ANTIBODY SPECIFICALLY BINDING WITH PD-L1 AND ANTIGEN-BINDING FRAGMENT OF ANTIBODY

Abstract
Provided are an antibody specifically binding to PD-L1 and an antigen-binding fragment thereof. The antibody and the antigen-binding fragment thereof have high specificity and stability, can specifically block a PD-L1-SIPRα pathway, do not cause hemagglutination within a certain concentration range, and thus can significantly suppress tumor growth in vivo.
Description
FIELD OF THE INVENTION

The invention relates to the research field of an antibody and the humanization transformation of the antibody, and in particular, the invention relates to an antibody capable of specifically binding to PD-L1 and an antigen-binding fragment thereof.


BACKGROUND OF THE INVENTION

Programmed death-1 (PD-1) is an immune checkpoint molecule, is mainly involved in the activation and regulation of T cells, and can regulate the strength and duration of an immune response. Under normal conditions, PD-1 can mediate and maintain the autoimmune tolerance of body tissues, prevent the excessive activation of the immune system from damaging self-tissues during inflammatory responses, and play a positive role in avoiding the occurrence of autoimmune diseases; under pathological conditions, it is involved in tumor immunity and the occurrence and development of various autoimmune diseases (Anticancer Agents Med Chem. 2015; 15(3):307-13; Hematol Oncol Stem Cell Ther. 2014 March; 7(1):1-17; Trends Mol Med. 2015 January; 21(1):24-33; Immunity. 2013 Jul. 25; 39(1):61-73; J Clin Oncol. 2015 Jun. 10; 33(17):1974-82.).


The ligands of PD-1 include PD-L1 (programmed death ligand 1) and PD-L2 (programmed death ligand 2). Its ligands belong to the B7 family, in which PD-L1 is inducibly expressed on the surface of various immune cells, including T cells, B cells, monocytes, macrophages, DC cells, endothelial cells, epidermal cells, etc., while PD-L2 is only inducibly expressed on some immune cells, including macrophages, DC cells, and B cells (Autoimmun Rev, 2013, 12(11): 1091-1100; Front Immunol, 2013, 4: 481; Nat Rev Cancer, 2012, 12(4): 252-264; Trends Mol Med. 2015 January; 21(1):24-33.).


PD-L1 is highly expressed on the surface of a variety of tumor cells, including melanoma, lung cancer, kidney cancer, breast cancer, ovarian cancer, cervical cancer, bladder cancer, esophageal cancer, gastric cancer, pancreatic cancer, intestinal cancer, etc. PD-L2 is highly expressed on B-cell lymphoma. Tumor cells bind to PD-1 on T cells through highly expressed PD-L1 or PD-L2, and transmit immunosuppressive signals, causing the body to develop immune tolerance to tumor cells and thereby, facilitating the growth and metastasis of tumor cells. High expression of PD-1 ligands is closely related to poor prognosis and drug resistance of tumor patients (Hematol Oncol Stem Cell Ther. 2014 March; 7(1):1-17.). Studies have further found that the expression of PD-1 is upregulated on the surface of T cells, especially the surface of T cells infiltrated in tumor cells, which is also closely related to poor prognosis (Trends Mol Med. 2015 January; 21(1):24-33.).


Several studies have shown that antibodies blocking the PD-1/PD-Ls signaling pathways have anti-tumor effects. Clinically, PD-1/PD-Ls blocking antibodies have the following characteristics: firstly, the efficacy of PD-1/PD-Ls blocking antibodies is not limited to a certain tumor type, contrarily, a potent and durative anti-tumor efficacy presents in a broad spectrum of tumors; secondly, the safety of PD-1/PD-Ls blocking antibodies is good, and it will not cause side effects common in some chemotherapy drugs and targeted drugs such as fatigue, leukopenia, baldness, diarrhea, skin rash, etc., but will only cause some immune-related side effects. The anti-PD-1 antibody nivolumab has been marketed for the treatment of advanced melanoma, non-small cell lung cancer, renal cell carcinoma and lymphoma, etc. Pembrolizumab has been marketed for the treatment of advanced melanoma, non-small cell lung cancer and lymphoma, etc. The anti-PD-L1 antibody Atezolizumab has been marketed for the treatment of advanced non-small cell lung cancer and urothelial cancer, durvalumab has been marketed for the treatment of advanced non-small cell lung cancer and urothelial carcinoma, and avelumab has been marketed for the treatment of advanced Merkel cell carcinoma.


There remains a need in the art to develop more effective antibodies that specifically bind to PD-L1.


