Antibody binding specifically to CD40 and use thereof

Information

  • Patent Grant
  • 11905331
  • Patent Number
    11,905,331
  • Date Filed
    Friday, November 10, 2017
    6 years ago
  • Date Issued
    Tuesday, February 20, 2024
    2 months ago
Abstract
The present invention relates to an anti-CD40 antibody binding specifically to CD40 and use thereof and, particularly, provides an anti-CD40 antibody or an antigen-binding fragment thereof, and a pharmaceutical composition comprising the same antibody or fragment as an effective ingredient for prevention and/or treatment of cancer, cancer metastasis, infection, and/or immune deficiency diseases.
Description
TECHNICAL FIELD

The present disclosure relates to an anti-CD40 antibody binding specifically to CD40 and a use thereof and, more particularly, to an anti-CD40 antibody or an antigen-binding fragment thereof and a composition comprising the same as an effective ingredient for prevention and/or treatment of cancer, cancer metastasis, infection, and/or immune deficiency diseases.


BACKGROUND ART

CD40 is a costimulatory protein molecule belonging to the tumor necrosis factor (TNF)-receptor superfamily found on antigen presenting cells (APC) including dendritic cells, B cells, and macrophages. The binding of the ligand CD154 (CD40L) on TH cells to CD40 activates antigen presenting cells. A variety of immune reactions including cytokine production, the upstream regulation of costimulatory molecules (e.g., CD86), and enhanced antigen presentation and B cell proliferation is accompanied by CD4-mediated APC activation. CD40 can also be expressed by endothelial cells, smooth muscle cells, fibroblasts, and epithelial cells.


Agonistic anti-CD40 antibodies constitute one of the most effective classes of these reagents. CD40 is a cell-surface member of the tumor necrosis factor superfamily expressed on antigen presenting cells (APC) such as dendrocytes, B cells and macrophages. Preclinical studies with anti-CD40 agonists suggest that triggering CD40 with crosslinking antibodies on antigen presenting cells (APC) can substitute for CD4 T cell help, normally provided via a CD40 ligand, and facilitate the activation as well as expansion of CD8 effector T cells. In addition, CD40-activated macrophages may also exert direct tumoricidal functions


PRIOR ART DOCUMENT



  • WO 2016/069919 A1 (May 6, 2016)



DETAILED DESCRIPTION OF THE INVENTION
Technical Problem

An embodiment provides an anti-CD40 antibody or an antigen-binding fragment thereof, which specifically recognizes CD40. The anti-CD40 antibody or the antigen-binding fragment thereof is characterized by recognizing a site different from the binding site between CD40 and the ligand thereof. CD40L. The anti-CD40 antibody or the antigen-binding fragment thereof is an agonist for CD40. In addition, the anti-CD40 antibody or the antigen-binding fragment thereof may not interfere with CD40/CD40L interaction by recognizing a site different from the binding site between CD40 and the ligand thereof. CD40L.


The anti-CD40 antibody or the antigen-binding fragment thereof may comprise:


heavy-chain complementarity determining regions (CDRs) or a heavy-chain variable region containing the heavy-chain complementarity determining regions (CDRs), the heavy-chain complementarity determining regions comprising: a polypeptide (CDR-H1) inclusive of the amino acid sequence of SEQ ID NO: 17 (GYGFTNYL) or SEQ ID NO: 33 (NYLIE); a polypeptide (CDR-H1) inclusive of the amino acid sequence of SEQ ID NO: 17 (GYGFTNYL) or SEQ ID NO: 33 (NYLIE); a polypeptide (CHR-H2) inclusive of the amino acid sequence of SEQ ID NO: 18 (INPGYGGV) or SEQ ID NO: 34 (VINPGYGGVNYNEKFKG); and a polypeptide (CHR-H3) inclusive of the amino acid sequence of SEQ ID NO: 19 (GGSGFAF); and


light-chain complementarity-determining regions (CDRs) or a light-chain variable region containing the light-chain complementarity-determining regions, light-chain complementarity-determining regions comprising: a polypeptide (CDR-L1) inclusive of the amino acid sequence of SEQ ID NO: 20 (QDISNH) or SEQ ID NO: 35 (RASQDISNHLN); a polypeptide (CDR-L2) inclusive of the amino acid sequence of SEQ ID NO: 32 (STS(Xaa)n; wherein n means a number of any amino acid Xaa and may be an integer of 0 (Xaa is null) or 1 to 4; for example, a polypeptide having the amino acid sequence of SEQ ID NO: 21 (STS) or SEQ ID NO: 36 (STSRLHS)); and a polypeptide (CDR-L3) inclusive of the amino acid sequence of SEQ ID NO: 22 (QQGNTLP).


Another embodiment provides a hybridoma producing the anti-CD40 antibody.


Another embodiment provides a nucleic acid molecule coding for a heavy-chain complementarity-determining region, a heavy-chain variable region, or a heavy chain of an anti-CD40 antibody.


Another embodiment provides a nucleic acid coding for a light-chain complementarity-determining region, a light-chain variable region, or a light chain of an anti-CD40 antibody.


Another embodiment provides a recombinant vector carrying a nucleic acid molecule coding for a heavy-chain complementarity-determining region, a heavy-chain variable region, or a heavy chain of the anti-CD40 antibody and a nucleic acid molecule coding for a light-chain complementarity-determining region, a light-chain variable region, or a light chain of the anti-CD40 antibody together, or recombinant vectors carrying the nucleic acid molecules, respectively.


Another embodiment provides a recombinant cell anchoring the recombinant vector or the recombinant vectors thereat.


Another embodiment provides a pharmaceutical composition comprising an anti-CD40 antibody or an antigen-binding fragment thereof as an effective ingredient for preventing and/or treating a disease.


Another embodiment provides a method of preventing and/or treating a disease, the method comprising administering an anti-CD40 antibody or an antigen-binding fragment thereof to a subject in need of preventing and/or treating the disease.


Another embodiment provides a use of an anti-CD40 antibody or an antigen-binding fragment thereof in preventing and/or treating a disease or in preparing a composition for prevention and/or treatment of a disease.


The disease may be selected from cancer, cancer metastasis, infection, and immune deficiency disease.


Technical Solution

Provided herein are an anti-CD40 antibody specifically recognizing CD40 and a use thereof. The anti-CD40 antibody is characterized by recognizing a site different from the binding site between CD40 and its ligand CD40L and may exhibit agonistic activity for CD40. In addition, the anti-CD40 antibody or the antigen-binding fragment thereof may not interfere with CD40/CD40L interaction by recognizing a site different from the binding site between CD40 and its ligand CD40L. Herein, the expression “does not interfere with the CD40/CD40L interaction” is intended to mean not inhibiting the CD40/CD40L interaction at a significant level and to encompass some inhibition that does not incur a significant inhibitory effect.


CD40, which is a member of the tumor necrosis factor (TNF)-receptor superfamily, is a 45-50 kDa cell-surface antigen found on surfaces of normal and tumor human B cells, dendrocytes, monocytes, macrophages, CD8+ T cells, endothelial cells, mononuclear and epithelial cells, and many tumors (for example, solid tumors) including lung, breast, ovarian, bladder, and rectal cancers. CD40 is expressed in activated T cells, activated platelets, inflammatory vascular smooth muscle cells, synovium in rheumatoid arthritis, dermal fibroblasts, and non-lymphoma cells.


The CD40 acting as an antigen to the antibody provided herein may be derived from mammals, for example, human-derived CD40 (e.g., NCBI accession numbers NP_001241.1, NP_001289682.1, NP_690593.1, NP_001309351.1, and NP_001309350.1). There is an extracellular region among the five isoforms of human-derived CD40 and the anti-CD40 antibody provided in the present disclosure recognizes and/or binds the extracellular region.


Binding between CD40 and its homologous ligand CD40L (or CD154) incites B-cell proliferation and differentiation to plasma cells, antibody production, antibody isotype switching, and memory B-cell development. CD40L, which is a ligand for CD40, also known as CD154, is a 32-33 kDa transmembrane protein composed of two subunits 18 kDa and 31 kDa, respectively and is present in a soluble form, which is biologically active.


Interaction between CD40 and CD40L induces humoral and cell-mediated immune responses.


CD40 upregulates the above-mentioned ligand-receptor interaction to activate other antigen-presenting cells (APC) including monocytes, B cells and dendrocytes (DC). CD40 signaling on monocytes and DC enhances survival as well as secretion of cytokines (IL-1, IL-6, IL-8, IL-10, IL-12, TNF-α, and MIP-la). CD40 ligation to these APCs also induces upregulation of costimulatory molecules such as ICAM-1, LFA-3, CD80, and CD86. CD40 ligand activation is one of the signals that play a critical role in completely maturing DC to APC effective for driving T cell activation.


As used herein, the term “antibody” generically denotes a substance that is made with the stimulation of an antigen in the immune system or a protein that binds specifically to a certain antigen and has no particular limitations to kinds thereof. The antibodies may be those that are produced in a non-natural manner, for example, in a recombinant or synthetic manner. The antibodies may be animal antibodies (e.g., mouse antibodies, etc.), chimeric antibodies, humanized antibodies, or human antibodies. The antibodies may be monoclonal or polyclonal.


Unless stated otherwise, the antibody in the present disclosure may be understood to encompass an antigen-binding fragment thereof that possesses antigen binding capacity. The term “complementarity-determining regions (CDRs)”, as used herein, means part of the variable region in an antibody, which confers binding specificity for an antigen. In this regard, the foregoing antigen-binding fragment of an antibody may be an antibody fragment comprising one or more of the complementarity-determining regions.


An embodiment of the present disclosure provides an anti-CD40 antibody or an antigen-binding fragment thereof that specifically recognize or binds to CD40.


The site on CD40 that is recognized or bound by the anti-CD40 antibody or the antigen-binding fragment thereof may be different from a site where CD40L interacts with or binds to CD40. The anti-CD40 antibody or the antigen-binding fragment thereof may have an agonist function for CD40. The anti-CD40 antibody or the antigen-binding fragment thereof may not interfere with CD40/CD40L interaction. In addition, the anti-CD40 antibody or the antigen-binding fragment thereof may have TAM (tumor associated macrophage) polarization and cancer cell apoptosis activity.


In a concrete embodiment, the anti-CD40 antibody or the antigen-binding fragment thereof may comprise, as the complementarity-determining region (CDR) in the heavy chain, a polypeptide (CDR-H1) comprising the amino acid sequence of SEQ ID NO: 17 (GYGFTNYL) or SEQ ID NO: 33 (NYLIE), a polypeptide (CDR-H2) comprising the amino acid sequence of SEQ ID NO: 18 (INPGYGGV) or SEQ ID NO: 34 (VINPGYGGVNYNEKFKG), and a polypeptide (CDR-H3) comprising the amino acid sequence of SEQ ID NO: 19 (GGSGFAF).


In addition, the anti-CD40 antibody or the antigen-binding fragment thereof may comprise, as the complementarity-determining region in the light chain, a polypeptide (CDR-L1) comprising the amino acid sequence of SEQ ID NO: 20 (QDISNH) or SEQ ID NO: 35 (RASQDISNHLN), polypeptide (CDR-L2) comprising the amino acid sequence of SEQ ID NO: 32 (STS(Xaa)n; wherein n means a number of any amino acid Xaa and may be an integer of 0 (Xaa is null) or 1 to 4; for example, SEQ ID NO: 21 (STS) or SEQ ID NO: 36 (STSRLHS)), and a polypeptide comprising the amino acid sequence (CDR-L3) of SEQ ID NO: 22 (QQGNTLP).


Therefore, the anti-CD40 antibody or the antigen-binding fragment thereof may comprise: a heavy chain complementarity-determining region or a heavy chain variable region inclusive of the heavy chain complementarity-determining region, the heavy chain complementarity-determining region comprising a polypeptide (CDR-H1) comprising the amino acid sequence of SEQ ID NO: 17 (GYGFTNYL) or SEQ ID NO: 33 (NYLIE), a polypeptide (CDR-H2) comprising the amino acid sequence of SEQ ID NO: 18 (INPGYGGV) or SEQ ID NO: 34 (VINPGYGGVNYNEKFKG), and a polypeptide (CDR-H3) comprising the amino acid sequence of SEQ ID NO: 19 (GGSGFAF); and a light chain complementarity-determining region or a light chain variable region inclusive of the light chain complementarity-determining region, the light chain complementarity-determining region comprising a polypeptide (CDR-L1) comprising the amino acid sequence of SEQ ID NO: 20 (QDISNH) or SEQ ID NO: 35 (RASQDISNHLN), a polypeptide (CDR-L2) comprising the amino acid sequence of SEQ ID NO: 32 (STS(Xaa)n; wherein n means a number of any amino acid Xaa and may be an integer of 0 (Xaa is null) or 1 to 4; for example, SEQ ID NO: 21 (STS) or SEQ ID NO: 36 (STSRLHS)), and a polypeptide (CDR-L3) comprising the amino acid sequence of SEQ ID NO: 22 (QQGNTLP).


The heavy-chain variable region may comprise the amino acid sequence of SEQ ID NO: 1, 23, 24, 25, 26, 27, or 28 (refer to Tables 1 and 6).


The light-chain variable region may comprise the amino acid sequence of SEQ ID NO: 3, 29, or 30 (refer to Tables 1 and 6).


When a medical treatment on humans is conducted therewith, animal-derived antibodies produced by immunizing non-immune animals with a desired antigen generally invoke an immune rejection response. In the interests of suppressing such immune rejection, a chimeric antibody has been developed. Chimeric antibodies are prepared using a genetic engineering technique in which constant regions of animal-derived antibodies causing an anti-isotype response are replaced by those of human antibodies. Although significantly improved in anti-isotype response when compared to their original animal-derived antibodies, chimeric antibodies still retain the potential risk of side effects of anti-idiotype responses because of the animal-derived amino acids incorporated into the variable region thereof. Humanized antibodies were developed to reduce such side effects. Humanized antibodies are produced by grafting complementarity determining regions (CDRs), critical to antigen binding, of variable regions of chimeric antibodies to a human antibody framework.


