WO2020173431A2 - 包含抗cd47抗体的制剂及其制备方法和用途 - Google Patents

包含抗cd47抗体的制剂及其制备方法和用途 Download PDF

Info

Publication number
WO2020173431A2
WO2020173431A2 PCT/CN2020/076611 CN2020076611W WO2020173431A2 WO 2020173431 A2 WO2020173431 A2 WO 2020173431A2 CN 2020076611 W CN2020076611 W CN 2020076611W WO 2020173431 A2 WO2020173431 A2 WO 2020173431A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
preparation
liquid
antibody preparation
cancer
Prior art date
Application number
PCT/CN2020/076611
Other languages
English (en)
French (fr)
Other versions
WO2020173431A3 (zh
Inventor
谢瑞霞
马丽强
汪音爵
周凯松
Original Assignee
信达生物制药(苏州)有限公司
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 信达生物制药(苏州)有限公司 filed Critical 信达生物制药(苏州)有限公司
Priority to KR1020217030704A priority Critical patent/KR20210134926A/ko
Priority to CA3131307A priority patent/CA3131307A1/en
Priority to CN202080014878.4A priority patent/CN113453719A/zh
Priority to EP20763678.8A priority patent/EP3932426A4/en
Priority to JP2021549889A priority patent/JP2022521624A/ja
Priority to US17/433,780 priority patent/US20220143180A1/en
Priority to AU2020227770A priority patent/AU2020227770A1/en
Publication of WO2020173431A2 publication Critical patent/WO2020173431A2/zh
Publication of WO2020173431A3 publication Critical patent/WO2020173431A3/zh

