hybridomas producing HD4, HD6, HD8 and HD10 are FERM BP-7771, FERM BP_7772, FERM BP-7773, and FERM BP-7774, respectively. Research Institute Patent Biological Deposit Center (Ibaraki, Japan) It is deposited internationally at Tsukuba Sakai Higashi 1-chome 1 1 Central 6). The hybridomas producing HD3 and HD7 have been deposited internationally as FERM BP-08534 and FERM BP-08536 on October 31, 2003, respectively. Furthermore, this patent also includes a human monoclonal antibody against CD40, preferably KM281-1-10, M281-2-10, which are anti-CD40 antagonist antibodies described in International Publication No. WO2002 / 088186 pamphlet. -1-2, M283-5, KM225-2-56, KM292- 1-24, KM341_6_9, 4D11, 5H10, 11E1, 5G3, 3811, 3411, 3417, F4_465, and KM302_1, M341-, which are anti-CD40 agonist antibodies 1-19, KM643-4-11, 2053, 2105, 3821, 3822, 285, 110, 115, F1-102, F2-103, F5_77, F5-157. Of these, Hypridoma clones KM302_1, KM281_1_10, and M281-2-10-1-2 were dated May 9, 2001 as clones KM341-1-19 as FERM BP_7578, FERM BP-75 79, and FERM BP-7580, respectively. And 4D11 dated September 27, 2001 as FERMBP-7759 and FERMBP-7758, respectively, and clone 2105 dated April 17, 2002 as FERMBP-8024. An international deposit has been made at Tsukuba Sakai Higashi 1-Chome 1-Chome 1-Chuo 6) under the Budapest Treaty. In addition, plasmids with variable regions of F2-103, F5-77 and F5-157 of heavy chain and light chain were ATCC PTA_3302 (F2_103 heavy chain) and ATCC PTA-3 303 (F2) as of April 19, 2001, respectively. -103 light chain), ATCC PTA_3304 (F5_77 heavy chain), ATCC PTA_3305 (F5_77 light chain), A TCC PTC-3306 (F5_157 heavy chain), and ATCC PTA_3307 (F5_157 light chain) as hybridoma clone F1-102 and F4-465 was dated April 24, 2001 as ATCC PTA-3337 and ATCC PTA-3338, respectively (American Type Culuture Collection, 10801 University Blvd., Manassas, Virginia, USA). Virginia 20110-2209 Manassas 10801 (University Benovard) has been deposited internationally under the Budapest Treaty. The lyophilized preparation of the present invention is stable when any of the above antibodies is used.
[0057] 本発明による凍結乾燥製剤は、有効成分として上記の抗体及び各種添加剤を含 む溶液製剤を調製し、その後この溶液を凍結乾燥することによって調製できる。  [0057] The lyophilized preparation according to the present invention can be prepared by preparing a solution preparation containing the above-described antibody and various additives as active ingredients, and then lyophilizing the solution.
[0058] 本発明の好まし!/、実施形態によれば、凍結乾燥製剤は、抗体の他にポリオール、 結晶性物質を含む、さらに緩衝剤及び界面活性剤を含む。 [0059] 「凍結乾燥」「凍結乾燥された(lyophilized)」及び「凍結乾燥された(Freeze-dried)」 は、乾燥すべき物質がまず凍結されて、次に氷又は凍結溶媒が真空環境中で昇華 されて除去される工程を意味する。保存中の凍結乾燥品の安定性を増強するために 、凍結乾燥前製剤中に賦形剤を含有しても良い。凍結乾燥工程は公知の方法で実 施すること力 Sでさる。 [0058] According to a preferred embodiment of the present invention, the lyophilized preparation contains, in addition to the antibody, a polyol, a crystalline substance, and further contains a buffer and a surfactant. [0059] "Freeze-dried", "lyophilized" and "Freeze-dried" means that the substance to be dried is first frozen and then the ice or freezing solvent is placed in a vacuum environment. It means the process of sublimation and removal. In order to enhance the stability of the lyophilized product during storage, an excipient may be contained in the preparation prior to lyophilization. The freeze-drying process is performed by a known method with a force S.
[0060] 本発明に含まれる「添加剤」とは活性成分以外の凍結乾燥製剤に含まれる全ての 成分を含み、例えば緩衝剤、 pH調整剤、等張化剤、賦形剤、安定化剤、界面活性剤 、防腐剤などが含まれる。また一種の添加剤成分において二種類以上の効果を示す ものも含まれる。  [0060] The "additive" included in the present invention includes all components included in the lyophilized preparation other than the active component, such as a buffer, a pH adjuster, an isotonic agent, an excipient, and a stabilizer. , Surfactants, preservatives and the like. In addition, one type of additive component that exhibits two or more types of effects is also included.
[0061] 「緩衝剤」は酸塩基共役成分の作用により pHの変化を穏やかにする添加剤を指す 。緩衝剤濃度はその緩衝能力及び/又は所望の浸透圧に応じて約 lmmol/Lから約 5 0mmo/L、好ましくは 5mmol/Lから 20mmol/L、最も好適なのは約 10mmol/Lである。好 ましい緩衝剤はグルタミン酸塩 (例えばグルタミン酸ナトリウム)やクェン酸及び他の 有機酸緩衝液が含まれる。最も好ましくはグルタミン酸塩である。  [0061] "Buffer" refers to an additive that moderates changes in pH by the action of an acid-base conjugate component. The buffer concentration is about 1 mmol / L to about 50 mmo / L, preferably 5 mmol / L to 20 mmol / L, and most preferably about 10 mmol / L, depending on its buffer capacity and / or the desired osmotic pressure. Preferred buffering agents include glutamate (eg, sodium glutamate), citrate and other organic acid buffers. Most preferred is glutamate.
[0062] 「ポリオール」は複数のヒドロキシル基を有する物質であり、凍結乾燥製剤中では賦 形剤としても使用される。糖 (還元及び非還元糖)、糖アルコール及び糖酸を含む。こ こで好適なポリオールは、約 600KD未満(例えば約 120KDから約 400KDの範囲)であ る分子量を有する。 「還元糖」は金属イオンを低減することができる力、、又はタンパク 質中のリジンと他のアミノ基と共有的に反応できるへミアセタール基を含むものであり 、「非還元糖」は、還元糖のこれらの性質を有さないものである。還元糖の例としては フノレクトース、マンノース、マノレトース、ラタトース、ァラビノース、キシロース、リボース、 ラムノース、ガラクトース及びグルコースである。非還元糖の例としてはスクロース、ト レノヽロース、ソルボース、メレチトース及びラフイノース等を含む。糖アルコールの例と してはマンニトール、キシリトーノレ、エリスリトーノレ、スレイトール、ソルビトール及びダリ セロース等である。糖酸は、 L-ダルコン酸とその金属塩が含まれる。凍結乾燥製剤へ の還元糖の使用は酸化体の生成を促進するため好ましくない。また、凍結、乾燥に 伴うストレスに対してタンパク質を保護する目的でポリオールを使用する場合には凍 結及び乾燥の際に非晶質 (アモルファス)構造をとるものが望ましい。凍結の際に非 晶質構造をとるポリオールの例示的なものとして、スクロース、トレハロース、ソルビト ールなどがある。本発明のポリオールは好ましくは非還元糖かつ非晶質のポリオール であり、又は非還元糖かつ非晶質の糖若しくは糖アルコール等のポリオールである。 より好ましくはスクロース、トレハロース、ソルボース、メレチトース及びラフィノースなど の非還元糖かつ非晶質のポリオールであり、最も好ましくはスクロース及びトレハロー スである。また、本発明の凍結乾燥製剤は複数のポリオールを含んでいてもよい。 [0062] "Polyol" is a substance having a plurality of hydroxyl groups, and is also used as an excipient in lyophilized preparations. Contains sugars (reducing and non-reducing sugars), sugar alcohols and sugar acids. Suitable polyols herein have a molecular weight that is less than about 600 KD (eg, in the range of about 120 KD to about 400 KD). “Reducing sugar” includes a force capable of reducing metal ions, or a hemiacetal group capable of reacting covalently with other amino groups and lysine in protein. “Non-reducing sugar” It does not have these properties of sugar. Examples of reducing sugars are funolectose, mannose, manoleose, ratatose, arabinose, xylose, ribose, rhamnose, galactose and glucose. Examples of non-reducing sugars include sucrose, trenosose, sorbose, meletitose, and raffinose. Examples of sugar alcohols include mannitol, xylitol, erythritol, thritol, sorbitol, and daricerose. Sugar acids include L-darconic acid and its metal salts. The use of reducing sugars in lyophilized formulations is not preferred because it promotes the formation of oxidants. In addition, when a polyol is used for the purpose of protecting a protein against stress caused by freezing and drying, it is desirable to use an amorphous structure during freezing and drying. Non-freezing Illustrative examples of polyols having a crystalline structure include sucrose, trehalose, sorbitol and the like. The polyol of the present invention is preferably a non-reducing sugar and amorphous polyol, or a polyol such as a non-reducing sugar and amorphous sugar or sugar alcohol. More preferred are non-reducing sugar and amorphous polyols such as sucrose, trehalose, sorbose, meretitol and raffinose, and most preferred are sucrose and trehalose. Moreover, the freeze-dried preparation of the present invention may contain a plurality of polyols.
