[go: up one dir, main page]

WO2024240224A1 - Formulation comprenant un anticorps anti-ccr8 et son utilisation - Google Patents

Formulation comprenant un anticorps anti-ccr8 et son utilisation Download PDF

Info

Publication number
WO2024240224A1
WO2024240224A1 PCT/CN2024/094955 CN2024094955W WO2024240224A1 WO 2024240224 A1 WO2024240224 A1 WO 2024240224A1 CN 2024094955 W CN2024094955 W CN 2024094955W WO 2024240224 A1 WO2024240224 A1 WO 2024240224A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
polysorbate
ccr8
antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2024/094955
Other languages
English (en)
Chinese (zh)
Inventor
苏智端
童仁超
朱利
陈柳华
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zai Lab Shanghai Co Ltd
ZLIP Holding Ltd
Original Assignee
Zai Lab Shanghai Co Ltd
ZLIP Holding Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zai Lab Shanghai Co Ltd, ZLIP Holding Ltd filed Critical Zai Lab Shanghai Co Ltd
Publication of WO2024240224A1 publication Critical patent/WO2024240224A1/fr
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons

Definitions

  • the present invention relates to the field of antibody preparations. More specifically, the present invention relates to a pharmaceutical preparation comprising an antibody and/or an antigen-binding fragment thereof that specifically binds to CCR8 and the therapeutic and/or preventive use of the pharmaceutical preparation.
  • CCR8 As a chemokine receptor, CCR8 (Chemokine receptor 8) is a seven-transmembrane G protein-coupled receptor that is highly expressed in tumor-infiltrating Treg cells and expressed at lower levels in Treg cells in the thymus, spleen, and peripheral blood. CCR8 participates in the development of various tumors and mediates tumor immunity through its interaction with its ligand. The specific high expression of CCR8 on Treg cells in tumor sites indicates that CCR8 is a very good biomarker for Treg cells in tumor sites and a highly potential tumor immunity target.
  • Antibodies targeting human CCR8 can lead to Treg depletion in tumors and sensitize tumors to immune checkpoint therapies such as anti-PD-1 therapy.
  • Anti-CCR8 antibodies that specifically bind to CCR8 are described in, for example, PCT patent application PCT/CN2022/136276, the contents of which are hereby incorporated by reference herein.
  • Antibody formulations not only need to be formulated in a manner suitable for administration to a subject, but also need to maintain their stability during storage and subsequent use, especially in liquid solutions where proteins tend to decompose, aggregate, denature, or undergo undesirable chemical modifications, etc. Although some anti-CCR8 antibodies are known, there is still a need in the art for anti-CCR8 antibody formulations that are stable enough for transportation and storage while being suitable for administration.
  • the present invention meets the above needs by providing a stable pharmaceutical preparation containing an antibody that specifically binds to CCR8.
  • the present invention provides a liquid antibody formulation comprising (i) an anti-CCR8 antibody or an antigen-binding fragment thereof; (ii) a buffer, (iii) a stabilizer, and (iv) a surfactant.
  • the anti-CCR8 antibody is any antibody that binds to a CCR8 molecule (e.g., a human CCR8 molecule), such as a polyclonal antibody, a monoclonal antibody, or a combination of the two.
  • a CCR8 molecule e.g., a human CCR8 molecule
  • the anti-CCR8 antibody is a monoclonal antibody.
  • the anti-CCR8 antibody or its antigen-binding fragment reduces or prevents the binding of CCR8 to CCL1, and thus, among other effects, inhibits the recruitment of regulatory T cells (Tregs) and/or the recruitment of signaling factors such as ⁇ -inhibitor proteins.
  • the concentration of the anti-CCR8 antibody or its antigen-binding fragment in the liquid antibody formulation of the present invention is about 1-200 mg/mL. In another embodiment, the concentration of the anti-CCR8 antibody or its antigen-binding fragment in the liquid antibody formulation of the present invention is about 1 mg/mL to about 100 mg/mL. In other embodiments, the concentration of the anti-CCR8 antibody or its antigen-binding fragment in the liquid antibody formulation of the present invention is about 1, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90 or 100 mg/mL.
  • the buffer is selected from the group consisting of histidine-histidine hydrochloride buffer system, citric acid-sodium citrate buffer system, acetic acid-sodium acetate buffer system, and phosphate buffer system.
  • the buffer in the liquid antibody formulation is selected from the group consisting of histidine, histidine hydrochloride, and combinations thereof.
  • the concentration of the buffer in the liquid antibody formulation of the present invention is about 0.1-100 mM. In one embodiment, the concentration of the buffer in the liquid antibody formulation of the present invention is about 1-50 mM, for example, about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mM.
  • the stabilizer is selected from sugars, polyols, amino acids and combinations thereof.
  • the stabilizer is selected from sucrose, trehalose, mannitol, arginine hydrochloride and combinations thereof.
  • the concentration of the stabilizer in the liquid antibody formulation of the present invention is about 0.1-20% (w/v). In one embodiment, the concentration of the stabilizer in the liquid antibody formulation of the present invention is about 1-10% (w/v), such as about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9% or 10%. In one embodiment, the stabilizer is about 0.1-20% (w/v) of a stabilizer selected from sugars and polyol stabilizers and amino acid stabilizers and combinations thereof.
  • the surfactant is a nonionic surfactant.
  • the surfactant is, for example, selected from polysorbate surfactants, such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80, etc., poloxamer, polyethylene glycol or a combination thereof.
  • the surfactant is polysorbate 80.
  • the concentration of the surfactant in the liquid antibody formulation of the present invention is about 0.001-0.5% (w/v). In one embodiment, the concentration of the surfactant in the liquid antibody formulation of the present invention is about 0.01-0.1% (w/v), such as about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09 or 0.1%.
  • the pH of the liquid formulation is about 4 to 8. In one embodiment, the pH of the liquid formulation is, for example, about 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5 or 8.
  • the liquid antibody formulation of the present invention comprises an anti-CCR8 antibody or antigen-binding fragment, a buffer, a stabilizer, and a surfactant
  • the buffer is selected from a histidine-histidine hydrochloride buffer system, a citric acid-sodium citrate buffer system, an acetic acid-sodium acetate buffer system, a phosphate buffer system, and a combination thereof
  • the stabilizer is selected from sucrose, trehalose, mannitol, arginine hydrochloride, and a combination thereof
  • the surfactant is selected from a polysorbate surfactant such as polysorbate 20, polysorbate 40, polysorbate 60, or polysorbate 80, poloxamer, polyethylene glycol, and a combination thereof.
  • the pH of the antibody formulation is about 4-8, more preferably, about 5-7.
  • liquid antibody formulation of the present invention comprises
  • a buffer selected from the group consisting of a histidine-histidine hydrochloride buffer system, a citric acid-sodium citrate buffer system, an acetic acid-sodium acetate buffer system, a phosphate buffer system, and a combination thereof;
  • a stabilizer selected from sugars, polyols, amino acids, and combinations thereof;
  • the pH of the liquid preparation is about 4-8.
  • liquid antibody formulation of the present invention comprises
  • a buffer selected from the group consisting of a histidine-histidine hydrochloride buffer system, a citric acid-sodium citrate buffer system, an acetic acid-sodium acetate buffer system, a phosphate buffer system, and combinations thereof;
  • a stabilizer selected from sucrose, trehalose, mannitol, arginine hydrochloride, and combinations thereof;
  • a surfactant selected from a polysorbate surfactant such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80, poloxamer, polyethylene glycol, and combinations thereof;
  • the pH of the liquid preparation is about 5-7.
  • liquid antibody formulation of the present invention comprises
  • a buffer selected from the group consisting of a histidine-histidine hydrochloride buffer system, a citric acid-sodium citrate buffer system, an acetic acid-sodium acetate buffer system, a phosphate buffer system, and a combination thereof;
  • a stabilizer selected from sucrose, trehalose, mannitol, arginine hydrochloride, and combinations thereof;
  • a surfactant selected from a polysorbate surfactant such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80, poloxamer, polyethylene glycol, and combinations thereof;
  • the pH of the liquid preparation is about 5-7.
  • liquid antibody formulation of the present invention comprises
  • a buffer selected from the group consisting of a histidine-histidine hydrochloride buffer system, a citric acid-sodium citrate buffer system, an acetic acid-sodium acetate buffer system, a phosphate buffer system, and a combination thereof;
  • a nonionic surfactant for example, selected from polysorbate surfactants such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80, poloxamers, polyethylene glycols and combinations thereof;
  • the pH of the liquid preparation is about 4.5-7.5.
  • the liquid antibody formulation of the present invention comprises about 1-200 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 1-100 mM histidine-histidine hydrochloride buffer system, about 0.1%-20% sucrose, about 0.01-0.5% polysorbate 80, and a pH of about 5-7.
  • the liquid antibody formulation of the present invention comprises about 1-100 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 1-50 mM histidine-histidine hydrochloride buffer system, about 1%-10% sucrose, about 0.01-0.1% polysorbate 80, and a pH of about 5-7.
  • the liquid antibody formulation of the present invention comprises about 10-50 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 1-50 mM histidine-histidine hydrochloride buffer system, about 1%-10% sucrose, about 0.01-0.1% polysorbate 80, and a pH of about 5-7.
  • the liquid antibody formulation of the present invention comprises about 10-50 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 20 mM histidine-histidine hydrochloride buffer system, about 7% sucrose, about 0.02-0.06% polysorbate 80, and a pH of about 5-7.
  • the liquid antibody formulation of the present invention comprises about 50 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 20 mM histidine-histidine hydrochloride buffer system, about 7% sucrose, about 0.02-0.06% polysorbate 80, and a pH of about 5-7.
  • the liquid antibody formulation of the present invention comprises about 50 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 20 mM histidine-histidine hydrochloride buffer system, about 7% sucrose, about 0.04% polysorbate 80, and a pH of about 6.
  • the anti-CCR8 antibody or antigen-binding fragment thereof in the liquid formulation of the present invention can specifically bind to CCR8 with high affinity, for example, with a KD of 10 -7 M or less, preferably 10 -8 M to 10 -12 M.