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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to the field of antibody preparations. More specifically, the present invention relates to a pharmaceutical preparation containing a recombinant fully human anti-cluster of differentiation 47 (CD47) monoclonal antibody, especially a stable high-concentration antibody liquid preparation, and a method for preparing the pharmaceutical preparation, and The therapeutic and/or preventive use of the pharmaceutical preparation.
  • a pharmaceutical preparation containing a recombinant fully human anti-cluster of differentiation 47 (CD47) monoclonal antibody, especially a stable high-concentration antibody liquid preparation, and a method for preparing the pharmaceutical preparation, and The therapeutic and/or preventive use of the pharmaceutical preparation.
  • CD47 fully human anti-cluster of differentiation 47
  • CD47 Cluster of differentiation 47
  • IAP integrin-associated protein
  • CD47 fully human anti-cluster of differentiation 47
  • Monoclonal antibodies have a high degree of specificity for I barb, and are generally more effective than small molecule drugs.
  • Monoclonal antibody-based drugs has its own challenges.
  • Monoclonal antibodies are more complex than traditional organic and inorganic drugs, and generally have a degradation pattern similar to other protein-based biopharmaceuticals.
  • the degradation of antibody proteins can be divided into two main types: physical instability (involving changes in the high-level structure of proteins;) and chemical instability (involving various chemical modifications of proteins).
  • Chemical instability can be caused by deamidation, racemization, hydrolysis, oxidation, P-elimination or disulfide bond exchange, among which the most common are fragmentation, deamidation and oxidation.
  • Physical instability can be caused by denaturation, aggregation, precipitation or adsorption.
  • the degradation of monoclonal antibodies can occur at various stages, including the preparation, storage, and delivery of antibody preparations.
  • the biological activity of drugs based on monoclonal antibodies is closely related to their structure, conformation and chemical stability. Therefore, the development of antibody preparations is an important aspect of antibody product development and is often a key step for successful clinical production.
  • the stability of antibody drugs is an important indicator to ensure the effectiveness and safety of drugs. Obtaining a prescription that gives antibody drugs good stability is a key condition for drugs to maintain their effectiveness and safety during the shelf life. Therefore, the antibody preparation must be formulated in a way that not only makes the antibody suitable for administration to a subject, but also in a way that maintains its stability during storage and subsequent use. If the antibody is not properly formulated in the liquid, the antibody in the liquid solution tends to decompose, aggregate, or undergo undesirable chemical modification.
  • monoclonal antibodies are being developed at increased doses for the treatment of various indications.
  • cetuximab is administered at a dose of 250-400 mg/m 2
  • efalizumab is administered at a dose of approximately 1 mg/kg.
  • high-concentration stable monoclonal antibody liquid preparations is a direction in the art, in which the protein concentration needs to reach 100 mg/ml or higher.
  • the formulation form satisfies a wide range of dosage levels (typically, depending on the target indication, the amount of monoclonal antibody can be 0.1-20 mg/kg) and in multiple routes of administration (typically subcutaneous and Intravenous) flexibility of administration.
  • the inventors of the present application have designed two formulations: pre-prescription and prescription screening.
  • pre-prescription and prescription screening In the phase test, the effects of different pH, surfactants, stabilizers and antioxidants on the stability of anti-CD47 antibody preparations were investigated.
  • the present inventors have provided pharmaceutical preparations containing recombinant fully human anti-cluster of differentiation 47 (CD47) monoclonal antibodies, especially stable high-concentration antibody liquid preparations.
  • the present invention provides a liquid antibody preparation comprising (i) a recombinant fully human anti-CD47 monoclonal antibody (hereinafter also referred to as "anti-CD47 antibody”); (ii) a buffer, (iii) a stabilizer , And (iv) surfactants.
  • the anti-CD47 antibody is a recombinant fully human anti-cluster of differentiation 47 (CD47) monoclonal antibody disclosed in Chinese application CN201710759828.9 (submitted on August 29, 2017). For the purpose of this application, the entire content of the Chinese application is hereby incorporated by reference.
  • the anti-CD47 antibody comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region comprises the sequence of SEQ ID NO: 1 or a sequence having at least 90% identity thereto, and the light chain The variable region comprises the sequence of SEQ ID NO: 2 or a sequence that is at least 90% identical to it:
  • the anti-CD47 antibody comprises:
  • -ARGKTGSAA SEQIDNO: 5
  • GKTGSAA SEQIDNO: 12
  • the anti-CD47 antibody is an IgG4 antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises the sequence of SEQ ID NO: 9 or a sequence that is at least 90% identical to it, and wherein the The light chain comprises the sequence of SEQ ID NO: 10 or a sequence that is at least 90% identical to it:
  • the anti-CD47 antibody is the anti-CD47 monoclonal antibody ADI-26630 disclosed in Chinese application CN201710759828.9 (filed on August 29, 2017), which consists of the heavy chain sequence of SEQ ID NO: 9 and SEQ ID NO: 10 light chain sequence composition.
  • the antibody showed significant anti-tumor activity.
  • the antibody can inhibit tumor growth. The rate reaches about 100% or higher; and the rate of tumor disappearance can reach more than 60%.
  • the concentration of the anti-CD47 antibody in the liquid antibody preparation is about 1-150 mg/mL, for example, 20 mg/ml or more, especially 50 mg/ml or more, preferably 100 mg/ml or 120 mg/ml.
  • the concentration of the anti-CD47 antibody in the liquid antibody preparation may be about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 70, 75, 80, 85 mg/ml, or more than 90 mg/mL , For example, 90, 95, 100, 105, 110, or 120mg/ml.
  • the liquid antibody preparation of the present invention has a pH of 5.0-6.0, such as pH 5.2 ⁇ 0.2, pH 5.5 ⁇ 0.2, pH 5.7 ⁇ 0.2, preferably pH 5.5.
  • the liquid antibody formulation of the present invention contains about 5-100 mM buffer. In one embodiment, the concentration of the buffer in the liquid antibody preparation of the present invention is about 10 mM, 20 mM, 30 mM, 40 mM, or 50 mM. In one embodiment, the buffer is a histidine buffer. In a preferred embodiment, the liquid antibody preparation of the present invention contains about 10-20 mM, especially 10 mM histidine buffer. In still another embodiment, the histidine buffer is composed of histidine-histidine hydrochloride buffer system. In still another preferred embodiment, the liquid antibody preparation of the present invention contains 0.4 mg/ml histidine and 1.5 mg/ml histidine hydrochloride.
  • the liquid antibody formulation of the present invention contains a stabilizer, preferably the stabilizer is selected from sorbitol, sucrose, trehalose, arginine and combinations thereof, more preferably sorbitol or sorbitol and arginine The combination.
  • the liquid formulation of the present invention contains sorbitol as a stabilizer, preferably the concentration of sorbitol is about 1-10% w/v, for example, 1.5%, 2%, 2.5%, 3%, 3.5% , 4%, 4.5%, 5%, 5.5% or 6% w/v, or about 10-60mg/ml, for example about 15mg/ml, 20mg/ml, 25mg/ml, 30mg/ml, 35mg/ml, 40mg / ml, 45mg / ml, an amount of 50mg / ml, 55mg / ml is present, preferably from about 40mg / ml o in still another preferred embodiment, the liquid antibody formulations of the present invention comprises a combination of a stabilizer sorbitol and arginine.
  • the amount of sorbitol is about 10-30 mg/ml, especially 15 mg/ml, and the amount of arginine is 80-1 10 mM, especially about 100 mM.
  • the liquid antibody formulation of the present invention contains a stabilizer combination of about 15 mg/ml sorbitol and about 21.1 mg/ml arginine hydrochloride.
  • the liquid antibody preparation of the present invention contains a surfactant, and preferably, the surfactant is a non-ionic surfactant.
  • the surfactant is selected from polysorbate surfactants, preferably polysorbate-20.
  • the liquid antibody preparation of the present invention contains about 0.1-1 mg/ml, for example, 0.1-0.5 mg/ml polysorbate-20.
  • the liquid antibody preparation of the present invention contains about 0.1 to 0.4 mg/ml of polysorbate-20.
  • the amount of polysorbate-20 in the liquid antibody preparation of the present invention is about 0.3 mg/ml.
  • the liquid formulation of the present invention further contains an antioxidant. In yet another embodiment, the liquid formulation of the present invention does not contain antioxidants. In one embodiment, the antioxidant is edetic acid (EDTA) or a salt thereof, such as EDTA-2Na. In one embodiment, the liquid preparation is a stable liquid pharmaceutical preparation, preferably an injection. In one embodiment, the pharmaceutical formulation of the present invention is used for subcutaneous, intradermal, intramuscular, or intravenous injection. In a preferred embodiment, the liquid antibody preparation of the present invention comprises:
  • the pH of the liquid formulation is about 5.0-6.0, preferably pH 5.5 ⁇ 0.2 c
  • liquid antibody preparation of the present invention comprises
  • the pH of the liquid formulation is about 5.0-6.0, preferably pH 5.5 ⁇ 0.2 c
  • liquid antibody preparation of the present invention comprises:
  • the present invention provides a solid antibody preparation, which is obtained by subjecting the liquid antibody preparation of the present invention to a curing treatment.
  • the curing treatment is performed by, for example, a crystallization method, a spray drying method, or a freeze drying method.
  • the solid antibody preparation is, for example, a lyophilized preparation, for example, in the form of a lyophilized powder injection.
  • the solid antibody preparation can be reconstituted in a suitable solvent before use to form the reconstituted preparation of the present invention.
  • the reconstituted preparation is also a liquid antibody preparation of the present invention.
  • the appropriate solvent is selected from water for injection and organic solvent for injection, including but not limited to oil for injection, ethanol, propylene glycol, etc., or a combination thereof.
  • the liquid formulation of the present invention can be stored stably for a long time, especially at a high concentration (for example, 50 mg/ml or more, preferably 90-150 mg/ml, such as 100 mg/ml or 120 mg/ml) for stable storage, such as at least 24 months or longer .
  • the liquid formulation of the present invention can be heated at about -80°C to about 45°C, such as -80°C, about -30°C, about -20°C, about 0°C, about 5°C.
  • C about 25°C, about 35°C, about 38°C, about 40°C, about 42°C, or about 45°C, storage for at least 10 days, at least 20 days, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, It is stable for at least 12 months, at least 18 months, at least 24 months, at least 36 months, or longer.
  • the liquid formulation of the present invention can be stored stably for at least 24 months. In yet another embodiment, the liquid formulation of the invention is stable at at least 40°C. In yet another embodiment, the liquid formulation of the present invention is stable at about 2°C-8°C for at least 12 months, preferably at least 24 months. In one embodiment, the liquid formulation of the invention remains stable at room temperature or, for example, about 25°C for at least 3 months, preferably at least 6 months. In yet another embodiment, the liquid formulation of the invention remains stable at about 40°C for at least one month. In yet another embodiment, the liquid formulation of the present invention can be stable at a temperature of about 5°C to 40°C, such as at a temperature of 25°C, for at least 1 day, such as 3 days or 5 days, under shaking.
  • the stability of the preparation after storage can be indicated by detecting changes in the appearance, visible foreign matter, protein content, purity, and/or charge variants of the preparation. In one embodiment, it can be in a high-temperature accelerated test, for example, after storing at least 1 week, 2 weeks or preferably 1 month at 40°C ⁇ 2°C, or storing at least 1 at 25°C ⁇ 2°C After one month or two months, the stability of the liquid preparation of the present invention was tested. In one embodiment, the oscillation stability of the liquid formulation of the present invention is tested by an oscillation test.
  • the stability of the liquid formulation of the present invention is visually inspected, wherein the liquid formulation of the present invention remains a clear, colorless or slightly yellow liquid in appearance, free of foreign matter, flocculent, and precipitation. In one embodiment, by visual inspection under natural light, no visible foreign matter is present in the preparation. In one embodiment, after storage, the stability of the liquid formulation of the present invention is checked by measuring the change in protein content, wherein, for example, by ultraviolet spectrophotometry (UV) method, the protein content change rate is relative to the initial value on day 0 of storage Not more than 20%, preferably not more than 10%, such as 7-8%, preferably not more than 5%.
  • UV ultraviolet spectrophotometry
  • the stability of the liquid preparation of the invention is checked by measuring the change in turbidity of the liquid preparation of the invention, for example, by the OD350mm method.
  • the change value is not the same as the initial value on the 0th day of storage. It exceeds 0.04, more preferably does not exceed 0.03, and more preferably does not exceed 0.02.
  • the stability of the liquid preparation of the present invention is checked by measuring the change in purity of the liquid preparation of the present invention, wherein size exclusion high performance liquid chromatography (SEC-HPLC) is used to check the stability of the liquid preparation of the present invention.
  • SEC-HPLC size exclusion high performance liquid chromatography
  • the change value of the purity of the main peak does not exceed 10%, for example, does not exceed 5%, 4%, 3%, such as 1-2%, preferably does not exceed 1%, and preferably, the increase in the aggregate does not exceed 2 %, preferably not more than 1%, 0.5% or 0.1%.
  • the stability of the liquid preparation of the invention is checked by measuring the purity change of the liquid preparation of the invention, wherein the main peak is determined by the non-reducing sodium dodecyl sulfate capillary electrophoresis (CE-SDS) method.
  • CE-SDS non-reducing sodium dodecyl sulfate capillary electrophoresis
  • the change in purity does not decrease by more than 10%, for example, not more than 5%, 4%, 3%, preferably not more than 2%, 1%, 0.5% or 0.1%.
  • iCIEF imaging capillary isoelectric focusing electrophoresis
  • the change value of the charge variant (main component, acidic component or basic component) of the antibody relative to the initial value on the 0th day of storage does not exceed 30%, preferably does not exceed 20%, or not more than 10%, such as not more than 5%, 4%, 3%, or 2%, for example, in one embodiment, the change value of the main component or the acidic component is not more than 20%, and it is alkaline The change value of the composition does not exceed 3%.
  • after the formulation is stored for example, after storage at 2-8°C for at least 24 months, or after storage at room temperature for at least 3 months, or after storage at 40°C ⁇ 2°C for 1 month, It is stable and preferably has one or more of the following characteristics:
  • the percentage of antibody monomer in the preparation is greater than 90%, preferably greater than 95%, and preferably the aggregate increase does not exceed 2%;
  • the preparation has a purity of greater than 90%, preferably greater than 95%, measured by non-reduced CE-SDS;
  • the formulation is stored after storage, for example, after storage at 2-8°C for at least 24 months, or after storage at room temperature for at least 3 months, or after storage at 40°C ⁇ 2°C for 1 month , Is stable, and preferably has one or more of the following characteristics:
  • the turbidity change value does not exceed 0.04, preferably does not exceed 0.02;
  • the present invention provides a delivery device comprising the liquid antibody preparation or solid antibody preparation of the present invention.
  • the delivery device of the present invention is provided in the form of a pre-filled syringe containing the liquid antibody preparation or solid antibody preparation of the present invention, for example, for intravenous, subcutaneous, intradermal or intramuscular injection.
  • the present invention provides a method for delivering anti-CD47 antibodies to a subject, such as a mammal, comprising the step of administering the liquid antibody preparation or solid antibody preparation of the present invention to the subject, and the delivery is, for example, by using Pre-filled syringe delivery device implementation.
  • the present invention provides the use of the liquid antibody preparation or solid antibody preparation of the present invention to prepare a delivery device (eg, a pre-filled syringe) or a drug for treating CD47-related diseases in a subject, especially
  • a delivery device eg, a pre-filled syringe
  • a drug for treating CD47-related diseases in a subject especially
  • CD47-related cancers such as various blood tumors, such as lymphoma, such as Burkitt’s lymphoma.
  • Figure 1 shows that in the pH screening test described in the examples, antibody preparations of different pH (pH 5.0, 5.5, 6.0 and 6.5) were stored at a temperature of 40°C ⁇ 2°C for 0 days and 1 week , 2 weeks and 1 month later, the preparation turbidity measured by OD350nm method over time.
  • Figure 2 shows that in the pH screening test described in the examples, antibody preparations of different pH (pH 5.0, 5.5, 6.0 and 6.5) were stored at a temperature of 40°C ⁇ 2°C for 0 days and 1 week , 2 weeks and 1 month later, the main peak purity of the preparation measured by SEC-HPLC method over time.
  • Figure 3 shows the purity profile of the antibody preparation determined by the SEC-HPLC method after storing the antibody preparation with pH 6.5 at 40°C ⁇ 2°C for 1 month in the pH screening test described in the embodiment.
  • Figure 4 shows that in the stabilizer screening test described in the examples, antibody preparations (prescriptions 1-4) with different stabilizers were stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, and After 1 month, the main peak purity measured by SEC-HPLC method changes with time.
  • Figure 5 shows that in the stabilizer screening test described in the examples, antibody preparations with different stabilizers (prescriptions 1-4) were stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, and After 1 month, the main peak purity measured by the non-reducing CE-SDS method over time.
  • Figure 6 shows that in the stabilizer screening test described in the examples, antibody preparations with different stabilizers (prescriptions 1-4) were stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, and After 1 month, the main component of the charge variant measured by the iCIEF method changes over time.
  • Figure 7 shows that in the stabilizer screening test described in the examples, antibody formulations (prescriptions 1-4) with different stabilizers were stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, and After 1 month, the change graph of the acidic component of the charge variant measured by the iCIEF method over time.
  • Figure 8 shows that in the antioxidant screening test described in the examples, the antibody preparations with or without EDTA (prescriptions A and B) are stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, And 1 month later, the turbidity measured by the OD350nm method over time.
  • Figure 9 shows that in the antioxidant screening test described in the examples, the antibody preparations with or without EDTA (prescriptions A and B) are stored at 40°C ⁇ 2°C for 0 days, 1 week, 2 weeks, And 1 month later, the charge variants (principal component, acidic component, and alkaline component) measured by iCIEF method over time.
  • the charge variants principal component, acidic component, and alkaline component
  • the term “comprising” or “including” means to include the stated elements, integers or steps, but does not exclude any other elements, integers or steps.
  • the term “comprises” or “includes” is used, unless otherwise specified, it also covers the situation consisting of the mentioned elements, integers or steps.
  • an antibody variable region that "comprises” a specific sequence it is also intended to encompass the antibody variable region composed of the specific sequence.
  • antibody refers to a polypeptide comprising light chain and heavy chain immunoglobulin variable regions, which specifically recognize and bind to an antigen.
  • the antibody of the present invention is a full-length antibody, consisting of two heavy chains and two light chains, wherein each heavy chain is composed of a heavy chain variable region (abbreviated as VH herein) and a heavy chain constant region, each Light chain can be The variable region (abbreviated as VL herein) and the light chain constant region are composed.
  • the antibody of the present invention can also refer to an antigen-binding fragment of an antibody.
  • antibody preparation refers to a preparation that is in a form that allows the biological activity of the antibody as an active ingredient to be effectively exerted, and does not contain unacceptable toxicity to the subject to which the preparation is administered. Other components. Such antibody preparations are usually sterile.
  • pharmaceutically acceptable excipients are included in antibody preparations.
  • a "pharmaceutically acceptable" excipient is an agent that can be reasonably administered to the mammal under test so that an effective dose of the active ingredient used in the formulation can be delivered to the subject.
  • the concentration of the excipient is adapted to the mode of administration, for example, it may be acceptable for injection.
  • anti-CD47 antibody preparation also referred to herein simply as “the antibody preparation of the present invention” means a preparation containing an anti-CD47 antibody as an active ingredient and a pharmaceutically acceptable excipient. After the combination, the anti-CD47 antibody as the active ingredient is suitable for therapeutic or preventive administration to humans or non-human animals.
  • the antibody preparation of the present invention can be prepared, for example, as an aqueous liquid preparation, for example, a ready-to-use pre-filled syringe, or prepared as a lyophilized preparation, which is carried out by dissolving and/or suspending in a physiologically acceptable solution immediately before use Reconstitution (ie, reconstitution).
  • the anti-CD47 antibody formulation is in the form of a liquid formulation.
  • a “stable” antibody preparation means that the antibody in the preparation retains an acceptable degree of physical and/or chemical stability after storage under specific conditions. Although the antibody contained in the antibody preparation may not maintain 100% of its chemical structure after storage for a specific time, it usually maintains about 90%, about 95%, about 96%, about 97%, about 98% after storage for a specific time. Or about 99% of the structure or function of the antibody, the antibody preparation is considered “stable.” In some specific embodiments, the anti-CD47 antibody preparation of the present invention exhibits low to undetectable antibody aggregation or degradation or chemical modification during the manufacturing, preparation, transportation, and long-term storage, so that there is little or no antibody. The biological activity of the CD47 antibody is lost, showing high stability.
  • the anti-CD47 antibody preparation of the present invention substantially retains its physical and chemical stability after storage.
  • the liquid formulation of the present invention can be stable at room temperature or at 40°C for at least 1 month, and/or at 2-8°C for at least 24 months.
  • the stability can be measured at a selected temperature and selected storage time. For example, the storage time can be selected based on the expected shelf life of the formulation. Or an accelerated stability test can be used. In some embodiments, stability testing is performed by performing various stress tests on antibody preparations.
  • the formulated anti-CD47 antibody preparation can be filled into a 5 mL glass bottle for shaking stress to detect the shaking/shear stability of the antibody; or the formulated anti-CD47 antibody preparation can be filled into a glass vial for testing Antibody stability under high temperature stress. After a period of storage, the preparation does not show aggregation, precipitation, turbidity and/or denaturation; or shows very little aggregation, precipitation, turbidity and/or denaturation, it can be considered that the antibody "maintains its physical stability" in the preparation.
  • the accumulation of antibodies in the preparation can potentially lead to an increased immune response in the patient, leading to safety issues. Therefore, there is a need to minimize or prevent aggregation of antibodies in the formulation.
  • Light scattering methods can be used to determine visible aggregates in the formulation.
  • SEC can be used to determine soluble aggregates in preparations.
  • the stability of the preparation can be indicated by visually inspecting the appearance, color, and/or clarity of the preparation, or detecting the turbidity of the preparation by the OD350nm method, or measuring the purity of the preparation by the non-reducing CE-SDS method. In one embodiment, the stability of the formulation is measured by determining the percentage of antibody monomers in the formulation after storage at a specific temperature for a specific time, wherein the higher the percentage of antibody monomers in the formulation, the higher the stability of the formulation .
  • an "acceptable degree" of physical stability can mean that at least about 92% of the anti-CD47 antibody monomer is detected in the preparation after storage at a specific temperature for a specific time.
  • an acceptable degree of physical stability means at least about 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of the anti-CD47 antibody body.
  • the specific temperature at which the pharmaceutical preparation is stored can be any temperature from about -80°C to about 45°C, for example, when stored at about -80°C, about -30°C, or about -20°C , About 0°C, about 4°C-8°C, about 5°C, about 25°C, about 35°C, about 37°C, about 40°C, about 42°C, or about 45°C.
  • about 40°C ⁇ 2°C for 1 month at least about 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of anti-CD47 antibody monomers are detected.
  • the pharmaceutical preparation is considered stable.
  • the pharmaceutical preparations are considered To be stable. If after 9 months of storage at about 5°C, at least about 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of anti-CD47 antibody monomers are detected, the pharmaceutical preparations will be considered To be stable. After a period of storage, if the antibody in the preparation does not show a significant chemical change, it can be considered that the antibody "maintains its chemical stability" in the preparation. Most chemical instabilities result from the formation of covalently modified forms of antibodies (for example, charge variants of antibodies).
  • the stability of the formulation is measured by determining the percentage change in the charge variant of the antibody in the formulation after storage at a specific temperature for a specific time, wherein the smaller the change, the higher the stability of the formulation.
  • the "acceptable degree" of chemical stability can mean that the percentage change of the charge variant (such as the main component or acidic component or alkaline component) in the preparation after storage at a specific temperature for a specific time does not exceed 30%, such as 20% .
  • the storage temperature of the pharmaceutical preparation can be any temperature from about -80°C to about 45°C, for example, when stored at about -80°C, about -30°C, about -20°C, About 0°C, about 4°C-8°C, about 5°C, about 25°C, or about 45°C.
  • the pharmaceutical preparation can be considered stable.
  • the pharmaceutical preparation can also be regarded as stable.
  • the pharmaceutical preparation can also be considered stable.
  • the term "lyophilized preparation” refers to a composition obtained or obtainable by freeze-drying a liquid preparation. Preferably, it is a solid composition having a water content of less than 5%, preferably less than 3%.
  • reconstituted preparation refers to a liquid preparation obtained by dissolving and/or suspending a solid preparation (such as a lyophilized preparation) in a physiologically acceptable solution.
  • room temperature refers to a temperature of 15°C to 30°C, preferably 20°C to 27°C, and more preferably 25°C.
  • Stress conditions refer to environments that are chemically and/or physically unfavorable to the antibody protein, which can lead to unacceptable instability of the antibody protein.
  • High temperature stress refers to storing the antibody preparation at room temperature or even higher temperature (for example, 40°C ⁇ 2°C) for a period of time. Through the accelerated test of high temperature stress, the stability of the antibody preparation can be checked.
  • parenteral administration means a mode of administration other than enteral and local administration, usually by injection Or infusion methods, and include, but are not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, and joint Internal, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injections and infusions.
  • the stable anti-CD47 antibody formulations of the invention are administered to the subject parenterally.
  • the anti-CD47 antibody formulation of the present invention is administered to a subject by subcutaneous, intradermal, intramuscular or intravenous injection.
  • the present invention provides a stable liquid antibody preparation comprising (i) a recombinant fully human anti-CD47 monoclonal antibody; (ii) a buffer, (iii) a stabilizer, and (iv) a surfactant, the pH of the antibody preparation It is about 5.0-6.0.
  • the liquid antibody preparation of the present invention is in the form of an injection preparation.
  • an "anti-CD47 antibody” refers to an antibody that can bind to a CD47 molecule with sufficient affinity so that the antibody can be used as a therapeutic and/or preventive agent that targets the CD47 molecule.
  • the antibody of the present invention is a recombinant fully human antibody.
  • the terms "fully human antibody” or “human antibody” are used interchangeably herein, and refer to that the antibody includes variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences, and if the antibody Contains constant regions, which are also derived from human germline immunoglobulin sequences.
  • the human antibody of the present invention may include amino acids that are not encoded by human germline immunoglobulin sequences (for example, mutations introduced by random or point-specific mutagenesis in vitro or somatic mutations in vivo).
  • human antibody is not intended to include antibodies in which CDR sequences are derived from germlines of other mammalian species (eg, mice) and transplanted into human framework sequences.
  • recombinant human antibody includes all human antibodies that are prepared, expressed, produced or isolated by recombinant means. These recombinant human antibodies have framework regions and CDR regions derived from variable regions of human germline immunoglobulin sequences.
  • the recombinant human antibody can be subjected to in vitro mutagenesis (or in vivo somatic mutagenesis when a human Ig sequence transgenic animal is used), and the amino acid sequences of the VH and VL regions of the recombinant antibody thus obtained are Although derived from and related to human germline VH and VL sequences, they do not naturally exist in the human antibody germline repertoire in vivo.