[0063] 「結晶性物質」とは凍結の際に結晶構造をとるものであればよぐァラニン、グリシン 等の結晶性のアミノ酸等を含む。特に好ましくはァラニンであり、 5〜40mg/mL、好ま しくは 10〜30mg/mL、さらに好ましくは 15〜25mg/mL、特に好ましくは 20mg/mL含ま れる。 [0063] "Crystalline substance" includes crystalline amino acids such as alanine and glycine as long as they take a crystal structure upon freezing. Particularly preferred is alanine, which is contained in an amount of 5 to 40 mg / mL, preferably 10 to 30 mg / mL, more preferably 15 to 25 mg / mL, and particularly preferably 20 mg / mL.
[0064] ポリオール及び結晶性物質は賦形剤や等張化剤として作用し得る。本発明におい て「賦形剤」とは、凍結乾燥製剤のケーキ形状を維持する機能及び/又は凍結、乾 燥に伴うストレスに対してタンパク質を保護する機能をもつ物質をいう。ポリオール及 び結晶性物質の他、賦形剤のために使用される一般的な例には、乳糖がある。賦形 剤は長期保存時のタンパク質安定性を増強するという点で有用な性質を与える。ま た、これらの物質は等張化剤としても作用し得る。「等張化剤」は対象の製剤がヒトの 血液と本質的に同じ浸透圧を有するよう浸透圧を調整するものを指す。等張製剤は 約 250から 350mOsm/kgの浸透圧を有し、生理食塩水の浸透圧を 1とした浸透圧比は 約 1である事が望ましい。本発明において賦形剤として用いられるァラニンは、ケーキ 形状を維持し、ポリオールは、凍結、乾燥に伴うストレスに対してタンパク質を保護す  [0064] Polyols and crystalline substances can act as excipients and tonicity agents. In the present invention, the “excipient” refers to a substance having a function of maintaining the cake shape of a freeze-dried preparation and / or a function of protecting a protein against stress associated with freezing and drying. In addition to polyols and crystalline materials, a common example used for excipients is lactose. An excipient provides useful properties in that it enhances protein stability during long-term storage. These substances can also act as isotonic agents. An “isotonic agent” refers to an agent that adjusts the osmotic pressure so that the subject formulation has essentially the same osmotic pressure as human blood. It is desirable that the isotonic preparation has an osmotic pressure of about 250 to 350 mOsm / kg, and the osmotic pressure ratio is about 1, with the osmotic pressure of physiological saline being 1. The alanine used as an excipient in the present invention maintains the cake shape, and the polyol protects the protein against stress associated with freezing and drying.
[0065] 本発明において、スクロースのようなポリオールは、抗体凝集に対する安定化剤とし ても作用し、粒状物の無!、溶液に凍結乾燥製剤が復元する時間を短縮するのに重 要な役割も果たす。ポリオールは、製剤の所望の浸透圧に応じて変化する量で製剤 に加えられる。好ましくは復元後の凍結乾燥製剤は、等張であるが、高張又は低張 でもよい。 [0065] In the present invention, a polyol such as sucrose also acts as a stabilizer against antibody aggregation, and plays an important role in reducing the time required for reconstitution of a freeze-dried preparation in a solution without any particulate matter. Also fulfills. The polyol is added to the formulation in an amount that varies depending on the desired osmotic pressure of the formulation. Preferably, the reconstituted lyophilized formulation is isotonic but may be hypertonic or hypotonic.
[0066] 「界面活性剤」にはポリソルベート(例えばポリソルベート 20、 80など)又はポロキサ マー(例えばポロキサマー 188)のような非イオン性界面活性剤が含まれる。添加され る界面活性剤の量は、製剤化された抗体の凝集を低減し、及び/又は製剤中の粒子 の形成を最小化し、及び/又は容器へのタンパク質の吸着を低減するような量である 。本発明において界面活性剤として好ましくはポリソルベートを含み、最も好ましくは ポリソルベート 80を含む。界面活性剤は製剤中に、好ましくは 0.02mg/mLから O. lmg/ mL、最も好ましくは約 0.05mg/mLの量で存在しうる。 [0066] "Surfactants" include nonionic surfactants such as polysorbates (eg, polysorbate 20, 80, etc.) or poloxamers (eg, poloxamer 188). Added The amount of detergent is such that it reduces aggregation of the formulated antibody and / or minimizes the formation of particles in the formulation and / or reduces protein adsorption to the container. In the present invention, the surfactant preferably includes polysorbate, and most preferably includes polysorbate 80. The surfactant may be present in the formulation, preferably in an amount of 0.02 mg / mL to O. lmg / mL, most preferably about 0.05 mg / mL.
[0067] 凍結乾燥前組成物の例としては、約 lmg/mLから 60mg/mLの IgG抗体、約 lOmmol/ Lから 20mmol/Lのグルタミン酸ナトリウム(ρΗ5·5)、約 0.05mg/mLのポリソルベート 80、 及び賦形剤として 20mg/mLのァラニンと 5mg/mLから 50mg/mLのスクロースを含む製 剤である。上記凍結乾燥前製剤は凍結乾燥されて乾燥した安定な粉末となり、これ はヒトへの投与に適した粒状物を含まない溶液に容易に復元することができる。  [0067] Examples of pre-lyophilized compositions include about 1 mg / mL to 60 mg / mL IgG antibody, about 10 mmol / L to 20 mmol / L sodium glutamate (ρΗ5 · 5), about 0.05 mg / mL polysorbate 80 And 20 mg / mL alanine and 5 mg / mL to 50 mg / mL sucrose as excipients. The pre-lyophilized formulation is lyophilized to a dry, stable powder that can be easily reconstituted into a particulate-free solution suitable for human administration.
[0068] 本発明は抗体に結晶性物質 (最も好ましくはァラニン)と非還元糖及び非晶質のポ リオール (最も好ましくはスクロース及びトレハロース)を賦形剤として添加し、凍結乾 燥することで安定な凍結乾燥製剤を提供する。実質的には、結晶性物質からなる結 晶相でケーキ構造を維持し、抗体と非還元糖及び/又は非晶質のポリオール (最も 好ましくはスクロース及びトレハロース)からなるアモルファス相によって凍結及び乾 燥中のストレスからタンパク質を保護することで、安定な凍結乾燥製剤を導くことがで きる。  [0068] In the present invention, a crystalline substance (most preferably alanine), a non-reducing sugar and an amorphous polyol (most preferably sucrose and trehalose) are added to an antibody as excipients and freeze-dried. A stable lyophilized formulation is provided. In essence, the cake structure is maintained in a crystalline phase composed of a crystalline substance, and frozen and dried by an amorphous phase composed of antibodies and non-reducing sugars and / or amorphous polyols (most preferably sucrose and trehalose). By protecting the protein from moderate stress, a stable lyophilized formulation can be derived.
[0069] 本発明に用いられる賦形剤のァラニンとスクロースの配合比及び配合量は活性成 分である抗体濃度に応じ適宜調整される。  [0069] The blending ratio and blending amount of the alanine and sucrose excipients used in the present invention are appropriately adjusted according to the antibody concentration which is the active ingredient.
[0070] Rを凍結乾燥製剤中に存在するスクロースとァラニンの質量比(スクロース質量/ァ ラニン質量)又は濃度比 (スクロース濃度/ァラニン濃度)とした時、安定性が良好で ある Rは 0.25以上であり、好ましくは 0.5から 2.5であり、より好ましくは 1から 2であった。 最も好ましくは 1.5である。 [0070] When R is the mass ratio of sucrose to alanine (sucrose mass / alanine mass) or concentration ratio (sucrose concentration / alanine concentration) present in the lyophilized preparation, stability is good R is 0.25 or more Preferably 0.5 to 2.5, more preferably 1 to 2. Most preferably 1.5.
[0071] また Qを凍結乾燥製剤中に存在する抗体とァラニンの質量比 (抗体質量/ァラニン 質量)又は濃度比(抗体濃度/ァラニン濃度)とした時、 R=l.5の比率でスクロース/ ァラニンを含有する凍結乾燥製剤で安定性が良好である Qは 0.05から 3、好ましくは 0[0071] Further, when Q is the mass ratio of antibody to alanine (antibody mass / alanine mass) or concentration ratio (antibody concentration / alanine concentration) present in the lyophilized preparation, sucrose / Good stability with freeze-dried formulation containing alanine Q between 0.05 and 3, preferably 0
• 05カゝら 1.5である。 • 05 and 1.5.
[0072] 上述の Rと Qの組合せより、抗体、ァラニン及びスクロースからなる安定な凍結乾燥 性剤は、抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0.3〜20 : 0.5〜30である。また、抗体に対するァラニンとスクロースの合計質量の比は、 1 : 5/6 〜50である。 [0072] From the combination of R and Q described above, stable lyophilization comprising antibody, alanine and sucrose In the sexing agent, the ratio of the mass of the three components of antibody mass, alanine mass, and sucrose mass is 1: 0.3 to 20: 0.5 to 30. The ratio of the total mass of alanine and sucrose to the antibody is 1: 5 / 6-50.
[0073] 本発明の凍結乾燥製剤は、復元して用いられる。  [0073] The freeze-dried preparation of the present invention is used after being restored.