  • the anti-CCR8 antibody or antigen-binding fragment thereof in the liquid preparation of the present invention is an anti-CCR8 antibody or antigen-binding fragment thereof in the liquid preparation of the present invention
  • the anti-CCR8 antibody or antigen-binding fragment comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein
  • CDRs three complementary determining regions selected from any one of the VHs set forth in SEQ ID NOs: 1, 3, 4, 5, 6, 7, 11, 12, 26, 28, 30, 32, 34, 36, 37, 39, 41, 54-59, 61 and 66-70,
  • the anti-CCR8 antibody or antigen-binding fragment thereof in the liquid preparation of the present invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein:
  • the heavy chain variable region VH comprises or consists of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical, or 100% identical to an amino acid sequence selected from ID NO: 1, 3, 4, 5, 6, 7, 11, 12, 26, 28, 30, 32, 34, 36, 37, 39, 41, 54-59, 61 and 66-70;
  • the light chain variable region VL comprises or consists of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical, or 100% identical to an amino acid sequence selected from SEQ ID NO: 2, 8, 9, 10, 27, 29, 31, 33, 35, 38, 40, 42-53, 60 and 62-65.
  • the anti-CCR8 antibody or antigen-binding fragment thereof in the liquid formulation of the present invention comprises an IgG1 framework.
  • the antibody or antigen-binding fragment comprises one or more mutations in the Fc region, wherein the one or more mutations comprise S239D mutation, I332E mutation, L235V mutation, F243L mutation,
  • the one or more mutations enhance antibody-dependent cellular cytotoxicity (ADCC).
  • the liquid preparation of the present invention comprises about 1-200 mg/ml anti-CCR8 antibody or antigen-binding fragment, about 0.1-100 mM buffer, about 0.1-20% (w/v) sugar or polyol stabilizer or amino acid stabilizer or a combination thereof, about 0.01-0.5% (w/v) surfactant, and a pH of about 4.0-8.0, wherein the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2 and the LCDR3 of the anti-CCR8 antibody or antigen-binding fragment correspond to SEQ ID NO: 13, 14, 19, 16, 17 and 18 respectively.
  • the liquid of the invention is stable after storage, for example after storage at about 5°C ⁇ 3°C for at least 12 or 24 months, after storage at about 25°C ⁇ 2°C for at least 1, 2, 3 or 6 months, or after storage at 40°C ⁇ 2°C for 1 month, preferably having one or more of the following characteristics:
  • the main peak variation is less than 5% and/or the preparation has a purity greater than 95% as measured by SEC-HPLC;
  • the sum of the changes in the components of the protein in the formulation does not exceed 40% and/or the change in the main component does not exceed 20% relative to the initial value on storage day 0, for example, after storage at 40°C ⁇ 2°C for 1 month, the sum of the changes does not exceed about 40% or the change in the main component does not exceed about 20%, or after storage at 25°C for 2 months, the sum of the changes does not exceed about 20% or the change in the main component does not exceed about 15%;
  • the biological activity should be 70%-130% of the activity of the reference product.
  • the present invention provides a solid antibody formulation obtained by subjecting the liquid antibody formulation of the present invention to a solidification treatment.
  • the solidification treatment is implemented by, for example, crystallization, spray drying or freeze drying.
  • the solid antibody formulation is, for example, in the form of a lyophilized powder injection.
  • the solid antibody preparation can be reconstituted in an appropriate solvent before use to form a 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, an organic solvent for injection, including but not limited to oil for injection, ethanol, propylene glycol, etc., or a combination thereof.
  • the present invention provides a delivery device comprising the liquid antibody formulation or solid antibody formulation of the present invention as described in each embodiment of the present specification.
  • the delivery device of the present invention is provided in the form of a prefilled syringe containing the liquid antibody formulation or solid antibody formulation of the present invention as described in each embodiment of this specification, which is used for intravenous injection, subcutaneous injection or intramuscular injection, or is an infusion, such as for intravenous infusion.
  • the present invention relates to a method for delivering an anti-CCR8 antibody or antigen-binding fragment to a subject, such as a mammal, comprising the step of administering to the subject, such as a mammal, the liquid antibody formulation or solid antibody formulation of the present application as described in each embodiment of the specification by using a delivery device.
  • the present invention further provides a method for targeting a subpopulation of tumor infiltrating lymphocytes (TIL) in a subject, such as regulatory T cells (Tregs), in a subject, comprising administering to the subject a liquid antibody formulation or a solid
  • TIL tumor infiltrating lymphocytes
  • Regs regulatory T cells
  • the present invention further provides a use of a liquid antibody preparation or a solid antibody preparation of the present invention in preparing a delivery device or a drug for targeting a tumor infiltrating lymphocyte (TIL) subpopulation, such as regulatory T cells (Tregs), in a subject.
  • TIL tumor infiltrating lymphocyte
  • Regs regulatory T cells
  • the present invention further provides a method for treating cancer in a subject, comprising the step of administering to the subject the liquid antibody formulation or solid antibody formulation of the present invention, or a delivery device (e.g., a prefilled syringe) or a drug comprising the liquid antibody formulation or solid antibody formulation.
  • a delivery device e.g., a prefilled syringe
  • a drug comprising the liquid antibody formulation or solid antibody formulation.
  • the present invention further provides use of the liquid antibody formulation or solid antibody formulation of the present invention in the preparation of a delivery device or a medicament for treating cancer in a subject.
  • the above cancers include, for example, colorectal cancer, breast cancer, ovarian cancer, lung cancer (eg, non-small cell lung cancer), melanoma, liver cancer, kidney cancer, bladder cancer or pancreatic ductal adenocarcinoma.
  • FIG1 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of the present application in Example 2 with different formulations after being placed at 5 ⁇ 3° C. for 4 weeks.
  • FIG. 2 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 2 of the present application with different formulations after being placed at 25 ⁇ 2° C. for 4 weeks.
  • FIG3 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 2 of the present application with different formulations after being placed at 40° C. ⁇ 2° C. for 4 weeks.
  • FIG. 4 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 2 of the present application with different formulations after being placed at 5 ⁇ 3° C. for 4 weeks.
  • FIG5 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 2 of the present application with different formulations after being placed at 25° C. ⁇ 2° C. for 4 weeks.
  • FIG. 6 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 2 of the present application with different formulations after being placed at 40° C. ⁇ 2° C. for 4 weeks.
  • FIG. 7 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after being placed at 5 ⁇ 3° C. for 8 weeks.
  • FIG8 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after being placed at 25 ⁇ 2° C. for 8 weeks.
  • FIG. 9 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of the present application in Example 3 with different formulations after being placed at 40 ⁇ 2° C. for 4 weeks.
  • FIG. 10 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after repeated freezing and thawing.
  • FIG. 11 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after being placed at 5 ⁇ 3° C. for 8 weeks.
  • FIG. 12 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after being placed at 25 ⁇ 2° C. for 8 weeks.
  • FIG. 13 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after being placed at 40 ⁇ 2° C. for 4 weeks.
  • FIG. 14 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 3 of the present application with different formulations after repeated freezing and thawing.
  • FIG. 15 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 5 ⁇ 3° C. for 12 weeks.
  • FIG. 16 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 25 ⁇ 2° C. for 12 weeks.
  • FIG. 17 exemplarily shows the SEC-HPLC main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 40 ⁇ 2° C. for 4 weeks.
  • FIG. 18 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 5 ⁇ 3° C. for 12 weeks.
  • FIG. 19 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 25 ⁇ 2° C. for 12 weeks.
  • FIG. 20 exemplarily shows the icIEF main peak purity graph of the anti-CCR8 antibody preparations of Example 4 of the present application with different formulations after being placed at 40 ⁇ 2° C. for 4 weeks.
  • compositions of the present disclosure may comprise, consist essentially of, or consist of the disclosed components.
  • treatment is used herein, for example, with respect to a method of treating cancer, and generally includes administering a compound or composition to reduce the frequency of symptoms of a medical condition or delay its onset (e.g., cancer) relative to a subject not receiving the compound or composition. This can include reversing, reducing, or preventing symptoms, clinical signs, and underlying pathology of a disease in a manner that improves or stabilizes the subject's disease (e.g., regression of tumor growth).
  • the terms "subject,” “individual,” “host,” and “patient” are used interchangeably herein to refer to a mammal, e.g., a primate, preferably a higher primate, such as a human (e.g., a patient suffering from or at risk of suffering from a disorder described herein).
  • cancer refers to cells that exhibit relatively autonomous growth, so they exhibit cell proliferation with an abnormal growth phenotype characterized by a significant loss of control.
  • target cells for detection or treatment in the present application include precancerous (e.g., benign), malignant, pre-metastatic, metastatic and non-metastatic cells.
  • normal used in the context of “normal cells” refers to cells of an untransformed phenotype or the morphology of untransformed cells that exhibit the type of tissue being examined.
  • “Cancerous phenotype” generally refers to any of a variety of biological phenomena characteristic of cancer cells, which can vary with the type of cancer. Cancerous phenotypes are typically identified by abnormalities in aspects such as cell growth or proliferation (e.g., uncontrolled growth or proliferation), cell cycle regulation, cell mobility, cell-cell interactions or metastasis.
  • antibody preparation refers to a preparation that is in a form that permits the biological activity of the antibody as an active ingredient to be effective, and contains no additional components that are unacceptably toxic to a subject to which the preparation would be administered.
  • anti-CCR8 antibody preparation means a combination of at least one anti-CCR8 antibody as an active ingredient and at least one inactive ingredient. After such combination, the anti-CCR8 antibody as an active ingredient is suitable for therapeutic or prophylactic administration to humans or non-human animals.