One important aspect of the CDR grafting process in producing humanized antibodies is to choose a human antibody optimized for accepting CDRs of animal-derived antibodies. To this end, utilization is made of antibody databases, crystal structure analysis, molecular modeling techniques, etc. However, even when CDRs of animal-derived antibodies are grafted to an optimized human antibody framework, there may be amino acids that are positioned in the framework of the animal-derived antibody and have an influence on antigen binding, which leads, in many cases, to a reduction in the antigen binding force. Hence, the production of humanized antibodies may require additional antibody engineering technology for recovering the antigen binding force.


According to one embodiment, the antibody may be an animal antibody (e.g., mouse antibody), a chimeric antibody (e.g., mouse-human chimeric antibody), or a humanized antibody.


The chimeric antibody may comprise:


a heavy chain comprising a heavy-chain variable region of a mouse antibody and a constant region of a human immunoglobulin (IgG (IgG1, IgG2, IgG3, IgG4, and the like), IgM, IgA, IgD, or IgE) (the C-terminus of the heavy-chain variable region is linked to the N-terminus of the constant region); and


a light chain comprising a light-chain variable region and a kappa (κ)- or lambda (λ)-type constant region of a mouse antibody (the C terminus of the light-chain variable region is linked to the N terminus of the constant region).


In an embodiment, the chimeric antibody may comprise, as a heavy chain, a heavy-chain variable region of a mouse antibody and a constant region of human IgG, for example, human IgG1, human IgG2, human IgG3, or human IgG4. In an embodiment, the chimeric antibody may comprise, as a heavy chain, a heavy-chain variable region of a mouse antibody and a constant region of human IgG4.


The humanized antibody may comprise:


a heavy chain comprising a complementarity-determining region (CDR) of a mouse antibody and a framework (a variable domain region exclusive of CDR) and a heavy chain of a human immunoglobulin (IgG (IgG1, IgG2, IgG3, IgG4, and the like), IgM, IgA, IgD, or IgE); and


a light chain comprising a light-chain CDR and a kappa (κ)- or lambda (λ)-type framework and constant region of a mouse antibody.


In an embodiment, the humanized antibody may comprise, as a heavy chain, a heavy-chain CDR of a mouse antibody and a framework and constant region of a human IgG, for example, human IgG1, human IgG2, human IgG3, or human IgG4. In an embodiment, the humanized antibody may comprise, as a heavy chain, a heavy-chain CDR of a mouse antibody and a framework and constant region of a human IgG4.


An intact antibody has a structure composed of two full-length light chains and two full-length heavy chains where each light chain is linked to the heavy chain by disulfide bonds. A constant region is present in each of the heavy and the light chains. The heavy chain constant region is of a gamma (γ), mu (μ), alpha (α), delta (δ), or epsilon (ε) type, which can be further subclassified into gamma 1 (γ1), gamma 2 (γ2), gamma 3 (γ3), gamma 4 (γ4), alpha 1 (α1), or alpha 2 (α2). The light chain constant region is of either a kappa (κ) or lambda (λ) type.


As used herein, the term “heavy chain” is intended to encompass a full-length heavy chain composed of a variable region VH comprising an amino acid sequence sufficient to confer specificity for an antigen on the antibody, three constant region domains CH1, CH2, and CH3, a hinge, and a fragment of the full-length heavy chain. The term “light chain” refers to a full-length light chain composed of a variable region VL comprising an amino acid sequence sufficient to confer specificity for an antigen on the antibody, and a constant region CL, or a fragment of the full-length light chain.


The term “CDR (complementarity determining region)” denotes an amino acid sequence which resides in the heavy chain and the light chain hypervariable regions of an immunoglobulin. Each heavy and light chain has three CDRs (CDRH1, CDRH2, CDRH3, and CDRL1, CDRL2, CDRL3). The CDRs provide contact residues that play a major role in the binding of antibodies to antigens or epitopes. As used herein, the term “specifically binding” or “specifically recognizing” has the same meaning as is well known to one of ordinary skill in the art, and indicates that an antibody and an antigen specifically interact with each other to induce an immunological activity.


The term “antigen-binding fragment” refers to a fragment of the whole immunoglobulin structure which possesses a part of a polypeptide responsible for antigen binding. Examples of the antigen-binding fragment useful in the present disclosure comprise scFv, (scFv)2, Fab, Fab′ and F(ab′)2, but are not limited thereto.


Of the antigen-binding fragment, Fab is composed of one variable and one constant domain from the light chain, and one variable and the first constant (CH1) domain from the heavy chain, retaining one antigen-binding site.


A Fab′ fragment is different from Fab in that the Fab′ further comprises a hinge region having at least one cysteine residue at the C-terminus of the heavy chain CH1 domain.


A F(ab′)2 antibody forms as two Fab′ fragments are joined by a disulfide bond between the cysteine residues of the hinge region. A Fv fragment is a minimal antibody fragment composed only of variable domains from the heavy chain and the light chain. Recombinant techniques for producing the Fv are well known in the art.


In a two-chain Fv fragment, the heavy chain variable domains are associated with the light chain variable domains via a non-covalent bond. A single-chain Fv fragment has a structure in which a heavy chain variable domain and a light chain variable domain are covalently joined to each other via a covalent bond or directly at the C-terminus, so that it can form a dimer as in a two-chain Fv fragment.


The antigen-binding fragment may be obtained using a protease (for example, a whole antibody can be digested with papain to obtain Fab fragments, or can be digested with pepsin to obtain F(ab′)2 fragments), or may be prepared by a genetic recombinant technique.


The term “hinge region” refers to a region that is located between CH1 and CH2 regions in the heavy chains of an antibody, presenting flexibility to the antigen binding site in then antibody.


The anti-CD40 antibody may be a monoclonal antibody. Monoclonal antibodies may be prepared using a method well known in the art, for example, a phase display technique. Alternatively, the anti-CD40 antibody may be prepared as a mouse-derived monoclonal antibody by any conventional method.


Meanwhile, individual monoclonal antibodies can be screened on the basis of their CD40 binding capacity by using a typical ELISA (Enzyme-Linked ImmunoSorbent Assay) format. They may be analyzed for inhibitory activity by using a functional assay, such as competitive ELISA for examining molecular interactions between components within a complex, and cell-based assay. Then, the monoclonal antibody members selected based on potent inhibitory activity may be tested for their respective affinities to CD40 (Kd values).


Finally selected antibodies are composed of human immunoglobulin antibodies except for antigen binding portions and may be prepared as humanized antibodies. Preparation methods of humanized antibodies are well known in the art (Almagro, J. C. and Fransson, J., “Humanization of antibodies,” Frontiers in Bioscience, 13(2008), 1619-1633).


Another aspect provides a hybridoma producing the anti-CD40 antibody. In an embodiment, the hybridoma may be one deposited with accession number KCLRF-BP-00381.


Another aspect provides an anti-CD40 antibody produced by the hybridoma, or an antigen-binding fragment thereof.


Another aspect provides an anti-CD40 antibody or an antigen-binding fragment thereof, the anti-CD40 antibody comprising a heavy-chain complementarity-determining region (CDR-H1, CDR-H2, CDR-H3, or a combination thereof) or a light-chain complementarity-determining region (CDR-L1, CDR-L2, CDR-L3, or a combination thereof) in the anti-CD40 antibody produced by the hybridoma, or a combination thereof; or a heavy-chain variable region or a light-chain variable region in the anti-CD40 antibody produced by the hybridoma, or a combination thereof. In this regard, the complementarity-determining regions may be determined by all typical methods, for example, IMGT definition or Cabat definition, but not limited thereto.


Another aspect provides a pharmaceutical composition comprising the anti-CD40 antibody or the antigen-binding fragment thereof as an effective ingredient for preventing and/or treating disease. Another aspect provides a use of the anti-CD40 antibody or the antigen-binding fragment thereof in preventing and/or treating a disease or in preparing a composition for treatment and/or prevention of disease. Another aspect provides a method for preventing and/or treating a disease, the method comprising administering the anti-CD40 antibody or the antigen-binding fragment thereof in a therapeutically effective amount to a subject in need thereof. The method for preventing and/or treating a disease may further comprise identifying the subject in need of the prevention and/or treatment of the disease prior to the administration step. The disease may be selected from cancer, cancer metastasis, infection, and immune deficiency diseases.


The pharmaceutical composition may further comprise a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier may be one that is typically used for formulating a drug. Examples of the pharmaceutically acceptable carrier comprise lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, gelatin, calcium silicate, micro-crystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate, and mineral oil, but are not limited thereto. Also, the pharmaceutical composition may further comprise a typical additive selected from the group consisting of a diluent, an excipient, a lubricant, a humectant, a sweetener, a flavor enhancer, an emulsifier, a suspending agent, a preservative, and a combination thereof.


An effective amount of the pharmaceutical composition, or the antibody or the antigen-binding fragment thereof may be administered orally or parenterally. For parenteral administration, selection may be made of an intravenous, subcutaneous, intramuscular, intraperitoneal, intradermal, intranasal, intrapulmonary, intrarectal, or perilesional route. For oral administration, however, the pharmaceutical composition may be coated or formulated to protect the active ingredient from being degraded in the stomach because proteins or peptides are digested by pepsin. In addition, the administration may be performed with the aid of an instrument adapted for delivering the pharmaceutical composition to target cells.


The content of the anti-CD40 antibody or the antigen-binding fragment thereof in the pharmaceutical composition or the administration amount of the anti-CD40 antibody or the antigen-binding fragment thereof may be determined in consideration of various factors comprising the type of formulation, the patent's age, weight, and sex, the severity of the disorder being treated, diet, the time of administration, the interval of administration, the route of administration, the rate of excretion, and sensitivity. For example, the daily dose of the anti-CD40 antibody or the antigen-binding fragment thereof may be on the order of 0.001 to 1000 mg/kg, particularly on the order of 0.01 to 100 mg/kg, more particularly on the order of 0.1 to 50 mg/kg, and far more particularly on the order of 0.1 to 20 mg/kg, but is not limited thereto. A daily dose may be formulated into a unit dose form or distributed into separate dose forms or may be comprised within a multiple dose package.


The pharmaceutical composition may be administered in combination with a different drug such as an anticancer agent. In this regard, the dose and administration route of the pharmaceutical composition, and kinds of the different drug may be determined appropriately depending on states of patients.


The pharmaceutical composition may be formulated into solutions in oil or aqueous media, suspensions, syrup, emulsions, elixirs, powders, granules, tablets, or capsules, and in this context, a dispersant or a stabilizer may be further employed.


The subject to which the pharmaceutical composition is to be administered may be a mammal, examples of which comprise primates such as humans or monkeys, and rodents such as mice and rats.


The cancer may be a solid cancer or a blood cancer. The cancer may be selected from the group consisting of squamous cell carcinoma, small-cell lung cancer, non-small-cell lung cancer, adrenocarcinoma of lung, squamous cell carcinoma of lung, peritoneal cancer, skin cancer, skin or intraocular melanoma, rectal cancer, perianal cancer, esophagus cancer, small intestine cancer, endocrine gland cancer, parathyroid cancer, adrenal cancer, soft-tissue sarcoma, urethral cancer, leukemia (e.g., chronic or acute leukemia), lymphoma, hepatoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular adenoma, breast cancer, colon cancer, large intestine cancer, endometrial carcinoma or uterine carcinoma, salivary gland tumor, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, head or neck cancer, brain cancer, osteosarcoma, and a combination thereof, but is not limited thereto. The cancer may be a primary cancer or a metastatic cancer.


As used herein, the term “treatment of cancer” is intended to encompass all anticancer actions by which cancer symptoms are prevented from aggravation, alleviated, or reversed toward a better condition, such as inhibiting cancer cell proliferation, killing cancer cells, suppressing cancer metastasis, etc., or by which cancer is eliminated partially or entirely.


The specific binding of the anti-CD40 antibody or the antigen-binding fragment thereof to CD40 can be utilized to detect or identify CD40. Accordingly, another aspect of the present disclosure provides a composition for detection of CD40, the composition comprising the anti-CD40 antibody or the antigen-binding fragment thereof. Another aspect provides a method for detection of CD40, the method comprising the steps of: treating a biological sample with the anti-CD40 antibody or the antigen-binding fragment thereof; and examining whether an antigen-antibody reaction has occurred or not. In the detection method, an antigen-antibody reaction, if occurring, might lead to determining (deciding) the presence of CD40 in the biological sample. Therefore, the detection method may further comprise determining the presence of CD40 in the biological sample when the antigen-antibody reaction is detected after the identifying step. The biological sample may be selected from cells, tissues, or body fluids, obtained (isolated) from a human (e.g., a cancer patient), or from a culture thereof.


The examination of an antigen-antibody reaction may be carried out using a method well known in the art, for example, on the basis of an enzyme reaction, fluorescence, luminescence, and/or radiation. Examples of the method useful for examining the antigen-antibody reaction of the biological sample may comprise immunochromatography, immunohistochemistry, enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), enzyme immunoassay (EIA), fluorescence immunoassay (FIA), luminescence immunoassay (LIA), Western blotting, and microarray, but are not limited thereto.


Another aspect provides a polypeptide molecule comprising a heavy-chain complementarity-determining region, a light-chain complementarity-determining region, or a combination thereof; or a heavy-chain variable region, a light-chain variable region, or a combination thereof in the anti-CD40 antibody described above.