[0074] 「復元」とは、凍結乾燥製剤を溶液で再水和して、粒子の無!/、清澄化溶液にするこ とで「復元時間」はそれに必要な時間である。凍結乾燥製剤の重要な特徴は、製品 の復元時間又は再水和するのに必要な時間であり、短い時間が要求される。非常に 速い完全な再水和を可能にするために、高多孔性の構造を有するケーキが重要で ある。ケーキ構造は、タンパク質濃度、賦形剤の種類と濃度、及び凍結乾燥サイクル のプロセスパラメータを含む多くのパラメータの関数である。一般に復元時間は、タン パク質濃度が上昇すると上昇する。従って、復元時間が長い場合は、より濃縮された 溶液にタンパク質がより長時間暴露されるため、製品の品質を劣化させる。さらに使 用者の側からは、製品が完全に再水和されるまで投与することができない。これは、 完全に再水和させることにより、製品が粒状物を含まず、正しい用量が投与され、そ の無菌性が影響を受けていないことを確保するためである。すなわち、迅速な再水和 は、患者と医者にとって好都合である。  [0074] "Restoration" is the time required for rehydration of a lyophilized preparation with a solution to obtain a particle-free / clarified solution. An important feature of lyophilized formulations is the product reconstitution time or the time required to rehydrate, requiring a short time. A cake with a highly porous structure is important in order to allow very fast complete rehydration. Cake structure is a function of many parameters, including protein concentration, excipient type and concentration, and process parameters of the lyophilization cycle. In general, the recovery time increases with increasing protein concentration. Therefore, longer reversion times degrade product quality because proteins are exposed to more concentrated solutions for longer periods of time. In addition, the user cannot administer until the product is completely rehydrated. This is to ensure complete rehydration to ensure that the product is free of particulates, is administered at the correct dose, and its sterility is not affected. That is, rapid rehydration is advantageous for patients and physicians.
[0075] 溶液製剤は、適切な乾燥パラメータを使用して凍結乾燥することができる。凍結乾 燥製剤は、抗体の生物活性の安定性を保持し、ヒト被験体への投与を目的とした抗 体を、最終製品中での物理的及び化学的分解から守る。  [0075] Solution formulations can be lyophilized using appropriate drying parameters. The lyophilized formulation retains the stability of the biological activity of the antibody and protects the antibody intended for administration to human subjects from physical and chemical degradation in the final product.
[0076] 凍結乾燥製剤は、使用時に希釈剤(例えば注射用水)で再水和されて、粒状物を 含まない溶液を与える。復元した抗体溶液は、周囲温度で凍結乾燥ケーキを長期保 存した後も粒状物を含まない。復元した溶液は、被験体に非経口的に、好ましくは静 脈内又は筋肉内又は皮下投与される。  [0076] The lyophilized formulation is rehydrated at the time of use with a diluent (eg, water for injection) to give a solution free of particulate matter. The reconstituted antibody solution does not contain particulate matter after long-term storage of the lyophilized cake at ambient temperature. The reconstituted solution is administered parenterally, preferably intravenously, intramuscularly or subcutaneously to the subject.
[0077] 本発明の組成物は、免疫グロブリンを含有する試薬中のタンパク質凝集物と粒状 物の形成を最小にし、時間が経っても溶液中の抗体がその免疫活性を維持すること を確保する。組成物は、中性又は酸性 pHを有する緩衝液中に抗体、ァラニン、及び ポリオール及び界面活性剤、を含む、水性の凍結乾燥前製剤から調製される無菌の 薬剤学的に許容される凍結乾燥製剤を含む。本発明の組成物はさらに、ァラニン及 びポリオール以外の賦形剤及び/又は等張化剤を含有してもよい。 [0077] The composition of the present invention minimizes the formation of protein aggregates and particulates in an immunoglobulin-containing reagent and ensures that the antibody in solution maintains its immune activity over time. . The composition is a sterile, pharmaceutically acceptable lyophilized solution prepared from an aqueous pre-lyophilized formulation containing antibodies, alanine, and polyol and surfactant in a buffer having a neutral or acidic pH. Contains formulation. The composition of the present invention further comprises alanine and And excipients other than polyols and / or tonicity agents.
[0078] 製剤は、抗体での治療を必要としている哺乳動物、好ましくはヒトに、既知の方法、 例えばボーラスとしてもしくは一定時間にわたる連続注入による静脈内投与、筋肉内 、腹腔内、大脳内、皮下、関節内、滑液包内、くも膜下腔内、経口、局所的、又は吸 入経路により、投与される。好適な実施様態では、製剤は静脈内投与により哺乳動 物に投与される。このような目的には、製剤は、例えばシリンジを使用して、又は点滴 静注ラインを介して注入される。 [0078] The formulation is administered to mammals, preferably humans, in need of treatment with antibodies, in a known manner, for example, intravenously administered as a bolus or by continuous infusion over time, intramuscular, intraperitoneal, intracerebral, subcutaneous. Administered by intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes. In a preferred embodiment, the formulation is administered to the mammal by intravenous administration. For such purposes, the formulation is infused, for example, using a syringe or via an intravenous infusion line.
実施例  Example
[0079] 本発明を以下の実施例によってさらに詳しく説明する。本発明は次の実施例を参 照すれば十分に理解されるであろう。しかし、これら実施例は、発明の範囲を制限す るものではない。本発明の記載に基づき種々の変更、修飾が当業者には可能であり 、これらの変更、修飾も本発明に含まれる。  [0079] The present invention is further illustrated by the following examples. The invention will be more fully understood with reference to the following examples. However, these examples do not limit the scope of the invention. Various changes and modifications can be made by those skilled in the art based on the description of the present invention, and these changes and modifications are also included in the present invention.
[0080] 実施例 1 ァラニンと各種賦形剤の組み合わせ  Example 1 Combination of alanine and various excipients
抗体として国際公開第 WO2003/033538号パンフレットに記載されている HLA-DR に対する抗体 (抗 HLA-DR抗体、 IgGl)用い、凍結乾燥製剤を調製した。  A freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in International Publication No. WO2003 / 033538 as an antibody.
[0081] 本実施例では表 1に示した製剤を調製し、ァラニンと組み合わせる好適な賦形剤の 詳細な検討を行った。  [0081] In this example, the formulations shown in Table 1 were prepared, and detailed examination of suitable excipients to be combined with alanine was conducted.
[表 1]  [table 1]
本実施 ί Ιに供した製剤一1 £
Figure imgf000019_0001
This implementation ί One preparation for 製 剤1 £
Figure imgf000019_0001
[0082] (1)製剤検体の調製 [0082] (1) Preparation of preparation specimen
本検討に使用した試薬は、抗 HLA-DR抗体 (約 20mg/mL、国際公開第 W02003/00 33538号パンフレットに記載されている方法に従ってキリンビール社医薬生産技術研 究所 (2007年 7月 1日以降は、キリンファーマ株式会社生産技術研究所)にて調製)、 L-グルタミン酸ナトリウム一水和物(日本薬局方外医薬品規格)、ァラニン (Ala、 日本 薬局方)、トレハロース(Tor、 日本薬局方)、スクロース(Suc、 日本薬局方)、塩化ナト リウム(NaCl、 日本薬局方)、塩酸(日本薬局方)、ポリソルベート 80 (日本薬局方)、注 射用水(日本薬局方)を用いた。各製剤検体はあらかじめ原薬を含まなレ、プラセボ溶 液を作製し、抗 HLA-DR抗体溶液を NAPカラム(フアルマシアバイオテク社製)を用い て製剤プラセボ溶液と置換することにより調製した。またタンパク濃度は OD280匪吸 光度係数 ε = 1.4を用い換算し濃度を調整した。 The reagent used in this study was an anti-HLA-DR antibody (approximately 20 mg / mL, Kirin Brewery's Pharmaceutical Production Technology Laboratory (July 2007 1) according to the method described in International Publication No. W02003 / 00 33538 pamphlet. After that, prepared by Kirin Pharma Co., Ltd. Production Technology Laboratory), sodium L-glutamate monohydrate (Japanese Pharmacopoeia Standard), alanine (Ala, Japanese Pharmacopoeia), trehalose (Tor, Japan Pharmacy) ), Sucrose (Suc, Japanese Pharmacopoeia), sodium chloride Lithium (NaCl, Japanese Pharmacopoeia), hydrochloric acid (Japanese Pharmacopoeia), polysorbate 80 (Japanese Pharmacopoeia), and water for injection (Japanese Pharmacopoeia) were used. Each preparation specimen was prepared by preparing a placebo solution containing no drug substance in advance and replacing the anti-HLA-DR antibody solution with the preparation placebo solution using a NAP column (manufactured by Pharmacia Biotech). The protein concentration was converted using the OD280 匪 absorbance coefficient ε = 1.4 and adjusted.