  • the anti-CD47 antibody capable of high affinity, for example, 10- 7 M or less, preferably in 10_ 9 M to 10_ 1Q M K D of specificity It binds to human CD47, and thereby mediates a highly effective blocking effect on the binding of CD47 and its ligands.
  • the antibody CD47 antibody of the present invention comprises: SEQ ID NO: 1 or a heavy chain variable region (VH) having at least 90% identity; and SEQ ID NO: 2 or at least 90% Identical light chain variable region (VL).
  • a "variable region” or “variable domain” is a domain in the heavy or light chain of an antibody that participates in the binding of the antibody to its antigen.
  • the heavy chain variable region (VH) and light chain variable region (VL) can be further divided into hypervariable regions (HVR, also called complementarity determining regions (CDR)), with more conservative regions (ie , Framework area (FR)).
  • HVR hypervariable regions
  • FR Framework area
  • Each VH and VL is composed of three CDRs and 4 FRs, arranged in the following order from the amino terminal to the carboxy terminal: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the anti-CD47 antibody of the present invention comprises the HCDR1 of the heavy chain variable region of SEQ ID NO: 1.
  • CDR region or “CDR” (herein used interchangeably with hypervariable region “HVR”) are the amino acid regions in the variable region of an antibody that are mainly responsible for binding to an epitope.
  • the CDRs of the heavy and light chains are usually called CDR1, CDR2, and CDR3, and are numbered sequentially from the N-terminus.
  • the CDRs located in the variable domain of the antibody heavy chain are called HCDR1, HCDR2, and HCDR3, and the CDRs located in the variable domain of the antibody light chain are called LCDR1, LCDR2, and LCDR3.
  • CDR complementarity determining region
  • CDR Kabat complementarity determining region
  • Chothia refers to the position of structural loops (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)).
  • AbM HVR It is a compromise between Kabat HVR and Chothia structural loops and is used by Oxford Molecular's AbM antibody modeling software. "Contact" HVR is based on the analysis of available complex crystal structures. HVR can also be determined based on having the same Kabat numbering position as a reference CDR sequence (such as an exemplary CDR disclosed herein).
  • the CDR of the antibody of the present invention is bounded by the Kabat rule, as shown in Table A(1) below.
  • the CDR of the antibody of the present invention determines the boundary by combining Kabat, AbM, Chothia, and empirical factors.
  • HCDR1 is the sequence of positions H27-H35 in the kabat numbering system
  • HCDR3 is the sequence of positions H93-H102 in the kabat numbering system
  • HCDR2 and LCDR1, LCDR2 and LCDR3 are determined by Kabat rules .
  • the CDR boundaries of the variable region of the same antibody obtained based on different assignment systems may be different. That is, the CDR sequences of the variable regions of the same antibody defined under different assignment systems are different. Therefore, when it comes to defining antibodies with specific CDR sequences defined in the present invention, the scope of the antibodies also covers antibodies whose variable region sequences include the specific CDR sequences, but due to the application of different schemes (for example, Different assignment system rules or combinations) cause the claimed CDR boundary to be different from the specific CDR boundary defined in the present invention.
  • Antibodies with different specificities have different CDRs.
  • the CDR is different from antibody to antibody, there are only a limited number of amino acid positions within the CDR that directly participate in antigen binding.
  • the minimum overlap area can be determined, thereby providing the "minimum binding unit" for antigen binding.
  • the minimum binding unit can be a sub-part of the CDR.
  • the structure of the antibody and protein folding can determine the residues of the rest of the CDR sequence. Therefore, the present invention also considers any CDR variants given herein. For example, in a CDR variant, the amino acid residue of the smallest binding unit can remain unchanged, while the remaining CDR residues defined by Kabat or Chothia can be replaced by conservative amino acid residues.
  • the anti-CD47 antibody of the present invention may comprise a heavy chain variable region (VH) having at least 90%, 95% or 98% or higher identity with SEQ ID NO: 1; and/or with SEQ ID NO: 1 ID NO: 2
  • VH heavy chain variable region
  • VL light chain variable region
  • sequence identity refers to the degree of sequence identity on a nucleotide-by-nucleotide or amino acid-by-amino-acid basis in the comparison window.
  • the “percentage of sequence identity” can be calculated in the following way: Compare the two optimally aligned sequences in the comparison window to determine the presence of the same nucleic acid base (for example, A, T, C, G, I) in the two sequences ) Or the same amino acid residue (for example, Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys, and Met) to obtain the number of matching positions, divide the number of matching positions by the total number of positions in the comparison window (ie, window size), And multiply the result by 100 to produce the percent sequence identity.
  • the same nucleic acid base for example, A, T, C, G, I
  • the same amino acid residue for example, Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys, and Met
  • the optimal alignment for determining the percent sequence identity can be achieved in a variety of ways known in the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine the appropriate parameters for sequence alignment, including any algorithm required to achieve maximum alignment within the full-length sequence being compared or within the target sequence region.
  • the VH sequence of the antibody of the present invention has no more than 10, preferably no more than 5, 4 or 3 different residues compared to SEQ ID NO:1, preferably the different residues are Conservative amino acid substitutions.
  • the VL sequence of the antibody of the present invention has no more than 10, preferably no more than 5, 4, or 3 different residues compared to SEQ ID NO: 2, preferably the different residues are Conservative amino acid substitutions.
  • Constant substitution refers to an amino acid change that results in the substitution of a certain amino acid with a chemically similar amino acid. It is well known in the art to provide conservative substitution tables of functionally similar amino acids.
  • the conservatively substituted residues are derived from conservative substitution table X below, preferably the preferred substituted residues shown in Table X.
  • the antibodies of the invention are antibodies in the form of IgG4.
  • Antibody in the form of IgG refers to the form of IgG to which the constant region of the heavy chain of the antibody belongs.
  • the heavy chain constant regions of all antibodies of the same type are the same, and the heavy chain constant regions of antibodies of different types are different.
  • an antibody in the form of IgG4 means that the Ig domain of its heavy chain constant region is the Ig domain of IgG4.
  • the anti-CD47 antibody of the present invention is the anti-CD47 monoclonal antibody ADI-26630 disclosed in Chinese application CN201710759828.9 (August 29, 2017), which has the heavy chain of SEQ ID NO: 9 and The light chain of SEQ ID NO: 10.
  • the anti-CD47 antibody is recombinantly expressed in CHO cells and purified IgG4 type antibody.
  • the antibody in the liquid formulation of the present invention exhibits significant anti-tumor activity.
  • the antibody in a mouse tumor model transplanted with human Burkitt’s lymphoma cells, such as intraperitoneal injection of 5 mg/kg, once every two days for two weeks, the antibody can lead to a tumor growth inhibition rate of about 50% or higher , Such as 100%; and/or tumor disappearance rate reaches more than 60%.
  • the amount of the antibody or antigen-binding fragment thereof contained in the antibody preparation of the present invention can be changed according to the specific purpose characteristics of the preparation, the specific environment, and the specific purpose of using the preparation.
  • the antibody formulation is a liquid formulation, which may contain about 1-150 mg/mL, preferably about 10-100 mg/mL, for example, about 15, 20, 25, 30, 35, 40, 45 , 50, 55, 60 mg/mL anti-CD47 antibody.
  • the present invention relates to a preparation having a high concentration of anti-CD47 antibody, for example, containing 50-150 mg/mL of anti-CD47 antibody. It is known in the art that such high-concentration antibody formulations can be diluted before injection, for example, if a lower antibody concentration is required for a specific therapeutic or prophylactic intervention or when treating patients of lower body weight including children. A suitable concentration can be 25 mg/mL or 10 mg/mL. Alternatively, the original formulation can be produced at such a low concentration.
  • the buffer is an agent that can maintain the pH of the solution within an acceptable range.
  • the buffering agent used in the preparation of the present invention can control the pH of the preparation of the present invention in the pH range of about 5.0-6.0, for example, about 5.2-5.8 pH, preferably 5.3-5.7.
  • the antibody preparation of the present invention has a pH of about 5.0, 5.2, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9 or 6.0, preferably a pH of about 5.5.
  • the buffer used in the present invention is a histidine buffer, such as a buffer system composed of histidine and histidine hydrochloride.
  • the concentration of the buffer in the antibody preparation of the present invention is about 5-100 mM, preferably about 10-50 mM, for example, about 10-20 mM.
  • the buffer is about 10-20 mM histidine buffer.
  • Suitable stabilizers for use in the present invention may be selected from sugars, polyols and amino acids and combinations thereof.
  • Sugars used as stabilizers include, but are not limited to, sucrose and trehalose.
  • the polyol used as a stabilizer includes but is not limited to sorbitol.
  • the amino acids used as stabilizers include but are not limited to arginine.
  • the liquid formulation of the present invention contains sucrose as a stabilizer.
  • the amount of sucrose in the liquid preparation of the present invention may be about 50-100 mg/ml, preferably 70-90 mg/ml, for example 80 mg/ml.
  • the liquid formulation of the present invention contains trehalose as a stabilizer.
  • the amount of trehalose in the liquid preparation of the present invention may be about 50-100 mg/ml, preferably 70-90 mg/ml, for example 80 mg/ml.
  • the liquid formulation of the present invention contains arginine as a stabilizer.
  • the amount of arginine in the liquid preparation of the present invention may be about 80-1 10mM, especially about 100mM, for example about 21.1 mg/ml arginine hydrochloride.
  • the liquid formulation of the present invention contains sorbitol as a stabilizer.
  • the amount of sorbitol in the liquid preparation of the present invention may be about 10-100 mg/ml, preferably 20-70 mg/ml, for example 30-60 mg/ml.
  • sorbitol may be present in an amount of about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65 or 70 mg/ml, preferably in an amount of about 40 mg/ml.
  • the liquid formulation of the present invention contains a combination of sorbitol and arginine as a stabilizer.
  • sorbitol may be present in an amount of about 5-60 mg/ml, preferably 10-30 mg/ml, such as 10-20 mg/ml, for example, 5, 10, 12, 15, 17, 20, 22, 25mg/ml.
  • arginine may be present in an amount of about 80-1 10mM, especially about 100mM.
  • the liquid preparation of the present invention contains about 10-20 mg/ml sorbitol and about 10-30 mg/ml arginine hydrochloride. More preferably, the liquid preparation of the present invention contains about 15 mg/ml sorbitol and about 21.1 mg/ml arginine hydrochloride.
  • the term "surfactant” refers to an organic substance with an amphiphilic structure; that is, they are composed of groups with opposite solubility tendencies, usually oil-soluble chain and water-soluble ionic groups. group.
  • the surfactant in the liquid formulation of the present invention is a nonionic surfactant, for example, alkyl poly(ethylene oxide).
  • Specific nonionic surfactants that can be included in the formulation of the present invention include, for example, polysorbates, such as polysorbate-20, polysorbate-80, polysorbate-60, or polysorbate-40; Nick waits.
  • the liquid formulation of the present invention contains polysorbate-20 as a surfactant.
  • the amount of the surfactant contained in the antibody preparation of the present invention can be changed according to the specific purpose characteristics of the preparation, the specific environment, and the specific purpose of using the preparation.
  • the formulation may contain about 0.01-5 mg/ml, preferably about 0.1-2 mg/ml, such as about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0 mg /ml of polysorbate-20, preferably about 0.3 mg/ml of polysorbate-20.
  • the antibody liquid preparation of the present invention may or may not contain other excipients.
  • the antibody liquid formulation of the present invention contains EDTA or a salt thereof.
  • the antibody liquid formulation of the present invention does not contain EDTA or its salt.
  • the antibody liquid formulation of the present invention added with EDTA or its salt has considerable stability compared to the corresponding formulation without EDTA or its salt.
  • excipients may also be used in the formulation of the present invention.
  • the excipients include, for example, flavoring agents, antimicrobial agents, sweeteners, antistatic agents, antioxidants, bright stocks and the like.
  • These and other known pharmaceutical excipients and/or additives suitable for the formulation of the present invention are well known in the art, for example, listed in "The Handbook of Pharmaceutical Excipients, 4th Edition, Rowe et al., eds., American Pharmaceuticals Association (2003); and Remington: the Science and Practice of Pharmacy, 21st edition, edited by Gennaro, Lippincott Williams & Wilkins (2005)''.
  • the present invention provides stable formulations containing antibodies.
  • the antibodies used in the formulations of the present invention can be prepared using techniques known in the art for antibody production. For example, antibodies can be produced recombinantly. In a preferred embodiment, the antibody of the invention is recombinantly produced in CHO cells.
  • Example 4 Tugcu et al. (Maximizing productivity of chromatography steps for purification of monoclonal antibodies, Biotechnology and Bioengineering 99 (2008) 599-613.) describe the use of ion exchange chromatography (anion IEX and/or cation CEX chromatography) after the protein A capture step Monoclonal antibody three-column purification method. Kelley et al.
  • recombinantly produced monoclonal antibodies can be purified by conventional purification methods to provide pharmaceutical substances with sufficient reproducibility and moderate purity for the preparation of antibody preparations.
  • a commercially available protein concentration filter such as Amicon's ultrafiltration device can be used to concentrate the supernatant from the expression system.
  • the antibody can be purified using methods such as chromatography, dialysis, and affinity purification.
  • Protein A is suitable as an affinity ligand for the purification of IgGl, IgG2 and IgG4 type antibodies.
  • Other antibody purification methods, such as ion exchange chromatography can also be used. After obtaining antibodies of sufficient purity, preparations containing antibodies can be prepared according to methods known in the art.
  • the following steps can be used for preparation: (1) After fermentation, the fermentation broth is centrifuged to clarify and remove impurities such as cells to obtain the supernatant; (2) affinity chromatography (for example, specific for IgGl, IgG2 and IgG4 antibodies) Affinity protein A column) capture antibody; (3) virus inactivation; (4) purification and purification (usually CEX cation exchange chromatography can be used) to remove impurities in the protein; (4) virus filtration (to make the virus titer Reduce, for example, 4 logl0 or more); (5) Ultrafiltration/diafiltration (can be used to replace the protein in a preparation buffer that is conducive to its stability and concentrate to a suitable concentration for injection). See, for example, B. Minow, P. Rogge, K. Thompson, BioProcess International, Vol. 10, No. 6, 2012, pp. 48-57. III. Analytical method of preparation
  • antibodies may undergo aggregation, degradation or chemical modification, resulting in antibody heterogeneity (including size heterogeneity and charge heterogeneity), aggregates and fragments, etc., thereby affecting the quality of antibody preparations. Therefore, it is necessary to monitor the stability of antibody preparations.
  • Various methods are known in the art that can be used to test the stability of antibody preparations. For example, methods such as non-reduced CE-SDS and SEC-HPLC can be used to analyze the purity of antibody preparations and evaluate the aggregation level of antibodies; Capillary isoelectric focusing (cIEF), imaging capillary isoelectric focusing (iCIEF), and Ion exchange chromatography (IEX), etc., analyze charge variants in antibody preparations.
  • cIEF Capillary isoelectric focusing
  • iCIEF imaging capillary isoelectric focusing
  • IEX Ion exchange chromatography
  • the appearance of the preparation can be visually inspected to quickly determine the stability of the preparation.
  • the OD350nm method can also be used to detect changes in the turbidity of the preparation, which can give information about the amount of soluble and insoluble aggregates.
  • ultraviolet spectrophotometry UV method
  • UV method can be used to detect changes in the protein content of the preparation.
  • the non-reduced CE-SDS method is a method for measuring the purity of monoclonal antibodies using capillary as the separation channel.
  • CE-SDS protein migration is driven by the surface charge caused by SDS binding, and the surface charge is proportional to the molecular weight of the protein. Since all SDS-protein complexes have similar mass-to-charge ratios, electrophoretic separation based on molecular size or hydrodynamic radius can be achieved in the molecular sieve gel matrix of the capillary. This method has been widely used to monitor the purity of denatured intact antibodies.
  • the test sample is mixed with SDS sample buffer and iodoacetamide.
  • the mixture can be incubated at 68-72°C for about 10-15 minutes, and the supernatant centrifuged after cooling to room temperature for analysis.
  • a UV detector is used to detect the migration of the protein to obtain an electrophoresis spectrum.
  • the purity of the antibody preparation can be calculated as the percentage of the peak area of the main IgG peak to the sum of all peak areas.
  • Size exclusion high performance liquid chromatography is another important method for monoclonal antibody standards and quality control. This method is mainly based on the molecular size or hydrodynamic radius difference to separate molecules.
  • SEC-HPLC antibodies can be separated into three main forms: high molecular weight form (HMMS), main peak (mainly antibody monomer), and low molecular weight form (LMMS).
  • HMMS high molecular weight form
  • LMMS low molecular weight form
  • Antibody purity can be calculated as the percentage of the main peak area on the chromatogram of the sum of all peak areas.
  • the SEC-HPLC method the percentage of antibody monomers in the preparation product can be measured, and the content information of soluble aggregates and shears can be given.
  • Imaging capillary isoelectric focusing electrophoresis can be used to analyze the charge heterogeneity of monoclonal antibodies. This method can provide quantitative distribution of charge variants.
  • iCIEF achieves molecular separation based on the charge difference (apparent pi value) of molecules in the pH gradient.
  • the separation column is usually a short capillary (for example, a silica capillary with a length of 5 cm and an inner diameter of 100 pm).
  • the protein is focused in the capillary column under high voltage, and the focusing is performed by a whole column imaging detection system operating at 280 nM. Real-time online monitoring.
  • One advantage of this technology is that the whole-column detection system can simultaneously record various charge variants of antibody samples.
  • the sample is mixed with urea and icIEF buffer, where the buffer contains methylcellulose, pi molecular weight standard and ampholytes c. Then, it can be used in an iCIEF analyzer such as iCE280 analyzer (Protein Simple, Santa Clara, CA), using an iCIEF column, such as an iCIEF column assembled by ProtionSimple, after the sample is focused for a certain period of time, the absorbance at 280 nm is measured to obtain a spectrum of the focused mAb charge variant.
  • iCIEF analyzer such as iCE280 analyzer (Protein Simple, Santa Clara, CA)
  • an iCIEF column such as an iCIEF column assembled by ProtionSimple
  • the protein-related peaks eluted before the main peak are classified as acidic components; in contrast, the protein-related peaks eluted after the main peak are classified as basic components.
  • the relative amounts of the main component, acidic component, and basic component can be expressed as a percentage of the total peak area.
  • the charge variant of the antibody in the antibody preparation can also be determined by cation exchange high performance liquid chromatography (CEX-HPLC).
  • CEX-HPLC cation exchange high performance liquid chromatography
  • Accelerated stability studies can be used to check the stability properties of products, which is conducive to the screening of stable pharmaceutical formulations.
  • the formulation sample can be placed at an elevated temperature, for example, about 40°C ⁇ 2°C ⁇ 25°C ⁇ 2°C for accelerated stability studies.
  • Detection indicators can include appearance, visible foreign matter, protein content, turbidity, purity (SEC-HPLC method, non-reduced CE-SDS method) and charge variants (iCIEF method).
  • a shaking test can be performed to examine the shaking/shear stability of the formulation.
  • the preparation samples are dispensed into vials, stoppered and capped, then set out, and perform an oscillation test, such as shaking at 650 r/min for 3-5 days, and then check the appearance, protein content, and
  • the efficacy or biological activity of the antibody can be tested.
  • the ability of the antibody in the preparation to bind to its antigen can be tested.
  • Those skilled in the art know that a variety of methods can be used to quantify the specific binding of an antibody to an antigen, such as immunoassay tests, ELISA, and the like.
  • the anti-CD47 antibody preparation of the present invention is stable. In one embodiment, after storage at about 25°C, 37°C, 40°C, or 45°C for at least 1 month or 2 months, for example, after storage at 40°C ⁇ 2°C for 1 month
  • the purity of the anti-CD47 antibody in the antibody preparation of the present invention is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more. Resistance chromatography or by non-reduced CS-SDS.
  • At least 50%, preferably at least 55% of the anti-CD47 antibody in the antibody preparation of the present invention is in a non-alkaline and non-acidic form (ie, a main peak or a main charge form), as determined by imaging capillary focus electrophoresis.
  • the antibody preparation of the present invention comprising an anti-CD47 antibody of the present invention can be used to treat, ameliorate or prevent various CD47-related diseases or disorders.
  • a "CD47-related disease or disorder” herein refers to a disease or disorder that can be treated (e.g., improved) or prevented with the anti-CD47 antibody of the present invention. Any disease or condition that can benefit from treatment with the antibody of the present invention is applicable to the present invention.
  • CD47 is a pluripotent molecule that plays an important role in the process of tumor cells evading immune surveillance. Blocking the CD47 signaling pathway can effectively stimulate the phagocytosis of macrophages on tumor cells, which can be used for tumor immunotherapy. Therefore, the preparations of the present invention containing anti-CD47 antibodies are particularly useful for treating, improving or preventing CD47-related cancers.
  • the cancer includes, for example, but not limited to: various hematological tumors and solid tumors, such as leukemia, including acute lymphoblastic leukemia, B-lymphocytic chronic leukemia and acute lymphocytic leukemia; non-Hodgkin's lymphoma; bone tumor; leiomyosarcoma ; Astrocytoma; Breast cancer; Ovarian cancer; Glioblastoma. See, for example, International Journal of Oncology, November 2013, Vol. 40, No. 11, pages 817-819.
  • the antibody preparations of the present invention are used to treat, ameliorate, or prevent CD47-related lymphoma, such as Burkitt's lymphoma.
  • the present invention also provides the use of the preparation of the present invention in the preparation of a medicament, wherein the medicament is used to deliver anti-CD47 antibodies to mammals, or to treat, prevent or ameliorate one or more of the aforementioned diseases and disorders.
  • the mammal is a human.
  • the antibody formulation of the present invention can be administered to a subject or patient in various ways.
  • administration can be by infusion or by syringe.
  • the present invention provides a delivery device (for example, a syringe), which includes the antibody preparation of the present invention (for example, a pre-filled syringe).
  • the patient will receive an effective amount of anti-CD47 antibody as the main active ingredient, that is, an amount sufficient to treat, ameliorate or prevent the target disease or condition.
  • the therapeutic effect may include reducing physical symptoms.
  • the optimal effective amount and concentration of antibodies for any particular subject will depend on Due to a variety of factors, including the patient’s age, weight, health status and/or gender, the nature and extent of the disease, the activity of a specific antibody, the clearance rate of the specific antibody, and also any possible combinations administered with the antibody preparation Other treatments.
  • the effective amount delivered can be determined within the judgment of the clinician.
  • the effective dose may be about 0.005 mg/kg body weight to about 50 mg/kg body weight, or about 0.1 mg/kg body weight to about 20 mg/kg body weight.
  • the application of known antibody-based drugs can provide certain guidance.
  • the dosage can be a single dose schedule or a multiple dose schedule.
  • pre-prescription and prescription screening are designed Two-stage test.
  • the materials and methods used in the test are as follows: Materials and methods
  • TSK-gel SuperSW mAb HR (7.8x300 mm, 4(j,m) type analytical column
  • TSK-gel SuperSW (6.0x40 mm, 4(j,m) guard column)
  • Sample tray temperature about 10°C
  • CE-SDS Non-reducing sodium dodecyl sulfate capillary electrophoresis
  • the non-reduced CE-SDS measurement can be performed as follows: Add approximately sample (protein content: 1mg/ml) into 14 ul non-reduced sample buffer (including 700 ul pH 6.5 sample buffer, add 31.3 ul 250mM NEM (N -Ethyl maleimide)), add 28 ul ultrapure water, mix well, and then incubate at 70°C for 10 minutes. After incubation, the samples were cooled to room temperature and then transferred to 96-well plates.
  • CE-SDS separation was performed on LabChip GXIITouch (PerkinElmer), using a protein chip.
  • the analysis method is HT Antibody Analysis 200, and the sample tray type is BioRad 96 HSP-96xx (Sip 4 mm). The protein migration is monitored by fluorescence.
  • Imaging capillary isoelectric focusing electrophoresis (icIEF method)
  • the icIEF test can be performed as follows: Dilute (or desalt) the antibody sample to approximately 1 mg/ml. Take 20 (J sample) and add 780 icIEF buffer, which contains urea, arginine (Protein Simple), pi marker standard (Protein Simple, Santa Clara, CA) and Pharmalytes (GE Healthcare Bio-Science, Pittsburgh, PA) ). On the Maurice C. Analyzer (Protein Simple, Santa Clara, CA), the FC-coated iCIEF column was used to generate the imaging capillary isoelectric focusing spectrum. The sample was focused for a total of 8 minutes, using Empower version 3 software (Waters, Milford) , MA), integrate the absorbance of the 280nm focused protein. Example 1. Preparation and purification of anti-CD47 antibodies
  • the anti-CD47 antibody ADI-26630 was prepared and purified according to CN201710759828.9.
  • the antibody consists of the heavy chain sequence of SEQ ID NO: 9 and the light chain sequence of SEQ ID NO: 10, and is an IgG4 type antibody.
  • the antibody was recombinantly expressed in CHO cells and purified.
  • the sample used for the pre-prescription test purified by CEX (Cation Exchange Chromatography), the sample has a protein content of 12.2mg/ml.
  • CEX Cation Exchange Chromatography
  • pass CEX cation exchange chromatography After purification to obtain a protein content of 15.7 mg/ml, it was concentrated to 100 mg/ml by diafiltration.
  • Example 2 Pre-prescription test
  • the determination standard that the quality of the sample detection index value has not changed compared with the initial value is set to determine whether the sample has changed. See Table 2 for details.
  • N/ A means not tested or not applicable, the same below.
  • Test items include appearance, protein content, turbidity and purity (SEC-HPLC method, non-reduced CE-SDS method). See Table 7 for the specific test plan.
  • the formulation formulation was determined to be histidine-histidine hydrochloride buffer system, the pH was selected as 5.5, and the surfactant was Polysorbate 20. On this basis, the following prescription screening test was carried out.
  • a total of 4 prescriptions were designed, and the detailed prescription information is shown in Table 9.
  • the protein concentration of each prescription was diluted to about 120 mg/ml, and polysorbate 20 was added to make the final concentration 0.3 mg/ml.
  • the detection indicators are appearance, visible foreign matter, protein content, turbidity, purity (SEC-HPLC method, non-reduced CE-SDS method) and charge variant (iCIEF method).
  • Prescription 1 0.4 mg/ml histidine, 1.5 mg/ml histidine hydrochloride, 40 mg/ml sorbitol, 0.3 mg/ml polysorbate 20, pH 5.5 prescription 2 0.4 mg/ml histidine, 1.5 mg /ml histidine hydrochloride, 80 mg/ml sugar, 0.3 mg/ml polysorbate 20, pH 5.5 prescription 3 0.4 mg/ml histidine, 1.5 mg/ml histidine hydrochloride, 80 mg/ml seaweed Sugar, 0.3 mg/ml polysorbate 20, pH 5.5 prescription 4 0.4 mg/ml histidine, 1.5 mg/ml histidine hydrochloride, 21.1 mg/ml arginine hydrochloride, 0.3 mg/ml polysorbate vinegar 20, pH5.5 detailed test conditions and sampling plan are shown in Table 10.
  • the judgment standard that the quality of the sample detection index value has not changed compared with the initial value is set to determine whether the sample has changed, see Table 11 for details.
  • the sample was divided into two parts, one part was added with EDTA-2Na to make the final concentration 0.01 mg/ml; the other part was not added with EDTA-2Na.
  • the protein concentration of each prescription is about 100 mg/ml.
  • the detection indicators are appearance, visible foreign matter, protein content, turbidity, purity (SEC-HPLC method, non-reduced CE-SDS method) and charge variant (iCIEF method). Table 16. Prescription information for antioxidant screening test
  • test index data shows that the two prescriptions have good stability and there is no difference between the prescriptions, that is, whether to add EDTA-2Na has little effect on product stability.
  • prescription B was finally selected as the final prescription of the antibody preparation.
  • the antibody protein is stable at pH 5.0 ⁇ 6.0; the surfactant polysorbate 20 can ensure that the product has good oscillation stability; among the stabilizers, sorbitol has a better effect and can more effectively inhibit the protein at 40
  • the high temperature antioxidant test found that there is no need to add EDTA-2Na in the prescription; in addition, adding an appropriate amount of arginine can reduce the viscosity of high-concentration protein products.
  • the preferred preparation scheme is: 100 mg/ml recombinant fully human anti-differentiation antigen cluster 47 (CD47) monoclonal antibody, 0.4 mg/ml histidine, 1.5 mg/ml histidine hydrochloride, 15.0 mg/ml ml sorbitol, 21.1 mg/ml arginine hydrochloride, 0.3 mg/ml polysorbate 20, pH 5.5.
  • CD47 fully human anti-differentiation antigen cluster 47