The polypeptide may be used as a precursor in constructing an antibody or may be comprised as a constituent in a protein scaffold (e.g., peptibody) having a structure analogous to that of an antibody, a bispecific antibody, and a multispecific antibody.


Another aspect provides a nucleic acid coding for a heavy-chain complementarity-determining region, a heavy-chain variable region, or a heavy chain in the anti-CD40 antibody.


Another aspect provides a nucleic acid molecule coding for a light-chain complementarity-determining region, a light-chain variable region, or a light chain in the anti-CD40 antibody.


Another aspect provides a recombinant vector carrying a nucleic acid molecule coding for a heavy-chain complementarity-determining region, a heavy-chain variable region, or a heavy chain of the anti-CD40 antibody and a nucleic acid molecule coding for a light-chain complementarity-determining region, a light-chain variable region, or a light chain of the anti-CD40 antibody together, or recombinant vectors carrying the nucleic acid molecules respectively.


Another aspect provides a recombinant cell harboring the recombinant vector thereat.


The term “vector” refers to a means for expressing a target gene in a host cell, as exemplified by a plasmid vector, a cosmid vector, and a viral vector such as a bacteriophage vector, an adenovirus vector, a retrovirus vector, and an adeno-related virus vector. The recombinant vector may be constructed from plasmids frequently used in the art (for example, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX series, pET series, and pUC19), phages (for example, λgt4λB, λ-Charon, λΔz1, and M13) or viruses (for example, SV40, etc.) by manipulation.


In the recombinant vector, the nucleic acid molecule may be operatively linked to a promoter. The term “operatively linked” is intended to pertain to a functional linkage between a nucleotide sequence of interest and an expression regulatory sequence (for example, a promoter sequence). When being “operatively linked, the regulatory element can control the transcription and/or translated of the nucleotide of interest.


The recombinant vector may be constructed typically as a cloning vector or an expression vector. For recombinant expression vectors, a vector typically available for expressing a foreign protein in plant, animal, or microorganism cells may be employed. Various methods well known in the art may be used for the construction of recombinant vectors.


For use in hosts, such as prokaryotic or eukaryotic cells, the recombinant vector may be constructed appropriately. For example, when a vector is constructed as an expression vector for us in a prokaryotic host, the vector typically comprises a strong promoter for transcription (e.g., a pLκλ promoter, a CMV promoter, a trp promoter, a lac promoter, a tac promoter, a T7 promoter, etc.), a ribosomal binding site for initiating translation, and transcriptional/translational termination sites. On the other hand, an expression vector for use in a eukaryotic host comprises an origin of replication operable in a eukaryotic cell, such as an f1 origin of replication, an SV40 origin of replication, a pMB1 origin of replication, an adeno origin of replication, an AAV origin of replication, and a BBV origin of replication, but is not limited thereto. In addition, the expression vector typically comprises a promoter derived from genomes of mammalian cells (for example, metallothionein promoter) or from mammalian viruses (for example, adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus promoter, and tk promoter of HSV), and a polyadenylation sequence as a transcription termination sequence.


Another aspect provides a recombinant cell harboring the recombinant vector thereat.


The recombinant cell may be prepared by introducing the recombinant vector into a suitable host cell. So long as it allows for the sequential cloning and expression of the recombinant vector in a stable manner, any host cell known in the art may be employed in the present disclosure. Examples of the prokaryotic host cell available for the present disclosure comprise E. coli JM109, E. coli BL21, E. coli RR1, E. coli LE392, E. coli B, E. coli X 1776, E. coli W3110, Bacillus spp. such as Bacillus subtilis and Bacillus thuringiensis, and enterobacteriaceae strains such as Salmonella typhimurium, Serratia marcescens and various Pseudomonas species. Eukaryotic host cells to be transformed may be, but not limited to, Saccharomyce cerevisiae, insect cells, and animal cells, such as Sp2/0, CHO (Chinese hamster ovary) K1, CHO DG44, PER.C6, W138, BHK, COS-7, 293, HepG2, Huh7, 3T3, RIN, and MDCK.


Using a method well known in the art, the nucleic acid molecule or a recombinant vector carrying the same may be introduced (incorporated) into a host cell. This transformation may be carried out using a CaCl2) or electroporation method when the host cell is prokaryotic. For eukaryotic host cells, the genetic introduction may be achieved using, but not limited to, microinjection, calcium phosphate precipitation, electroporation, liposome-mediated transfection, or particle bombardment.


To select a transformed host cell, advantage may be taken of the phonotype attributed to a selection marker according to a method known in the art. For example, when the selection marker is a gene resistant to a certain antibiotic, the host cells may be grown in the present of the antibiotic in a medium to select a transformant of interest.


Another aspect provides a method for producing an anti-CD40 antibody, the method comprising a step of expressing the nucleic acid molecule or a recombinant vector carrying the same in a host cell. The production method may comprise culturing a recombinant cell harboring the recombinant vector thereat, and optionally isolating and/or purifying the antibody from the culture medium.


Advantageous Effects

The present disclosure provides an anti-CD40 antibody that binds specifically to CD40, has an agonistic function for CD40, and does not interfere with CD40/CD40L interaction, thereby allowing more effective treatment of cancer.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a graph showing binding affinity of anti-CD40 monoclonal antibody 5C2 to CD40 (rCD40: recombinant human CD40; rCD40L: recombinant antigen human CD40L).



FIG. 2 is a graph showing binding affinity of 3 anti-CD40 5C2 chimeric antibodies to CD40 (Chimeric IgG1: a chimeric antibody comprising human IgG1 constant region; Chimeric IgG2: a chimeric antibody comprising human IgG2 constant region; Chimeric IgG4: a chimeric antibody comprising human IgG4 constant region; CP870,893: control antibody; (−): without antibody treatment).



FIG. 3 is a graph showing binding affinity of 8 humanized 5C2 antibodies to human CD40 (VH1VL1: a combination of heavy chain VH1 and light chain VL1; VH2VL1: a combination of heavy chain VH2 and light chain VL1; VH3VL1: a combination of heavy chain VH3 and light chain VL1; VH5VL1: a combination of heavy chain VH5 and light chain VL1; VH1VL2: a combination of heavy chain VH1 and light chain VL2; VH2VL2: a combination of heavy chain VH2 and light chain VL2; VH3VL2: a combination of heavy chain VH3 and light chain VL2; VH5VL2: a combination of heavy chain VH5 and light chain VL2; (−): antibody-untreated group; Positive: chimeric 5C2 antibody comprising human IgG4 constant region).



FIG. 4 shows the results of flow cytometry to examine the effects of anti-CD40 monoclonal antibody 5C2 on CD40/CD40L interaction.



FIG. 5 is a graph showing the expression level of CD80 in dendrocytes treated with monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 6 is a graph showing the expression level of CD86 in dendrocytes treated with monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 7 is a graph showing the expression level of CD83 in dendrocytes treated with monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 8 is a graph showing the expression level of CD80 when cells are co-treated with Toll like receptor ligand (Lipopolysaccharid; LPS) and monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (antibody-untreated group).



FIG. 9 is a graph showing the expression level of CD83 when cells are co-treated with Toll like receptor ligand (Lipopolysaccharid; LPS) and monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (antibody-untreated group).



FIG. 10 is a graph showing the expression level of CD86 when cells are treated with human IgG1 chimeric antibody (chimeric IgG1), human IgG2 chimeric antibody (chimeric IgG2), and human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 11 is a graph showing the expression level of CD83 when cells are treated with human IgG1 chimeric antibody (chimeric IgG1), human IgG2 chimeric antibody (chimeric IgG2), and human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 12 is a graph showing the expression level of CD86 when cells are co-treated with interferon gamma together with human IgG1 chimeric antibody (chimeric IgG1), human IgG2 chimeric antibody (chimeric IgG2), and human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 13 is a graph showing the expression level of CD83 when cells are co-treated with interferon gamma together with human IgG1 chimeric antibody (chimeric IgG1), human IgG2 chimeric antibody (chimeric IgG2), and human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 14 is a graph showing the expression level of CD86 and CD83 in dendrocytes which are treated with human IgG2 chimeric antibody (chimeric IgG2) or human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2 and co-cultured with PBMC, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 15 is a graph showing the ratio (%) of activated CD69-positive CD8 cells in dendrocytes which are treated with human IgG2 chimeric antibody (chimeric IgG2) or human IgG4 chimeric antibody (chimeric IgG4) of monoclonal antibody 5C2 and co-cultured with PBMC, wherein the expression level is indicated as a relative value to that of a control group (no treat: antibody-untreated group).



FIG. 16 is a graph showing Ramos tumor cell line apoptosis efficacy (%) induced by chimeric 5C2 antibodies (Human IgG1: irrelevant human IgG1; c5C2 IgG1: IgG1: chimeric 5C2 antibody comprising human IgG1 constant region; c5C2 IgG2: chimeric 5C2 antibody comprising human IgG2 constant region; c5C2: chimeric 5C2 antibody comprising human IgG4 constant region; CP870,893: a control antibody; Rituximab: a control antibody).



FIG. 17 is a graph showing Raji tumor cell line apoptosis efficacy (%) induced by humanized 5C2 antibodies (7 humanized 5C2 antibodies, VH1VL2, VH2VL1, VH2VL2, VH3VL1, VH3VL2, VH5VL1, VH5VL2; (−): antibody-untreated group; Human IgG: irrelevant human IgG1; CP870,893: a control antibody).



FIG. 18 is a graph showing T-cell activation level by 5C2 antibody under super-antigen SEB condition, wherein the T-cell activation level was obtained by treating PBMC with 30 ng/mL of SEB (Staphylococcal Enterotoxin B), to non-specifically induce T-cell activation, followed by treating with each of humanized 5C2 antibodies, and then, measuring the amount of IFN-gamma which is secreted by the treatment of humanized 5C2 antibodies (4 humanized 5C2 antibodies; (−): antibody-untreated group; Human IgG: irrelevant human IgG1; CP870,893: a control antibody).



FIG. 19 is a graph showing the change in tumor volume in xenograft mice models when they are treated with chimeric 5C2 IgG2 antibody alone, wherein the results are shown according to the number of days after antibody injection.



FIG. 20 is a graph showing the change in tumor volume in xenograft mice models co-grafted with human dendrocyte and PBMC when they are treated with chimeric 5C2 IgG2 antibody, wherein the results are shown according to the number of days after antibody injection.





MODE FOR CARRYING OUT THE INVENTION

Hereinafter, the present invention will be described in detail with reference to examples. These examples are only for illustrating the present invention more specifically, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples.


Example 1: Production of Anti-CD40 Antibody

1-1. Mouse Antibody Preparation


1-1-1. Preparation of Monoclonal Antibody-Producing Cell


Splenocytes from a Balb/c mouse injected with a human recombinant protein CD40 antigen were fused to the myeloma cell line X63-Ag8.653 (ATCC®, PTA-8431) from an 8-azaguanine-resistant mouse. To this end, Balb/c mice were each injected with 100 μg of the human CD40 recombinant antigen every two weeks for six weeks to induce an immune reaction. On day 3 after the final inoculation, splenocytes were isolated and suspended. According to Koeler and Milstein's method (Koeler & Milstein 1975), 108splenocytes and 107 myeloma cells were fused to each other using 50% polyethylene glycol 400 in DMEM (Dulbeco's modified Eagle's medium). The fused cells were washed and then resuspended in a DMEM medium supplemented with 20% fetal bovine serum, 100 μM hypoxanthine, 0.44 μM aminopterin, and 16 μM thymidine (HAT culture medium). The cells ere plated into 96-well plates and cultured at 37° C. in an incubator supplied with 5% CO2.


When colonies were observed two weeks later, the supernatant was taken and analyzed using ELISA (Enzyme-Linked ImmunoSorbent Assay) to examine whether to bind to CD40.


For a positive group, selection was made of wells in which 105 or more cells per well were formed in a single moiety. After being taken from the wells where colonies were formed with the supernatant measured to have a high antibody titer, cells were subcloned according to limiting dilution assay to obtain monoclonal cells having a high antibody titer. A supernatant was taken from the monoclonal cell culture and stored until the subsequent experiment.


1-1-2. Selection of Monoclonal Cell Producing Antibody to Human CD40


Human recombinant CD40 (R&D systems, Cat. No.: P25942) was plated in an amount of 100.0 ng per well into a MaxiSorp ELISA plate and reacted at 37° C. for one hour for antibody coating, followed by blocking by incubation with 200 μL of 1× blocking solution (Sigma) per well at 37° C. for one hour.


The monoclonal cell culture was added at a density of 100 μL/well, incubated at 37° C. for one hour, and washed three times with phosphate buffered saline before treatment with the secondary antibody goat anti-mouse IgG-HRP. Incubation at 37° C. for 30 minutes was followed by three rounds of washing with phosphate buffered saline. Then, TMB Single Solution (Life Technologies, Cat. No: 002023) was added in an amount of 100 μl per well and incubated at room temperature for 5-10 min in a dark place. Color development was stopped with 100 μL of 1.0 N sulfuric acid and absorbance at 450 nm was measured.


As a result, the monoclonal cell line 5C2 that produced an antibody reacting specifically with the recombinant antigen human CD40 could be selected. The cell line thus obtained was deposited Nov. 2, 2016, with the Korean Cell Line Research Foundation (KCLRF), located in Yongon-Dong, Chongno-Gu, Seoul, under accession number KCLRF-BP-00381.


1-1-3. Production of Monoclonal Antibody from Selected Monoclonal Cell Line


The established cell line 5C2 was cultured in a 10% fetal bovine serum-supplemented RPMI medium at 37° C. for two weeks under a 5% CO2 condition.