[0083] 各製剤検体はクリーンベンチ内で 0.22 mフィルター(ミリポア社製)を用い無菌濾 過を行い、 5mLガラスバイアル(日本薬局方に適合)に lmLずつ充填を行った。充填 後、ゴム栓で半打栓を行!/ \ 日本真空技術社製の凍結乾燥機を用レ、て凍結乾燥を 行った。また分析検体の希釈及び分析のブランクのための抗 HLA-DR抗体を含まな V、溶液(プラセボ)をクリーンベンチ内で 0.22 μ mボトルトップフィルター(ナルゲン社) を用い無菌濾過を行った。  [0083] Each preparation specimen was aseptically filtered using a 0.22 m filter (Millipore) in a clean bench, and filled in 5 mL glass vials (conforming to the Japanese Pharmacopoeia). After filling, half-plugging was performed with a rubber plug! / \ Freeze-drying was performed using a freeze dryer manufactured by Nippon Vacuum Technology Co., Ltd. Aseptic dilution of V and solution (placebo) containing anti-HLA-DR antibody for dilution of analysis specimen and analysis blank was performed using 0.22 μm bottle top filter (Nalgen) in a clean bench.
[0084] (2)試験条件  [0084] (2) Test conditions
本実施例において安定性評価を行うため、以下の条件に従い各製剤検体にストレ スを与えた。  In order to evaluate the stability in this example, each preparation specimen was stressed according to the following conditions.
[0085] 熱安定性試験: 40°C又は 25°Cに制御されたインキュベータ(TABAI ESPEC社製)に 1 箇月間及び 3箇月間保存した。また各検体は熱ストレス負荷後分析開始まで 4°Cに制 御された低温庫に保存した。  [0085] Thermal stability test: Stored in an incubator (manufactured by TABAI ESPEC) controlled at 40 ° C or 25 ° C for 1 month and 3 months. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after heat stress.
[0086] (3)分析方法 [0086] (3) Analysis method
外観目視:蛍光灯下、肉眼でケーキ形状を調べ、外観図を記述した。  Visual appearance: The shape of the cake was examined with the naked eye under a fluorescent lamp, and the appearance was described.
[0087] 以下の分析については、外観目視後、シリンジで注射用水を lmL加え再溶解し、 分析を行った。 [0087] For the following analysis, after visual inspection, 1 mL of water for injection was added with a syringe and dissolved again.
[0088] 不溶性異物及び濁り:白色光源の直下、約 5000ルクスの明るさの位置で、肉眼で不 溶性異物及び濁りの有無を調べた。  [0088] Insoluble foreign matter and turbidity: The presence of insoluble foreign matter and turbidity was examined with the naked eye at a position of about 5,000 lux directly under a white light source.
[0089] サイズ排除 HPLC検定 (SEC):重合体含量及び分解物含量はサイズ排除高速液体ク 口マトグラフ法により算出した。必要に応じて検体を lmg/mLに希釈し 20 Lの注入を 雰囲気温度で行った。分離は TSKgel G3000 SWXL 30cm X 7.8mm (東ソ一社製)カラ ムを使用し、移動相としては 20mmol/Lリン酸ナトリウム、 500mmol/L塩化ナトリウム pH 7.0を用い 0.5mL/分の流量で分析時間は 30分、検出を 215nmで行った。なお主ピー クより前に溶出するものを重合体、後に溶出するものを分解物と定義した。 [0089] Size exclusion HPLC assay (SEC): The polymer content and degradation product content were calculated by the size exclusion high-speed liquid chromatography method. Samples were diluted to lmg / mL as necessary and 20 L injections were performed at ambient temperature. For separation, use TSKgel G3000 SWXL 30cm X 7.8mm (manufactured by Tosohichi Co., Ltd.) column, and use 20mmol / L sodium phosphate and 500mmol / L sodium chloride pH 7.0 as mobile phase and analyze at 0.5mL / min flow rate. The time was 30 minutes and detection was performed at 215 nm. The main P The polymer eluted before the catalyst was defined as the polymer, and the polymer eluted after the product was defined as the decomposition product.
[0090] 陽イオン交換 HPLC検定 (IEC):デアミド体含量は陽イオン交換高速液体クロマトダラ フ法により算出した。 5mg/mLに適宜希釈を行った検体を 60 じ注入した。分離カラム として TSKgel BIOAssist S (東ソ一社製)を用い 280nmで検出を行った。移動相として Aに対して 20mM酒石酸 pH4.5、 Bに対して 20mM酒石酸、 1M塩化ナトリウム pH4.5を用 い、最適なグラジェント条件を用い分析を行った。なお、主ピークより前方に溶出した 劣化物をデアミド体と定義した。  [0090] Cation exchange HPLC assay (IEC): Deamide content was calculated by the cation exchange high performance liquid chromatographic method. Sixty specimens diluted appropriately to 5 mg / mL were injected. Detection was performed at 280 nm using TSKgel BIOAssist S (manufactured by Tosoh Corporation) as a separation column. As mobile phase, 20 mM tartaric acid pH 4.5 for A and 20 mM tartaric acid, 1 M sodium chloride pH 4.5 for B were used, and the analysis was performed using the optimum gradient conditions. Degraded substances that eluted ahead of the main peak were defined as deamides.
[0091] 疎水 HPLC検定 (HIC):酸化体の含量は疎水クロマトグラフ法により算出した。検体処 理液(20mmol/Lリン酸ナトリウム、 0.984mol/L硫酸アンモニゥム、 7.5mmol/Lメチォ二 ン)を用いて lmg/mLに適宜希釈を行った検体を 20 じ注入した。分離カラムとして TS gel Buty卜 NPR (東ソ一社製)を用いて 215nmで分析及び検出を行った。移動相は A として 20mmol/Lリン酸ナトリウム、 ρΗ6·5を用い、 Βとして 20mmol/Lリン酸ナトリウム、 1 .2mol/L硫酸アンモニゥム、 ρΗ6·5を用い、最適なグラジェント条件を用い分析を行つ た。なお主ピークより前方に溶出した劣化物を酸化体と定義した。  [0091] Hydrophobic HPLC assay (HIC): The content of oxidant was calculated by hydrophobic chromatography. Twenty samples, each appropriately diluted to 1 mg / mL, were injected using a sample processing solution (20 mmol / L sodium phosphate, 0.984 mol / L ammonium sulfate, 7.5 mmol / L methanoline). Analysis and detection were performed at 215 nm using TS gel Buty NPR (manufactured by Tosoh Corporation) as a separation column. The mobile phase is 20 mmol / L sodium phosphate, ρΗ6 · 5 as A, and 20 mmol / L sodium phosphate, 1.2 mol / L ammonium sulfate, ρΗ6.5 as Β, and the analysis is performed using the optimal gradient conditions. I went. In addition, the deteriorated substance eluted ahead of the main peak was defined as an oxidant.
[0092] 還元 SDS-PAGE検定:必要に応じて検体溶液を 400 g/mLに希釈した。検体溶液に ¾DS Sample Buffer (Invitrogen社製)、 sample Reducing Agent (Invitrogen¾¾)、 H Oを 加え、 95°Cで 5分間加熱し、還元検体溶液とした。泳動糟に電気泳動用の Tris-Glyci ne SDS Running buffer (Invitrogen社製)を満たした。試料溶液 5 Lを 4-20%Tris_Gly cine Gel (Invitrogen社製)にアプライし、電気泳動を約 125V定電圧で、試料溶液に含 まれるブロモフエノールブルーの青色がゲルの下端付近に移動するまで行った。泳 動終了後のゲルを銀染色し検出した。なお検体及び劣化体のおおよその分子量を 判定するために分子量マーカー(97KDa、 66KDa、 45KDa、 30KDa、 20KDa、 14 Da のタンパク質を含む)を同時に泳動する。  [0092] Reduction SDS-PAGE assay: The sample solution was diluted to 400 g / mL as necessary. ¾DS Sample Buffer (Invitrogen), sample Reducing Agent (Invitrogen¾¾), and H 2 O were added to the sample solution and heated at 95 ° C for 5 minutes to obtain a reduced sample solution. The electrophoresis basket was filled with Tris-Glycine SDS Running buffer (manufactured by Invitrogen) for electrophoresis. Apply 5 L of the sample solution to 4-20% Tris_Glycine Gel (Invitrogen) and perform electrophoresis at a constant voltage of about 125 V until the blue color of bromophenol blue contained in the sample solution moves near the bottom of the gel. went. The gel after swimming was detected by silver staining. In addition, in order to determine the approximate molecular weight of the specimen and the degraded substance, molecular weight markers (including 97 KDa, 66 KDa, 45 KDa, 30 KDa, 20 KDa, and 14 Da proteins) are run simultaneously.
[0093] 非還元 SDS-PAGE検定:必要に応じて検体溶液を 400 μ g/mLに希釈した。検体溶液 に LDS Sample Buffer (Invitrogen社製)、 H 0を加え、非還元検体溶液とした。泳動糟 に電気泳動用の Tris-Acetate SDS Running buffer (Invitrogen社製)を満たした。試料 溶液 5 Lを 3-8%Tris_Acetate Gel (Invitrogen社製)にアプライし、電気泳動を約 125 V定電圧で、試料溶液に含まれるブロモフエノールブルーの青色がゲルの下端付近 に移動するまで行った。泳動終了後のゲルを銀染色し検出した。なお検体及び劣化 体のおおよその分子量を判定するために分子量マーカー(220KDa、 116KDa、 97.4 Da、 63.3KDa、 54.4 Da, 36.5KDaのタンパク質を含む)を同時に泳動する。 [0093] Non-reducing SDS-PAGE assay: The sample solution was diluted to 400 μg / mL as necessary. LDS Sample Buffer (Invitrogen) and H0 were added to the sample solution to obtain a non-reduced sample solution. The electrophoresis gel was filled with Tris-Acetate SDS Running buffer (Invitrogen) for electrophoresis. Apply 5 L of the sample solution to 3-8% Tris_Acetate Gel (Invitrogen), perform electrophoresis at a constant voltage of about 125 V, and the blue color of bromophenol blue in the sample solution is near the bottom of the gel. Went to move on. The gel after electrophoresis was detected by silver staining. In addition, in order to determine the approximate molecular weight of the specimen and the degraded product, molecular weight markers (including 220 KDa, 116 KDa, 97.4 Da, 63.3 KDa, 54.4 Da, and 36.5 KDa proteins) are run simultaneously.