Landscapes

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

Abstract

本发明涉及包含抗CD47抗体的制剂,尤其涉及包含抗体、缓冲剂、稳定剂和表面活性剂的药物制剂。此外,本发明还涉及这些制剂的疾病治疗或预防用途。

Description

包含抗 CD47抗体的制剂及其制备方法和用途 技禾领域
本发明涉及抗体制剂领域。 更具体而言, 本发明涉及包含重组全人源抗分化抗原簇 47(CD47)单克隆抗体的药物制剂, 尤其是稳定的高浓度抗体液体制剂, 以及用于制备所述药 物制剂的方法, 以及所述药物制剂的治疗和 /或预防用途。 背景技术
分化抗原簇 47 (CD47) , 也被称为整联蛋白相关蛋白 (IAP) , 是免疫球蛋白超家族成 员。 不同的研究表明, 几乎所有的肿瘤细胞和组织都高表达 CD47。 目前已经报道了多个抗 CD47抗体被开发用于各种肿瘤和 /或癌症的治疗中。
在 CN201710759828.9的中国申请 (2017年 8月 29递交) 中, 公开了一系列重组全人源 抗分化抗原簇 47(CD47)单克隆抗体。 这些抗体能够改善巨噬细胞的呑噬作用, 并具有显著的 抗肿瘤活性, 能够显著抑制肿瘤的生长, 甚至能使得肿瘤完全消失。 此外, 这些抗体还表现 出显著降低的血细胞凝集作用, 因此在临床治疗中将具有显著降低的副作用。
单克隆抗体对 I巴标具有高度的特异性, 并且一般而言比小分子药物更为有效。 然而, 基 于单克隆抗体的药品开发具有其自身的挑战性。 单克隆抗体比传统的有机和无机药物更为复 杂, 并且通常具有与其它蛋白质类生物药相似的降解模式。 广义上, 抗体蛋白的降解可以分 为两种主要的类型: 物理不稳定性(:涉及蛋白质高级结构的改变;)和化学不稳定性(:涉及蛋白质 的各种化学修饰)。 化学不稳定性可以由脱酰胺、 外消旋、 水解、 氧化、 P-消除或二硫键交换 等引起, 其中最常见的是片段化、 脱酰胺和氧化。 物理不稳定性可以因变性、 聚集、 沉淀或 吸附等引起。
单克隆抗体的降解可以发生在各个阶段, 包括抗体制剂的配制、 存储和递送过程中。 而 基于单克隆抗体的药品的生物学活性与其结构、 构象和化学稳定性密切相关。 因此, 抗体制 剂的开发是抗体产品开发的一个重要方面, 并常常是成功临床生产的一个关键步骤。 抗体药 品的稳定性是保证药品有效性和安全性的一个重要指标。 获得赋予抗体药品良好稳定性的制 剂处方是药品在货架期内保持其有效性和安全性的关键条件。 因此, 抗体制剂必须以不仅使 抗体适于施用给受试者的方式调配, 还要以在储存以及后续使用期间维持其稳定性的方式来 调配。 如果抗体没有适当地在液体中得以调配, 则液体溶液中的该抗体倾向于分解、 聚集或 发生不希望的化学修饰等。
目前, 单克隆抗体正在以增高的剂量被开发用于各种适应症的治疗。 例如 cetuximab 以 250-400mg/m2的剂量施用; efalizumab以大约 lmg/kg的剂量的施用。 此外, 考虑施用的方便 性和患者依从性, 对皮下注射抗体制剂的需求也日益增多。 因此, 开发高浓度的稳定单克隆 抗体液体制剂是本领域的一个方向, 其中蛋白质浓度需要达到 100mg/ml或更高。
除了满足稳定性和纯度等质量要求外, 抗体制剂的灵活性也是生物医药领域考虑的一个 因素。 有利地是, 制剂形式满足在宽范围的剂量水平上(典型地, 取决于目标适应症, 单克隆 抗体的用量可以为 0.1-20mg/kg)以及在多种施用途径上 (典型地是皮下和静脉内) 给药的灵 活性。
因此, 在本领域中对于含有抗 CD47 抗体的足够稳定新药物制剂存在需要, 尤其是对适 用于抗体高浓度 (如大约 100mg/ml) 的稳定液体制剂存在需要。 发明概述
为了开发重组全人源抗 CD47 单克隆抗体的长期稳定储存的制剂处方, 确保产品在有效 期内 (例如, 至少 24个月) 的质量可控, 本申请发明人设计了处方前和处方筛选两个阶段的 试验, 考察了不同 pH、表面活性剂、稳定剂以及抗氧化剂对抗 CD47抗体制剂稳定性的影响。 通过该深入的研究, 本发明人提供了含有重组全人源抗分化抗原簇 47(CD47)单克隆抗体的药 物制剂, 尤其是稳定的高浓度抗体液体制剂。
在一个方面, 本发明提供了一种液体抗体制剂, 其包含 (i) 重组全人源抗 CD47单克隆 抗体 (以下也简称“抗 CD47抗体”); (ii) 缓冲剂, (iii) 稳定剂, 和 (iv) 表面活性剂。
i一个实施方案中, 抗 CD47抗体为在中国申请 CN201710759828.9 (2017年 8月 29递 交) 中公开的重组全人源抗分化抗原簇 47(CD47)单克隆抗体。 为本申请的目的, 该中国申请 的全部内容特此并入本文作为参考。 在一个实施方案中, 抗 CD47抗体包含重链可变区和轻 链可变区, 其中重链可变区包含 SEQ ID NO: 1的序列或与其具有至少 90%同一性的序列, 且 轻链可变区包含 SEQIDNO: 2的序列或与其具有至少 90%同一性的序列:
QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYIYYSGSTNYNPSLKSRVT ISVDTSKNQFSLKLSSVTAADTAVYYCARGKTGSAAWGQGTLVTVSS (SEQIDNO: 1) :
DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQKPGKAPKLLIYAASSLQSGVPSRFSGSGS GTDFTLTISSLQPEDFATYYCQQTVSFPITFGGGTKVEIK (SEQIDNO: 2) 。
在一个实施方案中, 所述抗 CD47抗体包含:
-GSISSYYWS (SEQIDNO: 3) 或 SYYWS (SEQIDNO: 11) 的重链 VHCDR1; -YIYYSGSTNYNPSLKS (SEQIDNO: 4) 的重链 VHCDR2;
-ARGKTGSAA (SEQIDNO: 5) 或 GKTGSAA (SEQIDNO: 12) 的重链 VHCDR3; -RASQGISRWLA (SEQIDNO: 6) 的轻链 VLCDR1;
-AASSLQS (SEQIDNO: 7) 的轻链 VLCDR2; 和
-QQTVSFPIT (SEQIDNO: 8) 的轻链 VLCDR3。
在一个实施方案中, 所述抗 CD47抗体是包含重链和轻链的 IgG4抗体, 其中所述重链包 含 SEQ ID NO: 9的序列或与其具有至少 90%同一性的序列, 且其中所述轻链包含 SEQ ID NO:10的序列或与其具有至少 90%同一性的序列:
QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYIYYSGSTNYNPSLKSRVT
ISVDTSKNQFSLKLSSVTAADTAVYYCARGKTGSAAWGQGTLVTVSSASTKGPSVFPLAPCSRST
SESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKT
YTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCV
VVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYK
CKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWE
SNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS
LSLG (SEQID NO: 9)
DIQMTQSPSSVSASVGDRVTITCRASQGISRWLAWYQQKPGKAPKLLIYAASSLQSGVPSRFSGSGS GTDFTLTISSLQPEDFATYYCQQTVSFPITFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVC LLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYAC EVTHQGLSSPVTKSFNRGEC(SEQ ID NO: 10)。
优选地, 所述抗 CD47抗体是中国申请 CN201710759828.9 (于 2017年 8月 29递交) 中公 开的抗 CD47单克隆抗体 ADI-26630, 该抗体由 SEQ ID NO: 9的重链序列和 SEQ ID NO: 10 的轻链序列组成。 在移植人源伯基特氏淋巴瘤细胞的小鼠肿瘤模型中, 该抗体表现出显著的 抗肿瘤活性, 例如腹腔注射 5 mg/kg, 两天一次连续两周, 该抗体可以导致肿瘤生长抑制率达 到约 100%或更高; 且肿瘤消失比例可以达到 60%以上。 在一个实施方案中, 液体抗体制剂中的抗 CD47 抗体的浓度为约 1-150 mg/mL, 例如 20mg/ml以上, 尤其是 50mg/ml以上, 优选 100mg/ml或 120mg/ml。 例如, 液体抗体制剂中 的抗 CD47抗体的浓度可以是约 15、 20、 25、 30、 35、 40、 45、 50、 55、 60、 70、 75、 80、 85mg/ml、 或 90mg/mL以上, 例如, 90, 95, 100, 105, 110, 或 120mg/ml。
在一个实施方案中, 本发明的液体抗体制剂具有 pH5.0-6.0, 例如 pH5.2±0.2, pH5.5±0.2, pH5.7±0.2, 优选 pH5.5。
在一个实施方案中, 本发明的液体抗体制剂包含约 5-100mM的缓冲剂。 在一个实施方案 中, 本发明的液体抗体制剂中的缓冲剂的浓度为约 10mM, 20mM, 30mM, 40mM, 或 50mM。 在一个实施方案中, 所述缓冲剂为组氨酸缓冲剂。 在一个优选实施方案中, 本发明液体抗体 制剂包含约 10-20mM, 尤其是 10mM组氨酸缓冲剂。 在再一实施方案中, 组氨酸缓冲剂由组 氨酸-盐酸组氨酸缓冲体系构成。 在再一优选实施方案中, 本发明液体抗体制剂包含 0.4mg/ml 组氨酸和 1.5mg/ml盐酸组氨酸。
在一个实施方案中,本发明的液体抗体制剂包含稳定剂,优选地所述稳定剂选自山梨醇、 蔗糖、 海藻糖、 精氨酸及其组合, 更优选为山梨醇或山梨醇和精氨酸的组合。 在一个优选实 施方案中, 本发明液体制剂包含山梨醇作为稳定剂, 优选地山梨醇的浓度为约 l-10%w/v, 例 如, 1.5%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, 5%, 5.5% 或 6%w/v, 或者以约 10-60mg/ml, 例如 约 15mg/ml, 20mg/ml, 25mg/ml, 30mg/ml, 35mg/ml, 40mg/ml, 45mg/ml, 50mg/ml, 55mg/ml的量 存在, 优选约 40mg/mlo 在再一优选实施方案中, 本发明的液体抗体制剂包含山梨醇和精氨 酸的稳定剂组合。 优选地, 在所述组合中, 山梨醇的量为约 10-30mg/ml, 尤其是 15mg/ml, 精 氨酸的量为 80-1 10mM, 尤其是约 lOOmM。 在再一优选实施方案中, 本发明的液体抗体制剂 包含约 15 mg/ml山梨醇和约 21.1mg/ml盐酸精氨酸的稳定剂组合。
在一个实施方案中, 本发明的液体抗体制剂包含表面活性剂, 优选地, 所述表面活性剂 是非离子型表面活性剂。 在一个实施方案中, 所述表面活性剂选自聚山梨酯类表面活性剂, 优选为聚山梨酯 -20。 在一个实施方案中, 本发明的液体抗体制剂包含约 0.1-1 mg/ml , 例如 0.1-0.5mg/ml聚山梨酯 -20。 在一个优选的实施方案中, 本发明的液体抗体制剂包含约 0.1-0. 4 mg/ml 的聚山梨酯 -20。 在再一优选实施方案中, 本发明的液体抗体制剂中聚山梨酯 -20 的量 为约 0.3mg/ml。
在一个实施方案中, 本发明液体制剂还包含抗氧化剂。 在再一实施方案中, 本发明液体 制剂不包含抗氧化剂。在一个实施方案中,抗氧化剂是依地酸 (EDTA)或其盐,例如 EDTA-2Na 在一个实施方案中, 所述液体制剂为稳定的液体药物制剂, 优选为注射剂。 在一个实施 方案中, 本发明的药物制剂用于皮下、 皮内、 肌内、 或静脉注射。 在一个优选的实施方案中, 本发明的液体抗体制剂包含:
(i) 约 10-150 mg/mL的抗 CD47抗体;
(ii) 约 5-50mM的组氨酸缓冲剂;
(iii) 约 l-10%w/v山梨醇, 或约 l-5%w/v山梨醇和约 80-1 10mM精氨酸的稳定剂组合, 和
(iv) 约 0.01-0.1% w/v的聚山梨酯 -20 ;
其中所述液体制剂的 pH为约 5.0— 6.0, 优选地 pH5.5±0.2c
在一个优选的实施方案中, 本发明的液体抗体制剂包含
(i ) 90-1 50 mg/mL, 尤其是 90-120mg/ml的抗 CD47抗体;
(ii) 约 10-20mM组氨酸缓冲剂;
(iii) 约 20-60mg/ml (例如, 40-50mg/ml)山梨醇, 或约 5-40 mg/ml (例如, 10-20 mg/ml) 山梨醇和约 80-1 10mM (例如约 100mM)精氨酸的组合, 和
(iv) 约 0.1-0.4 mg/ml的聚山梨酯 -20 ;
其中所述液体制剂的 pH为约 5.0— 6.0, 优选地 pH5.5±0.2c
在一个优选的实施方案中, 本发明的液体抗体制剂包含:
(i) 约 100 mg/mL的抗 CD47抗体; (ii) 约 0.4mg/ml组氨酸和约 1.5mg/ml盐酸组氨酸;
(iii) 约 15.0 mg/ml山梨醇和 21. lmg/ml盐酸精氨酸, 和
(iv) 约 0.3mg/ml的聚山梨酿 -20 ;
其中所述液体制剂的 pH为约 5.5。 另一方面, 本发明提供了一种固体抗体制剂, 其是通过将本发明的液体抗体制剂经 固化处理而获得的。 所述固化处理是通过例如结晶法、 喷雾干燥法、 冷冻干燥法实施 的。 在一个优选的实施方案中, 所述固体抗体制剂例如是冻干制剂, 例如冻干粉针剂 形式。 固体抗体制剂可在使用前, 通过重构于适当的溶媒中, 形成本发明的重构制剂。 所 述重构制剂也是一种本发明的液体抗体制剂。 在一个实施方案中, 所述适当的溶媒选自注射 用水、 注射用有机溶剂, 包括但不限于注射用油、 乙醇、 丙二醇等, 或其组合。 本发明的液体制剂可以长期稳定储存, 尤其是以高浓度 (例如 50mg/ml 以上, 优选 90-150mg/ml, 例如 100mg/ml或 120mg/ml ) 稳定储存, 例如至少 24个月或更长时间。 在一 个实施方案中, 本发明的液体制剂可以在约 -80°C至约 45°C, 例如 -80°C、 约 -30°C、 约 -20°C、 约 0°C、 约 5°C、 约 25°C、 约 35°C、 约 38°C、 约 40°C、 约 42°C或约 45°C的条件下, 储存至 少 10天、 至少 20天、 至少 1个月、 至少 2个月、 至少 3个月、 至少 4个月、 至少 5个月、 至少 6个月、 至少 7个月、 至少 8个月、 至少 9个月、 至少 10个月、 至少 1 1个月、 至少 12 个月、 至少 18个月、 至少 24个月, 至少 36个月, 或更长时间, 是稳定的。
在一个实施方案中, 本发明的液体制剂可以稳定储存至少 24个月。 在再一实施方案中, 本发明的液体制剂在至少 40°C是稳定的。在再一实施方案中,本发明的液体制剂在约 2°C-8°C 保持稳定至少 12个月, 优选至少 24个月。 在一个实施方案中, 本发明的液体制剂在室温或 例如约 25°C保持稳定至少 3个月, 优选至少 6个月。 在再一实施方案中, 本发明的液体制剂 在约 40°C保持稳定至少 1个月。 在再一实施方案中, 本发明的液体制剂在约 5°C至 40°C的温 度, 例如在 25°C的温度, 在振荡下可以保持稳定至少 1天, 例如 3天或 5天。
在一个实施方案中, 可以通过检测制剂的外观、 可见异物、 蛋白含量、 纯度、 和 /或电荷 变异体的变化, 来指示储存后制剂的稳定性。 在一个实施方案中, 可以在高温加速试验中, 例如在 40°C±2°C储存至少 1周、 2周或优选地 1个月后, 或在 25°C±2°C储存至少 1个月或 2 个月后, 检测本发明液体制剂的稳定性。 在一个实施方案中, 通过振荡试验检测本发明液体 制剂的振荡稳定性。
在一个实施方案中, 在储存后, 通过目视检查本发明液体制剂的稳定性, 其中本发明液 体制剂在外观上保持为澄明、 无色或微黄色的液体, 无异物、 无絮状物及沉淀。 在一个实施 方案中, 在自然光下目视检查, 制剂中无可见异物存在。 在一个实施方案中, 在储存后, 通 过测定蛋白含量变化, 检查本发明液体制剂的稳定性, 其中例如通过紫外分光光度 (UV)法, 相对于储存第 0天的初始值, 蛋白含量变化率不超过 20%, 优选不超过 10%, 例如 7-8%, 优 选不超过 5%。 在一个实施方案中, 在储存后, 通过测定本发明液体制剂的浊度变化, 检查本 发明液体制剂的稳定性, 其中例如通过 OD350mm法检测, 相对于储存第 0天的初始值, 变 化值不超过 0.04, 更优选地不超过 0.03, 更优选地不超过 0.02。 在一个实施方案中, 在储存 后, 通过测定本发明液体制剂的纯度变化, 检查本发明液体制剂的稳定性, 其中通过尺寸排 阻高效液相色谱法 (SEC-HPLC),相对于储存第 0天的初始值,主峰纯度的变化值不超过 10%, 例如不超过 5%、 4%、 3%、 例如 1-2%, 优选不超过 1%, 且优选地, 聚体的增幅不超过 2%, 优选地不超过 1%、 0.5%或 0.1%。 在一个实施方案中, 在储存后, 通过测定本发明液体制剂 的纯度变化, 检查本发明液体制剂的稳定性, 其中通过非还原型十二烷基硫酸钠毛细管电泳 (CE-SDS)法, 主峰纯度的变化值下降不超过 10%, 例如不超过 5%、 4%、 3%、 优选不超过 2%、 1%、0.5%或 0.1%。在一个实施方案中,在储存后,通过成像毛细管等电聚焦电泳 (iCIEF ) 检测本发明液体制剂的稳定性, 其中相对于储存第 0天的初始值, 抗体的电荷变异体 (主成 分、 酸性组分或碱性组分) 的变化值不超过 30%, 优选地不超过 20%、 或不超过 10%, 例如 不超过 5%、 4%、 3%、 或 2%, 例如, 在一个实施方案中, 主成分或酸性组分的变化值不超 过 20%, 且碱性组分的变化值不超过 3%。
在一个实施方案中, 制剂在储存后, 例如在 2-8°C储存至少 24个月后, 或在室温储 存至少 3个月后, 或在 40°C±2°C储存 1个月后, 是稳定的, 优选地具有如下特征之一或 多项:
(i) 通过 SEC测量, 制剂中抗体单体的百分数大于 90%, 优选大于 95%, 且优选地 聚体增幅不超过 2% ;
(ii) 通过非还原型 CE-SDS测量, 制剂具有大于 90%的纯度, 优选大于 95%的纯度;
(iii) 通过 iCIEF测量, 相对于储存第 0天的初始值, 制剂中抗 CD47抗体的至少 50%, 优 选至少 55%是非碱性及非酸性形式, 且优选地抗体的酸性组分电荷变异体的增加不超过 20%。
在再一实施方案中, 制剂在储存后, 例如在 2-8°C储存至少 24个月后, 或在室温储 存至少 3个月后, 或在 40°C±2°C储存 1个月后, 是稳定的, 优选地具有以下特征之一或 多项:
(i) 外观澄明, 无可见异物;
(ii) 用紫外分光光度 (UV)法检测, 相对于储存第 0天的初始值, 蛋白含量变化率不超过
10%;
(iii) 用 OD350mm法检测, 相对于储存第 0天的初始值, 浊度变化值不超过 0.04, 优选地 不超过 0.02 ;
(iv) 用 SEC-HPLC检测, 相对于储存第 0天的初始值, 主峰纯度的变化值不超过 5%、 优 选不超过 1% ;
(v) 用非还原型 CE-SDS检测, 相对于储存第 0天的初始值, 主峰纯度的变化值下降不超 过 5%、 优选不超过 3% ;
(vi) 用 iCIEF检测, 相对于储存第 0天的初始值, 制剂中抗 CD47抗体的至少 50%, 优选至 少 55%是非碱性及非酸性形式, 且优选地抗体的酸性组分电荷变异体的增加不超过 20%。 在一个方面, 本发明提供了一种递送装置, 其包含本发明的液体抗体制剂或固体抗体制 剂。 在一个实施方案中, 本发明的递送装置以包含本发明的液体抗体制剂或固体抗体制剂的 预填装注射器形式提供, 例如用于静脉内、 皮下、 皮内或者肌内注射。
在再一方面, 本发明提供向受试者, 例如哺乳动物递送抗 CD47抗体的方法, 包括给予 所述受试者本发明的液体抗体制剂或固体抗体制剂的步骤, 所述递送是例如通过使用预填装 注射器的递送装置实施的。
在再一方面, 本发明提供本发明的液体抗体制剂或固体抗体制剂的用途, 用于制备在受 试者中治疗 CD47相关疾病的递送装置 (如, 预填装注射器) 或药物, 特别地用于治疗 CD47 相关癌症, 例如各种血液肿瘤, 如淋巴瘤, 例如伯基特氏淋巴瘤。
本发明的其它实施方案将通过参阅此后的详细说明而清楚明了。 附图简述
结合以下附图一起阅读时, 将更好地理解以下详细描述的本发明的优选实施方案。 出于 说明本发明的目的, 图中显示了目前优选的实施方案。 然而, 应当理解本发明不限于图中所 示实施方案的精确安排和手段。
图 1显示, 在实施例所述的 pH筛选试验中, 将不同 pH (pH5.0, 5.5, 6.0和 6.5) 的抗体制剂 在 40°C±2°C的温度条件下储存 0天、 1周、 2周和 1个月后, 用 OD350nm法测定的制剂浊度随时 间的变化图。 图 2显示, 在实施例所述的 pH筛选试验中, 将不同 pH (pH5.0, 5.5, 6.0和 6.5) 的抗体 制剂在 40°C±2°C的温度条件下储存 0天、 1周、 2周和 1个月后, 用 SEC-HPLC法测定的制剂 主峰纯度随时间的变化图。