After being sterilized by filtration, the cell line culture was loaded into HiTrap Protein G HP column (GE Healthcare, Cat. No.: 17-0405-03) equilibrated with Protein G equilibration buffer (20 mM phosphate, pH7.4), washed by flowing the equilibration buffer through the column, and recovered with an elution buffer (20 mM Citric acid pH3.0). The recovered antibody was named 5C2 (mouse antibody) and dialyzed against phosphate buffered saline to exchange the buffer for use in subsequent tests.


1-1-4. Assay for Specificity of Monoclonal Anti-CD40 Antibody


For assay for the specificity of the anti-CD40 5C2 antibody (mouse antibody), reactivity against a recombinant antibody human CD40 (rCD40; R&D systems, Cat. No.: P25942), a recombinant antigen human CD40L (Recombinant Human sCD40 Ligand; Peprotech, Cat. No.: 310-02; amino acid sequence: MQKGDQNPQI AAHVISEASS KTTSVLQWAE KGYYTMSNNL VTLENGKQLT VKRQGLYYIY AQVTFCSNRE ASSQAPFIAS LWLKSPGRFE RILLRAANTH SSAKPCGQQS IHLGGVFELQ PGASVFVNVT DPSQVSHGTG FTSFGLLKL (SEQ ID NO: 31)), and a human IgG (Dinona) was analyzed using ELISA. The three antigens were each diluted at a density of 1.0 μg/mL in phosphate buffered saline and the dilution was plated at 100 μL/well into microplates and incubated at 37° C. for one hour to coat the plates therewith. Then, a 1× blocking solution (Sigma) was added in an amount of 200 μL per well and incubated at 37° C. for one hour to block the antigens.


Anti-CD40 antibody 5C2 and anti-CD40L 5C8 (ATCC®, ATCC HB-10916; positive control) were each diluted to a density of 1.0 μg/mL in phosphate buffered saline, and the dilutions were added in an volume of 100 μL per well and incubated at 37° C. for one hour, washed three times with phosphate buffered saline, and treated with the secondary antibody goat anti-mouse IgG-HRP. After 30 min of incubation at 37° C., the wells were washed three times with phosphate buffered saline, added with TMB Single Solution (Life Technologies, Cat. No: 002023) in an amount 100 μL per well, and incubated at room temperature for 5-10 min in a dark place. The color development was stopped with 100 μL of 1.0 N sulfuric acid and absorbance at 450 nm was measured.


The results obtained are depicted in FIG. 1. In FIG. 1, the y axis represents absorbance. As shown in FIG. 1, 5C2 antibody was identified to bind specifically to the human CD40 antigen because of no cross reactivity for CD40L or human IgG.


1-2. Preparation of Chimeric Antibody


On the basis of the amino acid sequence of the produced anti-CD40 mouse antibody 5C2, an anti-CD40 chimeric antibody was prepared.


1-2-1. Plasmid Construction


For use in expressing an anti-CD40 chimeric antibody, a heavy-chain expressing plasmid and a light-chain expressing plasmid were constructed, separately. Both the heavy-chain and light-chain expressing plasmids were based on the pcDNA3.4 vector (Invitrogen).


Heavy- and light-chain variable region-encoding cDNA for antibody expression were cloned using Ig-Primer sets (Novagen), inserted into pCR2.1 vector (Invitrogen, Cat. No.: K200001), and identified by sequencing. A mouse antibody gene was identified with the aid of the IMGT site (www.imgt.org).


The amino acid sequences of heavy- and light-chain variable regions in the mouse 5C2 monoclonal antibody and the nucleotide sequence of the coding gene therefor are as follows.









TABLE 1







Amino acid sequence of Variable Region in Anti-CD40 Mouse 5C2


Monoclonal Antibody










Amino acid sequence
Coding gene (DNA) sequence





Heavy-
QVQMLQSGTELVRPGT
CAGGTACAGATGCTGCAGAGCGGAACTGAACTG


chain
SVKVSCKASGYGFTNY
GTTAGACCTGGTACTAGCGTTAAGGTCAGCTGTA


variable
LIEWVKQRPGQGLEWIG
AGGCTAGCGGATACGGTTTCACCAACTACCTGAT


region
VINPGYGGVNYNEKFK
CGAATGGGTCAAGCAGAGGCCAGGACAAGGTTT


sequence
GKAILTADKSSSTAYMH
GGAGTGGATTGGAGTGATTAACCCCGGGTATGG


(VH)
LTSLTSDDSAVYFCARG
GGGCGTGAATTACAATGAGAAGTTTAAAGGCAA




GSGFAFWGOGTLVTVS

AGCCATACTGACCGCAGACAAATCAAGTAGTAC



T (SEQ ID NO: 1)
CGCCTATATGCACCTGACATCTTTGACATCTGAC




GATTCTGCCGTGTATTTTTGCGCCCGGGGCGGGA




GTGGCTTTGCTTTTTGGGGCCAGGGCACACTTGT




GACTGTGTCTACA (SEQ ID NO: 2)





Light-
DIQMTQTTSSLSASLGQ
GACATCCAAATGACCCAAACCACCTCCTCACTTT


chain
RVTISCRASQDISNHLN
CCGCATCTCTTGGACAAAGAGTCACCATCTCCTG


variable
WYQQKPNGTVRLLISST
FAGGGCAAGTCAAGACATCTCCAACCACCTCAAC


region

SRLHSGVPSRFSGSGSG

FGGTACCAGCAGAAGCCAAACGGAACTGTTAGG


sequence
TDYSLTISNLEQEDIATY
FTGTTGATCTCCAGCACCTCACGTTTGCACTCAG


(VL)
FCQQGNTLPWTFGGGT
GAGTACCATCACGATTCAGCGGTAGTGGTTCTGG



KLEIK (SEQ ID NO: 3)
FACAGATTACAGCTTGACCATTAGCAACCTGGAG




CAGGAGGATATTGCTACCTACTTCTGCCAGCAGG




GCAATACCCTGCCTTGGACATTTGGGGGGGGCAC




AAAACTGGAAATTAAG (SEQ ID NO: 4)









(CDR1, CDR2, and CDR3 (according to the IMGT definition) are sequentially underlined)


In order to allow the variable region-encoding cDNAs and constant region-encoding cDNA to be expresses as sequential amino acid sequences in antibodies, respective gene fragments in which the cloned variable region-encoding nucleotide sequences were linked to nucleotide sequences coding for the constant region (heavy chain) of known human IgG1, human IgG2, and human IgG4 (S228P, serine at position 228 was substituted with proline) and the kappa constant region (light chain) were synthesized (Bioneer). As such, the synthesized heavy- and light-chain expressing genes were digested with restriction enzymes Xho I and EcoR1. The heavy chain and light chain gene fragments thus obtained were ligated to respective pcDNA3.4 vectors to construct antibody-expressing vectors for three heavy chains and one light chain. The amino acid sequences of the heavy and light chains in the three chimeric antibodies thus constructed and nucleotide sequences coding therefor are summarized in Table 2, below:









TABLE 2







Amino acid sequences of heavy and light chains in three chimeric


antibodies and nucleotide sequence coding therefor


(bolds represent variable regions)










Amino acid sequence
Nucleotide sequence (cDNA)





Heavy

QVQMLQSGTELVRP


CAGGTACAGATGCTGCAGAGCGGAACTGAACTG



chain of

GTSVKVSCKASGYGF


GTTAGACCTGGTACTAGCGTTAAGGTCAGCTGTA



chimeric

TNYLIEWVKQRPGQ


AGGCTAGCGGATACGGTTTCACCAACTACCTGAT



IgG1 Ab

GLEWIGVINPGYGGV


CGAATGGGTCAAGCAGAGGCCAGGACAAGGTTT



(comprising

NYNEKFKGKAILTAD


GGAGTGGATTGGAGTGATTAACCCCGGGTATGG



human

KSSSTAYMHLTSLTS


GGGCGTGAATTACAATGAGAAGTTTAAAGGCAA



IgG1

DDSAVYFCARGGSGF


AGCCATACTGACCGCAGACAAATCAAGTAGTAC



Constant

AFWGQGTLVTVSTA


CGCCTATATGCACCTGACATCTTTGACATCTGAC



region)
STKGPSVFPLAPSSKST

GATTCTGCCGTGTATTTTTGCGCCCGGGGCGGG




SGGTAALGCLVKDYF

AGTGGCTTTGCTTTTTGGGGCCAGGGCACACTT




PEPVTVSWNSGALTSG

GTGACTGTGTCTACAGCTTCAACTAAGGGACCAA




VHTFPAVLQSSGLYSL
GCGTATTCCCACTTGCTCCATCTAGCAAGAGCACTA



SSVVTVPSSSLGTQTYI
GCGGAGGAACAGCTGCTTTGGGGTGTTTGGTAAAG



CNVNHKPSNTKVDKK
GATTACTTTCCCGAACCTGTTACCGTGAGCTGGAAC



VEPKSCDKTHTCPPCP
AGCGGGGCTTTGACAAGTGGCGTTCATACATTTCCT



APELLGGPSVFLFPPKP
GCCGTTTTGCAAAGCAGCGGCTTGTATAGCTTGAGC



KDTLMISRTPEVTCVV
TCTGTTGTTACCGTTCCAAGCTCATCTCTGGGCACA



VDVSHEDPEVKFNWY
CAAACATACATCTGCAACGTGAACCACAAGCCCTC



VDGVEVHNAKTKPRE
AAACACCAAGGTGGACAAGAAGGTGGAGCCAAAG



EQYNSTYRVVSVLTV
TCTTGCGACAAGACCCACACCTGTCCACCTTGTCCA



LHQDWLNGKEYKCK
GCCCCTGAACTCCTGGGGGGCCCTTCAGTTTTTCTC



VSNKALPAPIEKTISKA
TTTCCTCCTAAACCTAAAGATACACTCATGATCAGT



KGQPREPQVYTLPPSR
CGGACCCCTGAAGTTACCTGTGTGGTGGTCGATGTG



DELTKNQVSLTCLVK
TCTCATGAAGATCCTGAAGTCAAGTTTAACTGGTAT



GFYPSDIAVEWESNGQ
GTGGACGGCGTGGAGGTGCATAATGCCAAGACCAA



PENNYKTTPPVLDSDG
GCCTCGGGAGGAGCAATATAATTCTACCTATCGCGT



SFFLYSKLTVDKSRWQ
CGTCTCTGTCCTCACCGTCCTGCATCAGGACTGGCT



QGNVFSCSVMHEALH
GAATGGCAAAGAGTATAAGTGCAAAGTCAGTAACA



NHYTQKSLSLSPGK
AAGCCCTCCCCGCCCCCATAGAGAAAACCATTAGT



(SEQ ID NO: 5)
AAAGCCAAAGGGCAGCCCCGCGAGCCCCAGGTCTA




TACACTGCCCCCCAGTAGAGACGAGCTGACAAAGA




ATCAGGTGTCTCTGACATGCCTGGTGAAAGGCTTTT




ATCCCTCTGACATTGCCGTCGAGTGGGAGTCTAATG




GGCAGCCCGAGAATAATTATAAGACAACACCCCCC




GTGCTGGACAGTGACGGCTCATTTTTCCTGTATTCA




AAACTGACAGTGGACAAAAGTCGGTGGCAGCAGGG




GAATGTGTTTTCATGCAGTGTCATGCACGAGGCCCT




CCACAATCACTATACCCAGAAATCTCTGAGTCTCTC




TCCTGGGAAATGA (SEQ ID NO: 6)





Heavy

QVQMLQSGTELVRP


CAGGTACAGATGCTGCAGAGCGGAACTGAACTG



chain of

GTSVKVSCKASGYGF


GTTAGACCTGGTACTAGCGTTAAGGTCAGCTGTA



IgG2

TNYLIEWVKQRPGQ


AGGCTAGCGGATACGGTTTCACCAACTACCTGAT



chimeric

GLEWIGVINPGYGGV


CGAATGGGTCAAGCAGAGGCCAGGACAAGGTTT



Ab

NYNEKFKGKAILTAD


GGAGTGGATTGGAGTGATTAACCCCGGGTATGG



(comprising

KSSSTAYMHLTSLTS


GGGCGTGAATTACAATGAGAAGTTTAAAGGCAA



human

DDSAVYFCARGGSGF


AGCCATACTGACCGCAGACAAATCAAGTAGTAC



IgG2

AFWGQGTLVTVSTA


CGCCTATATGCACCTGACATCTTTGACATCTGAC



constant
STKGPSVFPLAPCSRST

GATTCTGCCGTGTATTTTTGCGCCCGGGGCGGG



region)
SESTAALGCLVKDYFP

AGTGGCTTTGCTTTTTGGGGCCAGGGCACACTT




EPVTVSWNSGALTSG

GTGACTGTGTCTACAGCTTCCACCAAGGGCCCATC




VHTFPAVLQSSGLYSL
CGTGTTCCCTCTGGCCCCATGTTCTAGGTCTACATCT



SSVVTVPSSNFGTQTY
GAGAGCACCGCCGCCCTCGGCTGTCTGGTGAAGGA



TCNVDHKPSNTKVDK
TTATTTCCCCGAGCCCGTGACCGTGTCTTGGAACAG



TVERKCCVECPPCPAP
CGGAGCCCTGACTAGCGGAGTGCACACCTTCCCAG



PVAGPSVFLFPPKPKD
CTGTGCTGCAGAGCTCCGGCCTGTACAGCCTCTCTT



TLMISRTPEVTCVVVD
CTGTGGTGACCGTGCCCTCTAGCAACTTCGGAACAC



VSHEDPEVQFNWYVD
AGACCTACACATGTAACGTGGATCACAAGCCTTCC



GVEVHNAKTKPREEQ
AACACCAAGGTGGATAAGACCGTGGAGAGAAAGTG



FNSTFRVVSVLTVVHQ
CTGTGTGGAGTGCCCTCCATGTCCTGCCCCACCTGT



DWLNGKEYKCKVSN
GGCTGGACCTTCTGTGTTTCTGTTCCCTCCAAAGCC



KGLPAPIEKTISKTKGQ
AAAGGATACCCTGATGATCAGCAGAACTCCTGAGG



PREPQVYTLPPSREEM
TGACCTGTGTGGTGGTGGACGTGAGCCACGAGGAT



TKNQVSLTCLVKGFYP
CCTGAGGTGCAGTTTAACTGGTACGTGGATGGCGTG



SDIAVEWESNGQPENN
GAGGTGCATAACGCTAAGACAAAGCCTAGGGAGGA



YKTTPPMLDSDGSFFL
GCAGTTTAACAGCACCTTCAGAGTGGTGAGCGTGCT



YSKLTVDKSRWQQGN
GACCGTGGTGCACCAGGATTGGCTGAACGGCAAGG



VFSCSVMHEALHNHY
AGTATAAGTGTAAGGTGTCTAACAAGGGCCTGCCA



TQKSLSLSPGK (SEQ
GCCCCTATTGAGAAGACCATCAGTAAGACCAAGGG



ID NO: 7)
ACAGCCTAGGGAGCCTCAGGTGTACACCCTGCCTCC




TTCCAGAGAGGAGATGACAAAGAACCAGGTGAGCC




TGACCTGTCTGGTGAAGGGCTTCTACCCTAGCGATA




TCGCCGTGGAGTGGGAGAGCAACGGCCAGCCTGAG




AACAACTACAAGACCACCCCACCTATGCTGGACAG




CGATGGCTCTTTCTTCCTGTACTCTAAGCTGACCGT




GGACAAGAGCAGATGGCAGCAGGGCAACGTGTTTT




CTTGTTCTGTGATGCACGAGGCCCTGCACAACCACT




ACACCCAGAAGTCTCTGTCTCTGTCTCCAGGCAAGT




GA (SEQ ID NO: 8)