[0094] 浸透圧比:自動浸透圧測定装置(アークレー社製 OSMO STATION OM-6050)を用 V、て浸透圧測定を行い、同時に測定した生理食塩水の浸透圧に対する比を計算し た。 pH :自動 pH測定装置 (メトラー 'トレド社製 MP-230など)を用いて pH測定を行った 。測定開始時に pH4、 pH7、 pH9の標準溶液を用い校正を行った後測定を行った。  [0094] Osmotic pressure ratio: V was measured using an automatic osmotic pressure measuring apparatus (OSMO STATION OM-6050 manufactured by ARKRAY, Inc.), and the ratio of the measured physiological saline to the osmotic pressure was calculated. pH: pH was measured using an automatic pH measuring device (Metler 'MP-230 manufactured by Toledo Co., Ltd.). Measurement was performed after calibration using standard solutions of pH 4, pH 7, and pH 9 at the start of measurement.
[0095] 水分測定:自動水分気化装置(平沼産業社製 LE-20SA)を接続した微量水分測定 装置(平沼産業社製 AQ-2000)を用いて水分量測定を行い、含湿度の計算した。  [0095] Moisture measurement: The moisture content was measured using a trace moisture analyzer (AQ-2000, Hiranuma Sangyo Co., Ltd.) connected to an automatic moisture vaporizer (LE-20SA, Hiranuma Sangyo Co., Ltd.), and the moisture content was calculated.
[0096] (4)結果及び考察  [0096] (4) Results and discussion
図 1は各凍結乾燥製剤を 25°C及び 40°Cで保存した時の重合体の変化を示した。重 合体量はサイズ排除クロマトグラフにより測定した。ァラニンのみを賦形剤とした製剤 では 40°C 1箇月で約 5%、 40°C3箇月で約 9%の重合体増加が確認された。ァラニンと 塩化ナトリウムを賦形剤とした製剤では、再溶解後の肉眼による目視検査で異物、濁 りが確認され、重合体の増加も大きく不安定であった。トレハロース、スクロースと組み 合わせた製剤では 40°C3箇月保存においても重合体の増加は確認されず非常に安 定であった。ァラニンに上記で用いたポリオ一ノレ(トレハロース、スクロース)を糸且み合 わせることで劇的に重合体を抑制することが明らかとなり、安定性が高いことが示され た。また、その他の分析項目(ケーキ形状、異物、濁り、分解物、不溶性微粒子、デァ ミド体、酸化体)においてもァラニンとポリオールを組み合わせた製剤は熱ストレス負 荷前後でほとんど変化は認められず、安定であった。  Figure 1 shows the change in polymer when each lyophilized formulation was stored at 25 ° C and 40 ° C. The amount of polymer was measured by size exclusion chromatography. In the formulation using only alanine as an excipient, a polymer increase of about 5% at 40 ° C for 1 month and about 9% at 40 ° C for 3 months was confirmed. In the formulation using alanine and sodium chloride as excipient, foreign matter and turbidity were confirmed by visual inspection with the naked eye after redissolving, and the increase in polymer was also largely unstable. The formulation combined with trehalose and sucrose was very stable with no increase in polymer even after storage at 40 ° C for 3 months. It was revealed that the polymer was dramatically suppressed by stringing polanolin (trehalose, sucrose) used above with alanine, indicating a high stability. In addition, in other analysis items (cake shape, foreign matter, turbidity, decomposed product, insoluble fine particles, amide, and oxidant), a combination of alanine and polyol shows almost no change before and after thermal stress loading. It was stable.
[0097] 本実施例より抗体を安定化させる凍結乾燥製剤の賦形剤としてァラニンとポリオ一 ルの組み合わせが好適であることが判明した。好ましくは非晶質のポリオール、さらに 好ましくは非還元かつ非晶質のポリオール、最も好ましくはポリオールとしてトレハロ ース、スクロースを用いることが望ましい。  [0097] From this example, it was found that a combination of alanine and polyol was suitable as an excipient of a lyophilized preparation for stabilizing an antibody. It is desirable to use an amorphous polyol, more preferably a non-reducing and amorphous polyol, and most preferably, trehalose or sucrose as the polyol.
[0098] 実施例 2 スクロースーァラニン比  [0098] Example 2 Sucrose-alanin ratio
実施例 1でァラニンと組み合わせるポリオールとしてトレハロース又はスクロースが 最も好ましいことが明らかとなった。 [0099] 本実施例ではポリオールとしてスクロースを用い、ァラニン濃度を一定とし、ァラニン と混合するスクロース濃度を変化させ、重合体の抑制に最適な濃度比率 Rの検討を 行った。ここで Rは凍結乾燥生成物中に存在するスクロース質量/ァラニン質量を表 す。 In Example 1, it was found that trehalose or sucrose is most preferable as the polyol combined with alanine. In this example, sucrose was used as the polyol, the concentration of alanine was kept constant, the concentration of sucrose mixed with alanine was changed, and the optimum concentration ratio R for polymer suppression was examined. Here, R represents the mass of sucrose / alanine present in the lyophilized product.
[0100] 抗体として国際公開第 WO2003/033538号パンフレットに記載されて!/、る HLA-DR に対する抗体 (抗 HLA-DR抗体、 IgGl)を用い、凍結乾燥製剤を調製した。  [0100] A freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in International Publication No. WO2003 / 033538 as an antibody.
[0101] 本実施例では表 2に示した製剤を調製し、ァラニンと組み合わせるスクロースの好 適な濃度の詳細な検討を行った。  [0101] In this example, the preparations shown in Table 2 were prepared, and a detailed examination of an appropriate concentration of sucrose to be combined with alanine was conducted.
[表 2]  [Table 2]
本実施例に供した製剤 -ΪΕ  Formulation used in this example -ΪΕ
Figure imgf000023_0001
Figure imgf000023_0001
[0102] (1)材料と方法 [0102] (1) Materials and methods
本実施例に用いた材料及び分析方法は実施例 1に示したものと同様である。  The materials and analysis methods used in this example are the same as those shown in Example 1.
[0103] (2)試験条件 [0103] (2) Test conditions
実施例 1に示した方法と同様に、 25°C又は 40°Cに制御されたインキュベータ(TABA I ESPEC社製)に 1箇月間及び 3箇月間保存し、熱安定性試験を実施した。また各検 体は熱ストレス負荷後分析開始まで 4°Cに制御された低温庫に保存した。  Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TABA I ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each specimen was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after thermal stress loading.
[0104] (3)結果及び考察 [0104] (3) Results and discussion
図 2は各凍結乾燥製剤を 25°C及び 40°Cで保存した時の重合体の変化を示した。重 合体量はサイズ排除クロマトグラフにより測定した。混合するスクロース質量増加に伴 い重合体生成を抑制することが明らかとなり、 R=0.25から R=2.5の範囲で安定であるこ とを示した。さらに SDS-PAGE分析においてもサイズ排除クロマトグラフで得られた結 果と同様の傾向が認められる泳動像が得られた。  Figure 2 shows the change in polymer when each lyophilized formulation was stored at 25 ° C and 40 ° C. The amount of polymer was measured by size exclusion chromatography. It was clarified that the polymer formation was suppressed as the mass of sucrose mixed increased, indicating that it was stable in the range of R = 0.25 to R = 2.5. Furthermore, in the SDS-PAGE analysis, an electrophoretic image showing the same tendency as that obtained by the size exclusion chromatograph was obtained.
[0105] 本実施例で行なった検討のいずれの濃度においてもケーキが形成され、特に R=l. 5以下できれいなケーキを形成し、長期保存に耐え得る形状を示した。またその他の 分析項目(異物、濁り、不溶性微粒子、デアミド体、酸化体)においても熱ストレス負 荷前後における変化はほとんど確認されず、安定であった。 [0105] A cake is formed at any of the concentrations studied in this example, particularly R = l. A beautiful cake was formed at 5 or less and showed a shape that could withstand long-term storage. The other analysis items (foreign matter, turbidity, insoluble fine particles, deamide, and oxidant) were stable with almost no change observed before and after thermal stress loading.
[0106] 本実施例にお!/、て抗体を安定化させる凍結乾燥製剤の賦形剤としてァラニンと組 み合わせる最適なスクロース質量比率 R (スクロース質量/ァラニン質量)が明らかと なった。安定性が良好である Rは 0.25以上であり、好ましくは 0.5から 2.5であり、より好 ましくは 1から 2であり、さらに好ましくは 0.75から 2であった。最も好ましくは 1.5であった [0106] In this example, the optimum sucrose mass ratio R (sucrose mass / alanine mass) to be combined with alanine as an excipient of a freeze-dried preparation for stabilizing the antibody was revealed. R having good stability was 0.25 or more, preferably 0.5 to 2.5, more preferably 1 to 2, and still more preferably 0.75 to 2. Most preferably it was 1.5
[0107] 実施例 3 抗体濃度の影響検討 [0107] Example 3 Effect of antibody concentration
抗体として国際公開第 WO2003/033538号パンフレットに記載されている HLA-DR に対する抗体 (抗 HLA-DR抗体、 IgGl)を用い凍結乾燥製剤を調製した。  A freeze-dried preparation was prepared using an antibody against HLA-DR (anti-HLA-DR antibody, IgGl) described in WO2003 / 033538 pamphlet as an antibody.