  • the antibody preparation of the present invention can be sterile, homogeneous and/or isotonic, and the antibody preparation can be directly prepared as a liquid preparation in aqueous form, for example, a ready-to-use prefilled syringe, or prepared as a lyophilized preparation, and reconstituted (i.e., reconstituted) by dissolving and/or suspending in a physiologically acceptable solution immediately before use.
  • the anti-CCR8 antibody preparation is in the form of a liquid preparation, a lyophilized preparation, or a reconstituted preparation.
  • lyophilized preparation refers to a composition obtained or obtainable by freeze-drying a liquid preparation.
  • substitution refers to the process of dissolving and/or suspending a solid formulation (eg, a lyophilized formulation) in a physiologically acceptable solution to obtain a liquid formulation.
  • the anti-CCR8 antibody preparation of the present invention exhibits low to undetectable levels of antibody aggregation or degradation or chemical modification during manufacturing, preparation, transportation and long-term storage, thereby having little or no loss of biological activity of the anti-CCR8 antibody, and exhibiting high stability.
  • the antibodies contained in the antibody preparation do not maintain their chemical structure 100% after storage for a specific period of time, generally after storage for a specific period of time, if the chemical structure is maintained, If about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% of the antibody structure or function is retained, the antibody formulation is considered “stable.”
  • the anti-CCR8 antibody formulation of the invention substantially retains its physical and chemical stability after storage.
  • the pharmaceutical composition After a long period of storage, if the pharmaceutical composition shows no visible signs of precipitation, aggregation and/or denaturation (i.e., discoloration or loss of transparency) or measurable signs (e.g., using size exclusion chromatography (SEC) or UV light scattering), it is considered to retain its physical stability at a given time point.
  • SEC size exclusion chromatography
  • UV light scattering e.g., UV light scattering
  • chemical stability when a protein such as an antibody is formulated into a pharmaceutical composition, it is considered to retain its chemical stability at a given time point if the measurement of chemical stability indicates that the protein (i.e., antibody) retains the biological activity of interest in the pharmaceutical composition.
  • Methods for monitoring changes in chemical stability include, but are not limited to, methods for detecting chemically altered forms of proteins using, for example, SDS-PAGE, SEC and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry; and those related to changes in molecular charge using, for example, whole-column imaging capillary isoelectric focusing (iCIEF).
  • methods for detecting chemically altered forms of proteins using, for example, SDS-PAGE, SEC and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry
  • iCIEF whole-column imaging capillary isoelectric focusing
  • the stability of the preparation is measured by measuring the percentage of non-aggregated and non-decomposed antibodies in the preparation at a selected temperature within a specific time period, wherein the higher the percentage of non-aggregated and non-decomposed antibodies in the preparation, the higher the stability of the preparation.
  • the percentage of non-aggregated and non-decomposed antibodies can be measured by size exclusion chromatography (e.g., size exclusion high performance liquid chromatography).
  • an acceptable degree of stability means that at least about 92% of non-aggregated and non-degraded anti-CCR8 antibodies are detected in the formulation after storage at a specific temperature for a specific time.
  • an acceptable degree of stability means at least about 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% non-aggregated and non-degraded anti-CCR8 antibodies after storage at a specific temperature for at least 2 weeks, at least 28 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, at least 12 months, at least 18 months, at least 24 months, or more.
  • the specific temperature at which the pharmaceutical formulation is stored can be any temperature between about -80°C and about 45°C, for example, 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, about 35°C, about 37°C, about 40°C, about 42°C, or about 45°C.
  • the stable anti-CCR8 antibody formulations of the invention are administered parenterally to a subject.
  • parenteral administration includes, but is not limited to, subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intracranial, intrathecal, and intraperitoneal injections, and any similar injection or infusion techniques.
  • the anti-CCR8 antibody formulation of the present invention is administered subcutaneously to a subject.
  • the dosage and frequency of administration are determined based on age, sex, patient condition, other drugs taken concurrently, adverse reactions, and other factors considered by the clinician.
  • the stable liquid antibody formulation of the present invention comprises: (i) an anti-CCR8 antibody or an antigen-binding fragment thereof; (ii) a buffer; (iii) a stabilizer, and (iv) a surfactant, and the pH of the anti-CCR8 antibody formulation is about 4.0-8.0.
  • the antibody preparation of the present invention may include an anti-CCR8 antibody or an antigen-binding fragment thereof that specifically binds to a CCR8 molecule.
  • CCR8 is known to be a seven-transmembrane G protein-coupled receptor that belongs to a chemokine receptor protein.
  • Anti-CCR8 antibodies as CCR8 are disclosed in the prior art. For example, CN113260381A. The entire text of the document is incorporated herein by reference.
  • anti-CCR8 antibody refers to an antibody that is capable of binding to the CCR8 molecule with sufficient affinity so that the antibody can be used as a therapeutic and/or prophylactic agent targeting the CCR8 molecule.
  • the binding of an antibody or antigen-binding fragment to CCR8 can be characterized or expressed by any method known in the art. For example, binding can be characterized by binding affinity, which can be the strength of the interaction between the antibody or antigen-binding fragment and the antigen. Binding affinity can be determined by any method known in the art, such as an in vitro binding assay.
  • the binding affinity of an antibody or antigen-binding fragment disclosed herein can be determined when assayed in an in vitro binding assay using cells expressing CCR8.
  • the equilibrium dissociation constant ( KD ) of an anti-CCR8 antibody is ⁇ 1 ⁇ M, ⁇ 100nM, ⁇ 10nM, ⁇ 1nM, ⁇ 0.1nM, ⁇ 0.01nM, or ⁇ 0.001nM.
  • the anti-CCR8 antibody can specifically bind to CCR8 with high affinity, for example, with a KD of 10-7 M or less, preferably 10-8 M to 10-12 M.
  • antigen-binding fragments of anti-CCR8 antibody are also included.
  • antibodies are composed of an antigen binding site (Fab) and a crystallizable fragment (Fc).
  • Fab can bind to a specific antigen, thereby determining the specificity and affinity of the antibody.
  • the Fc region consists of two heavy chains and is used to regulate the activity of immune cells. Depending on the type of antibody, each heavy chain in this region consists of 2 or 3 constant domains.
  • the Fc segment can bind to Fc receptors (Fc ⁇ RI, Fc ⁇ RII, Fc ⁇ RIII) expressed on the surface of immune cells, complement (C1q) and FcRn in the blood, thereby activating immune effects to clear foreign substances.
  • the Fc segment determines the effector function of the antibody, including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • CDR region or “CDR” is those parts of the variable region of the heavy or light chain of an antibody that are arranged side by side in three-dimensional space to form an antigen binding surface. CDR is primarily responsible for binding to an antigen epitope.
  • the CDRs of the heavy and light chains are usually referred to as CDR1, CDR2, and CDR3, and are numbered sequentially from the N-terminus.
  • the CDRs located within the variable domain of the heavy chain of an antibody are referred to as HCDR1, HCDR2, and HCDR3, while the CDRs located within the variable domain of the light chain of an antibody are referred to as LCDR1, LCDR2, and LCDR3, which are located between relatively conservative framework regions (FRs).
  • both the light and heavy chain variable domains include FR-1 (or FR1), CDR-1 (or CDR1), FR-2 (FR2), CDR-2 (CDR2), FR-3 (or FR3), CDR-3 (CDR3), and FR-4 (or FR4).
  • the positions of CDRs and framework regions can be determined using a variety of definitions well known in the art, such as Kabat, Chothia, and AbM (see, e.g., Johnson et al., Nucleic Acids Res., 29:205-206 (2001); Chothia and Lesk, J. Mol.
  • the scope of the antibodies also covers antibodies whose variable region sequences comprise the specific CDR sequences but whose declared CDR boundaries differ from the specific CDR boundaries defined herein due to the application of a different scheme (e.g., a different assignment system rule or combination).
  • conservative substitution means replacing the original amino acid with a new amino acid that does not substantially change the chemical, physical and/or functional properties of the antibody or fragment, such as its binding affinity to CCR8.
  • common conservative substitutions of amino acids are well known in the art, such as substitutions of amino acid residues with similar side chains.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • non-polar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • ⁇ -branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • one or more amino acid residues of an anti-CCR8 antibody can be replaced with other amino acid residues from the same side chain family, and the altered antibody can be tested for function, particularly binding properties to the same CCR8 molecule.
  • Changes in charge presentation on the CDR surface are expected to affect the interface of the antibody with the solvent, and therefore, non-conservative amino acid residue substitutions have unpredictable effects on maintaining or improving the stability of the antibody in solution.
  • antibody or antigen-binding fragment includes, but is not limited to, polyclonal, monoclonal, monovalent, bispecific, multispecific, recombinant, heterologous, heterohybrid, human antibodies, humanized antibodies, chimeric antibodies, single-chain antibodies, nanobodies, and anti-idiotypic (anti-Id) antibodies, Fab fragments, Fab' fragments, F(ab') 2 fragments, fragments produced by Fab expression libraries, Fd, Fv, disulfide-linked Fv (dsFv).
  • Antibodies can be of any isotype/class (e.g., IgG, IgE, IgM, IgD, IgA, and IgY) or subclass (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2).
  • the antigen-binding fragment refers to a molecule different from an intact antibody, which comprises a portion of an intact antibody and binds to the antigen to which the intact antibody binds.
  • Humanized antibodies refer to chimeric antibodies that contain amino acid residues of complementary determining regions other than human and amino acid residues of framework regions of human antibodies.
  • humanized antibodies will contain substantially all of at least one, usually two variable domains, wherein all or substantially all of the CDR regions correspond to those of non-human immunoglobulins, and all or substantially all of the FR regions are regions of human immunoglobulin sequences.