图 3显示, 在实施例所述的 pH筛选试验中, 将 pH6.5的抗体制剂在 40°C±2°C储存 1个月 后, 通过 SEC-HPLC法测定的抗体制剂纯度图谱。
图 4显示, 在实施例所述的稳定剂筛选试验中, 将具有不同稳定剂的抗体制剂 (处方 1-4) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过 SEC-HPLC法测定的主峰纯度随 时间的变化图。
图 5显示, 在实施例所述的稳定剂筛选试验中, 将具有不同稳定剂的抗体制剂 (处方 1-4) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过非还原型 CE-SDS法测定的主峰纯 度随时间的变化图。
图 6显示, 在实施例所述的稳定剂筛选试验中, 将具有不同稳定剂的抗体制剂 (处方 1-4) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过 iCIEF法测定的电荷变异体主成分 随时间的变化图。
图 7显示, 在实施例所述的稳定剂筛选试验中, 将具有不同稳定剂的抗体制剂 (处方 1-4) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过 iCIEF法测定的电荷变异体酸性组 分随时间的变化图。
图 8显示, 在实施例所述的抗氧化剂筛选试验中, 将添加或不添加 EDTA的抗体制剂 (处方 A和 B) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过 OD350nm法测定的浊度 随时间的变化图。
图 9显示, 在实施例所述的抗氧化剂筛选试验中, 将添加或不添加 EDTA的抗体制剂 (处方 A和 B) 置于 40°C±2°C储存 0天、 1周、 2周、 和 1月后, 通过 iCIEF法测定的电荷变 异体 (主成分、 酸性组分和碱性组分) 随时间的变化图。 发明详述
在详细描述本发明前, 应了解, 本发明不受限于所述的特定方法及实验条件, 因为所述 方法以及条件是可以改变的。 另外, 本文所用术语仅是供说明特定实施方案之用, 而不意欲 为限制性的。 定义
除非另有定义, 否则本文中使用的所有技术和科学术语均具有与本领域一般技术人员通 常所理解的含义相同的含义。 为了本发明的目的, 下文定义了以下术语。
术语“约”在与数字数值联合使用时意为涵盖具有比指定数字数值小 5%的下限和比指定数 字数值大 5%的上限的范围内的数字数值。
术语“和 /或”当用于连接两个或多个可选项时, 应理解为意指可选项中的任一项或可选项 的任意两项或多项。
如本文中所用, 术语“包含”或“包括”意指包括所述的要素、 整数或步骤, 但是不排除任意 其他要素、 整数或步骤。 在本文中, 当使用术语“包含”或“包括”时, 除非另有指明, 否则也 涵盖由所述及的要素、 整数或步骤组成的情形。 例如, 当提及“包含”某个具体序列的抗体可 变区时, 也旨在涵盖由该具体序列组成的抗体可变区。 在本文中, 术语“抗体”是指包含轻链和重链免疫球蛋白可变区的多肽, 所述免疫球蛋白 可变区特异性识别并结合抗原。 优选地, 本发明的抗体是全长抗体, 由两条重链和两条轻链 组成, 其中每条重链由重链可变区(本文中缩写为 VH)和重链恒定区组成, 每条轻链由轻链可 变区 (本文中缩写为 VL)和轻链恒定区组成。 在一些实施方案中, 本发明的抗体也可以指抗体 的抗原结合片段。 术语“抗体制剂”指一种制备物, 所述制备物处于允许作为活性成分的抗体的生物活性可 以有效发挥的形式, 并且不含有对于待施用该制剂的受试者而言具有不可接受毒性的其它组 分。 这类抗体制剂通常是无菌的。 通常, 抗体制剂中包含可药用赋形剂。 “可药用”赋形剂是 可以合理地施用至受试哺乳动物以便制剂中所用活性成分的有效剂量可以递送至受试者的试 剂。 赋形剂的浓度与施用模式相适应, 例如可以是注射可接受的。
术语“抗 CD47抗体制剂”, 在本文中也简称为“本发明的抗体制剂”, 意指包含抗 CD47抗体 作为活性成分和可药用赋形剂的制备物。 经所述组合后, 作为活性成分的抗 CD47抗体适于治 疗性或预防性施与人类或非人类动物。 本发明的抗体制剂可以例如制备成水性形式的液体制 剂, 例如, 即用式预填装注射器, 或者制备成冻干制剂, 在即将使用前通过溶解和 /或悬浮于 生理可接受的溶液中进行重构 (即, 复溶) 。 在一些实施方案中, 抗 CD47抗体制剂是液体制 剂形式。
“稳定”抗体制剂是制剂中的抗体在储存于特定条件下之后保有可接受程度的物理稳定 性和 /或化学稳定性。 尽管抗体制剂中所含的抗体在储存特定时间之后可能不会 100%维持其 化学结构, 但通常在储存特定时间之后维持约 90%、 约 95%、 约 96%、 约 97%、 约 98%或约 99%的抗体结构或功能, 则认为抗体制剂是“稳定的”。 在一些具体的实施方案中, 本发明的 抗 CD47抗体制剂在制造、 制备、 运输和长期储存过程中表现出低至检测不到的抗体聚集或 降解或化学修饰, 从而极少或甚至是没有抗 CD47抗体的生物活性损失, 表现出高度稳定性。 在一些实施方案中, 本发明的抗 CD47抗体制剂在储存后, 基本上保留其物理和化学稳定性。 优选地, 本发明液体制剂可以在室温或在 40°C稳定至少 1 个月, 和 /或在 2-8°C稳定至少 24 个月。
本领域已知多种分析技术可以用于测定蛋白质的稳定性, 参见例如 Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs (1991) and Jones, A. Adv. Drug Delivery Rev. 10: 29-90 (1993)。 可以在选定的温度和选定的储存时间测量 稳定性。 例如, 可以基于预期的制剂货架期来选择储存时间。 或者可以使用加速稳定性试验。 在一些实施方案中, 通过对抗体制剂进行各种胁迫测试来进行稳定性测试。 这些测试可以代 表调配的抗体制剂在制造、 储存或运输期间可能遭遇到的极端条件, 也可以代表在非制造、 储存或运输期间可能使抗体制剂中的抗体的不稳定性加速的条件。 例如, 可以将经调配的抗 CD47抗体制剂充填至 5 mL玻璃瓶中用于振荡胁迫以检测抗体的振荡 /剪切稳定性;或者可以 将经调配的抗 CD47抗体制剂充填至玻璃小瓶中以检验在高温胁迫下的抗体稳定性。 经一段储存时间后, 制剂不显示聚集、 沉淀、 混浊和 /或变性; 或显示非常少的聚集、 沉 淀、 混浊和 /或变性, 则可以认为抗体在制剂中“保持其物理稳定性”。 由于制剂中抗体的聚集 可以潜在地导致患者增加的免疫反应, 从而导致安全性问题。 因此, 需要使在制剂中的抗体 聚集最小化或防止聚集。 光散射法可以用于测定制剂中的可见聚集物。 S EC可以用于测定制 剂中的可溶性聚集物。 此外, 可以通过目视检查制剂的外观、 颜色和 /或澄清度、 或者通过 OD350nm法检测制剂的浊度、 或者通过非还原型 CE-SDS法测定制剂的纯度, 来指示制剂的 稳定性。 在一个实施方案中, 通过测定在特定温度下储存特定时间之后制剂中的抗体单体的 百分比来测量制剂的稳定性, 其中制剂中的抗体单体的百分比越高, 则制剂的稳定性越高。
“可接受程度的” 物理稳定性可以表示于特定温度下储存特定时间之后, 在制剂中检测 到至少约 92%的抗 CD47抗体单体。 在一些实施方案中, 在特定温度储存至少 2周、 至少 28天、 至少 1个月、 至少 2个月、 至少 3个月、 至少 4个月、 至少 5个月、 至少 6个月、 至 少 7个月、 至少 8个月、 至少 9个月、 至少 10个月、 至少 1 1个月、 至少 12个月、 至少 18 个月、 至少 24个月或更久后, 可接受程度的物理稳定性表示至少约 92%、 93 %、 94%、 95 %、 96%、 97%、 98 %、 99%的抗 CD47抗体单体。 当评估物理稳定性时, 药物制剂储存的 特定温度可为约 -80°C至约 45°C的任一温度, 例如储存于约 -80°C、 约 -30°C、 约 -20°C、 约 0°C、 约 4°C-8°C、 约 5°C、 约 25°C、 约 35°C、 约 37°C、 约 40°C、 约 42°C或约 45°C。 例如, 若储 存于约 40°C±2°C 1个月之后, 检测到至少约 92%、 93 %、 94%、 95 %、 96%、 97%、 98 %、 99%的抗 CD47抗体单体, 则药物制剂视为是稳定的。 若储存于约 25°C 2个月之后, 检测到 至少约 92%、 93 %、 94%、 95 %、 96%、 97%、 98 %、 99%的抗 CD47抗体单体, 则药物制 剂视为是稳定的。 若储存于约 5°C 9个月之后, 检测到至少约 92%、 93 %、 94%、 95 %、 96 %、 97%、 98 %、 99%的抗 CD47抗体单体, 则药物制剂视为是稳定的。 经一段储存时间后, 如果制剂中的抗体不显示显著的化学改变, 则可以认为抗体在制剂 中“保持其化学稳定性”。 大多数化学不稳定性源自于形成了抗体的共价修饰形式(例如, 抗 体的电荷变异体)。 例如由天冬氨酸异构化、 N和 C末端修饰, 可以形成碱性变异体; 由脱酰 胺化、 唾液酸化和糖化, 可以产生酸性变异体。 化学稳定性可以通过检测和 /或定量抗体的化 学改变形式来评估。例如, 可以通过阳离子交换色谱(CEX)或成像毛细管等电聚焦电泳(icIEF) 检测制剂中抗体的电荷变异体。 在一个实施方案中, 通过测定在特定温度下储存特定时间之 后制剂中抗体的电荷变异体百分比变化值来测量制剂的稳定性, 其中该变化值越小, 则制剂 的稳定性越高。
“可接受程度”的化学稳定性可以表示于特定温度下储存特定时间之后制剂中电荷变异 体 (例如主成分或酸性组分或碱性组分) 的百分比变化值不超过 30%, 例如 20%。 在一些实 施方案中, 在特定温度储存至少 2周、 至少 28天、 至少 1个月、 至少 2个月、 至少 3个月、 至少 4 个月、 至少 5个月、 至少 6个月、 至少 7个月、 至少 8个月、 至少 9个月、 至少 10个月、 至少 1 1 个月、 至少 12个月、 至少 18个月、 至少 24个月或更久后, 可接受程度的化学稳定性可以表现 为酸性组分电荷变异体的百分比变化值不超过约 25%、 20%、 15%、 10%、 5%、 4%、 3%、 2%、 或 1%。 当评估化学稳定性时, 储存药物制剂的温度可为约 -80°C至约 45°C的任一温度, 例如 储存于约 -80°C、 约 -30°C、 约 -20°C、 约 0°C、 约 4°C-8°C、 约 5°C、 约 25°C或约 45°C。 例如, 若 在储存于 5°C 24个月之后,酸性组分电荷变异体的百分比变化值少于约 25%、 24%、 23%、 22%、 21%、 20%、 19%、 18%、 17%、 16%、 15%、 14%、 13%、 12%、 10%、 9%、 8%、 7%、 6%、 5%、 4%、 3%、 2%、 1%、 0.5%或 0.1%, 则药物制剂可被视为是稳定的。 若在储存于 25°C 2 个月后, 酸性组分电荷变异体的百分比变化值少于约 20%、 19%、 18%、 17%、 16%、 15%、 14%、 13%、 12%、 10%、 9%、 8%、 7%、 6%、 5%、 4%、 3%、 2%、 1%、 0.5%或 0.1%, 则药 物制剂亦可被视为是稳定的。 若在储存于 40°C 1个月之后, 酸性组分电荷变异体的百分比变 化值少于约 25%、 24%、 23%、 22%、 21%、 20%、 19%、 18%、 17%、 16%、 15%、 14%、 13%、 12%、 10%、 9%、 8%、 7%、 6%、 5%、 4%、 3%、 2%、 1%、 0.5%或 0.1%, 则药物制剂亦可 被视为是稳定的。 术语“冻干制剂”是指通过液体制剂的冷冻干燥处理得到或能够得到的组合物。 优选地, 其为具有少于 5%、 优选少于 3%水含量的固体组合物。
术语“重构制剂”是指将固体制剂 (例如冻干制剂) 溶解和 /或悬浮于生理可接受的溶液中 得到的液体制剂。
文中使用的术语“室温”是指 15°C至 30°C、 优选 20°C至 27°C、 更优选 25°C的温度。
“胁迫条件”是指在化学和 /或物理上不利于抗体蛋白的环境, 所述环境可以导致不可接受 的抗体蛋白失稳定。“高温胁迫”是指,将抗体制剂置于室温或甚至于更高温度(:例如 40°C±2°C) 储存一段时间。 通过高温胁迫加速试验, 可以检查抗体制剂的稳定性。
如本文所使用, 术语“肠胃外施用”意指肠内和局部给药以外的给药方式, 通常通过注射 或输注方式, 并且包括但不限于, 静脉内、 肌内、 动脉内、 鞘内、 囊内、 眶内、 心内、 皮内、 腹膜内、 经气管、 皮下、 表皮下(subcuticular)、 关节内、 囊下、 蛛网膜下、 脊柱内、 硬膜外和 胸骨内注射以及输注。 在一些实施方案中, 本发明的稳定抗 CD47抗体制剂肠胃外施用于受 试者。 在一个实施方案中, 本发明的抗 CD47抗体制剂以皮下、 皮内、 肌内或静脉内注射方 式施用于受试者。
I. 抗体制剂
本发明提供稳定的液体抗体制剂, 其包含 (i) 重组全人源抗 CD47单克隆抗体; (ii) 缓 冲剂, (iii) 稳定剂, 和 (iv) 表面活性剂, 所述抗体制剂的 pH为约 5.0-6.0。 在一个优选方 案中, 本发明的液体抗体制剂是注射制剂形式。
(i) 抗 CD47抗体
“抗 CD47抗体”是指这样的抗体, 所述抗体能够以足够的亲和力结合 CD47分子, 以致 所述抗体可以用作靶向 CD47分子的治疗剂和 /或预防剂。
本发明的抗体是重组全人源抗体。 术语“全人源抗体”或“人抗体”在本文中可以互换使用, 指该抗体包括其中构架区和 CDR区二者均源自人种系免疫球蛋白序列的可变区,而且如果抗 体含有恒定区, 恒定区也源自人种系免疫球蛋白序列。 本发明的人抗体可包括不由人种系免 疫球蛋白序列编码的氨基酸(例如, 通过体外随机或点特异诱变或体内体细胞突变引入的突 变)。 然而, 如本文所使用的, 术语“人抗体”不意欲包括其中的 CDR序列衍生自其他哺乳动 物物种(如, 小鼠)的种系而移植入人构架序列的抗体。 如本文所用, 术语“重组人抗体”包括所 有通过重组方式制备、 表达、 产生或分离的人抗体。 这些重组人抗体具有构架区和 CDR区源 自人种系免疫球蛋白序列的可变区。 然而, 在某些实施方案中, 可以对重组人抗体进行体外 诱变(或使用人 Ig序列转基因动物时为体内体细胞诱变), 由此得到的重组抗体的 VH和 VL 区的氨基酸序列, 尽管源自人种系 VH和 VL序列并与之相关、 但是并不天然存在于体内的 人抗体种系库中。
在一些实施方案中, 如通过生物光干涉法测量, 所述抗 CD47抗体能够以高的亲和力, 例如以 10-7M或更小、 优选地以 10_9 M至 10_1Q M的 KD特异性结合人 CD47, 并由此介导对 CD47及其配体结合的高效阻断作用。
在一些实施方案中, 本发明抗体 CD47抗体包含: SEQ ID NO: 1或与之具有至少 90%同 一性的重链可变区(VH);和 SEQ ID NO: 2或与之具有至少 90%同一性的轻链可变区(VL)。“可 变区”或“可变结构域”是抗体的重链或轻链中参与抗体与其抗原的结合的结构域。 一般, 重链 可变区(VH)和轻链可变区(VL)可以进一步再划分为高变区(HVR,又称作互补决定区(CDR)), 其间插有较保守的区域 (即, 构架区(FR)) 。 每个 VH和 VL由三个 CDR和 4个 FR组成, 从氨基端到羧基端以如下顺序排列: FR1 , CDR1, FR2, CDR2, FR3, CDR3, FR4。
在一些实施方案中,本发明的抗 CD47抗体包含 SEQ ID NO: 1的重链可变区的 HCDR1、
2和 3序列和 SEQ ID NO : 2的轻链可变区的 LCDR1、 2和 3序列。 “互补决定区”或“CDR 区”或“CDR” (在本文中与超变区“HVR”可以互换使用) , 是抗体可变区中主要负责与抗原表 位结合的氨基酸区域。 重链和轻链的 CDR通常被称作 CDR1、 CDR2和 CDR3, 从 N-端开始 顺序编号。 位于抗体重链可变结构域内的 CDR被称作 HCDR1、 HCDR2和 HCDR3, 而位于 抗体轻链可变结构域内的 CDR被称作 LCDR1、 LCDR2和 LCDR3。 本领域公知多种用于在 一个给定的 VH或 VL氨基酸序列中确定其 CDR序列的方案。例如, Kabat互补决定区(CDR) 是基于序列变异性确定的并且是最常用的(Kabat等人, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.(1991)) 。 而 Chothia指的是结构环的位置(Chothia和 Lesk, J. Mol. Biol. 196:901-917(1987))。 AbM HVR 是 Kabat HVR和 Chothia结构环之间的折中, 并且由 Oxford Molecular的 AbM抗体建模软件 使用。“接触性”(Contact) HVR基于对可获得的复杂晶体结构的分析。 HVR也可以基于与参考 CDR序列(例如本文公开的示例性 CDR)具有相同的 Kabat编号位置而确定。
在一个实施方案中, 本发明抗体的 CDR通过 Kabat规则确定边界, 例如下文表 A(l) 所
TF
在一个实施方案中, 本发明抗体的 CDR通过结合 Kabat、 AbM、 Chothia及经验性等综 合因素确定边界。 例如下文表 A(2) 所示: HCDR1是在 kabat编号系统中位置 H27-H35的序 列, HCDR3是在 kabat编号系统中位置 H93 -H102的序列, 以及 HCDR2和 LCDR1、 LCDR2 和 LCDR3通过 Kabat规则确定。
然而, 应该注意, 基于不同的指派系统获得的同一抗体的可变区的 CDR的边界可能有所 差异。 即不同指派系统下定义的同一抗体可变区的 CDR序列有所不同。 因此, 在涉及用本发 明定义的具体 CDR序列限定抗体时, 所述抗体的范围还涵盖了这样的抗体, 其可变区序列包 含所述的具体 CDR序列, 但是由于应用了不同的方案 (例如不同的指派系统规则或组合) 而 导致其所声称的 CDR边界与本发明所定义的具体 CDR边界不同。
具有不同特异性 (即, 针对不同抗原的不同结合位点) 的抗体具有不同的 CDR。 然而, 尽管 CDR在抗体与抗体之间是不同的, 但是 CDR内只有有限数量的氨基酸位置直接参与抗 原结合。 使用 Kabat, Chothia, AbM、 Contact和 North方法中的至少两种, 可以确定最小重叠 区域,从而提供用于抗原结合的“最小结合单位”。最小结合单位可以是 CDR的一个子部分。 正如本领域技术人员明了,通过抗体的结构和蛋白折叠,可以确定 CDR序列其余部分的残基。 因此, 本发明也考虑本文所给出的任何 CDR的变体。 例如, 在一个 CDR的变体中, 最小结 合单位的氨基酸残基可以保持不变, 而根据 Kabat或 Chothia定义的其余 CDR残基可以被保 守氨基酸残基替代。
表 A
Figure imgf000012_0001
在一些实施方案中,本发明的抗 CD47抗体可以包含与 SEQ ID NO: 1具有至少 90%, 95% 或 98%或更高同一性的重链可变区(VH); 和 /或与 SEQ ID NO: 2具有至少 90%、 95%或 98% 或更高同一性的轻链可变区(VL)。 在本文中, “序列同一性”是指在比较窗中以逐个核苷酸或 逐个氨基酸为基础的序列相同的程度。 可以通过以下方式计算“序列同一性百分比”: 将两条 最佳比对的序列在比较窗中进行比较, 确定两条序列中存在相同核酸碱基(例如, A、 T、 C、 G、 I)或相同氨基酸残基(例如, Ala、 Pro、 Ser、 Thr、 Gly、 Val、 Leu、 lie、 Phe、 Tyr、 Trp、 Lys、 Arg、 His、 Asp、 Glu、 Asn、 Gin、 Cys 和 Met)的位置的数目以得到匹配位置的数目, 将匹配位置的数目除以比较窗中的总位置数(即, 窗大小), 并且将结果乘以 100, 以产生序列 同一性百分比。 为了确定序列同一性百分数而进行的最佳比对, 可以按本领域已知的多种方 式实现, 例如, 使用可公开获得的计算机软件如 BLAST、 BLAST-2、 ALIGN 或 Megalign (DNASTAR)软件。 本领域技术人员可以确定用于比对序列的适宜参数, 包括为实现正在比较 的全长序列范围内或目标序列区域内最大比对所需要的任何算法。
在一些实施方案中, 本发明抗体的 VH序列与 SEQ ID NO : 1相比具有不超过 10个, 优 选地不超过 5个、 4个或 3个不同残基, 优选地所述不同残基为保守氨基酸替代。 在一些实 施方案中, 本发明抗体的 VL序列与 SEQ ID NO: 2相比具有不超过 10个, 优选地不超过 5 个、 4个或 3个不同残基, 优选地所述不同残基为保守氨基酸替代。“保守性取代”是指导致某 个氨基酸置换为化学上相似的氨基酸的氨基酸改变。 提供功能上相似氨基酸的保守性置换表 是本领域熟知的。 在本发明任一实施方案中, 在一个优选的方面, 保守取代残基来自以下的 保守替代表 X, 优选地为表 X中所示优选置换残基。
表 X
Figure imgf000013_0001
在一些实施方案中, 本发明的抗体是 IgG4形式的抗体。 “IgG形式的抗体”是指抗体的重 链恒定区所属于的 IgG形式。 所有同一型的抗体的重链恒定区都是相同的, 不同型的抗体之 间的重链恒定区不同。 例如, IgG4形式的抗体是指其重链恒定区 Ig结构域为 IgG4的 Ig结构 域。