[0108] 本実施例は表 3に示した製剤を調製し、実施例 2で得られた抗体を安定化させるの に最適なスクロース/ァラニン処方 (R=1.5)が抗体濃度の異なる製剤に与える影響を 評価した。ここで Qをァラニン質量に対する抗体質量の比とした。すなわち、 Qは凍結 乾燥製剤中の抗体質量/ァラニン質量を表す。 [0108] In this example, the preparations shown in Table 3 were prepared, and the sucrose / alanine formulation (R = 1.5) optimal for stabilizing the antibody obtained in Example 2 was given to the preparations having different antibody concentrations. The impact was evaluated. Where Q is the ratio of antibody mass to alanine mass. That is, Q represents the antibody mass / alanine mass in the lyophilized preparation.
[表 3] 本突施例に供した製剤-一 W  [Table 3] Formulations used in this project-One W
Figure imgf000024_0001
Figure imgf000024_0001
[0109] (1)材料と方法 [0109] (1) Materials and methods
本実施例に用レ、た材料及び分析方法は実施例 1に示したものと同様である。  The materials, materials and analysis methods used in this example are the same as those shown in Example 1.
[0110] (2)試験条件 [0110] (2) Test conditions
実施例 1に示した方法と同様に、 25°C又は 40°Cに制御されたインキュベータ(TAB AI ESPEC社製)に 1箇月間及び 3箇月間保存し、熱安定性試験を実施した。また各 検体は熱ストレス負荷後分析開始まで 4°Cに制御された低温庫に保存した。  Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TAB AI ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after the heat stress.
[0111] (3)結果及び考察 図 3は各凍結乾燥製剤を 25°C及び 40°Cで保存した時の重合体の変化を示した。重 合体量はサイズ排除クロマトグラフにより測定した。抗体濃度が最も高濃度である (抗 体量が最も多い) Q=3の製剤について、 40°C3箇月保存で重合体約 2%であった。一 般的に抗体濃度が高くなると重合体生成量が増加し、不安定になることが知られて いる。実施例 1の抗体濃度 10mg/mL、ァラニン単独処方の製剤について、 40°C3箇月 保存での重合体量が約 9%であったことを考慮すると、十分に重合体生成を抑制して V、ることは明らかである。なお抗体濃度が最も低濃度である(抗体量が最も少な!/、) Q =0.05の製剤についてはストレス負荷前後における変化はほとんど確認されず、安定 であった。またその他の分析項目(異物、濁り、分解物、デアミド体、酸化体)におい てもストレス負荷前後における変化はほとんど確認されず、安定であった。 [0111] (3) Results and discussion Figure 3 shows the polymer changes when each lyophilized formulation was stored at 25 ° C and 40 ° C. The amount of polymer was measured by size exclusion chromatography. The formulation with the highest antibody concentration (the highest amount of antibody) Q = 3 was about 2% polymer when stored at 40 ° C for 3 months. In general, it is known that as the antibody concentration increases, the amount of polymer produced increases and becomes unstable. Considering that the antibody concentration in Example 1 was 10 mg / mL and the formulation of alanine alone was about 9% when stored at 40 ° C for 3 months, the polymer formation was sufficiently suppressed by V, Obviously. The formulation with the lowest antibody concentration (the least antibody amount! /,) Q = 0.05 was stable with almost no change observed before and after stress loading. In other analysis items (foreign substances, turbidity, decomposition products, deamides, and oxidants), almost no change was observed before and after stress loading, and the analysis was stable.
[0112] 本実施例において Q=0.05から 3に対し、 R=1.5の比率でァラニン/スクロースを含有 することで安定な凍結乾燥製剤を得ることが明らかとなった。すなわち安定な凍結乾 燥製剤は、抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0.3〜 20 : 0.5〜30の範囲にある凍結乾燥製剤で得られることを示す。抗体質量に対するァ ラニン質量とスクロース質量の合計で表した場合 (抗体質量を 1とする)、 1 : 5/6〜50 であり、好ましくは 1 :;!〜 20、さらに好ましくは 1 :;!〜 10、特に好ましくは 1 : 5である。  [0112] In this example, it was revealed that a stable lyophilized preparation can be obtained by containing alanine / sucrose at a ratio of R = 1.5 for Q = 0.05 to 3. That is, a stable freeze-dried preparation is obtained with a freeze-dried preparation in which the ratio of the mass of the three components of antibody mass, alanine mass, and sucrose mass is in the range of 1: 0.3 to 20: 0.5 to 30. When expressed as the sum of alanine mass and sucrose mass relative to antibody mass (antibody mass is 1), it is 1: 5 / 6-50, preferably 1:;!-20, more preferably 1:;! To 10, particularly preferably 1: 5.
[0113] 実施例 4 他の抗体での検討  [0113] Example 4 Investigation with other antibodies
本検討では実施例 2で得られた抗体を安定化させるのに最適なァラニン/スクロー ス処方(R=1.5)について、国際公報第 WO0563981号パンフレットに公開されている C D40に対する組換え完全ヒ HgG4抗体 4D11 (抗 CD40抗体)を用いて製剤の安定性 を検討した。  In this study, an optimal alanine / sucrose formulation (R = 1.5) for stabilizing the antibody obtained in Example 2 was used for recombinant complete HgG4 against CD40 published in International Publication No. WO0563981. The stability of the preparation was examined using antibody 4D11 (anti-CD40 antibody).
[0114] 本実施例は表 4に示した製剤を調製し、抗体の種類が製剤の安定性に与える影響 を評価した。  [0114] In this example, the preparations shown in Table 4 were prepared, and the effect of the type of antibody on the stability of the preparation was evaluated.
[表 4]  [Table 4]
本実施钶に洪した製剤一 W  Formulations that have fallen into this implementation
原体 度 ί几体 ¾ 赋 剤 界面活性铜 pH Active ingredient 几几 体 ¾ 赋 Surfactant 铜 pH
1 0.05rng/mし 1 0.05rng / m
抗 HLA-DR抗体 20mg/mL 30mg/mし  Anti-HLA-DR antibody 20mg / mL 30mg / m
10mg/mL ァラニン ポリソルべ一ト 5.5 10 mg / mL alanine polysorbate 5.5
2 スクロ一ス 2 Scroll
抗 CD40抗体 80 [0115] (1)材料と方法 Anti-CD40 antibody 80 [0115] (1) Materials and methods
本実施例に用レ、た材料及び分析方法は実施例 1に示したものと同様である。 R= 1.5 、 Q=0.5であり、抗体質量に対するァラニン及びスクロースの質量の比は、 1 : 5であつ た。  The materials, materials and analysis methods used in this example are the same as those shown in Example 1. R = 1.5, Q = 0.5, and the ratio of alanine and sucrose mass to antibody mass was 1: 5.
[0116] (2)試験条件  [0116] (2) Test conditions
実施例 1に示した方法と同様に、 25°C又は 40°Cに制御されたインキュベータ(TAB AI ESPEC社製)に 1箇月間及び 3箇月間保存し、熱安定性試験を実施した。また各 検体は熱ストレス負荷後分析開始まで 4°Cに制御された低温庫に保存した。  Similarly to the method shown in Example 1, the samples were stored in an incubator (manufactured by TAB AI ESPEC) controlled at 25 ° C or 40 ° C for 1 month and 3 months, and a thermal stability test was performed. Each sample was stored in a low-temperature chamber controlled at 4 ° C until the start of analysis after the heat stress.
[0117] (3)結果及び考察  [0117] (3) Results and discussion
図 4は各凍結乾燥製剤を 40°Cで保存した時の重合体の変化(Initial比:凍結乾燥前 の重合体量で割った値であり、凍結乾燥前の値を 1とした)を示した。重合体量はサイ ズ排除 HPLCにより測定した。抗 CD40抗体においても、賦形剤としてァラニンとスクロ ースを用いた製剤では保存期間における重合体生成の抑制が確認され、温度及び 時間に関わらず本製剤中の抗体は安定に保持された。またその他の分析項目(異物 、濁り、分解物、デアミド体、酸化体)においても熱ストレス負荷前後における変化は ほとんど確認されず、安定であった。  Figure 4 shows the change in polymer when each lyophilized preparation was stored at 40 ° C (Initial ratio: value divided by the amount of polymer before lyophilization, with the value before lyophilization being 1). It was. The amount of polymer was measured by size exclusion HPLC. Also in the anti-CD40 antibody, in the preparation using alanine and sucrose as excipients, suppression of polymer formation during the storage period was confirmed, and the antibody in this preparation was stably maintained regardless of temperature and time. In addition, the other analysis items (foreign matter, turbidity, degradation products, deamides, and oxidants) were stable with almost no change observed before and after thermal stress loading.
[0118] 本実施例においてァラニン及びスクロースを賦形剤とする安定な凍結乾燥製剤は 抗体の種類によらず安定であることが判明した。  [0118] In this example, it was found that a stable lyophilized preparation using alanine and sucrose as excipients is stable regardless of the type of antibody.
[0119] 本明細書で引用した全ての刊行物、特許および特許出願をそのまま参考として本 明細書にとり入れるものとする。  [0119] All publications, patents, and patent applications cited herein are incorporated herein by reference in their entirety.