  • Humanized antibodies may also contain at least a portion of an immunoglobulin constant region (Fc), typically a constant region of a human immunoglobulin, such as human IgG1. Humanized antibodies can be used when administered to humans for therapeutic purposes, etc., because their antigenicity is reduced in humans.
  • Fc immunoglobulin constant region
  • specific binding means that an antibody or its antigen-binding fragment forms a relatively stable complex with an antigen under physiological conditions.
  • Specific binding is characterized by an equilibrium dissociation constant of at least about 1 ⁇ 10-6 M or less (e.g., the smaller the KD , the tighter the binding).
  • the KD value is preferably 1 ⁇ 10-7 M or less, more preferably 1 ⁇ 10-8 M or less, and further preferably 1 ⁇ 10-9 M to 10-12 M.
  • the percent homology between two amino acid sequences is equal to the percent identity between the two sequences.
  • Sequence alignment can be performed using various methods in the art to determine the percent amino acid sequence identity, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or MEGALIGN (DNASTAR) software.
  • One skilled in the art can determine the appropriate parameters for measuring the comparison, including any algorithm required for obtaining the maximum comparison over the entire length of the compared sequence.
  • the amount of the antibody or antigen-binding fragment thereof contained in the antibody preparation of the present invention may vary with the specific purpose characteristics of the preparation, the specific environment, and the specific purpose for which the preparation is used.
  • the concentration of the anti-CCR8 antibody or antigen-binding fragment in the liquid preparation is about 1 to about 200 mg/mL, preferably about 1-100 mg/mL, such as about 1, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90 or 100 mg/mL.
  • the anti-CCR8 antibody formulations of the invention described in each embodiment herein are stable.
  • the purity of the anti-CCR8 antibody in the antibody formulation of the invention is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more, as determined by size exclusion chromatography.
  • At least 50% of the anti-CCR8 antibody in the antibody formulation of the invention is in a non-basic and non-acidic form (i.e., the main peak or the main charge form), as determined by iCIEF.
  • the anti-CCR8 antibody or antigen-binding fragment described in the present application comprises a light chain and/or a heavy chain, which specifically binds to CCR8 and/or blocks the binding of CCL1 to CCR8.
  • the anti-CCR8 antibody or antigen-binding fragment described in the present application comprises a light chain and/or a heavy chain, and a crystallizable fragment (Fc) region operably linked to the antibody or antigen-binding fragment, wherein the antibody or antigen-binding fragment specifically binds to CCR8 and/or blocks the binding of CCL1 to CCR8.
  • the liquid antibody formulation provided by the present application comprises an anti-CCR8 antibody or antigen-binding fragment, wherein the anti-CCR8 antibody or antigen-binding fragment comprises a light chain and/or a heavy chain, and a crystallizable fragment (Fc) region comprising a mutation in at least one amino acid position operably linked to the antibody or antigen-binding fragment, wherein the antibody or antigen-binding fragment specifically binds to CCR8 and/or blocks the binding of CCL1 to CCR8, and wherein the Fc Mutations in the mitochondrial region enhance antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • the crystallizable fragment (Fc) region comprises at least a portion of a human immunoglobulin constant region (Fc) with or without any mutations.
  • the mutation comprises an S239D mutation, an I332E mutation, an L235V mutation, an F243L mutation, an R292P mutation, a Y300L mutation, a P396L mutation, or a combination thereof.
  • the antibody of the present application is an IgG1, IgG2, IgG3 or IgG4 isotype.
  • the antibody of the present application comprises the Fc domain of wild-type human IgG1.
  • the antibody of the present application comprises at least a portion of the Fc domain of human IgG1 with S239D/I332E mutation or L235V/F243L/R292P/Y300L/P396L mutation (according to the EU numbering system).
  • the crystallizable fragment (Fc) region comprises at least a portion of human IgG1 Fc (wild type, SEQ ID NO: 119), or human IgG1 Fc including S239D/I332E mutations (SEQ ID NO: 121), or at least a portion of human IgG1 Fc including L235V/F243L/R292P/Y300L/P396L mutations (SEQ ID NO: 120), wherein the mutations enhance antibody-dependent cellular cytotoxicity (ADCC), thereby potentially better promoting the depletion of Tregs.
  • ADCC antibody-dependent cellular cytotoxicity
  • the crystallizable fragment (Fc) region comprises an amino acid sequence selected from SEQ ID NOs: 119-121:
  • the antibody or antigen-binding fragment comprises a light chain CDR (LCDR or LCDR1-3).
  • the light chain CDR can be the complementary determining region of the light chain of the antibody or antigen-binding fragment.
  • the light chain CDR may comprise a continuous sequence of amino acid residues, or two or more continuous sequences of amino acid residues, which are separated by non-complementary determining regions such as framework regions, and are optionally located on its flanks.
  • the light chain CDR comprises two or more light chain CDRs, which may be referred to as LCDR1, LCDR2, etc.
  • the light chain CDR comprises three light chain CDRs, which may be referred to as LCDR1, LCDR2, and LCDR3, respectively.
  • a group of CDRs present on a common light chain may be collectively referred to as LCDR.
  • the antibody or antigen-binding fragment comprises a heavy chain CDR (HCDR or HCDR1-3).
  • Chain CDR can be the complementary determining region of the heavy chain of antibody or antigen binding fragment.
  • Heavy chain CDR can comprise continuous amino acid residue sequence, or two or more continuous amino acid residue sequence, they are separated by non-complementary determining region such as framework region and are optionally located at its flank.
  • heavy chain CDR comprises two or more heavy chain CDR, it can be referred to as HCDR1, HCDR2 etc.
  • heavy chain CDR comprises three heavy chain CDR, it can be referred to as HCDR1, HCDR2, HCDR3 respectively.
  • a group of CDR present on common heavy chain can be collectively referred to as HCDR.
  • the antibody or antigen binding fragment specifically binds to CCR8.
  • CCR8 can also refer to an ortholog, homolog, codon optimized form, truncated form, fragmented form, mutant form or any other known derivative form of the CCR8 sequence.
  • CCR8 can be human CCR8, mouse CCR8 or cynomolgus monkey CCR8.
  • Binding specificity can be determined by complementary determining region or CDR such as light chain CDR or heavy chain CDR. In many cases, binding specificity is determined by light chain CDR and heavy chain CDR. Compared with other reference antigens, a given combination of heavy chain CDR and light chain CDR provides a given binding pocket that gives greater affinity and/or specificity to CCR8. In some embodiments, antibodies or antigen binding fragments reduce or even prevent the combination of CCR8 and CCL1, and therefore, among other effects, suppress the recruitment of regulatory T cells and/or signal factors such as ⁇ -inhibitor proteins.
  • the antibody or antigen-binding fragment may comprise any of the sequences listed in Table 1. Additionally or alternatively, the antibody or antigen-binding fragment may comprise a sequence that is at least 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the sequences listed in Tables 1-3, including any ranges having such values as endpoints (e.g., 60%-85%).
  • the antibody or antigen-binding fragment comprises vH and vL selected from the following, or any combination of LCDR and HCDR.
  • anti-CCR8 antibodies that can be included in the antibody formulations of the present invention are, for example, novel anti-CCR8 antibodies that specifically bind to CCR8 as described in PCT patent application PCT/CN2022/136276, Hu149-1, Hu149-2, Hu149-3, Hu149-4, Hu149-5, Hu149-6, Hu149-7, Hu149-8, Hu149-9, 11-24, Hu149 -11G1, Hu149-11G1m, Hu149-11G1x, Hu149-12G1m, Hu149-12G1x, Hu348-1, Hu348-2, Hu348-3, Hu348-4, Hu348-5, Hu348-6, Hu348-7, Hu348-8, Hu348-9, Hu348-4-m2G1, Hu348-4-m2G1m and Hu34 8-4-m2G1x, 191E12H8, 234G9C12, 273H2G6, 348E2D11, 149F2C10, 153D4G2, 160D1E3, 164G10D
  • variable region comprises wild-type human IgG1 Fc (SEQ ID NO: 119), or human IgG1 Fc including S239D/I332E mutations (SEQ ID NO: 121, expressed as a G1x suffix), or human IgG1 Fc including L235V/F243L/R292P/Y300L/P396L mutations (SEQ ID NO: 120, expressed as a G1m suffix).
  • the anti-CCR8 antibody or antigen-binding fragment in the antibody preparation described in the present application comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein
  • CDRs three complementary determining regions selected from any one of the VHs set forth in SEQ ID NOs: 1, 3, 4, 5, 6, 7, 11, 12, 26, 28, 30, 32, 34, 36, 37, 39, 41, 54-59, 61, and 66-70,
  • the HCDR1 in the antibody preparation described in the present application comprises a sequence selected from SEQ ID NO: 13 and 115
  • the HCDR2 comprises a sequence selected from SEQ ID NO: 14, 89 and 114
  • the HCDR3 comprises a sequence selected from SEQ ID NO: 15 and 19-22
  • the LCDR1 comprises a sequence selected from SEQ ID NO: 16, 90 and 102-106
  • the LCDR2 comprises a sequence selected from SEQ ID NO: 17
  • the LCDR3 comprises a sequence selected from SEQ ID NO: 18 and 23-25.
  • the HCDR1 comprises a sequence selected from SEQ ID NO: 83 and 117
  • the HCDR2 comprises a sequence selected from SEQ ID NO: 84, 116 and 118
  • the HCDR3 comprises a sequence of SEQ ID NO: 85;
  • the LCDR1 contains the sequence of SEQ ID NO:86
  • LCDR2 contains the sequence of SEQ ID NO:87
  • LCDR3 contains the sequence of SEQ ID NO:88.
  • the HCDR1, HCDR2 and HCDR3 in the antibody formulation of the present invention are an amino acid sequence selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14 and SEQ ID NO: 15; SEQ ID NO: 13, SEQ ID NO: 14 and SEQ ID NO: 19; SEQ ID NO: 13, SEQ ID NO: 14 and SEQ ID NO: 20; SEQ ID NO: 13, SEQ ID NO: 14 and SEQ ID NO: 21; SEQ ID NO: 13, SEQ ID NO: 14 and SEQ ID NO: 22; SEQ ID NO: 71, SEQ ID NO: 72 and SEQ ID NO: 73; SEQ ID NO: 71, SEQ ID NO: 77 and SEQ ID NO: 78; SEQ ID NO: 71, SEQ ID NO: 72 and SEQ ID NO: 81; SEQ ID NO: 83, SEQ ID NO: 84 and SEQ ID NO: 85; SEQ ID NO: 13, SEQ ID NO: 89 and SEQ ID NO: 15; SEQ ID NO: 13
  • the LCDR1, the LCDR2 and the LCDR3 are an amino acid sequence selected from the following group: SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 18; SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 23; SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 24; SEQ ID NO: 16, SEQ ID NO: 17 and SEQ ID NO: 25; SEQ ID NO: 74, SEQ ID NO: 75 and SEQ ID NO:76; SEQ ID NO:79, SEQ ID NO:80 and SEQ ID NO:76; SEQ ID NO:79, SEQ ID NO:75 and SEQ ID NO:82; SEQ ID NO:86, SEQ ID NO:87 and SEQ ID NO:88; SEQ ID NO:90, SEQ ID NO:17 and SEQ ID NO:18; SEQ ID NO:94, SEQ ID NO:95 and SEQ ID NO:96; SEQ ID NO :79, SEQ ID NO:75 and SEQ ID NO:76; S