在一个优选的实施方案中, 本发明的抗 CD47抗体是中国申请 CN201710759828.9 (2017 年 8月 29) 中公开的抗 CD47单克隆抗体 ADI-26630, 其具有 SEQ ID NO:9的重链和 SEQ ID NO: 10的轻链。 在一个实施方案中, 该抗 CD47抗体是由 CHO细胞重组表达产生并经纯化 的 IgG4型抗体。 优选地, 在本发明液体制剂中所述抗体表现出显著的抗肿瘤活性。 例如, 在 移植人源伯基特氏淋巴瘤细胞的小鼠肿瘤模型中,例如腹腔注射 5 mg/kg,两天一次连续两周, 该抗体可以导致肿瘤生长抑制率达到约 50%或更高, 例如 100% ; 和 /或肿瘤消失率达到 60% 以上。 本发明的抗体制剂中所包含的抗体或其抗原结合片段的量可随着制剂的特定目的特性、 特定环境、 和使用制剂的特定目的而改变。 在一些实施方案中, 抗体制剂为液体制剂, 其可 含有约 1-150 mg/mL, 优选地为约 10-100 mg/mL, 例如, 约 15、 20、 25、 30、 35、 40、 45、 50、 55、 60 mg/mL抗 CD47抗体。 在一个实施方案中, 本发明涉及具有高浓度的抗 CD47抗 体的制剂, 例如, 含有 50-150 mg/mL的抗 CD47抗体。 本领域已知这样的高浓度抗体制剂可 以在注射前进行稀释, 例如, 如果特定的治疗性或预防性干预需要较低的抗体浓度或者当治 疗包括儿童的较小体重患者时。 适合的浓度可以是 25 mg/mL或 10 mg/mL。 备选地, 可以以 这样的低浓度生产原始制剂。
(ii) 缓冲剂
缓冲剂是可以将溶液的 pH维持在可接受范围的试剂。在一些实施方案中, 用于本发明制 剂中的缓冲剂可以将本发明制剂的 pH控制在大约 5.0-6.0的 pH范围, 例如约 5.2-5.8的 pH, 优选地 5.3-5.7的 pH。 在一个具体的实施方案中, 本发明的抗体制剂具有约 5.0、 5.2、 5.4、 5.5, 5.6, 5.7, 5.8, 5.9或 6.0的 pH, 优选地约 5.5的 pH。 优选地, 用于本发明的缓冲剂是组 氨酸缓冲剂, 例如由组氨酸和盐酸组氨酸组成的缓冲体系。
在一个实施方案中, 本发明的抗体制剂中缓冲剂的浓度为约 5-100mM, 优选地为约 10-50mM, 例如, 约 10-20 mM。 优选地, 缓冲剂为约 10-20mM的组氨酸缓冲剂。
(iii) . 稳定剂
用于本发明的合适的稳定剂可以选自糖、 多元醇和氨基酸及其组合。 对于作为稳定剂的 糖包括但不限于蔗糖和海藻糖。 对于作为稳定剂的多元醇包括但不限于山梨醇。 对于作为稳 定剂的氨基酸包括但不限于精氨酸。
在一个实施方案中, 本发明液体制剂包含蔗糖作为稳定剂。 蔗糖在本发明液体制剂中的 量可以是约 50-100mg/ml, 优选地 70-90mg/ml, 例如 80mg/ml。
在一个实施方案中, 本发明液体制剂包含海藻糖作为稳定剂。 海藻糖在本发明液体制剂 中的量可以是约 50-100mg/ml, 优选地 70-90mg/ml, 例如 80mg/ml。
在一个实施方案中, 本发明液体制剂包含精氨酸作为稳定剂。 精氨酸在本发明液体制剂 中的量可以是约 80-1 10mM, 尤其是约 100mM, 例如约 21.1 mg/ml盐酸精氨酸。
在一个实施方案中, 本发明液体制剂包含山梨醇作为稳定剂。 山梨醇在本发明液体制剂 中的量可以是约 10-100mg/ml, 优选地 20-70mg/ml, 例如 30-60mg/ml。 例如, 山梨醇可以以 约 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65或 70mg/ml的量存在, 优选地以约 40mg/ml的量 存在。
在一个实施方案中,本发明液体制剂包含山梨醇和精氨酸的组合作为稳定剂。该组合中, 山梨醇可以以约 5-60mg/ml, 优选地 10-30mg/ml, 例如 10-20mg/ml 的量存在, 例如可以为 5, 10, 12, 15, 17, 20, 22, 25mg/ml。 在该组合中, 精氨酸可以以约 80-1 10mM, 尤其是约 lOOmM 的量存在。 优选地, 本发明液体制剂包含约 10-20mg/ml的山梨醇和约 10-30mg/ml的盐酸精 氨酸。 更优选地, 本发明液体制剂包含约 15mg/ml的山梨醇和约 21.1mg/ml的盐酸精氨酸。
(iv) . 表面活性剂
如本文所使用的, 术语“表面活性剂”是指具有两亲结构的有机物质; 即, 它们由相反的 溶解性倾向的基团所组成, 通常是油溶性的经链和水溶性的离子基团。 在一个实施方案中, 本发明的液体制剂中的表面活性剂是非离子型表面活性剂, 例如, 烷基聚(环氧乙烯)。 可包括在本发明制剂中的特定非离子型表面活性剂包括, 例如聚山梨酯, 诸如聚山梨酯 -20、 聚山梨酯 -80、 聚山梨酯 -60、 或聚山梨酯 -40 ; 普洛尼克等。 在一个优选实 施方案中, 本发明的液体制剂中包含聚山梨酯 -20作为表面活性剂。
本发明抗体制剂中所含的表面活性剂的量可随制剂的特定目的特性、 特定环境、 和使用 制剂的特定目的而改变。 在优选的一些实施方案中, 制剂可含有约 0.01-5 mg/ml, 优选地为 约 0.1-2 mg/ml, 例如约 0.2、 0.3、 0.4、 0.5、 0.6、 0.7、 0.8、 0.9、 1.0 mg/ml的聚山梨酯 -20, 优选地约 0.3mg/ml的聚山梨酯 -20。
Cv)其它赋形剂
本发明的抗体液体制剂中可以包含或不包含其它赋形剂。 在一个实施方案中, 本发明的 抗体液体制剂包含 EDTA或其盐。 在另一实施方案中, 本发明的抗体液体制剂不包含 EDTA 或其盐。 在一个实施方案中, 添加 EDTA或其盐的本发明抗体液体制剂, 与不添加 EDTA或 其盐的相应制剂相比, 具有相当的稳定性。
出于其他考虑, 也可在本发明制剂中使用其它的赋形剂。 所述赋形剂包括, 例如, 调味 剂、 抗微生物剂、 甜味剂、 抗静电剂、 抗氧化剂、 明股等等。 这些和另外已知的药物赋形剂 和 /或适用于本发明制剂的添加剂是本领域公知的, 例如, 列出于“The Handbook of Pharmaceutical Excipients,第 4版, Rowe等人编, American Pharmaceuticals Association(2003); 和 Remington: the Science and Practice of Pharmacy, 第 21版, Gennaro编, Lippincott Williams & Wilkins(2005)’’。
II. 制剂的制备
本发明提供包含抗体的稳定制剂。 在本发明制剂中使用的抗体可以使用本领域已知的用 于生产抗体的技术进行制备。 例如, 可以重组制备抗体。 在一个优选的实施方案中, 本发明 的抗体在 CHO细胞中重组制备。
抗体作为药物的活性成分的应用现在已经很广泛。 用于将治疗性抗体纯化至药用级的技 术是本领域公 口的。 例 4 Tugcu 等(Maximizing productivity of chromatography steps for purification of monoclonal antibodies, Biotechnology and Bioengineering 99(2008) 599-613.)描述 在蛋白 A捕获步骤后使用离子交换色谱(阴离子 IEX和 /或阳离子 CEX色谱)的单克隆抗体三 柱纯化方法。 Kelley 等(Weak partitioning chromatography for anion exchange purification of monoclonal antibodies, Biotechnology and Bioengineering 101 (2008) 553-566)描述了两柱纯化 法, 其中在蛋白 A亲和色谱后使用弱分配阴离子交换树脂。
一般, 重组产生的单克隆抗体可以利用常规的纯化方法纯化, 以提供具有足够的可重复 性和适度纯度的药物物质用于抗体制剂的配制。 例如, 在抗体从重组表达细胞分泌至培养基 中后, 可以使用商业可得的蛋白浓缩过滤器例如 Amicon 的超滤装置, 浓缩来自该表达系统 的上清液。 之后, 可以使用例如色谱、 透析和亲和纯化等方式进行抗体的纯化。 蛋白 A适应 于作为亲和配体用于纯化 IgGl,IgG2和 IgG4型抗体。 也可以使用其它抗体纯化方法, 例如离 子交换色谱。在获得足够纯度的抗体后, 可以按照本领域已知的方法, 制备包含抗体的制剂。
例如, 可以采用如下步骤进行制备: ( 1)在发酵结束后将发酵液离心澄清去除细胞等杂质 以获得上清;(2) 使用亲和层析(例如对 IgGl, IgG2和 IgG4型抗体具有特异亲和力的蛋白 A柱) 捕获抗体;(3)进行病毒灭活; (4)精制纯化(一般可以采用 CEX 阳离子交换层析), 以去除蛋白 中的杂质; (4)病毒过滤 (使病毒滴度降低例如 4 1ogl0以上) ; (5)超滤 /渗滤 (可以用于将蛋 白置换于利于其稳定的制剂缓冲液中并浓缩至合适的浓度供注射用) 。 参见例如, B. Minow, P. Rogge, K. Thompson, BioProcess International, Vol. 10, No. 6, 2012, pp. 48-57。 III. 制剂的分析方法
在抗体制剂的储存过程中, 抗体可能会发生聚集、 降解或化学修饰, 导致抗体异质性 (包 括大小异质性和电荷异质性) 以及聚集物和片段等, 从而影响抗体制剂的质量。 因此, 有必 要进行抗体制剂稳定性的监测。 在本领域中已知多种方法可以用于检测抗体制剂的稳定性。 例如, 可以通过非还原型 CE-SDS和 SEC-HPLC等方法, 分析抗体制剂的纯度和评估抗体的 聚集水平; 可以通过毛细管等电聚焦电泳 (cIEF)、 成像毛细管等电聚焦电泳 (iCIEF)和离子交 换色谱 (IEX) 等, 分析抗体制剂中的电荷变异体。 此外, 可以通过目视检测制剂外观, 快 速地判断制剂的稳定性。 也可以使用 OD350nm 法检测制剂的浊度改变, 该方法可以给出有 关可溶性和不溶性聚集物量的信息。 此外, 可以使用紫外分光光度法 (UV法)检测制剂中的蛋 白质含量变化。
非还原型 CE-SDS 法是一种以毛细管为分离通道进行的单克隆抗体纯度测定方法。 在 CE-SDS中, 蛋白迁移由 SDS结合引起的表面电荷来驱动, 而该表面电荷与蛋白质的分子量 成正比。 由于所有的 SDS-蛋白质复合物都具有相似的质量-电荷比, 故可以在毛细管的分子 筛凝胶基质中, 实现基于分子的大小或流体动力学半径的电泳分离。 该方法已经被广泛地用 于监测变性的完整抗体的纯度。 一般, 在非还原 CE-SDS法中, 供试样品与 SDS样品缓冲液 和碘乙酰胺混合。 之后, 混合物可以于 68-72°C孵育约 10-15分钟, 冷却至室温后离心的上清 液用于分析。采用紫外检测器检测蛋白的迁移, 获得电泳谱图。抗体制剂纯度可以计算为 IgG 主峰的峰面积占所有峰面积之和的百分比。 关于 CE-SDS 法的进一步描述, 可以参见例如 Richard R.等, Application of CE SDS gel in development of biopharmaceutical antibody-based products, Electrophoresis, 2008, 29, 3612-3620。
尺寸排阻高效液相色谱法, 即 SEC-HPLC法, 是用于单克隆抗体标准和质控的另一重要 方法。 该方法主要依据分子的尺寸大小或流体动力学半径差异来进行分子的分离。 通过 SEC-HPLC,抗体可以分离出三种主要形式:高分子量形式 (HMMS)、主峰 (主要是抗体单体)、 和低分子量形式 (LMMS ) 。 抗体纯度可以计算为色谱图上主峰面积占所有峰面积之和的百 分比。 通过 SEC-HPLC法, 可以测量制剂产品中抗体单体的百分数, 给出可溶性聚集物和剪 切物的含量信息。 关于 SEC-HPLC 法的进一步描述, 可以参见例如, J. Pharm. Scien., 83 : 1645-1650, (1994); Pharm. Res., 1 1 :485 (1994); J. Pharm. Bio. Anal., 15 : 1928 (1997); J. Pharm. Bio. Anal., 14: 1 133-1 140 (I986)。此外,也可以 4见例如, R. Yang等, High resolution separation of recombinant monoclonal antibodies by size exclusion ultra-high performance liquid chromatography (SE-UHPLC), Journal of Pharmaceutical and Biomedical Analysis (2015), http://dx.doi.Org/10.1016/i .ipba.2015.02.032 : 和 Alexandre Goyon 等, Protocols for the analytical characterization of therapeutic monoclonal antibodies. I- Non-denaturing chromatographic techniques, Journal of Chromatography , http://dx.doi.Org/10.1016/j .jchromb.2017.05.010。
成像毛细管等电聚焦电泳 (iCIEF)可以用于分析单克隆抗体的电荷异质性。 该方法可以提 供电荷变异体的定量分布情况。 iCIEF基于分子在 pH梯度中的电荷差异 (表观 pi值)来实现分 子分离的目的。 在 iCIEF中, 分离柱通常是短毛细管 (例如, 5cm长 , 100pm内径的二氧化硅毛 细管), 蛋白质在高电压下在毛细管柱中聚焦, 并通过在 280nM 操作的全柱成像检测系统对 聚焦进行实时在线监测。 该技术的一个优点是, 可以通过该全柱检测系统同时记录抗体样品 的各种电荷变异体。 一般而言, 在 icIEF中, 将样品与尿素和 icIEF缓冲液混合, 其中所述缓 冲液含有甲基纤维素、 pi分子量标准和 ampholytes c 然后, 可以在 iCIEF分析仪例如 iCE280 分析仪 (Protein Simple, Santa Clara, CA)上,使用 iCIEF柱例如 ProtionSimple组装的 iCIEF柱, 在样品聚焦一定时间后, 测定 280nm的吸光度, 获得聚焦 mAb电荷变异体的谱图。 在 iCEIF 谱图中, 在主峰 (即主成分)之前洗脱的蛋白相关峰被分类为酸性组分; 相对地, 在主峰之后 洗脱的蛋白相关峰被分类为碱性组分。 主成分、 酸性组分和碱性组分的相对量可以表示为占 总峰面积的百分数。 关于 iCIEF的进一步描述, 可以参见例如, Salas-Solano 0等, Robustness of iCIEF methodology for the analysis of monoclonal antibodies: an interlaboratory study, J Sep Sci. 2012 Nov;35(22):3124-9. doi: 10.1002/jssc.201200633. Epub 2012 Oct 15;和 Dada 00 等, Characterization of acidic and basic variants of IgGl therapeutic monoclonal antibodies based on non-denaturing IEF fractionation, Electrophoresis. 2015 Nov;36(21-22):2695-2702. doi:
10.1002/elps.201500219. Epub 2015 Sep 18.
也可以通过阳离子交换高效液相色谱法 (CEX-HPLC) 测定抗体制剂中抗体的电荷变异 体。在该测定法中,以比主峰的保留时间更早从 CEX-HPLC柱洗脱出的峰被标记为“酸性峰”, 而那些以比主峰的保留时间更晚从 CEX-HPLC柱洗脱出的峰被标记为“碱性峰”。
加速稳定性研究可以用于检查产品的稳定性性质,有利于筛选稳定药物制剂形式。例如, 可以将制剂样品放置于升高的温度,例如约 40°C±2°C^ 25°C±2°C条件下进行加速稳定性研究。 检测指标可以包括外观、可见异物、蛋白含量、 浊度、纯度(SEC-HPLC法、非还原型 CE-SDS 法) 和电荷变异体 (iCIEF法) 。
可以进行振荡试验, 考察制剂的振荡 /剪切稳定性。 例如, 将制剂样品分装至西林瓶, 加 塞轧盖后放样,进行振荡试验,例如 650 r/min振荡 3-5天, 之后检测制剂的外观、蛋白含量、
;虫度和纯度。
此外, 可以检测抗体的功效或生物活性。 例如, 可以检测制剂中抗体与其抗原的结合能 力。 本领域技术人员已知多种方法可以用于定量抗体与抗原的特异性结合, 例如免疫测定试 验, ELISA等。
本发明的抗 CD47抗体制剂是稳定的。 在一个实施方案中, 于约 25°C、 37°C、 40°C、 或 45°C储存至少 1个月或 2个月后, 例如, 在 40°C±2°C储存 1个月后, 本发明的抗体制剂中的 抗 CD47抗体纯度是至少 90%、 91%、 92%、 93%、 94%、 95%、 96%、 97%、 98%、 或 99% 以上,如通过尺寸排阻色谱法或通过非还原型 CS-SDS所测定。在一个实施方案中,于约 25°C、 n°C、 40°C、 或 45°C储存至少 1个月或 2个月后, 例如, 在 40°C±2°C储存 1个月后, 本发 明的抗体制剂中抗 CD47抗体的至少 50%, 优选至少 55%是非碱性及非酸性形式(亦即, 主峰 或主要电荷形式), 如通过成像毛细管聚焦电泳法所测定。
IV. 制剂的用途
本发明的包含抗 CD47 抗体的本发明的抗体制剂可以用于治疗、 改善或预防多种 CD47 相关疾病或病症。“CD47相关疾病或病症”在本文中指可以用本发明抗 CD47抗体进行治疗(例 如改善)或预防的疾病或病症。任何可以得益于本发明抗体治疗的疾病或病症都适用于本发明。
CD47是一个多能分子, 在肿瘤细胞逃避免疫监视过程中扮演重要角色。 阻断 CD47信号 通路能够有效激发巨噬细胞对肿瘤细胞的呑噬作用, 从而用于肿瘤免疫治疗。 因此, 包含抗 CD47抗体的本发明制剂尤其可用于治疗、 改善或预防 CD47相关癌症。所述癌症包括例如但 不限于: 各种血液肿瘤和实体瘤, 例如白血病, 包括急性籁样白血病, B 淋巴细胞慢性白血 病和急性淋巴细胞白血病; 非霍奇金淋巴瘤; 骨籁瘤; 平滑肌肉瘤; 星形细胞瘤; 乳腺癌; 卵巢癌; 胶质母细胞瘤。 参见例如, 国际肿瘤学杂志, 2013年 11月 40卷第 11期, 817-819 页。 在一些实施方案中, 本发明的抗体制剂用于治疗、 改善或预防与 CD47相关的淋巴瘤, 例如伯基特淋巴瘤。
本发明也提供本发明的制剂在制备药物中的用途, 其中所述药物用于向哺乳动物递送抗 CD47抗体, 或用于治疗、 预防或改善上述疾病和病症中的一种或多种。 优选地, 哺乳动物是 人。
可以以多种途径将本发明的抗体制剂施用于受试者或患者。 例如, 施用可以通过输注或 通过注射器进行。 因此, 在一个方面, 本发明提供了一种递送装置 (例如注射器) , 其包括 本发明的抗体制剂 (例如, 预填装注射器) 。 患者将接受有效量的抗 CD47抗体作为主要活 性成分, 即足以治疗、 改善或预防目的疾病或病症的量。
治疗效果可包括减少生理症状。 用于任何特定受试者的抗体的最佳有效量和浓度将取决 于多种因素, 包括患者的年龄、 体重、 健康状况和 /或性别、 疾病的性质和程度、 特定抗体的 活性, 身体对其清除率, 并且也包括与所述抗体制剂组合施用的任何可能的其它治疗。 对于 具体的情况,所递送的有效量可以在临床医师的判断范围内来确定。取决于待治疗的适应症, 有效剂量可为约 0.005 mg/kg体重至约 50 mg/kg体重,或约 0.1 mg/kg体重至约 20 mg/kg体重。 在这方面, 已知的基于抗体的药物的应用可以提供一定的指导。 剂量可以是单剂量方案或多 剂量方案。 描述以下实施例以辅助对本发明的理解。 不意在且不应当以任何方式将实施例解释成限 制本发明的保护范围。 实施例
为了开发出重组全人源抗分化抗原簇 47(CD47)单克隆抗体注射液长期稳定储存的制剂 处方, 确保产品在有效期内 (至少 24个月) 的质量可控, 设计了处方前和处方筛选两个阶段 的试验。 试验所用材料和方法如下: 材料和方法
1.1.制剂研究中使用的化学品
名称 级别 产地及品牌 货号 符合标准
组氨酸 药用级 德国 Merck 1.04352.1000 Ph Eur、 JP、 USP
盐酸组氨酸 药用级 德国 Merck 1.04354.0500 Ph Eur、 BP、 JP
盐酸精氨酸 药用级 德国 Merck 1.01544.1000 Ph Eur、 BP、 JP、 USP
山梨醇 药用级 法国罗盖特 H20110265 Ch.P、 Ph Eur、 USP
海藻糖 药用级 美国 Pfanstiehl T-104-4 USP/NF、 EP、 JP
蔗糖 药用级 德国 Merck 1.00892.9050 Ph Eur、 USP
聚山梨酯 20 药用级 德国 Merck 8.17072.1000 Ph Eur、 JPE、 NF
盐酸 药用级 德国 Merck 1.00314.2508 Ph Eur、 BP, JP, NF
1 2 使用的仪器设备
设备名称 产地及品牌 型号
多通道微量分光光度计 美国 Thermo Nanodrop 8000
澄明度检测仪 天津天大天发 YB-2
紫外分光光度计 曰本岛津 UV-1800
SEC-HPLC分析仪 美国 Waters 2695
非还原型 CE-SDS分析仪 美国 Caliper Labchip GX Touch HT
icIEF分析仪 美国 Protein simple Maurice
1.3 . 制剂稳定性的检测项目和检测方法