Heavy

QVQMLQSGTELVRP


CAGGTACAGATGCTGCAGAGCGGAACTGAACTG



chain of

GTSVKVSCKASGYGF


GTTAGACCTGGTACTAGCGTTAAGGTCAGCTGTA



IgG4

TNYLIEWVKQRPGQ


AGGCTAGCGGATACGGTTTCACCAACTACCTGAT



chimeric

GLEWIGVINPGYGGV


CGAATGGGTCAAGCAGAGGCCAGGACAAGGTTT



Ab

NYNEKFKGKAILTAD


GGAGTGGATTGGAGTGATTAACCCCGGGTATGG



(comprising

KSSSTAYMHLTSLTS


GGGCGTGAATTACAATGAGAAGTTTAAAGGCAA



human

DDSAVYFCARGGSGF


AGCCATACTGACCGCAGACAAATCAAGTAGTAC



IgG4

AFWGQGTLVTVSTA


CGCCTATATGCACCTGACATCTTTGACATCTGAC



constant
STKGPSVFPLAPCSRST

GATTCTGCCGTGTATTTTTGCGCCCGGGGCGGG



region)
SESTAALGCLVKDYFP

AGTGGCTTTGCTTTTTGGGGCCAGGGCACACTT




EPVTVSWNSGALTSG

GTGACTGTGTCTACAGCTTCCACCAAGGGCCCCTC




VHTFPAVLQSSGLYSL
CGTGTTCCCTCTCGCCCCTTGCTCCAGATCCACCTCC



SSVVTVPSSSLGTKTY
GAGTCTACCGCCGCTCTGGGCTGCCTGGTCAAGGAC



TCNVDHKPSNTKVDK
TACTTCCCCGAGCCTGTGACCGTGTCTTGGAACTCT



RVESKYGPPCPPCPAP
GGCGCACTGACCAGCGGCGTGCACACCTTCCCTGCC



EFLGGPSVFLFPPKPK
GTGCTGCAGTCCTCCGGCCTGTACTCCCTGTCCTCC



DTLMISRTPEVTCVVV
GTCGTGACCGTGCCCTCCTCCAGCCTGGGCACCAAG



DVSQEDPEVQFNWYV
ACCTACACCTGTAACGTGGACCACAAGCCCTCCAA



DGVEVHNAKTKPREE
CACCAAGGTGGACAAGCGGGTGGAATCTAAGTACG



QFNSTYRVVSVLTVLH
GCCCTCCCTGCCCCCCCTGCCCTGCCCCTGAATTTCT



QDWLNGKEYKCKVS
GGGCGGACCTTCCGTGTTCCTGTTCCCCCCAAAGCC



NKGLPSSIEKTISKAKG
CAAGGACACCCTGATGATCTCCCGGACCCCCGAAG



QPREPQVYTLPPSQEE
TGACCTGCGTGGTGGTGGACGTGTCCCAGGAAGAT



MTKNQVSLTCLVKGF
CCCGAGGTCCAGTTCAATTGGTACGTGGACGGCGT



YPSDIAVEWESNGQPE
GGAAGTGCACAACGCCAAGACCAAGCCCAGAGAG



NNYKTTPPVLDSDGSF
GAACAGTTCAACTCCACCTACCGGGTGGTGTCCGTG



FLYSRLTVDKSRWQE
CTGACCGTCCTGCACCAGGACTGGCTGAACGGCAA



GNVFSCSVMHEALHN
AGAGTACAAGTGCAAGGTGTCCAACAAGGGCCTGC



HYTQKSLSLSLGK
CCTCCAGCATCGAAAAGACCATCTCCAAGGCCAAG



(SEQ ID NO: 9)
GGCCAGCCCCGCGAGCCTCAGGTGTACACCCTGCC




CCCTAGCCAGGAAGAGATGACCAAGAACCAGGTGT




CCCTGACCTGTCTCGTCAAAGGCTTCTACCCCTCCG




ATATCGCCGTGGAATGGGAGTCCAACGGCCAGCCC




GAGAACAACTACAAGACCACCCCCCCTGTGCTGGA




CTCCGACGGCTCCTTCTTTCTGTACTCTCGGCTGACC




GTGGATAAGAGCCGGTGGCAGGAAGGCAACGTCTT




CTCCTGCTCCGTGATGCACGAGGCCCTGCACAACCA




CTATACCCAGAAGTCCCTGTCCCTGAGCCTGGGCAA




ATGA (SEQ ID NO: 10)





Light

DIQMTQTTSSLSASL


GACATCCAAATGACCCAAACCACCTCCTCACTTT



chain

GQRVTISCRASQDISN


CCGCATCTCTTGGACAAAGAGTCACCATCTCCTG





HLNWYQQKPNGTVR


TAGGGCAAGTCAAGACATCTCCAACCACCTCAAC





LLISSTSRLHSGVPSR


TGGTACCAGCAGAAGCCAAACGGAACTGTTAGG





FSGSGSGTDYSLTISN


TTGTTGATCTCCAGCACCTCACGTTTGCACTCAG





LEQEDIATYFCQQGN


GAGTACCATCACGATTCAGCGGTAGTGGTTCTG





TLPWTFGGGTKLEIK


GTACAGATTACAGCTTGACCATTAGCAACCTGGA




RTVAAPSVFIFPPSDEQ

GCAGGAGGATATTGCTACCTACTTCTGCCAGCA




LKSGTASVVCLLNNFY

GGGCAATACCCTGCCTTGGACATTTGGGGGGGG




PREAKVQWKVDNAL

CACAAAACTGGAAATTAAGCGGACTGTTGCTGCTC




QSGNSQESVTEQDSKD
CATCTGTTTTTATATTTCCTCCCAGCGACGAGCAGC



STYSLSSTLTLSKADY
TGAAAAGCGGCACTGCCTCTGTGGTGTGTCTGCTGA



EKHKVYACEVTHQGL
ATAATTTTTACCCCCGGGAAGCCAAAGTCCAGTGG



SSPVTKSFNRGEC
AAGGTGGATAATGCCCTCCAGTCTGGGAACAGTCA



(SEQ ID NO: 11)
GGAAAGTGTGACAGAACAGGATAGTAAGGACTCTA




CTTATAGCCTCTCTTCTACACTGACTCTGTCAAAGG




CCGACTATGAGAAGCATAAAGTGTATGCCTGCGAG




GTGACACATCAGGGCCTGAGTTCACCCGTGACAAA




ATCTTTTAACCGCGGCGAGTGCTGA (SEQ ID NO: 12)









For use as a positive control antibody, the anti-CD40 human antibody CP870,893 (U.S. Pat. No. 7,338,660 B2; 21.4.1 antibody) was also synthesized. A pcDNA3.4 expression vector was constructed in the same manner. For use as a control, an antibody was constructed using the sequence information of CP870,893 antibody (U.S. Pat. No. 7,338,660 B2; 21.4.1 antibody) and named CP870,893 antibody analogue. The amino acid sequences and coding nucleotide sequences used to construct the CP870,893 antibody analogue are summarized in Table 3, below:









TABLE 3







Amino acid and nucleotide sequences of anti-CD40 human antibody


CP870,893 (21.4.1 antibody) (signal sequences underlined and


variable regions in bold)










Amino acid sequence
Nucleotide sequence (cDNA)





Heavy

MDWTWRILFLVAAATGA


ATGGACTGGACCTGGAGGATCCTCTTCTTGGTGGCAG



chain

HS
QVQLVQSGAEVKKP


CAGCCACAGGAGCCCACTCC
CAGGTGCAGCTGGTGC





GASVKVSCKASGYTFTG


AGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTC





YYMHWVRQAPGQGLE


AGTGAAGGTCTCCTGCAAGGCTTCTGGATACACCT





WMGWINPDSGGTNYAQ


TCACCGGCTACTATATGCACTGGGTGCGACAGGCC





KFQGRVTMTRDTSISTA


CCTGGACAAGGGCTTGAGTGGATGGGATGGATCA





YMELNRLRSDDTAVYY


ACCCTGACAGTGGTGGCACAAACTATGCACAGAAG





CARDQPLGYCTNGVCS


TTTCAGGGCAGGGTCACCATGACCAGGGACACGTC





YFDYWGQGTLVTVSSAS


CATCAGCACAGCCTACATGGAGCTGAACAGGCTGA




TKGPSVFPLAPCSRSTSES

GATCTGACGACACGGCCGTGTATTACTGTGCGAGA




TAALGCLVKDYFPEPVTV

GATCAGCCCCTAGGATATTGTACTAATGGTGTATG




SWNSGALTSGVHTFPAVL

CTCCTACTTTGACTACTGGGGCCAGGGAACCCTGG




QSSGLYSLSSVVTVPSSNF

TCACCGTCTCCTCAGCCTCCACCAAGGGCCCATCGG




GTQTYTCNVDHKPSNTK
FCTTCCCCCTGGCGCCCTGCTCCAGGAGCACCTCCGA



VDKTVERKCCVECPPCPA
GAGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTAC



PPVAGPSVFLFPPKPKDTL
FTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGGCG



MISRTPEVTCVVVDVSHE
CTCTGACCAGCGGCGTGCACACCTTCCCAGCTGTCCT



DPEVQFNWYVDGVEVHN
ACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTG



AKTKPREEQFNSTFRVVS
ACCGTGCCCTCCAGCAACTTCGGCACCCAGACCTACA



VLTVVHQDWLNGKEYKC
CCTGCAACGTAGATCACAAGCCCAGCAACACCAAGGT



KVSNKGLPAPIEKTISKTK
GGACAAGACAGTTGAGCGCAAATGTTGTGTCGAGTGC



GQPREPQVYTLPPSREEM
CCACCGTGCCCAGCACCACCTGTGGCAGGACCGTCAG



FK
FCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATG



NQVSLTCLVKGFYPSDIA
ATCTCCCGGACCCCTGAGGTCACGTGCGTGGTGGTGG



VEWESNGQPENNYKTTPP
ACGTGAGCCACGAAGACCCCGAGGTCCAGTTCAACTG



MLDSDGSFFLYSKLTVDK
GTACGTGGACGGCGTGGAGGTGCATAATGCCAAGAC



SRWQQGNVFSCSVMHEA
AAAGCCACGGGAGGAGCAGTTCAACAGCACGTTCCGT



LHNHYTQKSLSLSPGK
GTGGTCAGCGTCCTCACCGTTGTGCACCAGGACTGGC



(SEQ ID NO: 13)
FGAACGGCAAGGAGTACAAGTGCAAGGTCTCCAACA




AAGGCCTCCCAGCCCCCATCGAGAAAACCATCTCCAA




AACCAAAGGGCAGCCCCGAGAACCACAGGTGTACAC




CCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAG




GTCAGCCTGACCTGCCTGGTCAAAGGCTTCTACCCCA




GCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGC




CGGAGAACAACTACAAGACCACACCTCCCATGCTGGA




CTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCG




FGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTC




ATGCTCCGTGATGCATGAGGCTCTGCACAACCACTAC




ACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAATGA




(SEQ ID NO: 14)





Light

MRLPAOLLGLLLLWFPGS


ATGAGGCTCCCTGCTCAGCTCCTGGGGCTCCTGCTGCT



chain

RC
DIQMTQSPSSVSASVG


CTGGTTCCCAGGTTCCAGATGC
GACATCCAGATGAC





DRVTITCRASQGIYSWL


CCAGTCTCCATCTTCCGTGTCTGCATCTGTAGGAG





AWYQQKPGKAPNLLIY


ACAGAGTCACCATCACTTGTCGGGCGAGTCAGGGT





TASTLQSGVPSRFSGSGS


ATTTACAGCTGGTTAGCCTGGTATCAGCAGAAACC





GTDFTLTISSLQPEDFAT


AGGGAAAGCCCCTAACCTCCTGATCTATACTGCAT





YYCQQANIFPLTFGGGT


CCACTTTACAAAGTGGGGTCCCATCAAGGTTCAGC





KVEIKRTVAAPSVFIFPPS


GGCAGTGGATCTGGGACAGATTTCACTCTCACCAT




DEQLKSGTASVVCLLNNF

CAGCAGCCTGCAACCTGAAGATTTTGCAACTTACT




YPREAKVQWKVDNALQS

ATTGTCAACAGGCTAACATTTTCCCGCTCACTTTCG




GNSQESVTEQDSKDSTYS

GCGGAGGGACCAAGGTGGAGATCAAACGAACTGTG




LSSTLTLSKADYEKHKVY
GCTGCACCATCTGTCTTCATCTTCCCGCCATCTGATGA



ACEVTHQGLSSPVTKSFN
GCAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCTGC



RGEC (SEQ ID NO: 15)
FGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTG




GAAGGTGGATAACGCCCTCCAATCGGGTAACTCCCAG




GAGAGTGTCACAGAGCAGGACAGCAAGGACAGCACC




TACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAG




ACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCAC




CCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTC




AACAGGGGAGAGTGTTAG (SEQ ID NO: 16)









1-2-2. Chimeric Anti-CD40 Antibody Production


For chimeric antibody production, expression vectors carrying three heavy chains (respectively comprising constant regions of human IgG1, human IgG2, and human IgG4) and one light chain (inclusive of the kappa constant regions) were introduced into the EXPICHO™ Expression system (Thrmofisher, Cat. No.: A29133) in a transient transformation manner to produce IgG1 chimeric antibody (inclusive of the constant region of human IgG1), IgG2 chimeric antibody (inclusive of the constant region of human IgG2), and IgG4 chimeric antibody (inclusive of the constant region of human IgG4).