  • the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2 and the LCDR3 in the antibody preparation of the present invention are an amino acid sequence selected from the following groups: SEQ ID NO: 13, 14, 15, 16, 17 and 18; SEQ ID NO: 13, 14, 19, 16, 17 and 18; SEQ ID NO: 13, 14, 20, 16, 17 and 18; SEQ ID NO:13, 14, 21, 16, 17 and 18; SEQ ID NO:13, 14, 22, 16, 17 and 18; SEQ ID NO:13, 14, 15, 16, 17 and 23; SEQ ID NO:13, 14, 15, 16, 17 and 24; SEQ ID NO:13, 14, 15, 16, 17 and 25; SEQ ID NO:13, 14, 19, 16, 17 and 23; SEQ ID NO:13, 14, 15, 16, 17 and 24; D NO:13, 14, 20, 16, 17 and 23; SEQ ID NO:13, 14, 21, 16, 17 and 23; SEQ ID NO:13, 14, 22, 16, 17 and 23; SEQ ID NO:13, 89, 15, 90, 17 and 18; SEQ ID NO:13, 89,
  • the HCDR1, the HCDR2, the HCDR3, the LCDR1, the LCDR2 and the LCDR3 in the antibody preparation of the present invention are an amino acid sequence selected from the following groups: SEQ ID NO: 83, 84, 85, 86, 87 and 88; SEQ ID NO: 83, 116, 85, 86, 87 and 88; SEQ ID NO: 117, 116, 85, 86, 87 and 88; and SEQ ID NO: 83, 118, 85, 86, 87 and 88.
  • the anti-CCR8 antibody or antigen-binding fragment in the antibody preparation of the present invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein:
  • the heavy chain variable region VH comprises or consists of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical, or 100% identical to an amino acid sequence selected from ID NO: 1, 3, 4, 5, 6, 7, 11, 12, 26, 28, 30, 32, 34, 36, 37, 39, 41, 54-59, 61 and 66-70;
  • the light chain variable region VL comprises or consists of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical, or 100% identical to an amino acid sequence selected from SEQ ID NO: 2, 8, 9, 10, 27, 29, 31, 33, 35, 38, 40, 42-53, 60 and 62-65.
  • the anti-CCR8 antibody or antigen-binding fragment in the antibody preparation of the present invention comprises a heavy chain variable region VH and a light chain variable region VL, wherein the heavy chain variable region VH and the light chain variable region VL respectively comprise an amino acid sequence selected from the following groups: SEQ ID NO: 1 and 2; SEQ ID NO: 3 and 2; SEQ ID NO: 4 and 2; SEQ ID NO: 5 and 2; SEQ ID NO: 6 and 2; SEQ ID NO: 7 and 2; SEQ ID NO: 1 and 8; SEQ ID NO: 1 and 9; SEQ ID NO: 1 and 10; SEQ ID NO: 3 and 8; SEQ ID NO: 4 and 8; SEQ ID NO: 5 and 8; SEQ ID NO: 6 and 8; SEQ ID NO:11 and 8; SEQ ID NO:12 and 8; SEQ ID NO:34 and 35; SEQ ID NO:34 and 43; SEQ ID NO:34 and 44; SEQ ID NO:34 and 45; SEQ ID NO:34 and 46; SEQ ID NO
  • the heavy chain variable region VH and light chain variable region VL in the antibody preparation of the present invention respectively comprise amino acid sequences selected from the following groups: SEQ ID NO: 32 and 33; SEQ ID NO: 61 and 62; SEQ ID NO: 61 and 63; SEQ ID NO: 61 and 64; SEQ ID NO: 61 and 65; SEQ ID NO: 66 and 62; SEQ ID NO: 67 and 62; SEQ ID NO: 68 and 62; SEQ ID NO: 69 and 62; and SEQ ID NO: 70 and 62.
  • the antibody in the antibody preparation of the present invention can be selected from polyclonal antibodies, monoclonal antibodies, humanized antibodies, chimeric antibodies, monovalent antibodies, multivalent antibodies, bispecific antibodies, multispecific antibodies, and any combination thereof.
  • the antigen-binding fragment in the antibody preparation of the present invention may be scFv, Fab', single-chain Fab (scFab'), Fd or F(ab') 2 , sFC or VHH.
  • Solution pH and buffer are among the most important parameters affecting the physicochemical stability of biopharmaceuticals. pH can regulate the charge distribution on the protein surface, thereby affecting the mutual attraction and repulsion within and between biopharmaceutical molecules. It can also regulate the interaction between biopharmaceuticals and other small molecules such as excipients. Therefore, pH is a key factor in formulation optimization. Different buffers can also affect the stability of protein molecules.
  • buffer or pH adjusting agent is used to maintain acceptable pH level in liquid preparation, so as to maintain the stability of antibody.
  • Liquid preparation is usually adjusted to make pH remain in the scope of about 4-8, about 4.5-7.5, about 5.0-6.5 or about 5.3-6.3.
  • the scope between the above-mentioned pH also wishes to be a part of the present invention.For example, it is desirable to include the scope of the value of using the combination of the above-mentioned any value as the upper limit and/or lower limit.
  • the pH value of the liquid preparation is, for example, about 4,4.5,5,5.5,6,6.5,7,7.5 or 8. In one embodiment, the pH value of the liquid preparation is, for example, about 5-7.
  • Suitable buffers for use in the present invention include, but are not limited to, histidine-histidine hydrochloride buffer systems, citric acid-sodium citrate buffer systems, acetic acid-sodium acetate buffer systems, and phosphate buffer systems.
  • the buffer in the liquid antibody formulation is selected from histidine, histidine hydrochloride, and combinations thereof.
  • the concentration of the buffer included in the antibody formulation of the present invention is about 0.1-100 mM. In some embodiments, the concentration of the buffer included in the antibody formulation of the present invention is about 1-50 mM, such as about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 mM.
  • Suitable stabilizers for the present invention may be sugars, polyols and amino acids, and their mechanism of protecting proteins is "preferential exclusion". Such excipients are preferentially excluded from the surface of proteins, increasing the chemical potential energy of proteins. Since the surface area increases after protein denaturation and unfolding, the interaction with excipients increases, causing the chemical potential energy to increase, thereby increasing the activation energy required for protein conformational denaturation, making proteins naturally more resistant to sugars as stabilizers.
  • monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, etc.
  • disaccharides such as lactose, sucrose, trehalose, cellobiose, etc.
  • polysaccharides such as raffinose, melezitose, maltodextrin, dextran, starch, etc.
  • sugar alcohols such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol), etc., and combinations thereof.
  • the sugar can be sucrose, trehalose, mannitol, or a combination thereof.
  • the stabilizer may also be an amino acid stabilizer, such as arginine hydrochloride.
  • composition of the present invention may optionally further comprise a pharmaceutically acceptable surfactant.
  • surfactant is a class of amphiphilic molecules having both a hydrophilic group and a lipophilic group, which can change (generally reduce) the surface tension of a liquid or a substance that has an interfacial tension between two phases.
  • a "surfactant” can help improve the stability of any anti-CCR8 antibody formulation described herein by reducing the surface tension of the liquid or the interfacial tension between two phases.
  • a surfactant can reduce the aggregation of the formulated antibody and/or minimize the formation of particles in the formulation and/or reduce adsorption.
  • Surfactants can also improve the stability of antibodies during and after freeze/thaw cycles.
  • the surfactant in the liquid preparation of the present invention is a nonionic surfactant.
  • Nonionic surfactants can inhibit protein aggregation, and the mechanism may be that the surfactant tends to be arranged at the air-liquid interface, thereby causing the protein to leave the interface to inhibit protein aggregation denaturation.
  • nonionic surfactants that can be included in the formulations of the present invention include, for example, polysorbates, such as polysorbate 20, polysorbate 40, polysorbate 60, or polysorbate 80, poloxamers, polyethylene glycol, pluronics, or combinations thereof. In one embodiment, polysorbate 80 is preferred.
  • the amount of nonionic surfactant contained in the antibody formulation of the present invention can change with the specific purpose characteristics of the formulation, the specific environment, and the specific purpose of using the formulation.
  • the formulation can contain a concentration of about 0.001-0.5% (w/v), preferably about 0.01-0.1% (w/v), such as about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09 or 0.1% (w/v).
  • the liquid antibody preparation of the present invention adopts recombinant DNA technology to efficiently express specific anti-CCR8 antibodies or antigen-binding fragments thereof in the mammalian cell expression system CHO, and after cell culture, harvesting, virus inactivation, and high purification, specific auxiliary materials are added, and it is prepared by common preparation processes in the field such as sterilization filtration, filling, stoppering, capping and visual inspection.
  • the present invention obtains a solid antibody preparation by solidifying the liquid antibody preparation, wherein the solidification is performed by, for example, crystallization, spray drying or freeze drying, and the solid antibody preparation is, for example, in the form of a lyophilized powder injection.
  • the stability of the purified antibody composition produced by the process during storage is related to the quality attributes of the product. For example, aggregation, degradation or chemical modification of antibodies can lead to antibody heterogeneity, including size heterogeneity and charge heterogeneity, thereby affecting the quality of antibody drug products. Therefore, it is necessary to monitor the stability of the purified antibody composition produced by the process.
  • Product stability 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, CEX-HPLC method, etc.).
  • the efficacy or biological activity of the antibody can be detected.
  • the binding ability of the antibody in the preparation to its antigen CCR8 can be detected.
  • various methods can be used to quantify the specific binding of antibodies to antigens, such as immunoassays, ELISA, etc.
  • Stability studies generally include long-term stability studies, accelerated stability studies, and forced condition tests under actual storage conditions. Under accelerated and forced conditions, degradation is accelerated, and degradation products can be monitored at an earlier time point and in larger amounts. Therefore, accelerated and forced condition tests can be used to clarify the degradation pathway of the target antibody product, which helps to understand the nature (mechanism) of the degradation of the target antibody product; provide the stability of the product under deviation from the optimal storage conditions and extreme conditions.
  • the antibody composition sample is placed at an elevated temperature, such as about 40°C ⁇ 2°C, 25°C ⁇ 2°C, for forced or accelerated stability studies.
  • an oscillation experiment or a repeated freeze-thaw experiment can be performed to detect the stability properties of the product.
  • an oscillation experiment is performed at room temperature, away from light, and 650r/min is shaken for 1-5 days.
  • a product can be frozen below -30°C for one day and then melted at room temperature or in a water bath at 25°C as a freeze-thaw cycle, and a repeated freeze-thaw experiment is performed, in which 1-6 repeated freeze-thaw cycles can be implemented.
  • the antibody formulations according to the invention as described in the various embodiments herein are stable after storage, for example, at least 12 or 24 months at about 5°C ⁇ 3°C, at least 1, 2, 3 or 6 months at about 25°C ⁇ 2°C, or at least 1 month at 40°C ⁇ 2°C, preferably having one or more of the following characteristics:
  • the main peak variation is less than 5% and/or the preparation has a purity greater than 95% as measured by SEC-HPLC;
  • the sum of the changes in the components of the protein in the formulation does not exceed 40% and/or the change in the main component does not exceed 20% relative to the initial value on storage day 0, for example, after storage at 40°C ⁇ 2°C for 1 month, the sum of the changes does not exceed about 40% or the change in the main component does not exceed about 20%, or after storage at 25°C for 2 months, the sum of the changes does not exceed about 20% or the change in the main component does not exceed about 15%;
  • the biological activity should be 70%-130% of the activity of the reference product.
  • the formulation is stable under shaking and/or repeated freeze-thaw cycles.
  • the antibody preparation of the present invention comprising an anti-CCR8 antibody of the present invention can be used to treat, improve or prevent cancer-related diseases.
  • Cancer can be a blood cancer or a solid tumor.
  • Blood cancers include, but are not limited to, leukemia, lymphoma (e.g., non-Hodgkin's lymphoma) and myeloma.
  • Solid tumors include, but are not limited to, colorectal cancer (CRC), spleen cancer, breast cancer, non-small cell lung cancer (NSCLC), pancreatic cancer, melanoma, bile duct cancer, gallbladder cancer, thyroid cancer, sarcoma, kidney cancer, bladder cancer, uterine body cancer, ovarian cancer, lung cancer, prostate cancer, head and neck cancer, thymus cancer, liver cancer, testicular cancer, urothelial cancer, esophageal tumors and gastric tumors.
  • CRC colorectal cancer
  • NSCLC non-small cell lung cancer
  • pancreatic cancer melanoma
  • bile duct cancer gallbladder cancer
  • thyroid cancer sarcoma
  • kidney cancer bladder cancer
  • uterine body cancer ovarian cancer
  • lung cancer prostate cancer
  • head and neck cancer thymus cancer
  • testicular cancer urothelial cancer
  • esophageal tumors and gastric tumors gastric tumors
  • the antibody preparations of the invention are used to treat, ameliorate or prevent colorectal cancer, breast cancer, ovarian cancer, lung cancer (eg, non-small cell lung cancer), melanoma, liver cancer, kidney cancer, bladder cancer or pancreatic ductal adenocarcinoma.
  • lung cancer eg, non-small cell lung cancer
  • melanoma liver cancer
  • kidney cancer kidney cancer
  • bladder cancer pancreatic ductal adenocarcinoma.
  • a liquid antibody formulation or solid antibody formulation of the invention is administered to an individual in need thereof in combination with one or more other agents, such as a therapeutic antibody, such as an anti-PD-1 antibody.
  • the invention also provides a formulation of the invention as described herein in various embodiments for use as a medicament, for example, for delivering an anti-CCR8 antibody to a mammal, or for treating, preventing or ameliorating one or more of the above diseases and conditions.
  • the mammal is a human.
  • the antibody formulations of the invention can be administered to a subject or patient. Administration is typically by infusion or by syringe.
  • a delivery device e.g., a syringe
  • an antibody formulation of the invention e.g., a prefilled syringe
  • the present invention also provides a pre-filled syringe comprising the antibody formulation of the present invention, preferably for intravenous injection, subcutaneous injection or intramuscular injection, or an infusion, such as for intravenous infusion.
  • the liquid antibody formulation or solid antibody formulation of the invention is used to prepare a delivery device or pre-filled syringe or medicament for targeting a tumor infiltrating lymphocyte (TIL) subpopulation, such as regulatory T cells (Tregs), in a subject.
  • TIL tumor infiltrating lymphocyte
  • Regs regulatory T cells
  • the anti-CCR8 antibody or antigen-binding fragment in the antibody preparation of the present invention inhibits Tregs migration and/or eliminates Tregs by antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • the liquid antibody formulation or solid antibody formulation of the invention is used to prepare a delivery device or prefilled syringe or medicament for treating cancer in a subject, for example, colorectal cancer, breast cancer, ovarian cancer, lung cancer (e.g., non-small cell lung cancer), melanoma, liver cancer, kidney cancer, bladder cancer, or pancreatic ductal adenocarcinoma.
  • a delivery device or prefilled syringe or medicament for treating cancer in a subject, for example, colorectal cancer, breast cancer, ovarian cancer, lung cancer (e.g., non-small cell lung cancer), melanoma, liver cancer, kidney cancer, bladder cancer, or pancreatic ductal adenocarcinoma.
  • the pH meter is calibrated with three standard solutions (pH 4.01, 7.00, 9.21 respectively).
  • the calibration slope is required to be between 95.0% and 105.0%, and the calibration zero drift value is required to be between -60.0mv and 60.0mv. If the calibration value is not within the above range, it needs to be recalibrated. Aliquot 200 ⁇ L of sample to measure the pH value.
  • SEC-HPLC detection was performed using Thermo U3000 high performance liquid chromatography, TSKgel G3000SWXL gel column (5 ⁇ m, 7.8mm ⁇ 300mm), column temperature 25°C, injection plate temperature 2 ⁇ 8°C, mobile phase composition 50mM phosphate buffer, 300mM sodium chloride, pH 6.8 ⁇ 0.1, isocratic elution, flow rate 1.0ml/min, elution time 20min. Detection wavelength was 280nm, injection volume 100 ⁇ g.
  • the icIEF test uses ProteinSimple's iCE3 (or equivalent) capillary isoelectric focusing electrophoresis analyzer and ProteinSimple's fluorine-coated capillary assembly.
  • the amphoteric electrolytes are pharmalyte 3-10 and pharmalyte 8-10.5, the low pI marker is 7.05, the high pI marker is 9.50, and the sample plate temperature is 10°C. Focusing stage 1: 1500V focusing for 1 minute, focusing stage 2: 3000V focusing for 10 minutes.
  • the sample injection time is 60 seconds.
  • Reduction capillary gel electrophoresis uses UV or diode array detector, non-coated fused silica capillary (inner diameter 50um), dilute the test sample to 1 mg/ml with SDS sample buffer, take 95ul of the test solution (1mg/ml), add 5ul of 2-mercaptoethanol, and vortex to mix. Take 95ul of the blank control, add 5ul of 2-mercaptoethanol, vortex to mix, and detect the reduction blank control, and calculate the heavy chain and light chain percentages by area normalization method.
  • Reduction capillary gel electrophoresis uses UV or diode array detectors, and the capillary uses non-coated fused silica capillary (inner diameter 50um).
  • the test sample is diluted to 1 mg/ml with SDS sample buffer.
  • Prepare the solution by taking 95ul of the test sample solution (1mg/ml), adding 5ul of 0.8mol/L iodoacetamide aqueous solution, and vortexing. Take 95ul of the blank control, add 5ul of 0.8mol/L iodoacetamide aqueous solution, vortexing, and detect as a non-reduction blank control.
  • the main peak purity is calculated by area normalization method.
  • the MFI test uses the ProteinSimple 5200 instrument, Bot1 automatic sample addition, 1ml disposable conductivity pipette tip standardized sampling, particle detection range 1-70um, and injection volume 1.5ml.
  • Biological activity was determined using an antibody-mediated cytotoxicity (ADCC) cell assay.
  • Target cells and effector cells were thawed and cultured.
  • Target cells were added to a 96-well plate and incubated with the test article after gradient dilution. Effector cells were then added to a 96-well plate and incubated together. Luciferase was then added and fluorescence readings were taken after 5 minutes of incubation.
  • a four-parameter fitting model was applied to generate a fitting curve and obtain the EC50 (50% maximum effect concentration) value.
  • the relative activity of the test article was calculated as the EC50 of the reference article divided by the EC50 of the test article and multiplied by 100%.
  • Stable after storage for example, at about 5°C ⁇ 3°C for at least 12 or 24 months, at about 25°C ⁇ 2°C for at least 1, 2, 3 or 6 months, or at 40°C ⁇ 2°C for 1 month:
  • the main peak variation is less than 5% and/or the preparation has a purity greater than 95% as measured by SEC-HPLC;
  • the sum of the changes in the components of the protein in the formulation does not exceed 40% and/or the change in the main component does not exceed 20% relative to the initial value on storage day 0, for example, after storage at 40°C ⁇ 2°C for 1 month, the sum of the changes does not exceed about 40% or the change in the main component does not exceed about 20%, or after storage at 25°C for 2 months, the sum of the changes does not exceed about 20% or the change in the main component does not exceed about 15%;
  • the biological activity should be 70%-130% of the activity of the reference product.
  • variable region sequence features are summarized in Tables 1 to 3 above, and the constant region comprises wild-type human IgG1 Fc (SEQ ID NO: 119), or human IgG1 Fc comprising S239D/I332E mutations (SEQ ID NO: 121, represented by a G1x suffix), or human IgG1 Fc comprising L235V/F243L/R292P/Y300L/P396L mutations (SEQ ID NO: 120, represented by a G1m suffix).
  • This example uses a prescription prepared with antibody Hu149-11G1m as an example to evaluate the effect of buffer and its pH value on the stability of anti-CCR8 antibody.
  • Solution pH and buffer are one of the most important parameters affecting the physicochemical stability of biopharmaceuticals. pH can adjust the charge distribution on the protein surface, thereby affecting the mutual attraction and repulsion forces within and between biopharmaceutical molecules, and can also adjust the interaction between biopharmaceuticals and other small molecules such as excipients. Different buffers will also affect the stability of protein molecules. Taking a protein concentration of 10 mg/ml as an example, different buffers at different pH values are prepared according to Table 6.
  • the purified samples were replaced with the prepared buffers of each prescription, and then diluted with the corresponding buffers to the target concentration of 10 mg/ml to obtain each prescription, and then sterilized and filtered and dispensed into 2 ml vials.
  • the samples were placed at 5 ⁇ 3°C, 25 ⁇ 2°C/60% ⁇ 5%RH, and 40 ⁇ 2°C/75% ⁇ 5%RH for stability tests, and the appearance, pH, protein concentration, SEC-HPLC and icIEF were tested at the sampling points shown in the table below.
  • the sampling points and test items of the stability test samples are shown in Table 7.
  • Figures 1 to 3 show the SEC-HPLC main peak purity graphs of the formulations F1-F12 prepared by taking the antibody Hu149-11G1m in the above-mentioned antibodies as an example in the stability experiment under different temperature conditions in this embodiment. Among them, compared with T0, each formulation remained relatively stable after being placed under 5 ⁇ 3°C for 4 weeks, without obvious changes, and the sample purity in the phosphate buffer system was slightly lower than the purity in the other three buffer systems (see Figure 1).
  • Figures 4 to 6 show the icIEF main peak purity graphs of the formulations F1-F12 prepared in this embodiment using the antibody Hu149-11G1m in the above-mentioned antibodies as an example in the stability experiment under different temperature conditions. Compared with T0, the main peak purity of each formulation remained relatively stable after being placed at 5 ⁇ 3°C for 4 weeks, without showing a significant decrease (see Figure 4).