对抗体制剂检测了以下项目: (1) 检测外观以及是否存在可见异物; (2) 通过紫外法 (UV法)测定制剂中的蛋白质含量; (3) 通过 OD350nm法检测制剂的浊度; (4) 通过尺寸 排阻高效液相色谱法 (SEC-HPLC) 测定抗体制剂的纯度, 表示为主峰的面积占所有峰面积 之和的百分数; (5) 通过非还原型十二烷基硫酸钠毛细管电泳(非还原型 CE-SDS)测定抗体 制剂的纯度, 表示为主峰的面积占所有峰面积之和的百分数; (6) 通过成像毛细管等电聚焦 电泳法(icIEF法;)测定抗体制剂中电荷变异体,表示为主成分、酸性组分和碱性组分的百分数。 蛋白含量测定
使用紫外分光光度计(日本岛津生产, 型号 UV- 1800)测定样品中的蛋白质含量。
浊度测定
使用多通道微量分光光度计(美国 Thermo生产,型号 Nanodrop8000),测定样品在 350nm 的吸光度, 确定样品浊度。
可见异物检测
按照国家药典委员会, 中华人民共和国药典 (2015年版, 四部通则 0904“可见异物检查 法”) .北京:中国医药科技出版社. 2015中所记载的方法, 采用澄明度检测仪(天津天大天发生 产, 型号 YB-2), 检查样品中的可见异物。
尺寸排阻高效液相色谱法 (SEC-HPLC) 法
在用于分析抗体制剂的 SEC-HPLC法中, 可以使用以下参数:
色谱柱: TSK-gel SuperSW mAb HR (7.8x300 mm, 4(j,m) 型分析柱, TSK-gel SuperSW (6.0x40 mm, 4(j,m) 保护柱
流动相: 20 mmol/L磷酸缓冲液 + 200 mmol/L NaCl, pH6.8
流速: 0.5 ml/min
柱温度: 25°C
检测波长: 280 nm
进样体积: 50 [j]
进样盘温度: 约 10°C
运行时间: 30分钟
上样浓度: 2.0 mg/mL
非还原型十二烷基硫酸钠毛细管电泳(CE-SDS)法
可以按如下进行非还原型 CE-SDS测定: 将大约 的样品 (蛋白含量: lmg/ml)加入 14 ul 非还原型样品 Buffer ( 包括取 700 ul pH 6.5样品缓冲液, 加入 31.3 ul 250mM NEM(N-乙基顺丁烯二 酰亚胺)), 再加 28 ul超纯水, 混匀, 之后在 70°C孵育 10分钟。 孵育后, 样品冷却至室温, 然后 转移至 96孔板中。 CE-SDS分离在 LabChip GXIITouch(PerkinElmer)上进行, 使用 Protein芯片。 分 析方法选择 HT Antibody Analysis 200, 样品盘类型选择 BioRad 96 HSP-96xx (Sip 4 mm) 通过焚光 监测蛋白迁移。
成像毛细管等电聚焦电泳法(icIEF法)
可以如下进行 icIEF检测: 将抗体样品稀释(或脱盐)至大约 lmg/ml。取 20(J样品加入 780 icIEF缓冲液, 所述缓冲液含有尿素、 精氨酸(Protein Simple)、 pi marker标准(Protein Simple, Santa Clara, CA)和 Pharmalytes(GE Healthcare Bio-Science, Pittsburgh, PA)。在 Maurice C.分析仪 (Protein Simple, Santa Clara, CA)上, 使用 FC涂层 iCIEF柱, 产生成像毛细管等电聚焦谱图。 样 品聚焦总共 8分钟, 使用 Empower 版本 3软件(Waters, Milford, MA), 对 280nm聚焦蛋白的吸光 度进行积分。 实施例 1. 制备和纯化抗 CD47抗体
根据 CN201710759828.9所述制备和纯化了抗 CD47抗体 ADI-26630。 该抗体由 SEQ ID NO : 9的重链序列和 SEQ ID NO: 10的轻链序列组成, 为 IgG4型抗体。 简言之, 抗体在 CHO 细胞中重组表达并进行纯化。对于用于处方前试验的样品,通过 CEX(阳离子交换色谱)纯化, 样品具有 12.2mg/ml的蛋白含量。对于用于处方筛选试验的样品,通过 CEX(阳离子交换色谱) 纯化获得 15.7mg/ml的蛋白含量后, 通过渗滤浓缩至 100mg/ml。 实施例 2. 处方前试验
2.1. pH影响试验
2.1.1 pH影响试验步骤
配制 10 mM组氨酸, 5%山梨醇缓冲液, 分别将 pH调节为 5.0、 5.5、 6.0和 6.5。 用配制 完成的各个 pH缓冲液分别对小试样品进行超滤置换。 置换完成后, 将各样品的浓度稀释至 20 mg/ml c 过滤分装至西林瓶, 加塞轧盖后放样, 进行 40°C加速稳定性试验。 具体试验方案 见表 1。
_ 表 1. pH影响试验具体方案 _
样品名称 批号 试验条件 取样点 检测项目
pH5.0 20170527-01 外观、 蛋白含量、 浊度 pH5.5 20170527-02 0天、 1周、 2周、 和纯度 (SEC-HPLC pH
Figure imgf000020_0001
6.0 20170527-05 1月取样 非还原型 CE-SDS pH6.5 20170527-06
Figure imgf000020_0002
注: 上述时间点取样后均先放入超低温冰箱中冻存待检, 按需要化冻送检。
2.1.2 判断标准
根据对产品的认识以及仪器和方法的精密度, 设定了样品检测指标数值与初始值相比质 量未发生变化的判定标准, 用以判断样品是否发生了变化, 具体见表 2。
_ 表 2. 产品质量未发生变化的判断标准 _
检测项目 未发生变化的判定标准
外观 澄明、 无色或微黄色液体, 无异物, 无絮状物及沉淀 蛋白含量 (UV法) 变化率 $10%
油度 ( OD35Q nm法) 变化率 $10%
纯度 (SEC-HPLC法) 主峰纯度变化值 %
纯度 (非还原型 CE-SDS法) 主峰纯度变化值
Figure imgf000020_0003
2.1.3 pH影响试验结果
(1) 外观
在 40°C±2°C的条件下放置 1月, 各组样品外观均合格。
(2) 蛋白含量
在 40°C±2°C条件下, 各组样品的蛋白含量均未发生变化 (见表 3)。
表 3. pH影响试验蛋白含量结果 (UV法, mg/ml)
40°C±2°C
样品名称 0天
1周 2周 1月
pH5.0 18.8 18.5 18.8 18.7 pH5.5 20.4 20.6 21.0 20.9 pH6.0 21.4 21.3 21.3 21.3 pH6.5 _ 209 _ 207 21.1 21.1
(3 ) 浊度
在 40°C±2°C条件下,各组样品的浊度均升高,且 pH越高,变化速率越快(见表 4、图 1)。 表 4. pH影响试验浊度结果 (OD350nm法) 40°C±2°C
样品名称 0天
1周 2周 1月
pH5.0 0.043 0.043 0.047 0.052 pH5.5 0.056 0.061 0.067 0.077 pH6.0 0.065 0.073 0.078 0.101 pH6.5 0.063 0.075 0.088 0.102
(4) 纯度
纯度 (SEC-HPLC法): 40°C±2°C条件下加速 1个月, pH5.0、 pH5.5、 pH6.0各样品的纯 度均未发生明显变化, pH6.5条件下, 样品纯度下降, 变化原因主要是在加速后发生聚集。 且随着 pH升高, 样品初始纯度有降低趋势。 (见表 5、 图 2〜图 3)。
表 5. pH影响试验纯度结果 ( SEC-HPLC法)
40°C±2°C
样品名称 0天
1周 2周 1月 pH5.0 99.0 99.1 99.0 98.6 pH5.5 98.7 98.9 98.6 98.3 pH6.0 98.5 98.4 98.0 97.9 pH6.5 98.3 97.8 97.4 96.9 纯度 (非还原型 CE-SDS法): 40°C±2°C条件下加速 1个月, 各 pH条件下样品的纯度均 未发生明显变化。 (见表 6)。
表 6. pH影响试验纯度结果 (非还原型 CE-SDS法)
40°C±2°C
样品名称 0天
1周 2周 1月 pH5.0 98.8 N/A N/A 98.6 pH5.5 99.0 N/A N/A 98.1 pH6.0 99.0 N/A N/A 98.5 pH6.5 98.9 N/A N/A 97.7 注: N/A表示未检测或不适用, 下同。
pH影响试验结果表明随着 pH升高,样品浊度变化速率越快; pH6.5时,样品纯度降低, 发生聚集。 综合以上结果, pH 5.0〜 6.0间产品稳定性良好。
2.2. 表面活性剂影响试验
2.2.1 表面活性剂影响试验步骤
配制 10 mM组氨酸, 5%山梨醇, pH6.0缓冲液,超滤置换小试样品,浓度稀释至 20 mg/ml 后, 分别不添加聚山梨酯 20以及加入终浓度为 0.3 mg/ml聚山梨酯 20。 过滤分装至西林瓶, 加塞轧盖后放样, 进行振荡试验。 检测项目包括外观、 蛋白含量、 浊度和纯度 (SEC-HPLC 法、 非还原型 CE-SDS法) 。 具体试验方案见表 7。
_ 表 7. 表面活性剂筛选试验方案 _
样品名称 批号 试验条件及取样点
不添加聚山梨酿 20170527-03 振荡试验: 25°C、 650 r/min、 避光, 于第 0、 3、 5天 添加聚山梨酯 20 20170619 取样
2.2.2 判断标准
见 2 1.2节。
2.2.3 表面活性剂影响试验结果
650 r/min振荡 3天, 不添加聚山梨酯 20的样品发生混浊, 其他检测项目均不再进行检 测; 添加 0.3 mg/ml聚山梨酯 20样品振荡 5天各检测结果均无变化, 表明聚山梨酯 20能够保 证制剂具有良好的振荡稳定性。 具体结果见表 8。
表 8. 表面活性剂影响试验检测结果
检测项 不添加聚山梨酯 20 添加聚山梨酯 20
0天 3天 5天 0天 3天 5天 外观 合格 混油 混油 合格 合格 合格 可见异物 合格 N/A N/A 合格 合格 合格 蛋白含量 (UV法, mg/ml) N/A N/A N/A 20.8 21.1 20.8 浊度 (OD350 nm 法) N/A N/A N/A 0.062 0.066 0.067 纯度 (SEC-HPLC , %) N/A N/A N/A 98.2 98.3 98.3 纯度 (非还原型 CE-SDS法, %) N/A N/A N/A 99.4 N/A 99.4
综合处方前研究结果, 将制剂处方定为组氨酸-盐酸组氨酸缓冲体系, 选定 pH为 5.5 , 表 面活性剂为聚山梨酿 20, 在此基础上进行接下来的处方筛选试验。
实施例 3. 处方筛选试验
3.1 稳定剂筛选试验
3.1.1 稳定剂筛选试验步骤
共设计了 4个处方, 详细处方信息见表 9。 按照表 9配制各个处方的缓冲液, 将抗体蛋 白超滤置换到各自的处方溶液中。 置换完成后, 将各处方蛋白浓度稀释至约 120 mg/ml, 并加 入聚山梨酯 20,使终浓度为 0.3 mg/ml。过滤分装至西林瓶,加塞轧盖后在 40oC±2°C^ 25oC±2°C 条件下进行加速稳定性考察。检测指标为外观、可见异物、蛋白含量、浊度、纯度(SEC-HPLC 法、 非还原型 CE-SDS法) 和电荷变异体 (iCIEF法) 。
表 9. 稳定剂筛选试验备选处方信息表
序号 处方信息
处方 1 0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 40 mg/ml山梨醇, 0.3 mg/ml聚山梨醋 20, pH5.5 处方 2 0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 80 mg/ml蔑糖, 0.3 mg/ml聚山梨醋 20, pH5.5 处方 3 0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 80 mg/ml海藻糖, 0.3 mg/ml聚山梨 S旨 20, pH5.5 处方 4 0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 21.1 mg/ml盐酸精氨酸, 0.3 mg/ml聚山梨醋 20, pH5.5 详细试验条件及取样计划见表 10。
_ 表 10 试验条件及取样表 _
试验名称 试验方案及取样点
40°C加速试验 ~~ 40°C±2°C条件下放置, 于 0天、 1周、 2周和 1个月取样
25°C加速试验 25°C±2°C条件下放置, 于 0天、 1个月和 2个月取样
3.1.2 判定标准
根据对产品的认识以及仪器和方法的精密度, 设定了样品检测指标数值与初始值相比质 量未发生变化的判定标准, 用以判断样品是否发生了变化, 具体见表 1 1。
表 1 1. 质量未发生变化的判定标准
检测指标 未发生变化的标准
外观 澄明、 无色或微黄色液体, 无异物, 无絮状物及沉淀 可见异物 符合《中华人民共和国药典》 (2015年版, 四部) 通则 0904 蛋白含量 (UV法) 变化率 $10%
油度 ( OD35G nm法) 变化值 $0.02 检测指标 未发生变化的标准
纯度 (SEC-HPLC法) 主峰纯度变化值 $1%
纯度(非还原型 CE-SDS法) 主峰纯度变化值 $2%
电荷变异体 (iCIEF法) 各组分 (即主成分、 酸性组分及碱性组分) 变化值 $2%
3.1.3 稳定剂筛选试验结果
(1) 外观、 可见异物
在 40°C±2°C的条件下观察至 1个月, 25°C±2°C条件下观察至 2个月, 四组处方外观、 可 见异物均合格。
(2) 蛋白含量
在 40°C±2°C和 25°C±2°C条件下, 4组处方的蛋白含量均未发生变化 (见表 12) 。
表 12. 稳定剂筛选试验蛋白含量结果 (UV法, mg/ml)
40°C±2°C 25°C±2°C 样品名称 0天
1周 2周 1月 1月 2月 处方 1 120.0 120.0 123.9 123.4 126.0 1 17.1 处方 2 120.8 124.4 1 19.7 1 18.1 1 19.4 1 15.8 处方 3 120.5 125.5 121.3 126.0 126.0 1 18.4 处方 4 120.2 125.0 125.7 127.8 125.7 120.8
(3) 纯度
纯度 (SEC-HPL C法) : 40°C±2°C条件下 1个月, 处方 1 (山梨醇) 纯度未发生明显变 化, 其余处方均超出了拟定的判断标准, 主要表现为聚体增多, 但增长幅度均 < 2% ; 在 25°C±2°C条件下, 各处方纯度没有发生明显变化。 详见表 13、 图 4。
表 13. 稳定剂筛选试验纯度结果 (SEC-HPLC法, %)
40°C±2°C 25°C±2°C 样品名称 0天
1周 2周 1月 1 n_ 2 n 处方 1 98.0 97.8 97.8 97.9 97.8 97.7 处方 2 98.2 98.1 98.0 97.1 97.7 98.0 处方 3 98.3 97.7 97.8 97.2 97.7 97.9 处方 4 98.4 98.3 98.4 96.8 98.2 98.2 纯度 (非还原型 CE-SDS法) : 40°C±2°C条件下, 各处方纯度均有下降趋势, 处方间无 差异; 25°C±2°C条件下, 各处方纯度没有发生明显变化。 详见表 14、 图 5。
表 14. 稳定剂筛选试验纯度结果 (非还原型 CE-SDS法, %)
40°C±2°C 25°C±2°C 样品名称 0天
1周 _ 2 M _ \_ n_ 1月 2月 处方 1 98.9 N/A 98.0 97.4 99.1 98.7 处方 2 98.9 N/A 98.2 97.1 99.1 98.9 处方 3 99.1 N/A 98.2 97.4 99.1 98.9 处方 4 99.2 N/A 98.2 97.1 99.1 98.9
(4) 电荷变异体
40°C±2°C和 25°C±2°C条件下, 各处方电荷变异体-酸性组分和主成分均发生明显变化, 碱性组分变化较小, 各处方间趋势一致, 无明显差异, 且变化幅度均可接受。 (见表 15、 图 6~7) 表 15. 稳定剂筛选试验电荷变异体结果 (iCIEF法, %)
40°C±2°C 25°C±2°C 组分 样品名称 0天
1周 2周_ 1 ^ 1月 2月 处方 1 29.8 N/A 37.9 42.8 32.1 34.4 处方 2 30.3 N/A 36.9 46.2 30.3 33.7 酸性组分
处方 3 30.0 N/A 36.3 44.8 31.5 32.9 处方 4 29.2 N/A 36.0 46.3 30.8 32.1 处方 1 68.0 N/A 58.3 52.8 65.0 62.5 处方 2 67.5 N/A 59.3 49.7 66.7 63.2 主成分
处方 3 67.7 N/A 60.0 51.3 65.8 64.2 处方 4 68.4 N/A 60.2 50.1 66.4 65.0 处方 1 2.3 N/A 3.8 4.3 2.9 3.1 处方 2 2.3 N/A 3.7 4.1 2.9 3.1 碱性组分
处方 3 2.4 N/A 3.7 3.9 2.7 2.9 处方 4 2.4 N/A 3.8 3.7 2.8 2.9 从纯度 (SEC-HPLC法) 的检测结果看, 山梨醇有优势, 能够更有效抑制蛋白在 40°C高 温下聚体的产生, 其他检测项无明显差别; 考虑到有文献报道精氨酸能降低高浓度蛋白产品 6勺枯度 (Borwankar A U, Dear B J, Twu A, et al. Viscosity reduction of a concentrated monoclonal antibody with arginine HC1 and arginine glutamate [J]. Industrial & Engineering Chemistry Research, 2016, 55(43): 11225-11234.) , 故选取山梨醇和精氨酸的稳定剂组合处方用于下一轮抗氧化剂的考察。
3.2 抗氧化剂试验
3.2.1 抗氧化剂试验步骤
将样品分为两份,一份加入 EDTA-2Na使终浓度为 0.01 mg/ml ;另一份不添加 EDTA-2Na。 各处方蛋白浓度为约 100 mg/ml。 过滤分装至西林瓶, 加塞轧盖后在 40°C士 2°C条件下进行加 速稳定性考察, 于 1周、 2周和 1 月取样检测。 检测指标为外观、 可见异物、 蛋白含量、 浊 度、 纯度 (SEC-HPLC法、 非还原型 CE-SDS法) 和电荷变异体 (iCIEF法) 。 表 16. 抗氧化剂筛选试验处方信息
序号 处方信息
0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 15.0 mg/ml山梨醇, 21.1 mg/ml精氨酸, 0.3 mg/ml 处方 A
聚山梨醋 20, 0.01 mg/ml依地酸二納, pH5.5
0.4 mg/ml组氨酸, 1.5 mg/ml盐酸组氨酸, 15.0 mg/ml山梨醇, 21.1 mg/ml精氨酸, 0.3 mg/ml 处方 B
聚山梨酯 20, pH5.5
3.2.2 判定标准
见 3.1.2节。
3.2.3 抗氧化剂试验结果
在 40°C士 2°C条件下考察 1 个月, 两个处方样品的外观、 可见异物、 蛋白含量、 纯度 (SEC-HPLC法) 均未发生变化; 两个处方样品的浊度均升高, 变化速率保持一致; 纯度(非 还原型 CE-SDS法) 1个月均降低 2.1% ; 电荷变异体-主成分降低, 酸性组分升高, 变化速率 保持一致。 具体见表 17、 图 8和图 9。
各检测指标数据表明两个处方均有良好的稳定性, 且处方间无差异, 即是否添加 EDTA-2Na对产品稳定影响不大。 从处方简单化和安全化出发, 最终选定处方 B作为抗体制 剂的最终处方。
表 17. 抗氧化剂筛选试验检测结果 (40°C±2°C)
处方 A (添加 EDTA-2Na) 处方 B (无 EDTA-2Na) 检测项目
0天 1周 2周 1月 0天 1周 2周 1月 外观 合格 合格 合格 合格 合格 合格 合格 合格 可见异物 合格 合格 合格 合格 合格 合格 合格 合格 蛋白含量 (UV法, mg/ml) 99.3 97.4 99.0 95.6 102.9 100.6 101.6 95.4 油度 (OD35Q nm法) 0.212 0.224 0.227 0.246 0.222 0.223 0.224 0.245 电荷变异体 酸性组分 24.2 27.6 32.6 40.8 24.3 28.3 32.9 40.7
(iCIEF ;i, %) 主成分 73.7 69.7 64.0 55.0 73.7 69.0 63.6 55.1 碱性组分 2.0 2.7 3.4 4.2 1.9 2.7 3.4 4.2 纯度 (SEC-HPLC , %) 99.4 99.1 99.1 98.8 99.3 99.1 99.1 98.8 纯度(非还原型 CE-SDS,%)法, 99.2 98.9 98.4 97.1 99.1 98.5 98.2 97.0 结论
综合上述各个试验结果, 抗体蛋白在 pH 5.0〜 6.0时表现稳定; 表面活性剂聚山梨酯 20能 够保证产品具有良好的振荡稳定性; 稳定剂中山梨醇效果较好, 能更有效抑制蛋白在 40°C高 温下聚体的产生; 高温抗氧化试验发现处方中无需添加 EDTA-2Na ; 另外, 加入适量精氨酸 能降低高浓度蛋白产品的粘度。 由此, 确定优选的制剂方案为: 100 mg/ml重组全人源抗分化 抗原簇 47 (CD47) 单克隆抗体、 0.4 mg/ml 组氨酸、 1.5 mg/ml 盐酸组氨酸、 15.0 mg/ml 山 梨醇、 21.1 mg/ml盐酸精氨酸、 0.3 mg/ml聚山梨酯 20, pH 5.5。 以上描述了本发明的示例性实施方案, 本领域技术人员应当理解的是, 这些公开内容仅 是示例性的, 在本发明的范围内可以进行各种其它替换、 适应和修改。 因此, 本发明不限于 文中列举的具体实施方案。