The ExpiCHO-S cell line included within the EXPICHO™ Expression system kit (Termofisher, Cat. No.: A29133) was thawed and added to the Expression medium and incubated at 37° C. in a 8% CO2 atmosphere with shaking at 120 rpm to secure a necessary number of cells. After being harvested, the ExpiCHO-S cells were seeded at a density of 6×106 cells/mL in the Expression medium to a final volume of 150 mL. As indicated by the manual, 100 μg of the light chain vector and 50 μg of the heavy chain vector were mixed with OPTIPRO™ SFM and EXPIFECTAMINE™ CHO reagent to give a mixture in which the vectors were at a concentration of 1.0 μg/mL with the light chain and the heavy chain maintained at a ratio of 2:1. The vector mixture prepared for transformation was added to the ExpiCHO-S cell line and incubated at 37° C. for 24 hours in 8% CO2 atmosphere with shaking at 120 rpm. At 24 hours of incubation, the EXPICHO™ Enhancer and Feed were further added. On day five after incubation, the incubation condition was changed to 32° C., 5% CO2, and 120 rpm. In the new condition, the cells were incubated with the EXPICHO™ Feed for an additional 12 days.


The culture medium thus obtained was sterilized by filtration and loaded into HITRAP® Protein A HP column (GE Healthcare, Cat. No.: 11-0034-93) equilibrated with Protein A equilibration buffer (20 mM phosphate, pH7.4) and washed by flowing the equilibration buffer through the column, followed by antibody recovery with an elution buffer (20 mM Citric acid pH3.0). The recovered antibody was dialyzed against phosphate buffered saline to exchange the buffer for use in subsequent tests.


1-2-3. Assay for Antigen Binding Force of Chimeric Anti-CD40 Antibody


The three anti-CD40 chimeric antibodies IgG1, IgG2, and IgG4 constructed in Examples 1-2-2 were assayed for antigen reactivity by ELISA. A dilution of the recombinant antigen human CD40 (Sino Biological Inc., Cat. No.: 10774-H02H) at a concentration of 1.0 μg/mL in phosphate buffered saline was plated at 100 μL/well into microplates and incubated at 37° C. for one hour to coat the plates therewith. Then, a 1× blocking solution (Sigma) was added in an amount of 200 μL per well and incubated at 37° C. for one hour to block the antigen.


Each of the three anti-CD40 chimeric antibodies and the control antibody anti-CD40 human antibody CP870,893 analogue (21.4.1 antibody in Table 2) was serially diluted from a concentration of 10.0 μg/mL. The dilutions were added in an amount of 100 μL/well, incubated at 37° C. for one hour and washed three times with phosphate buffered saline. The antibodies were treated with the secondary antibody goat anti-human IgG F(ab′)2-HRP (Jackson Immunoresearch, Cat. No.: 109-035-006) at 37° C. for 30 min and washed three times with phosphate buffered saline. After reaction with 100 μL of TMB Single Solution (Life Technologies, Cat. No: 002023) per well at room temperature for 4-10 min in a dark place, the color development was stopped with 100 μL of 1.0 N sulfuric acid and then absorbance at 450 nm was measured.


Absorbance at 450 nm of the chimeric antibodies thus obtained are depicted in FIG. 2. ((−) denotes a control treated with no antibodies). As shown in FIG. 2, all the three anti-CD40 chimeric antibodies were found to have affinity for CD40 as high as or higher than that the control antibody.


1-3. Preparation of Humanized Antibody


On the basis of the amino acid sequence of the anti-CD40 mouse antibody 5C2, an anti-CD40 humanized antibody was constructed.


1-3-1. Selection of Recombinant Antibody Sequence by in Silico Humanization


Humanized antibody sequences that retained CDR region sequences of each of the heavy and light chains of mouse CD40 antibody 5C2 and in which on the basis of a germline sequence of a human antibody gene, sequences for the framework region were recombined were selected in an in-silico manner.


To begin with, heavy-chain CDRs (complementarity determining regions) and light-chain CDRs were determined on the basis of the heavy- and light-chain variable regions of the anti-CD40 mouse antibody 5C2 introduced in Table 1 (referring to IMGT/V-QUEST and are summarized in Table 4, below:












TABLE 4






CDR1
CDR2
CDR3















DeterminedaccordingtoIMGT/V-QUEST


(http://www.imgt.org/IMGT_vquest/share/textes/)










Heavy
GYGFTNYL (SEQ
INPGYGGV (SEQ ID NO:
GGSGFAF (SEQ


chain
ID NO: 17)
18)
ID NO: 19)


Light
QDISNH (SEQ ID
STS (SEQ ID NO: 21)
QQGNTLP (SEQ


chain
NO: 20)

ID NO: 22)










Determined according to Cabat definition


(http://www.bioinf.org.uk/abs/)










Heavy
NYLIE (SEQ ID NO:
VINPGYGGVNYNEKFKG
GGSGFAF (SEQ


chain
33)
(SEQ ID NO: 34)
ID NO: 19)


Light
RASQDISNHLN
STSRLHS (SEQ ID NO:
QQGNTLP (SEQ


chain
(SEQ ID NO: 35)
36)
ID NO: 22)









In Table 5 are summarized human antibody germline genes that were employed as the backbones of humanized recombinant antibody sequences because of the highest similarity in sequence to each of the heavy and light chains of mouse CD40 antibody 5C2:









TABLE 5







Human Antibody Germline Gene Used for


Humanization of Mouse 5C2 Antibody


Human Ab Germline










Heavy chain
Light chain







IGHV1-2*02
IGKV1-NL1*01



IGHV1-69*06
IGKV3-15*01



IGHV5-51*01











Six heavy-chain variable regions and two light-chain variable regions for humanized 5C2 antibodies were selected in an in-silico method using the human antibody germline gene sequences and are summarized in Table 6, below:









TABLE 6







5C2 humanized antibody variable region sequence selected in silico











Class
Sequence
SEQ ID NO:





Heavy
VH1
QVQLVQSGAEVKKPGASVKVSCKASGYGFTNYLIEWVRQAP
23


chain

GOGLEWIGVINPGYGGVNYNEKFKGRATLTADKSISTAYMEL



variable

SRLRSDDTAVYFCARGGSGFAFWGQGTLVTVSS



region
VH2
QVQLVQSGAEVKKPGASVKVSCKASGYGFTNYLIEWVRQAP
24




GQGLEWIGVINPGYGGVNYNEKFKGRVTLTADKSISTAYMEL





SRLRSDDTAVYFCARGGSGFAFWGOGTLVTVSS




VH3
QVQLVQSGAEVKKPGSSVKVSCKASGYGFTNYLIEWVRQAPG
25




QGLEWIGVINPGYGGVNYNEKFKGRATLTADKSTSTAYMELS





SLRSEDTAVYFCARGGSGFAFWGQGTLVTVSS




VH4
QVQLVQSGAEVKKPGSSVKVSCKASGYGFTNYLIEWVRQAPG
26




QGLEWIGVINPGYGGVNYNEKFKGRVTITADKSTSTAYMELSS





LRSEDTAVYFCARGGSGFAFWGQGTLVTVSS




VH5
EVQLVQSGAEVKKPGESVKISCKASGYGFTNYLIEWVROMPG
27




KGLEWIGVINPGYGGVNYNEKFKGQATLSADKSISTAYLQLSS





LKASDTAVYFCARGGSGFAFWGQGTLVTVSS




VH6
EVQLVQSGAEVKKPGESLKISCKASGYGFTNYLIEWVRQMPG
28




KGLEWIGVINPGYGGVNYNEKFKGQVTISADKSISTAYLOLSS





LKASDTAMYFCARGGSGFAFWGQGTLVTVSS






Light
VL1
DIQMTQSPSSLSASVGDRVTITCRASQDISNHLNWYQQKPGKA
29


variable

VKLLISSTSRLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYFC



region


QQGNTLPWTFGQGTKVEIK




Chain
VL2
EIVMTQSPATLSVSPGERATLSCRASQDISNHLNWYQQKPGOA
30




VRLLISSTSRLHSGIPARFSGSGSGTEYTLTISSLQSEDFAVYFCQ






QGNTLPWTFGQGTKVEIK










(CDR1, CDR2, are CDR3 (according to the IMGT definition) are sequentially underlined)


1-3-2. Expression and Analysis of Humanized Recombinant Antibody


The heavy-chain variable region sequences in-silico selected were linked to the human IgG2 constant region to complete heavy-chain sequences while the light-chain variable region sequences were linked to the kappa light-chain constant region to complete light-chain sequences. The selected amino acid sequences were converted into genetic sequences coding therefor and gene fragments having the sequences are synthesized (Cosmogenetech).


The coding gene fragments of heavy and light chains prepared above were transferred into respective pCDNA3.4 vectors. Four heavy chains (VH1, VH2, VH3, VH5) and two light chains (L1, L2) were combined to make eight humanized antibodies (VH1VL1: a combination of VH1 and VL1; VH2VL1: a combination of VH2 and VL1; VH3VL1: a combination of VH3 and VL1; VH5VL1: a combination of VH5 and VL1; VH6VL1: a combination of VH6 and VL1; VH1VL2: a combination of VH1 and VL2; VH2VL2: a combination of VH2 and VL2; VH3VL2: a combination of VH3 and VL2; VH5VL2: a combination of VH5 and VL2; VH6AVL2: a combination of VH6 and VL2). The antibodies were introduced to be expressed in CHO cells (Sigma, Cat. #: 85050302) with the aid of VIAFECT™ Transfection Reagent (Promega, Cat. No.: D4981) and incubated for an additional two day before taking the supernatants.


Each of the humanized antibody cultures was assayed for reactivity for a recombinant antigen human CD40 (rCD40; R&D systems, Cat. No.: P25942) by ELISA. For this assay, a dilution of rCD40 at a concentration of 1.0 μg/mL in phosphate buffered saline was plated at a density of 100 μL/well into microplates and incubated at 37° C. for one hour to coat the microplates with the antigen which was then blocked by adding a 1× blocking solution (Sigma) in an amount of 200 μl per well and incubating at 37° C. for one hour.


Following one hour of incubation with 100 μL of each of the humanized anti-CD40 antibody cultures at 37° C., the microplates were washed three times with phosphate buffered saline. Treatment with the secondary antibody goat anti-human IgG F(ab′)2-HRP (Jackson Immunoresearch, Cat. No.: 109-035-006) at 37° C. for 30 min was followed by three rounds of washing with phosphate buffered saline. Then, TMB Single Solution (Life Technologies, Cat. No: 002023) was added in an amount of 100 μl per well and incubated at room temperature for 5-10 min in a dark place. Color development was stopped with 100 μL of 1.0 N sulfuric acid and absorbance at 450 nm was measured.


Absorbances at 450 nm of the eight humanized antibodies are depicted in FIG. 3. As shown in FIG. 3, all the eight humanized antibodies were observed to have high reactivity for CD40.


Example 2: Effect of Anti-CD40 Antibody 5C2 on CD40/CD40L Binding Affinity

A blocking test was performed so as to examine whether the 5C2 antibody (mouse antibody) has an influence on interaction between CD40 and CD40L in the human body.


For buffer change, the 5C2 antibody and the isotype control antibody (Dinona, Cat. No.: 88020R) were each dialyzed against 0.1M sodium bicarbonate buffer and 1.0 mg of each antibody solution was prepared. A solution of FITC Isomer I (Invitrogen, Cat. No.: F1906) at a concentration of 2.6 mg/mL in DMSO (Sigma-Aldrich, Cat. No.: 276855) was added in an amount of 20.0 μL to each of the antibody solutions. After FITC conjugation by stirring at room temperature for 2 hours, the resulting antibody solutions were dialyzed against phosphate buffered saline to obtain FITC-labeled antibodies.


Then, 5×106 cells of Ramos cell line (ATCC, ATCC CRL-1596), which expresses human CD40, was incubated at room temperature for 30 min with 100 μL of a dilution of the recombinant antigen CD40L (Peprotech, Cat. No.: 310-02) at a concentration of 15.0 μg/mL. After washing with phosphate buffered saline, the cells were reacted with 1.0 μg/mL of each of the prepared FITC-labeled 5C2 and FITC-labeled isotype antibodies at room temperature for 15 min. The cells were washed again with phosphate buffered saline and then subjected to fluorescent immunoassay using flow cytometry (Stratedigm, S1000EXi).