  • Common protein protectants or stabilizers include sugars, polyols, and amino acids, whose mechanism of protecting proteins is "preferential exclusion". These excipients are preferentially excluded from the surface of proteins, increasing the chemical potential energy of proteins. As the surface area increases after protein denaturation and unfolding, the interaction with excipients increases, causing the chemical potential energy to increase, thereby increasing the activation energy required for conformational denaturation of proteins, shifting the balance between the native conformation and the denatured conformation of proteins toward the native conformation, thereby increasing the conformational stability of proteins.
  • Nonionic surfactants can inhibit protein aggregation.
  • the mechanism may be that surfactants tend to be arranged at the air-liquid interface, thereby causing proteins to leave the interface to inhibit protein aggregation and denaturation.
  • Commonly used surfactants include polysorbate 80 and polysorbate 20. The surfactants in this embodiment are preferably used for subsequent experiments.
  • the corresponding buffer was used to dilute the target concentration to obtain each prescription sample, sterilized and filtered, and dispensed into 2 ml vials. Stability tests were performed under 5 ⁇ 3°C, 25 ⁇ 2°C/60% ⁇ 5%RH, 40 ⁇ 2°C/75% ⁇ 5%RH, and freeze-thaw conditions to test appearance, pH, protein concentration, SEC-HPLC, icIEF, and MFI. The sampling points and test items for stability test samples are shown in Table 9.
  • the pH test results and protein concentrations at different sampling points of each formulation did not change significantly in the 8-week 5 ⁇ 3°C, 8-week 25 ⁇ 2°C, and 4-week 40 ⁇ 2°C stability tests, indicating that the pH and protein concentrations were relatively stable.
  • the pH test results and protein concentrations at different sampling points of each formulation did not change significantly compared with T0, indicating that the pH and protein concentrations were relatively stable.
  • FIGS 7 to 10 show the SEC-HPLC main peak purity graphs of the formulations F1-F4 prepared in this embodiment using the antibody Hu149-11G1m in the above-mentioned antibodies as an example in the stability test under different temperature conditions and the repeated freeze-thaw stability test.
  • the SEC-HPLC main peak purity of each formulation containing different excipients did not change significantly, indicating that the SEC-HPLC purity of each formulation selected by the excipients was relatively stable after being stored at 5 ⁇ 3°C and 25 ⁇ 2°C for 8 weeks (see Figures 7 and 8).
  • the SEC-HPLC main peak purity of each formulation showed a downward trend, and there was no significant difference between the formulations (see Figure 9).
  • Figures 11 to 14 show the icIEF main peak purity graphs of the formulations F1-F4 prepared in this embodiment using the antibody Hu149-11G1m in the above-mentioned antibodies as an example in the stability test under different temperature conditions and the repeated freeze-thaw stability test. After 8 weeks of observation under the stability test conditions of 5 ⁇ 3°C and 25 ⁇ 2°C, compared with T0, the main peak purities of the icIEF of each formulation containing different excipients did not show significant changes, and all remained in a relatively stable state, with no significant differences between the formulations (see Figures 11 and 12).
  • the insoluble particles of each formulation did not increase significantly. Compared with T0, under the high temperature condition of 40 ⁇ 2°C, the insoluble particles of each formulation increased to a certain extent. In the repeated freeze-thaw stability test, the insoluble particles of each formulation sample did not increase significantly, but were within the range specified in the pharmacopoeia, and there was no significant difference between the formulations.
  • polysorbate 80 As a surfactant, 20 mM histidine as a buffer, pH 6.0, taking the antibody Hu149-11G1m as an example with a protein concentration of 50 mg/ml, and 7% (w/v) sucrose as a stabilizer, the prescription design is shown in Table 10 below to investigate the effects of different surfactant concentrations on protein stability.
  • the corresponding buffer was used to dilute them to the target concentration to obtain the samples of each prescription. After sterilization and filtration, the samples were dispensed into 2 ml vials and placed under 5 ⁇ 3°C, 25 ⁇ 2°C/60% ⁇ 5%RH, 40 ⁇ 2°C/75% ⁇ 5%RH, and freeze-thaw conditions for stability tests, and the appearance, pH, protein concentration, SEC-HPLC, icIEF and MFI were tested. The sampling points and test items of the stability test samples are shown in Table 11.
  • 15-17 show the SEC-HPLC main peak purity graphs of the formulations F1-F3 prepared using the antibody Hu149-11G1m in the above antibodies as an example in the stability experiment under different temperature conditions in this example.
  • the insoluble particles of each prescription did not increase significantly, and all were within the prescribed range of the pharmacopoeia.
  • the insoluble particles of each prescription were within the prescribed range of the pharmacopoeia, among which the insoluble particles of F2 prescription (polysorbate concentration 0.04%) were slightly less than the other two prescriptions.
  • the insoluble particles of each prescription did not increase significantly, among which the insoluble particles of F2 prescription (polysorbate concentration 0.04% (w/v)) were less than the other two prescriptions, and slightly increased under repeated freeze-thaw conditions, all within the prescribed range of the pharmacopoeia.
  • polysorbate 80 As a surfactant, 20 mM histidine as a buffer, pH 6.0, taking the antibody Hu149-11G1m as an example with a protein concentration of 50 mg/ml, 7% (w/v) sucrose as a stabilizer, and polysorbate 80 as a surfactant (e.g. 0.04% (w/v)), according to ICH (The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use) Q5C (Stability Testing of Biotechnological/Biological Products) and the Guidelines for Stability Testing of Biological Products (Chinese Pharmacopoeia, current edition, Part IV, 9402), two batches of long-term, accelerated, high temperature and high humidity forced stability investigations were carried out.
  • ICH The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use
  • Q5C Stability Testing of Biotechnological/Biological Products
  • the molecular size heterogeneity of two batches of preparations was evaluated by SEC-HPLC under long-term, accelerated and high temperature and high humidity forced conditions. After storage for 12 months at 5°C ⁇ 3°C, no obvious change trend was observed, meeting the acceptance criteria; after storage for 6 months at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH, no obvious change trend was observed; after storage for 1 month at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH, the average decrease in monomer content did not exceed 5%, and no obvious change in polymer content was observed. The batch-to-batch consistency was good.
  • the molecular size heterogeneity of two batches of finished products was evaluated by non-reduced and reduced CE-SDS methods under long-term, accelerated and forced conditions of high temperature and high humidity. After 12 months of storage at 5°C ⁇ 3°C, no obvious trend of change was observed, and all batches met the acceptance criteria; after 6 months of storage at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH, no obvious trend of change was observed; after 1 month of storage at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH, the purity of intact IgG detected by non-reduced CE-SDS method was greater than 90%; the purity of (light chain + heavy chain) detected by reduced CE-SDS method decreased slightly. The consistency between batches was good.
  • the charge heterogeneity of two batches of finished products was evaluated by the icIEF method under long-term, accelerated and forced conditions of high temperature and high humidity. After storage for 12 months at 5°C ⁇ 3°C, no obvious trend of change was observed, which met the acceptance criteria; after storage for 6 months at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH, the main peak area percentage did not exceed 10%, which met the acceptance criteria. After storage for 1 month at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH, the main peak area percentage did not exceed 20%, which met the acceptance criteria. The batch-to-batch consistency was good.
  • the biological activity of the two finished product batches was evaluated by cell-based antibody-mediated cytotoxicity assay under long-term, accelerated and forced conditions of high temperature and high humidity. No significant changes were observed after storage at -5°C ⁇ 3°C for 12 months. All batches met the acceptance criteria; no obvious trend was observed after 6 months of storage at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH; no obvious trend was observed after 1 month of storage at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH. The batch-to-batch consistency was good.
  • the protein concentration of two batches of finished products was evaluated by UV absorbance under long-term, accelerated and forced conditions of high temperature and high humidity. No obvious trend of change was observed after 12 months of storage at 5°C ⁇ 3°C; no obvious trend of change was observed after 6 months of storage at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH; no obvious trend of change was observed after 1 month of storage at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH. The consistency between batches was good.
  • Appearance includes appearance description, solution color, chromaticity, clarity and visible foreign matter.
  • the solution appearance was tested visually under long-term, accelerated and high temperature and high humidity forced conditions.
  • the solution color, chromaticity, clarity and visible foreign matter of 2 batches of finished products were evaluated. After storage for 12 months at 5°C ⁇ 3°C, no obvious change trend was observed; after storage for 6 months at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH, no obvious change trend was observed; after storage for 1 month at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH, no obvious change trend was observed. The consistency between batches is good.
  • the pH of two batches of finished products was evaluated under long-term, accelerated, and forced conditions of high temperature and high humidity. No obvious trend was observed after 12 months of storage at 5°C ⁇ 3°C; no obvious trend was observed after 6 months of storage at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH; no obvious trend was observed after 1 month of storage at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH. The consistency between batches was good.
  • the insoluble particles of two batches of finished products were evaluated under long-term, accelerated and forced conditions of high temperature and high humidity. After 12 months of storage at 5°C ⁇ 3°C, no obvious trend of change was observed, and all batches met the acceptance criteria; after 6 months of storage at 25°C ⁇ 2°C, 60%RH ⁇ 5%RH, no obvious trend of change was observed; after 1 month of storage at 40°C ⁇ 2°C, 75%RH ⁇ 5%RH, no obvious trend of change was observed. The consistency between batches was good.