Claims

权 利 要 求 书
1. 一种具有 pH约为 5.06.0的液体抗体制剂, 例如, pH约为 5.5的液体抗体制剂, 包含
(i) 抗 CD47抗体;
(ii) 缓冲剂,
(iii) 稳定剂, 和
(iv) 表面活性剂,
其中, 所述抗 CD47抗体包含:
-GSISSYYWS (SEQIDNO: 3) 或 SYYWS (SEQIDNO: 11) 的重链 VHCDR1;
-YIYYSGSTNYNPSLKS (SEQIDNO: 4) 的重链 VHCDR2;
-ARGKTGSAA (SEQIDNO: 5) 或 GKTGSAA (SEQIDNO: 12) 的重链 VHCDR3;
-RASQGISRWLA (SEQIDNO: 6) 的轻链 VLCDR1;
-AASSLQS (SEQIDNO: 7) 的轻链 VLCDR2; 和
-QQTVSFPIT (SEQIDNO: 8) 的轻链 VLCDR3。
2. 权利要求 1的液体抗体制剂, 特征在于所述液体抗体制剂中的抗 CD47抗体的浓度为 约 1-150 mg/mL, 优选地为约 10-120mg/mL, 例如 20mg/ml, 更优选地为约 50-120mg/mL, 例如, 约 100 或 120mg/mL。
3. 根据权利要求 1或 2所述的液体抗体制剂, 特征在于所述液体抗体制剂中的缓冲剂为组 氨酸缓冲剂, 优选地, 所述缓冲剂的浓度为约 5-100mM, 优选地为约 10-50mM, 例如, 约 10或 20mM。
4. 根据权利要求 1-3 中任何一项所述的液体抗体制剂,特征在于所述稳定剂选自山梨醇、 蔗糖、 海藻糖、 精氨酸及其组合。
5. 根据权利要求 1-4中任何一项所述的液体抗体制剂,特征在于所述液体抗体制剂包含山 梨醇作为稳定剂,优选地山梨醇以约 10-80mg/ml,优选地约 30-50mg/ml,如 40mg/ml的量存在。
6. 根据权利要求 1-4中任何一项所述的液体抗体制剂, 特征在于液体抗体制剂包含山梨 醇和精氨酸的稳定剂组合, 优选地山梨醇的量为约 10-30mg/ml, 尤其是 15mg/ml, 精氨酸的 的量为 80-110mM, 尤其是约 lOOmM。
I 根据权利要求 1-6中任何一项所述的液体抗体制剂,特征在于所述液体抗体制剂中的表 面活性剂选自聚山梨酯类表面活性剂, 优选为聚山梨酯 -20。
8. 根据权利要求 1-7中任何一项所述的液体抗体制剂,特征在于所述表面活性剂的浓度为 约 0.1-1 mg/ml, 优选地为约 0.1-0.5mg/ml, 例如约 0.2、 0.3、 0.4、 0.5mg/ml。
9. 根据权利要求 1-8中任何一项所述的液体抗体制剂,特征在于所述液体制剂不包含依地 酸 (EDTA)或其盐。
10. 根据权利要求 1-9所述的液体抗体制剂,特征在于所述抗 CD47抗体包含重链可变区 VH和轻链可变区 VL, 其中重链可变区包含 SEQ ID NO: 1的序列或与其具有至少 90%, 95%, 98%或 99%同一性的序列, 且轻链可变区包含 SEQ ID NO: 2的序列或与其具有至少 90%, 95%, 98%或 99%同一性的序列。
II. 根据权利要求 1-10所述的液体抗体制剂, 特征在于所述抗 CD47抗体是 IgG4型 抗体,优选地包含 SEQ ID NO : 9或与之具有至少 90%, 95%, 98%或 99%同一性的重链序列 以及 SEQ ID NO: 10或与之具有至少 90%, 95%, 98%或 99%同一性的轻链序列。
12. 根据权利要求 1-11所述的液体抗体制剂, 特征在于所述抗 CD47抗体在 CHO细 胞中重组表达。
13. 根据权利要求 1-12所述的液体抗体制剂, 特征在于所述液体制剂为注射剂, 优选 用于皮下或静脉注射。
14. 根据权利要求 1所述的液体抗体制剂, 包含: (i) 90-150 mg/mL, 尤其是 90-120mg/ml的抗 CD47抗体;
(ii) 约 10-20mM组氨酸缓冲剂;
(iii) 约 20-60mg/ml (例如, 40-50mg/ml)山梨醇, 或约 5-40 mg/ml (例如, 10-20 mg/ml) 山梨醇和约 80-1 10mM (例如约 100mM)精氨酸的组合, 和
(iv) 约 0.1-0.4 mg/ml (例如, 约 0.3mg/ml)的聚山梨酿 -20 ;
其中所述液体制剂的 pH为 pH5.5±0.2, 优选地约 5.5。
15. 根据权利要求 1所述的液体抗体制剂, 其包含:
(i) 约 100 mg/mL的抗 CD47抗体;
(ii) 约 0.4mg/ml组氨酸和约 1.5mg/ml盐酸组氨酸;
(iii) 约 15.0 mg/ml山梨醇和约 21.1 mg/ml盐酸精氨酸, 和
(iv) 约 0.3mg/ml的聚山梨酿 -20 ;
其中所述液体制剂的 pH为约 5.5±0.2, 优选地约 5.5。
16. 根据权利要求 1 - 1 5中任何一项所述的液体抗体制剂, 其特征在于, 该制剂在 储存后, 例如在 2-8°C储存至少 24个月后, 或在室温储存至少 3个月后, 或在 40°C±2°C 储存 1个月后, 是稳定的, 优选地具有如下特征之一或多项:
(i) 通过 SEC测量, 制剂中抗体单体的百分数大于 90%, 优选大于 95%, 且优选地 聚体增幅不超过 2% ;
(ii) 通过非还原型 CE-SDS测量, 制剂具有大于 90%的纯度, 优选大于 95%的纯度;
(iii) 通过 iCIEF测量, 相对于储存第 0天的初始值, 制剂中抗 CD47抗体的至少 50%, 优 选至少 55%是非碱性及非酸性形式, 且优选地抗体的酸性组分电荷变异体的增加不超过 20%。
17. 一种固体抗体制剂, 其通过固化权利要求 1-16中任何一项所述的液体抗体制 剂而获得, 所述固体抗体制剂例如是冻干制剂, 更优选地是冻干粉针剂形式。
18. 递送装置, 其包含权利要求 1-16中任何一项的液体抗体制剂或权利要求 17的固体 抗体制剂。
19. 预填装注射器,其包含权利要求 1-16中任何一项的液体抗体制剂或权利要求 17的固 体抗体制剂, 用于静脉内注射或者肌内注射。
20. 根据权利要求 1-16中任何一项的液体抗体制剂或权利要求 17的固体抗体制剂的用途, 用于制备在受试者中诱导抗肿瘤活性或抗癌症活性的药物, 优选地所述肿瘤或癌症选自血液 肿瘤和实体瘤, 例如急性骨髓性白血病 (AML) 、 慢性骨髓性白血病 (CML) 、 急性淋巴细胞 白血病 (ALL) 、 慢性淋巴细跑性白血病 (CLL) 、 非霍奇金淋巴瘤 (NHL) 、 多发性骨髓瘤
(MM) 、 淋巴瘤、 乳腺癌、 头颈癌、 胃癌、 肺癌、 食管癌、 肠癌、 卵巢癌、 宫颈癌、 肝癌、 肾癌、 胰腺癌、 膀胱癌、 结直肠癌、 神经胶质瘤、 黑素瘤和其他实体瘤, 其中所述肿瘤或癌 症更优选是淋巴瘤, 如伯基特淋巴瘤。
PCT/CN2020/076611 2019-02-26 2020-02-25 包含抗cd47抗体的制剂及其制备方法和用途 WO2020173431A2 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
KR1020217030704A KR20210134926A (ko) 2019-02-26 2020-02-25 항-cd47 항체를 포함하는 제제, 이의 제조 방법 및 이의 용도
CA3131307A CA3131307A1 (en) 2019-02-26 2020-02-25 Formulation comprising anti-cd47 antibody, preparation method therefor and use thereof
CN202080014878.4A CN113453719A (zh) 2019-02-26 2020-02-25 包含抗cd47抗体的制剂及其制备方法和用途
EP20763678.8A EP3932426A4 (en) 2019-02-26 2020-02-25 PREPARATIONS WITH ANTI-CD47 ANTIBODY, PROCESS FOR THEIR PRODUCTION AND THEIR USE
JP2021549889A JP2022521624A (ja) 2019-02-26 2020-02-25 抗cd47抗体を含む製剤、その調製方法および使用
US17/433,780 US20220143180A1 (en) 2019-02-26 2020-02-25 Formulation comprising anti-cd47 antibody, preparation method therefor and use thereof
AU2020227770A AU2020227770A1 (en) 2019-02-26 2020-02-25 Preparations containing anti-CD47 antibody, and preparation method and use therefor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910141894.9 2019-02-26
CN201910141894 2019-02-26

Publications (2)

Publication Number Publication Date
WO2020173431A2 true WO2020173431A2 (zh) 2020-09-03
WO2020173431A3 WO2020173431A3 (zh) 2020-10-22

Family

ID=72239062

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/076611 WO2020173431A2 (zh) 2019-02-26 2020-02-25 包含抗cd47抗体的制剂及其制备方法和用途

Country Status (9)

Country Link
US (1) US20220143180A1 (zh)
EP (1) EP3932426A4 (zh)
JP (1) JP2022521624A (zh)
KR (1) KR20210134926A (zh)
CN (1) CN113453719A (zh)
AU (1) AU2020227770A1 (zh)
CA (1) CA3131307A1 (zh)
TW (1) TWI764097B (zh)
WO (1) WO2020173431A2 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022121966A1 (en) * 2020-12-10 2022-06-16 Wuxi Biologics (Shanghai) Co., Ltd. An antibody against p-cadherin and uses thereof
WO2023039778A1 (zh) * 2021-09-16 2023-03-23 上海美雅珂生物技术有限责任公司 抗体药物偶联物制剂及其用途
WO2023056971A1 (en) * 2021-10-09 2023-04-13 Hutchmed Limited Anti-cd47 antibody formulation
EP4094777A4 (en) * 2020-01-21 2024-01-24 Innovent Biologics Suzhou Co Ltd RECOMBINANT FULLY HUMAN MONOCLONAL ANTI-TIGITE ANTIBODIES, METHOD FOR THE PRODUCTION THEREOF AND USE THEREOF

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023246790A1 (zh) * 2022-06-21 2023-12-28 广东菲鹏制药股份有限公司 一种含抗cd47抗体或其抗原结合片段的制剂及其制备方法和应用

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007008895A (ja) * 2005-07-04 2007-01-18 Chugai Pharmaceut Co Ltd 抗cd47抗体とインテグリンリガンドとの併用
HUE060541T2 (hu) * 2010-05-14 2023-03-28 Univ Leland Stanford Junior Humanizált és kiméra monoklonális CD47 elleni ellenanyagok
US9216219B2 (en) * 2012-06-12 2015-12-22 Novartis Ag Anti-BAFFR antibody formulation
US9221908B2 (en) * 2012-12-12 2015-12-29 Vasculox, Inc. Therapeutic CD47 antibodies
JP2018535692A (ja) * 2015-09-21 2018-12-06 エラスムス ユニバーシティ メディカル センターErasmus University Medical Center 抗cd47抗体及び使用方法
CN106084052B (zh) * 2016-06-17 2019-12-27 长春金赛药业股份有限公司 抗cd47单克隆抗体及其应用
CN106117354B (zh) * 2016-06-24 2020-01-14 安徽未名细胞治疗有限公司 一种全人源抗CD47的全分子IgG抗体及其应用
WO2018075960A1 (en) * 2016-10-21 2018-04-26 Tioma Therapeutics, Inc. Therapeutic cd47 antibodies

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Remington: the Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"The Handbook of Pharmaceutical Excipients", 2003, AMERICAN PHARMACEUTICALS ASSOCIATION
ALEXANDRE GOYON ET AL.: "Protocols for the analytical characterization of thermal monoclonal antibodies, I-Non-denaturing chromatographic techniques", JOURNAL OF CHROMATOGRAPHY, Retrieved from the Internet <URL:http://dx.doi.org/10.1016/j.jchromb.2017.05.010>
B. MINOWP. ROGGEK. THOMPSON, BIOPROCESS INTERNATIONAL, vol. 10, no. 6, 2012, pages 48 - 57
BORWANKAR A UDEAR B JTWU A ET AL.: "Viscosity reduction of a concentrated monoclonal antibody with arginineHCl and arginineglutamate[J", INDUSTRIAL & ENGINEERING CHEMISTRY RESEARCH, vol. 55, no. 43, 2016, pages 11225 - 11234
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
DADA 00 ET AL.: "Characterization of acidic and basic variants of IgG1 therapeutic monoclonal antibodies based on non-denaturing IEF fractionation", ELECTROPHORESIS, vol. 36, no. 21-22, 18 September 2015 (2015-09-18), pages 2695 - 2702
J. PHARM. BIO. ANAL., vol. 14, 1986, pages 1133 - 1140
J. PHARM. BIO. ANAL., vol. 15, 1997, pages 1928
J. PHARM. SCIEN., vol. 83, 1994, pages 1645 - 1650
JONES, A, ADV. DRUG DELIVERY REV., vol. 10, 1993, pages 29 - 90
JOURNAL OF INTERNATIONAL ONCOLOGY, vol. 40, no. 11, November 2013 (2013-11-01), pages 817 - 819
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH, pages: 247 - 301
KELLEY ET AL.: "Weak partitioning chromatography for anion exchange purification of monoclonal antibodies", BIOTECHNOLOGY AND BIOENGINEERING, vol. 101, 2008, pages 553 - 566, XP002545519, DOI: 10.1002/it.21923
NATIONAL PHARMACOPOEIA COMMITTEE: "Pharmacopoeia of the People's Republic of China", vol. IV, 2015, CHINA MEDICAL SCIENCE PRESS, article "Test for Visible Particles"
PHARM. RES., vol. 11, 1994, pages 485
R. YANG ET AL.: "High resolution separation of recombinant monoclonal antibodies by size exclusion ultra-high performance liquid chromatography (SE-UHPLC", JOURNAL OF PHARMACEUTICAL AND BIOMEDICAL ANALYSIS, 2015, Retrieved from the Internet <URL:http://dx.doi.org/10.1016/i,ipba.2Q15.02.032>
RICHARD R ET AL.: "Application of CE SDS gel in development of biopharmaceutical antibody-based products", ELECTROPHORESIS, vol. 29, 2008, pages 3612 - 3620
SALAS-SOLANO O ET AL.: "Robustness of iCIEF methodology for the analysis of monoclonal antibodies: an interlaboratory study", J SEP SCI, vol. 35, no. 22, 15 October 2012 (2012-10-15), pages 3124 - 9

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4094777A4 (en) * 2020-01-21 2024-01-24 Innovent Biologics Suzhou Co Ltd RECOMBINANT FULLY HUMAN MONOCLONAL ANTI-TIGITE ANTIBODIES, METHOD FOR THE PRODUCTION THEREOF AND USE THEREOF
WO2022121966A1 (en) * 2020-12-10 2022-06-16 Wuxi Biologics (Shanghai) Co., Ltd. An antibody against p-cadherin and uses thereof
WO2023039778A1 (zh) * 2021-09-16 2023-03-23 上海美雅珂生物技术有限责任公司 抗体药物偶联物制剂及其用途
WO2023056971A1 (en) * 2021-10-09 2023-04-13 Hutchmed Limited Anti-cd47 antibody formulation

Also Published As

Publication number Publication date
EP3932426A4 (en) 2022-12-14
AU2020227770A1 (en) 2021-09-23
TWI764097B (zh) 2022-05-11
JP2022521624A (ja) 2022-04-11
CN113453719A (zh) 2021-09-28
US20220143180A1 (en) 2022-05-12
CA3131307A1 (en) 2020-09-03
WO2020173431A3 (zh) 2020-10-22
KR20210134926A (ko) 2021-11-11
TW202045136A (zh) 2020-12-16
EP3932426A2 (en) 2022-01-05

Similar Documents

Publication Publication Date Title
JP5631591B2 (ja) 安定な抗体製剤
TWI764097B (zh) 包含抗cd47抗體的製劑及其製備方法和用途
TWI761869B (zh) 包含抗cd47/pd-l1雙特異性抗體的製劑及其製備方法和用途
JP2023535433A (ja) Pd-l1/lag-3二重特異性抗体製剤およびその調製方法ならびに使用
CN114146174A (zh) 抗pd-l1/ox40双特异性抗体制剂及其制备方法和用途
TWI782397B (zh) 重組全人源抗tigit單株抗體製劑及其製備方法和用途
CN114007648B (zh) 包含抗lag-3抗体的制剂、其制备方法及其用途
US20230173069A1 (en) Formulation comprising anti-il-23p19 antibody, method for preparing same and use thereof
WO2022111612A1 (zh) 包含抗tigit/pd-1双特异性抗体的制剂及其制备方法和用途
CN114206382B (zh) 包含抗pd-1/her2双特异性抗体的制剂及其制备方法和用途
CN112675300A (zh) 包含抗gitr抗体的制剂及其制备方法和用途

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: 20763678

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 3131307

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021549889

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020227770

Country of ref document: AU

Date of ref document: 20200225

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20217030704

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020763678

Country of ref document: EP

Effective date: 20210927