The flow cytometry analysis results thus obtained are depicted in FIG. 4. As shown in FIG. 4, the fluorescence of FITC-labeled 5C2 was still generated although it was a little bit lower upon preincubation with the recombinant antigen CD40L than without CD40L because of the steric hinderance of the FITC-labeled 5C2. The results imply that the 5C2 antibody recognizes CD40 at a site different from the site of CD40/CD40L interaction.


Example 3: Assay for Activation of Dendrocyte

3-1. Preparation of Monocyte-Derived Dendrocyte


In order to examine whether the anti-CD40 antibody activates dendrocytes, monocyte-derived dendrocytes were prepared by differentiation and used for testing the efficacy of the anti-CD40 antibody.


Whole blood sampled from a healthy adult volunteer was transferred to an EDTA (ethylenediaminetetraacetic acid)-treated tube and mixed with one volume of phosphate buffered saline. The whole blood mixture was loaded on FICOLL® PAQUE PLUS (GE Healthcare, Cat. No.: 17-1440-03) and centrifuged at 700×g at room temperature for 30 min. After centrifugation, only the PBMC (peripheral blood mononuclear cell) layer was transferred to a new tube and added with about two volumes of phosphate buffered saline. Centrifugation at 4° C. at 700 g for 5 min formed a PBMC pellet. A suspension of the PBMC pellet in a 10% fetal bovine serum-supplemented RPMI medium was cultured at 37° C. for 4 hours in a cell culture dish (VOM plastic crop., Cat. No.: V100D) with 5% CO2.


After removal of non-adherent cells from the cell culture dish, a fresh 10% fetal bovine serum-supplemented RPMI was added to the dish. For differentiation to dendrocytes, the adherent cells were incubated with 5×103 unit/mL of each of rhGM-CSF (Recombinant Human Granulocyte Macrophage Colony Stimulating Factor; JW creagene) and rhIL-4 (JW creagene). On days 3 and 6 after incubation, the 10% fetal bovine serum-supplemented RPMI medium was changed with a fresh one, with each of rhGM-CSF and rhIL-4 added at a concentration of 5×103 unit/mL.


On day 7 of differentiation, the dendrocytes were detached with trypsin-EDTA buffer (Thermo, Cat. No.: R001100), washed with a 10% fetal bovine serum-supplemented RPMI medium, and aliquoted into 24- or 12-well cell culture dishes. The cells were treated with the anti-CD40 antibody and various reagents according to test purposes and cultured for an additional 2 to 5 days before analysis of changes in the dendrocytes


3-2. Activation of Dendrocyte by Mouse Monoclonal 5C2


5×105 cells of the monocyte-derived dendrocytes prepared in Example 3-1 were treated with 10 μg/mL of the mouse monoclonal antibody 5C2, together with the secondary antibody goat anti-mouse IgG (H+L) (Dionona) for crosslinking so as to amplify the efficacy.


After incubation with the antibody at 37° C. for 2 days in a 5% CO2 atmosphere, the dendrocytes were detached with trypsin-EDTA (Thermo, Cat. No.: R001100). Dendrocytes were identified by immunostaining with an anti-CD11c antibody-FITC (eBiosceince, Cat. No.: 11-0116-42). Degrees of activation of the dendrocytes were accounted for by expression levels of the co-stimulatory factors CD80, CD83, and CD86 which were measured as fluorescence intensities detected after staining the cells with each of anti-CD80 antibody-PECy5 (eBiosceince, Cat. No.: 15-0809-42), anti-CD86 antibody-PE (eBiosceince, Cat. No.: 12-0869-42), and anti-CD83 antibody-APC (eBiosceince, Cat. No.: 17-0839-42).


The fluorescence intensities obtained above are depicted as relative values to a control (not treated with the antibody, 100%) in FIG. 5 (relative expression level of CD80), FIG. 6 (relative expression level of CD86) and FIG. 7 (relative expression level of CD83) (in FIGS. 5 to 7, GAM denotes the secondary antibody goat anti-mouse IgG (H+L)). As shown in FIGS. 5 to 7, the 5C2 monoclonal antibody was found to induce the overexpression of the co-stimulatory factors CD80, CD83, and CD86, with the effect further enhanced by crosslinking with the secondary antibody.


3-3. Effect of Toll-like Receptor Ligand Used in Combination on Activation of Dendrocyte


Toll-like receptors (TLRs) are usually expressed on sentinel cells, such as macrophages and dendrocytes, which account for innate immunity. The proteins are responsible for the function of recognizing molecules derived from microbes and activating innate immunity. Lipopolysaccharide (LPS), known as a TLR ligand recognized by TLR-4, was used in combination with the 5C2 antibody in order to evaluate the activation of dendrocytes.


For this, 5×105 cells of the monocyte-derived dendrocytes prepared in Example 3-1 were incubated for two days with 1.0 ng/mL of LPS (Sigma-Aldrich, Cat. No.: L3024) in combination of 10 μg/mL of 5C2 and 20 μg/mL of a secondary antibody goat anti-Mouse IgG(H+L) (Dionona), followed by detaching the dendrocytes with trypsin-EDTA. The dendrocytes were stained with anti-CD11c-FITC (eBiosceince, Cat. No.: 11-0116-42).


Degrees of activation of the dendrocytes were accounted for by expression levels of the co-stimulatory factors CD80 and CD83 which were measured as fluorescence intensities detected after staining the cells with each of anti-CD80 antibody-PECy5 (eBiosceince, Cat. No.: 15-0809-42) and anti-CD86 antibody-PE (eBiosceince, Cat. No.: 12-0869-42).


The fluorescence intensities obtained above are depicted as relative values to a control (not treated with the antibody, 100%) in FIG. 8 (relative expression level of CD80) and FIG. 9 (relative expression level of CD86) (in FIGS. 8 and 9, GAM denotes the secondary antibody goat anti-mouse IgG (H+L)). As shown in FIGS. 8 and 9, LPS alone induced overexpression of CD80 and CD83, but the overexpression of CD80 and CD83 was remarkably enhanced by LPS in the condition of crosslinking 5C2.


3-4. Activation of Dendrocyte by Chimeric 5C2


Dendrocytes which were differentiated from 5×105 monocytes as described in Example 3-1 were treated with 10 μg/mL of each of the two chimeric 5C2 antibodies (chimeric IgG2 and chimeric IgG4) and the CP870,893 analogue (U.S. Pat. No. 7,338,660 B2; 21.4.1 antibody; refer to Table 3) in the absence of a second antibody. After incubation at 37° C. for 5 days in a 5% CO2 atmosphere, the dendrocytes were detached with trypsin-EDTA(Thermo, Cat. No.: R001100).


Dendrocytes were identified by immunostaining with an anti-CD11c antibody-FITC (eBiosceince, Cat. No.: 11-0116-42). Degrees of activation of the dendrocytes were accounted for by expression levels of the co-stimulatory factors CD80, CD83, and CD86 which were measured as fluorescence intensities detected after staining the cells with each of anti-CD80 antibody-PECy5 (eBiosceince, Cat. No.: 15-0809-42), anti-CD86 antibody-PE (eBiosceince, Cat. No.: 12-0869-42), and anti-CD83 antibody-APC (eBiosceince, Cat. No.: 17-0839-42).


The fluorescence intensities obtained above are depicted as relative values to a control (not treated with the antibody, 100%) in FIG. 10 (relative expression level of CD86) and FIG. 11 (relative expression level of CD83).


The three 5C2-derived chimeric antibodies were observed to be not much different in reactivity for human CD40 because of the identical variable region thereof (FIG. 2), but to differ in the activation of dendrocytes from one isotype to another. For the mouse 5C2 antibody, a crosslinking antibody was indispensable for inducing the overexpression of the co-stimulatory factors, but the chimeric antibodies in IgG2 or IgG4 type were found to elicit the overexpression of CD86 and CD83 on dendrocytes even in the absence of a crosslinking antibody. Further, the 5C2 IgG4 chimeric antibody exhibited higher activation of dendrocyte, compared to the Pfizer analogue CP870,893, which is known as the best inducer for the activation of dendrocytes.


3-5. Synergistic Effect of IFN-Gamma on Activation of Dendrocytes


Dendrocytes indispensably requires interferon gamma for inducing T cell activation to effectively attack foreign matter or cancer cells, and interferon gamma is known to be directly involved in dendritic differentiation.


In order to examine the effect of the 5C2 chimeric antibody on dendrocyte activation and cytokine change in the presence of interferon gamma (IFN), interferon gamma (Pepprotech, Cat. No.: 300-02) was added at a concentration of 1000 unit/mL to the 5×105 monocyte-derived dendrocyte prepared in Example 3-1.


To the resulting cells, each of the two chimeric 5C2 antibodies (chimeric IgG2 and chimeric IgG4) and the CP870,893 analogue (U.S. Pat. No. 7,338,660 B2; 21.4.1 antibody; refer to Table 3) was added at a concentration of 10.0 μg/mL.


The dendrocytes were incubated with the antibodies at 37° C. for 2 days in a 5% CO2 atmosphere and then detached with trypsin-EDTA (Thermo, Cat. No.: R001100). Dendrocytes were identified by immunostaining with an anti-CD11c antibody-FITC (eBiosceince, Cat. No.: 11-0116-42). Degrees of activation of the dendrocytes were accounted for by expression levels of the co-stimulatory factors CD86 and CD83 which were measured as fluorescence intensities detected after staining the cells with each of anti-CD86 antibody-PE (eBiosceince, Cat. No.: 12-0869-42) and anti-CD83 antibody-APC (eBiosceince, Cat. No.: 17-0839-42).


The fluorescence intensities obtained above are depicted as relative values to a control (not treated with the antibody, 100%) in FIG. 12 (relative expression level of CD86) and FIG. 13 (relative expression level of CD83) (N: treated with neither antibody nor interferon gamma, N+IFN: treated with interferon gamma alone, IgG2: treated with IgG2 chimeric antibody alone, IgG2+IFN: treated with IgG2 chimeric antibody and interferon gamma in combination, IgG4: treated with IgG4 chimeric antibody alone, IgG4+IFN: treated with IgG4 chimeric antibody and interferon gamma in combination, CP8: treated with CP870,893 analogue alone, and CP8+IFN: treated with CP870,893 analogue and interferon gamma in combination).


As shown in FIGS. 12 and 13, interferon gamma was observed to induce the overexpression of CD86 and CD83 and to elicit a synergistic effect, in combination with the chimeric antibodies, especially the IgG4 isotype chimeric antibody, on the expression of CD86 and CD83.


Example 4: Activation Effect of Dendrocyte on CD8 T Cell

After recognizing an external antigen, dendrocytes present a MHC/foreign antigen to T cells. Then, T cells recognize the MHC/foreign antigen and are activated to effectively remove the foreign antigen. In order to examine whether the anti-CD40 antibody provided in the present disclosure induces T cell activation, the Burkitt's lymphoma cell line Ramos was used as a foreign antigen in a T cell activation assay.


After being cultured in a 10% fetal bovine serum RPMI medium, the Ramos cell line (ATCC®, ATCC CRL-1596) were repetitively frozen and thawed twice and centrifuged. Absorbance of the supernatant (Ramos lysate) thus obtained was measured at 280 nm and arbitrarily set in the concentration unit “mg/mL”.


Dendrocytes which were differentiated from 5×105 monocytes as described in Example 3-1 were seeded at a density of 5×105 cells/well into 12-well culture dishes to which the prepared Ramos lysate was then added at a concentration of 20 μg/mL. For comparison of effects of the anti-CD40 antibodies, the cells were incubated with 10 μg/mL of each of the three chimeric 5C2 antibodies (chimeric IgG1, chimeric IgG2, and chimeric IgG4) and the CP870,893 analogue (U.S. Pat. No. 7,338,660 B2; 21.4.1 antibody; refer to Table 3). On the next day, PBMC (peripheral blood mononuclear cell) was separated from the whole blood from the donor of the dendrocytes and then co-cultured with the prepared dendrocytes.


Dendrocytes were identified by immunostaining with an anti-CD11c antibody-FITC (eBiosceince, Cat. No.: 11-0116-42). Degrees of activation of the dendrocytes were accounted for by expression levels of the co-stimulatory factors CD86 and CD83 which were measured as fluorescence intensities detected after staining the cells with each of anti-CD86 antibody-PE (eBiosceince, Cat. No.: 12-0869-42) and anti-CD83 antibody-APC (eBiosceince, Cat. No.: 17-0839-42).


The fluorescence intensities obtained above are depicted as relative values to a control (not treated with the antibody, 100%) in FIG. 14 (No treat: treated with no antibodies, IgG2: treated with IgG2 chimeric antibody, IgG4: treated with IgG4 chimeric antibody, CP870893: treated with CP870,893 analogue). When co-cultured with PBMC, dendrocytes increased in the expression levels of CD86 and CD83 in the presence of the chimeric antibodies of the monoclonal antibody 5C2 and a remarkable increase was observed particularly in the presence of the IgG4 chimeric antibody.


For T cell activation assay, PBMC co-cultured with dendrocytes were treated with the antibodies and immunostained with anti-CD8-FITC (Dinona, Cat. No.: 10143B) and anti-CD69-PE (eBioscience, Cat. No.: 12-0699-42) together. CD8-expressing cells (CD8-FITC positive cells) and CD69-expressing cells (CD69 positive cells) were selected with reference to the fluorescence intensities detected. Proportions (% based on numbers of cells) of CD69-positive cells in the selected CD8-FITC positive T cell population are depicted in FIG. 15. As shown in FIG. 15, proportions of activated CD69-positive CD8 cells increased with the application of the chimeric antibodies of the monoclonal antibody 5C2. Particularly, the proportion of activated CD69-positive CD8 was observed to peak upon treatment with the IgG4 chimeric antibody.