Landscapes

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

Abstract

L'invention concerne une formulation d'anticorps liquide, comprenant : (i) un anticorps anti-CCR8 ou un fragment de liaison à l'antigène; (ii) un tampon; (iii) un stabilisant; et (iv) un tensioactif. L'invention concerne en outre un procédé de traitement du cancer chez un sujet, comprenant l'étape d'administration à un sujet de la formulation d'anticorps liquide ou d'administration au moyen d'un dispositif d'administration (par exemple, une seringue pré-remplie) ou d'administration d'un médicament comprenant la formulation d'anticorps liquide.
PCT/CN2024/094955 2023-05-23 2024-05-23 Formulation comprenant un anticorps anti-ccr8 et son utilisation Pending WO2024240224A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2023095853 2023-05-23
CNPCT/CN2023/095853 2023-05-23

Publications (1)

Publication Number Publication Date
WO2024240224A1 true WO2024240224A1 (fr) 2024-11-28

Family

ID=93588951

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2024/094955 Pending WO2024240224A1 (fr) 2023-05-23 2024-05-23 Formulation comprenant un anticorps anti-ccr8 et son utilisation

Country Status (2)

Country Link
TW (1) TW202511293A (fr)
WO (1) WO2024240224A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2025186043A1 (fr) 2024-03-06 2025-09-12 Bayer Aktiengesellschaft Formulation pharmaceutique pour anticorps anti-ccr8

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113260381A (zh) * 2018-12-27 2021-08-13 盐野义制药株式会社 新型抗ccr8抗体
CN114929278A (zh) * 2020-01-06 2022-08-19 瓦西尼斯公司 抗ccr8抗体及其用途
US11427640B1 (en) * 2020-06-26 2022-08-30 Bayer Aktiengesellschaft CCR8 antibodies for therapeutic applications
WO2022241034A1 (fr) * 2021-05-12 2022-11-17 BioLegend, Inc. Anticorps anti-ccr8, fragments de liaison à l'antigène de ceux-ci, et agents et compositions et leurs procédés de fabrication et d'utilisation
US20220372127A1 (en) * 2019-04-02 2022-11-24 Joint Stock Company "Biocad" Aqueous pharmaceutical composition of an anti-il17a antibody and use thereof
US20230049152A1 (en) * 2021-07-14 2023-02-16 Genentech, Inc. Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
WO2023098888A1 (fr) * 2021-12-02 2023-06-08 Zai Lab (Shanghai) Co., Ltd Unité de liaison à l'antigène ccr8 et ses utilisations

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113260381A (zh) * 2018-12-27 2021-08-13 盐野义制药株式会社 新型抗ccr8抗体
US20220064312A1 (en) * 2018-12-27 2022-03-03 Shionogi & Co., Ltd. Novel Anti-CCR8 Antibody
US20220372127A1 (en) * 2019-04-02 2022-11-24 Joint Stock Company "Biocad" Aqueous pharmaceutical composition of an anti-il17a antibody and use thereof
CN114929278A (zh) * 2020-01-06 2022-08-19 瓦西尼斯公司 抗ccr8抗体及其用途
US11427640B1 (en) * 2020-06-26 2022-08-30 Bayer Aktiengesellschaft CCR8 antibodies for therapeutic applications
CN116096750A (zh) * 2020-06-26 2023-05-09 拜耳公司 抗ccr8抗体疗法:生物标志物和组合疗法
WO2022241034A1 (fr) * 2021-05-12 2022-11-17 BioLegend, Inc. Anticorps anti-ccr8, fragments de liaison à l'antigène de ceux-ci, et agents et compositions et leurs procédés de fabrication et d'utilisation
US20230049152A1 (en) * 2021-07-14 2023-02-16 Genentech, Inc. Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
WO2023098888A1 (fr) * 2021-12-02 2023-06-08 Zai Lab (Shanghai) Co., Ltd Unité de liaison à l'antigène ccr8 et ses utilisations

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2025186043A1 (fr) 2024-03-06 2025-09-12 Bayer Aktiengesellschaft Formulation pharmaceutique pour anticorps anti-ccr8

Also Published As

Publication number Publication date
TW202511293A (zh) 2025-03-16

Similar Documents

Publication Publication Date Title
KR102624564B1 (ko) 항-ctla4 항체 단독의, 및 프로그램화된 사멸 수용체 1 (pd-1) 항체와 조합된 항-ctla4 항체의 안정한 제제 및 그의 사용 방법
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
CN110913906A (zh) 抗lag3抗体的制剂和抗lag3抗体与抗pd-1抗体的共制剂
JP2020518600A (ja) 単独のおよびプログラム死受容体1(pd−1)抗体と組み合わされた抗tigit抗体の安定な製剤、ならびにその使用方法
KR20210089215A (ko) 항-lag3 항체 및 항-pd-1 항체의 공동-제제
TWI764097B (zh) 包含抗cd47抗體的製劑及其製備方法和用途
CA2875386A1 (fr) Formulation d'anticorps
CN114555118A (zh) 包含针对cd3和cd20的双特异性抗体的药物组合物及其用途
EP4151233A1 (fr) Préparation comprenant un anticorps anti-il-23p19, son procédé de préparation et son utilisation
TW202130367A (zh) 結合pd-1和pd-l1的雙特異性抗體的製劑及其用途
CN114146174B (zh) 抗pd-l1/ox40双特异性抗体制剂及其制备方法和用途
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
WO2024240224A1 (fr) Formulation comprenant un anticorps anti-ccr8 et son utilisation
TW202345902A (zh) 含有pd-1抗體的穩定高濃度精胺酸配製物及其使用方法
WO2020063668A1 (fr) Formulation contenant un anticorps anti-ox40, son procédé de préparation et son utilisation
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
CN115867579A (zh) 乐维利单抗的水性药用组合物及其用途
CN112675300A (zh) 包含抗gitr抗体的制剂及其制备方法和用途
US20250084168A1 (en) Stable high concentration sodium chloride formulations containing pd-1 antibody and methods of use thereof
RU2822192C2 (ru) Совместные составы анти-lag3 антитела и анти-pd-1 антитела
HK40059591A (en) Preparations containing anti-cd47 antibody, and preparation method and use therefor
HK40060532A (en) Formulations containing anti-cd47/pd-l1 bispecific antibody and preparation method therefor and use thereof
TW202541841A (zh) 用於治療c1s介導病症的包含抗體的醫藥組成物及其使用方法

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: 24810461

Country of ref document: EP

Kind code of ref document: A1