Claims

請求の範囲 The scope of the claims
[I] 抗体を含み、さらにァラニン及びポリオールを賦形剤として含む凍結乾燥製剤。  [I] A lyophilized preparation containing an antibody and further containing alanine and polyol as excipients.
[2] ポリオールが非還元糖であり、かつ非晶質のポリオールである請求項 1記載の凍結 乾燥製剤。  [2] The lyophilized preparation according to claim 1, wherein the polyol is a non-reducing sugar and is an amorphous polyol.
[3] ポリオールが非還元糖であり、かつ非晶質の糖である請求項 1記載の凍結乾燥製 剤。  [3] The freeze-dried preparation according to claim 1, wherein the polyol is a non-reducing sugar and is an amorphous sugar.
[4] ポリオールがスクロース又はトレハロースである請求項 3記載の凍結乾燥製剤。  [4] The lyophilized preparation according to claim 3, wherein the polyol is sucrose or trehalose.
[5] 抗体質量 1に対するァラニン及びポリオールの質量が、 5/6〜50である請求項;!〜 4 の!/、ずれか 1項に記載の凍結乾燥製剤。  [5] The mass of alanine and polyol with respect to antibody mass of 1 is 5/6 to 50;
[6] ポリオール質量/ァラニン質量比力 0.5〜2.5である請求項 5記載の凍結乾燥製剤6. The freeze-dried preparation according to claim 5, wherein the specific mass of polyol / alanine mass is 0.5 to 2.5.
Yes
[7] 抗体質量/ァラニン質量比が、 0.05〜3である請求項 5又は 6に記載の凍結乾燥製 剤。  [7] The freeze-dried product according to claim 5 or 6, wherein the antibody mass / alanine mass ratio is 0.05 to 3.
[8] 抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0·3〜20 : 0·5〜3 [8] The ratio of the three masses of antibody mass, alanine mass, and sucrose mass is 1: 0 · 3 to 20: 0 · 5 to 3
0である請求項 1〜7のいずれ力、 1項に記載の凍結乾燥製剤。 The lyophilized preparation according to any one of claims 1 to 7, which is 0.
[9] さらに、緩衝液を含む請求項;!〜 8のいずれか 1項に記載の凍結乾燥製剤。 [9] The lyophilized preparation according to any one of claims 8 to 8, further comprising a buffer solution.
[10] 緩衝液がグルタミン酸緩衝液である、請求項 9記載の凍結乾燥製剤。 [10] The lyophilized preparation according to claim 9, wherein the buffer is a glutamate buffer.
[I I] 緩衝液の濃度が 10mMである請求項 9又は 10に記載の凍結乾燥製剤。  [I I] The freeze-dried preparation according to claim 9 or 10, wherein the concentration of the buffer is 10 mM.
[12] さらに、界面活性剤を含む請求項;!〜 11のいずれか 1項に記載の凍結乾燥製剤。  [12] The freeze-dried preparation according to any one of claims 1 to 11, further comprising a surfactant.
[13] 界面活性剤がポリソルベートである、請求項 12記載の凍結乾燥製剤。 [13] The freeze-dried preparation according to claim 12, wherein the surfactant is polysorbate.
[14] 抗体を有効成分として含む医薬製剤である、請求項;!〜 13のいずれか 1項に記載 の凍結乾燥製剤。 [14] The freeze-dried preparation according to any one of claims;! To 13, which is a pharmaceutical preparation containing an antibody as an active ingredient.
[15] 抗体にァラニン及びポリオールを添加して凍結乾燥することを含む、安定な凍結乾 燥製剤を製造する方法。  [15] A method for producing a stable freeze-dried preparation comprising adding alanine and polyol to an antibody and freeze-drying.
[16] ポリオールが非還元糖であり、かつ非晶質のポリオールである請求項 15記載の凍 結乾燥製剤を製造する方法。 16. The method for producing a freeze-dried preparation according to claim 15, wherein the polyol is a non-reducing sugar and is an amorphous polyol.
[17] ポリオールが非還元糖であり、かつ非晶質の糖である請求項 15記載の凍結乾燥製 剤を製造する方法。 17. The method for producing a lyophilized product according to claim 15, wherein the polyol is a non-reducing sugar and is an amorphous sugar.
[18] ポリオールがスクロース又はトレハロースである請求項 17記載の凍結乾燥製剤を製 造する方法。 [18] The method for producing a freeze-dried preparation according to claim 17, wherein the polyol is sucrose or trehalose.
[19] 抗体質量 1に対するァラニン及びポリオールの質量が、 5/6〜50である請求項 15〜 [19] The mass of alanine and polyol with respect to antibody mass 1 is 5/6 to 50.
18のいずれか 1項に記載の凍結乾燥製剤を製造する方法。 19. A method for producing a freeze-dried preparation according to any one of 18 above.
[20] ポリオール質量/ァラニン質量比力 0.5〜2.5である請求項 19記載の凍結乾燥製 剤を製造する方法。 20. The method for producing a lyophilized product according to claim 19, wherein the specific mass of polyol / alanine mass is 0.5 to 2.5.
[21] 抗体質量/ァラニン質量比が、 0.05〜3である請求項 19又は 20に記載の凍結乾 燥製剤を製造する方法。  21. The method for producing a freeze-dried preparation according to claim 19 or 20, wherein the antibody mass / alanine mass ratio is 0.05 to 3.
[22] 抗体質量とァラニン質量とスクロース質量の 3成分の質量の比が 1 : 0.3〜20 : 0.5〜3[22] The ratio of the mass of antibody, alanine, and sucrose is 1: 0.3 to 20: 0.5 to 3
0である請求項 15〜21のいずれ力、 1項に記載の凍結乾燥製剤を製造する方法。 The method for producing a freeze-dried preparation according to any one of claims 15 to 21, which is 0.
PCT/JP2007/067481 2006-09-07 2007-09-07 Stable lyophilized pharmaceutical preparation comprising antibody WO2008029908A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2008533208A JPWO2008029908A1 (en) 2006-09-07 2007-09-07 Stable lyophilized pharmaceutical formulation containing antibody