Example 5: Assay for Apoptotic Efficacy

5-1. Assay for Apoptotic Effect of Chimeric 5C2 Antibody In order to identify the direct cytotoxicity of the 5C2 antibody against tumor cell lines, an apoptosis test was conducted. The Ramos cell line (ATCC®, ATCC CRL-1596) was washed with phosphate buffered saline, resuspended in a 5.0% fetal bovine serum-supplemented RPMI, and plated at a density of 2×105 cells/well into microplates. The cells were incubated with 1.0 μg/mL of each of the three anti-CD40 chimeric antibodies (IgG1, IgG2, and IgG4), the CP870,893 analogue, and Rituxan at 37° C. for 2 days in a 5% CO2 atmosphere. Cytotoxicity effects were determined by positive cell proportions (%) after staining FITC Annexin-V (BD Pharmingen, Cat. No.: 51-65874X) and 7-AAD (BD Pharmingen, Cat. No.: 51-68981E) according to the manufacturer's instruction. The test results are depicted in FIG. 16. As can be seen in FIG. 16, the 5C2 IgG2 isotype exhibited excellent anti-tumor cytotoxicity.


For use in examining the apoptotic efficacy of humanized antibody 5C2, the Raji Burkitt's lymphoma cell line (ATCC®, ATCC CCL-86) was washed with phosphate buffered saline, resuspended in a 3.0% fetal bovine serum-supplemented RPMI medium, and plated at a density of 2×105 cells/well into microplates. The cells were incubated with 10.0 μg/mL of each of seven humanized antibodies 5C2 IgG2 (VH1VL2, VH2VL1, VH2VL2, VH3VL1, VH3VL2, VH5VL1, and VH5VL2), the CP870,893 analogue, and the human IgG at 37° C. for two days in a 5% CO2 atmosphere. Subsequently, as described in Example 5-1, the cells were stained with FITC Annexin-V and 7-AAD to determine the apoptotic effect. The result is depicted in FIG. 17. As shown in FIG. 17, all the seven humanized antibodies were found to have higher apoptotic effects than the CP870,893 analogue.


Example 6: T Cell Activation by 5C2 Antibody in Presence of Super-Antigen Staphylococcal Enterotoxin B (SEB)

Because T cell activation is made in an antigen-specific manner, one antigen cannot induce the activation unless T cells have the same TCR. A super-antigen, such as SEB, which can activate T cells in a non-specific manner, should be employed in order to generally activate T cells in in vitro tests using PBMC.


In this test, SEB was applied to PBMC to establish a condition for T cell activation before examining whether treatment with 5C2 antibodies further enhanced an immune reaction.


After being isolated from the whole blood, from a healthy volunteer, PBMC was seeded at a density of 1×106 cells/well into microplates. Then, the cells were treated with 30 ng/mL of SEB, together with 10 μg/mL of each of five humanized antibodies 5C2 (VH2VL1, VH2VL2, VH3VL1, and VH3VL2), the CP870,893 analogue, and the human IgG at 37° C. for three days in a 5% CO2 atmosphere. The medium supernatant was taken and used to measure an amount of human IFN-gamma (Affymetrix, Cat. NO.: 88-7316-22) according to the manufacturer's instruction. The result thus obtained is depicted in FIG. 18. As can be seen FIG. 18, the amount of IFN-gamma produced upon treatment with the 5C2 humanized antibodies was two or three times larger than with the control, implying that 5C2 stimulation can further enhance an immune response.


Example 7: Assay for Anti-Tumor Effect of 5C2 in Animal Model

Being able to introduce CD8 T cell activation via dendrocytes, the 5C2 antibodies are expected to have an anti-tumor effector. In addition, the apoptotic effect of the 5C2 antibodies on CD40-expressing tumor cell lines allows the anticipation of an anti-tumor effector accompanied.


The 5C2 antibodies have no cross reactivity to mouse CD40. Thus, effects of the 5C2 antibodies were tested in two models: injected with a tumor cell line alone and in combination with human dendrocytes and peripheral blood mononuclear cell (PBMC).


The leukemia cell line Ramos (ATCC®, ATCC CRL-1596) was subcutaneously injected alone or human dendrocytes and PBMC to NSG (NOD.Cg-Prkdcscidll2rgtm1Wjl/SzJ) mice (the Jackson Laboratory, 25 grams, 6-8 weeks old).


In this test, dendrocytes were prepared by taking whole blood from a healthy adult volunteer, separating PBMC from the whole blood with the aid of FICOLL® PAQUE PLUS (GE Healthcare, Cat. No.: 17-1440-03), and differentiating the adherent cells in the presence of rhGM-CSF (Recombinant Human Granulocyte Macrophage Colony Stimulating Factor; JW creagene) and rhlL-4 (JW creagene), as in Example 3-1. On day 7 of differentiation, dendrocytes were detached, washed with phosphate buffered saline, and subcutaneously injected at a density of 5×105 cells/mouse to the mouse abdomen.


After being prepared by separation from whole blood taken from the same donor of dendrocytes, with the aid of FICOLL® PAQUE PLUS (GE Healthcare, Cat. No.: 17-1440-03), PBMC was washed with phosphate buffered saline and subcutaneously injected at a density of 2×106 cells/mouse to the mouse abdomen.


Subsequent to the cell injection, the chimeric 5C2 IgG2 antibody or phosphate buffered saline was intraperitoneally injected. At the time of one week later, the antibody or phosphate buffered saline was further injected once. The assay procedure is summarized in Table 7, below:









TABLE 7







Assay for Anti-Tumor Effector of 5C2 in Animal









Drug













Group
Tumor
Effector

Dose
# of



















Group
N
Tumor
Number
Route
Type
Number
Route
Type
(mg/Kg)
Dose
Route





Group 1
5
Ramos
1 × 107
S.C.
None
NA
S.C.
PBS
NA
2
I.P.


Group 2
5



None
NA

5C2
1.0


Group 3
5



DC/PBMC
5 × 105/2 × 106

PBS
NA


Group 4
5



DC/PBMC
5 × 105/2 × 106

5C2
1.0









After antibody administration, tumor volumes (mm3) were monitored.


Average values of the tumor volumes (mm3) that has changed with time are given in Table 8 and depicted in FIGS. 19 and 20.









TABLE 8







Average Tumor Volume (mm3)

















Time after












Ab administ.


(Day)
0
10
13
17
20
24
27
31
34
38




















Group 1
0
24.1
105.6
1090.0
2287.2
5912.3
11481.9





Group 2
0
44.5
111.0
141.2
591.5
1347.6
2816.7





Group 3
0
110.9
308.4
1109.0
2786.2
6073.3
12537.8





Group 4
0
2.8
5.2
23.2
38.4
0
0
0
0
96.2









As shown in Table 8 and FIG. 19, the 5C2 antibody exhibited the effect of delaying the growth of tumor in the animal model injected with tumor cells alone (Group 2). The apoptotic effect of the 5C2 antibody seems to contribute to the anti-tumor effect. In the animal model injected with the tumor cells together with dendrocytes and PBMC, as is understood from Table 8 and FIG. 20, the tumor rapidly grew to over 10,000 mm3 after 24 days for the control, but the tumor growth was perfectly suppressed until 34 days after administration of the 5C2 antibody (Group 4). After 34 days, tumor growth was observed in one mouse because the lives of the injected human dendrocytes and PBMC came to an end in the inventors' opinion.

Claims
  • 1. An anti-CD40 antibody or an antigen-binding fragment thereof, comprising the following complementarity determining region (CDRs): CDR-H1 comprising the amino acid sequence of SEQ ID NO: 17 or SEQ ID NO: 33,CDR-H2 comprising the amino acid sequence of SEQ ID NO: 18 or SEQ ID NO: 34,CDR-H3 comprising the amino acid sequence of SEQ ID NO: 19,CDR-L1 comprising the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 35,CDR-L2 comprising the amino acid sequence of SEQ ID NO: 21 or SEQ ID NO: 36, andCDR-L3 comprising the amino acid sequence of SEQ ID NO: 22.
  • 2. The anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1, comprising: a heavy-chain variable region comprising the amino acid sequence of SEQ ID NO: 1, 23, 24, 25, 26, 27, or 28; anda light-chain variable region comprising the amino acid sequence of SEQ ID NO: 3, 29, or 30.
  • 3. The anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1, wherein the anti-CD40 antibody is an animal antibody, a chimeric antibody, or a humanized antibody.
  • 4. The anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1, wherein the antigen-binding fragment is scFv, (scFv)2, Fab, Fab′, or F(ab′)2 of the anti-CD40 antibody.
  • 5. The anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1, having an agonist activity for CD40.
  • 6. A pharmaceutical composition comprising the anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1.
  • 7. A nucleic acid encoding the anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1.
  • 8. The nucleic acid according to claim 7, encoding either or both of: an amino acid sequence comprising SEQ ID NO: 17 or SEQ ID NO: 33, and SEQ ID NO: 18 or SEQ ID NO: 34, and SEQ ID NO: 19; andan amino acid sequence comprising SEQ ID NO: 20 or SEQ ID NO: 35, and SEQ ID NO: 21 or SEQ ID NO: 36, and SEQ ID NO: 22.
  • 9. The nucleic acid of claim 7, encoding either or both of: the amino acid sequence of SEQ ID NO: 1, 23, 24, 25, 26, 27, or 28; andthe amino acid sequence of SEQ ID NO: 3, 29, or 30.
  • 10. A recombinant vector comprising the nucleic acid of claim 8.
  • 11. A recombinant cell comprising the recombinant vector of claim 10.
  • 12. An anti-CD40 antibody or an antigen-binding fragment thereof, which comprise CDR-H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 of an anti-CD40 antibody produced by the hybridoma of accession number KCLRF-BP-00381.
  • 13. The anti-CD40 antibody or an antigen-binding fragment thereof according to claim 12, which comprise a heavy chain variable region and a light chain variable region of an anti-CD40 antibody produced by the hybridoma of accession number KCLRF-BP-00381.
  • 14. The anti-CD40 antibody or an antigen-binding fragment thereof according to claim 12, wherein the anti-CD40 antibody is produced by the hybridoma of accession number KCLRF-BP-00381.
  • 15. A hybridoma, deposited with accession number KCLRF-BP-00381, for producing an anti-CD40 antibody.
  • 16. A method of treating a disease, the method comprising administering the anti-CD40 antibody or the antigen-binding fragment thereof according to claim 1 to a subject in need of treating the disease, wherein the disease is selected from the group consisting of cancer and cancer metastasis.
  • 17. The method of claim 16, wherein the anti-CD40 antibody or the antigen-binding fragment thereof comprises: a heavy-chain variable region comprising the amino acid sequence of SEQ ID NO: 1, 23, 24, 25, 26, 27, or 28; anda light-chain variable region comprising the amino acid sequence of SEQ ID NO: 3, 29, or 30.
  • 18. A method of treating a disease, the method comprising administering an anti-CD40 antibody or an antigen-binding fragment thereof to a subject in need of treating the disease, wherein the disease is selected from the group consisting of cancer and cancer metastasis, wherein the anti-CD40 antibody is selected from the group consisting of: an anti-CD40 antibody produced by hybridoma of accession number KCLRF-BP-00381, or an antigen-binding fragment thereof;an anti-CD40 antibody comprising CDR-H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 of the anti-CD40 antibody produced by the hybridoma of accession number KCLRF-BP-00381, or an antigen-binding fragment thereof;and an anti-CD40 antibody comprising the heavy-chain and light-chain variable regions of the anti-CD40 antibody produced by the hybridoma of accession number KCLRF-BP-00381, or an antigen-binding fragment thereof.
Priority Claims (1)
Number Date Country Kind
10-2016-0150624 Nov 2016 KR national
PCT Information
Filing Document Filing Date Country Kind
PCT/KR2017/012747 11/10/2017 WO
Publishing Document Publishing Date Country Kind
WO2018/088850 5/17/2018 WO A
US Referenced Citations (8)
Number Name Date Kind
5801227 Fanslow, III et al. Sep 1998 A
7338660 Bedian et al. Mar 2008 B2
7449616 Pons et al. Nov 2008 B2
8496937 Schneider et al. Jul 2013 B2
8980257 Kaneda et al. Mar 2015 B2
20050169923 De Boer et al. Aug 2005 A1
20150309028 Jordan Oct 2015 A1
20170088605 Abend et al. Mar 2017 A1
Foreign Referenced Citations (10)
Number Date Country
2018202509 Apr 2018 AU
63489 Mar 2002 BG
2243531 Jul 1997 CA
10-2007-0012626 Jan 2007 KR
10-2008-0030958 Apr 2008 KR
10-2014-0009505 Jan 2014 KR
333602 Mar 1997 NZ
19970333602 Jun 2000 NZ
2012145673 Oct 2012 WO
2016069919 May 2016 WO
Non-Patent Literature Citations (4)
Entry
Juan C. Almagro et al., “Humanization of antibodies”, Frontiers in Bioscience, vol. 13, pp. 1619-1633, 2008.
Im Muno Geno Tics, “Welcome! to IMGT/V-QUEST”, http://www.imgt.org/IMGT_vquest/share/textes/, dicrectory release on Feb. 19, 2019.
Prof. Andrew C.R, “Antibodies”, Martin's Group at UCL, last modified on Feb. 5, 2019, http://www.bioinf.org.uk/abs/.
GenBank: AHX81815.1, “immunoglobulin heavy chain variable region, partial [Mus musculus]”, Apr. 2014.
Related Publications (1)
Number Date Country
20230192875 A1 Jun 2023 US