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006-243144 2006-09-07
JP2006243144 2006-09-07

Publications (1)

Publication Number Publication Date
WO2008029908A1 true WO2008029908A1 (en) 2008-03-13

Family

ID=39157331

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/067481 WO2008029908A1 (en) 2006-09-07 2007-09-07 Stable lyophilized pharmaceutical preparation comprising antibody

Country Status (3)

Country Link
JP (1) JPWO2008029908A1 (en)
TW (1) TW200826974A (en)
WO (1) WO2008029908A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010132626A (en) * 2008-12-08 2010-06-17 Kyorin Pharmaceut Co Ltd Tablet quickly disintegrating in oral cavity
WO2012028683A1 (en) 2010-09-02 2012-03-08 Novartis Ag Antibody gel system for sustained drug delivery
WO2012059598A2 (en) 2010-11-05 2012-05-10 Novartis Ag Methods of treating rheumatoid arthritis using il-17 antagonists
JP2013542938A (en) * 2010-10-12 2013-11-28 メルツ ファーマ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディト ゲゼルシャフト アウフ アクティーン Mammalian excipient-free preparation suitable for protein stabilization
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
JP7407118B2 (en) 2018-02-08 2023-12-28 アムジェン インコーポレイテッド Low pH pharmaceutical antibody preparation
WO2024100130A1 (en) * 2022-11-11 2024-05-16 Merck Patent Gmbh Thermostable vaccin formulations and process for preparing the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001503781A (en) * 1996-11-19 2001-03-21 ロシュ ダイアグノスティクス ゲゼルシャフト ミット ベシュレンクテル ハフツング Stable lyophilized pharmaceutical preparation of monoclonal or polyclonal antibodies
JP2005513110A (en) * 2001-12-21 2005-05-12 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Lyophilized preparation containing antibody against EGF receptor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001503781A (en) * 1996-11-19 2001-03-21 ロシュ ダイアグノスティクス ゲゼルシャフト ミット ベシュレンクテル ハフツング Stable lyophilized pharmaceutical preparation of monoclonal or polyclonal antibodies
JP2005513110A (en) * 2001-12-21 2005-05-12 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Lyophilized preparation containing antibody against EGF receptor

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010132626A (en) * 2008-12-08 2010-06-17 Kyorin Pharmaceut Co Ltd Tablet quickly disintegrating in oral cavity
WO2012028683A1 (en) 2010-09-02 2012-03-08 Novartis Ag Antibody gel system for sustained drug delivery
JP2013542938A (en) * 2010-10-12 2013-11-28 メルツ ファーマ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディト ゲゼルシャフト アウフ アクティーン Mammalian excipient-free preparation suitable for protein stabilization
US9173944B2 (en) 2010-10-12 2015-11-03 Merz Pharma Gmbh & Co. Kgaa Formulation suitable for stabilizing proteins, which is free of mammalian excipients
WO2012059598A2 (en) 2010-11-05 2012-05-10 Novartis Ag Methods of treating rheumatoid arthritis using il-17 antagonists
EP3542820A1 (en) 2010-11-05 2019-09-25 Novartis AG Methods of treating psoriatic arthritis using il-17 antagonists
EP3912640A1 (en) 2010-11-05 2021-11-24 Novartis AG Pharmaceutical compositions comprising secukinumab
EP4116325A1 (en) 2010-11-05 2023-01-11 Novartis AG Methods of treating rheumatoid arthritis using il-17 antagonists
JP7407118B2 (en) 2018-02-08 2023-12-28 アムジェン インコーポレイテッド Low pH pharmaceutical antibody preparation
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
WO2024100130A1 (en) * 2022-11-11 2024-05-16 Merck Patent Gmbh Thermostable vaccin formulations and process for preparing the same

Also Published As

Publication number Publication date
JPWO2008029908A1 (en) 2010-01-21
TW200826974A (en) 2008-07-01

Similar Documents

Publication Publication Date Title
US20240058263A1 (en) Formulations of antibody
RU2731418C2 (en) Stable pharmaceutical preparation based on the pd-1 antibody and its use in medicine
EP1712240B1 (en) Stable water-based medicinal preparation containing antibody
EP2648750B1 (en) Antibody formulation
JP7407118B2 (en) Low pH pharmaceutical antibody preparation
KR102307257B1 (en) Anti-il-4/anti-il-13 bispecific antibidy formulations
ES2791690T3 (en) Freeze-dried formulations
JP7204651B2 (en) Formulation of antibody-drug conjugate and method of lyophilization thereof
US9457089B2 (en) Highly concentrated aqueous protein solution with reduced viscosity
TW201347791A (en) Antibody formulation
JP6339578B2 (en) Lyophilized preparation containing GM-CSF neutralizing compound
CN112107538A (en) Stable aqueous antibody formulations
JP6769879B2 (en) Liquid formulation containing GM-CSF neutralizing compound
JP6346189B2 (en) Liquid formulation containing GM-CSF neutralizing compound
WO2008029908A1 (en) Stable lyophilized pharmaceutical preparation comprising antibody
US20090208509A1 (en) Pharmaceutical formulation of an antibody against IL-1R
KR20220113355A (en) Anti-Connexin Antibody Formulations
EP3434283A1 (en) Medicinal composition comprising peg anti-human ngf antibody fab' fragment
CN111683681B (en) Formulations comprising anti-OX 40 antibodies, methods of making, and uses thereof
RU2789476C2 (en) Production of antibody-drug conjugate and its lyophilization
KR20240016260A (en) Accelerated Manufacturing Method for Lyophilized Protein Formulations
ES2795993T3 (en) Anti-IL-4 / Anti-IL-13 Bispecific Antibody Formulations

Legal Events

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

Ref document number: 07806922

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008533208

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07806922

Country of ref document: EP

Kind code of ref document: A1