SUMMARY OF THE INVENTION

A first aspect of the invention relates to an isolated anti-human programmed death ligand 1 (PD-L1) antibody, an antigen-binding fragment, or a variant thereof, wherein said antibody or antigen-binding fragment thereof comprises a light chain variable region and/or a heavy chain variable region, wherein

    • the amino acid sequences of LCDR1, LCDR2, and LCDR3 of the light chain variable region are set forth in SEQ ID Nos. 20-22, respectively; and/or
    • the amino acid sequence of HCDR1 of the heavy chain variable region is set forth in SEQ ID No. 23; the amino acid sequence of HCDR2 of the heavy chain variable region has at least about 94% identity to any one of SEQ ID Nos. 24 and 45-47; the amino acid sequence of HCDR3 of the heavy chain variable region is set forth in SEQ ID No. 25; or
    • the amino acid sequences of the CDRs of the light chain variable region or the heavy chain variable region have at least 70% identity to the sequences set forth in SEQ ID Nos. 20-25 or 45-47 respectively and retain the biological activity of the corresponding parent sequences, such as at least 75%, 80%, 85%, 90% or 95% or higher identity; or
    • the amino acid sequences of the CDRs of the light chain variable region or the heavy chain variable region are variant sequences of the sequences set forth in SEQ ID Nos: 20-25 or 45-47 respectively obtained after deletion, substitution and/or addition of one or more amino acid residues which retain the biological activities of the corresponding parent sequences, such as 1, 2, 3 or more amino acid residues;
    • wherein the variant is one selected from the group consisting of chimeric antibodies, humanized antibodies, and fully human antibodies.


In some embodiments, the heavy chain constant region sequence of the antibody is the constant region sequence of one selected from the group consisting of human IgG1, IgG2, IgG3, IgG4, IgA, IgM, IgE, IgD, and/or, the light chain constant region sequence of the antibody is the constant region sequence of κ chain or λ chain; preferably, the heavy chain constant region sequence is the constant region sequence of IgG1 or IgG4, and/or the light chain constant region sequence is the constant region sequence of κ light chain.


In some embodiments, the amino acid sequence of the light chain variable region of a chimeric anti-PD-L1 antibody or a functional fragment thereof is as set forth in SEQ ID NO. 18; or has at least 70% identity to the sequence set forth in SEQ ID No. 18 and retains the biological activity of the corresponding parent sequence, such as at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% % or higher identity; or is a variant sequence of the sequence set forth in SEQ ID NO. 18 obtained by deleting, substituting and/or adding one or more amino acid residues which retains the biological activity of the corresponding parent sequence, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more amino acid residues; and/or the amino acid sequence of the heavy chain variable region is set forth in SEQ ID NO. 19; or has at least 70% identity to the sequence set forth in SEQ ID NO. 19 and retains the biological activity of corresponding parent sequence, such as at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or higher identity; or is a variant sequence of the sequence set forth in SEQ ID NO. 19 obtained by deleting, substituting and/or adding one or more amino acid residues that retains the biological activity of the corresponding parent sequence, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more amino acid residues;

    • the amino acid sequences of the light chain constant region and the heavy chain constant region of the chimeric anti-PD-L1 antibody or a functional fragment thereof are set forth in SEQ ID NO. 26 and SEQ ID No. 27 respectively; or have at least 70% identity to the sequences set forth in SEQ ID NO. 26 and SEQ ID No. 27 respectively and retain the biological activity of the corresponding parent sequences, such as at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.4%, 99.7% or higher identity; or are variant sequences of the sequences set forth in SEQ ID NO. 26 and SEQ ID No. 27 respectively obtained by deleting, substituting and/or adding one or more amino acid residues that retain the biological activity of the corresponding parent sequences, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more amino acid residues.


In some embodiments, the framework regions of the light chain variable region of the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof include FR-L1, FR-L2, FR-L3 and FR-L4, and the framework regions of the heavy chain variable include FR-H1, FR-H2, FR-H3 and FR-H4, and wherein,

    • the amino acid sequence of the FR-L1 is set forth in SEQ ID NO. 54;
    • the amino acid sequence of the FR-L2 is set forth in SEQ ID NO. 55, or an amino acid sequence as set forth in SEQ ID NO. 55 obtained by one of the following substitutions or any combination thereof:
    • the 2nd amino acid Y is substituted with I,
    • the 3rd amino acid Q is substituted with H,
    • the 9th amino acid A is substituted with S;
    • the amino acid sequence of the FR-L3 is set forth in SEQ ID NO. 56;
    • the amino acid sequence of the FR-L4 is set forth in SEQ ID NO. 57;
    • the amino acid sequence of the FR-H1 is set forth in SEQ ID NO. 58, or an amino acid sequence as set forth in SEQ ID NO. 58 obtained by one of the following substitutions or any combination thereof:
    • the 1st amino acid Q is substituted with E,
    • the 23rd amino acid K is substituted with T;
    • the amino acid sequence of the FR-H2 is set forth in SEQ ID NO. 59, or an amino acid sequence as set forth in SEQ ID NO. 59 obtained by the following substitution:
    • the 13th amino acid M is substituted with I;
    • the amino acid sequence of the FR-H3 is set forth in SEQ ID NO. 60, or an amino acid sequence as set forth in SEQ ID NO. 60 obtained by one of the following substitutions or any combination thereof:
    • the 2nd amino acid V is substituted with A,
    • the 8th amino acid E is substituted with T,
    • the 11th amino acid S is substituted with N,
    • the 31st amino acid A is substituted with G; and/or
    • the amino acid sequence of the FR-H4 is set forth in SEQ ID NO. 61.


In some embodiments, the amino acid sequence of the light chain variable region is as set forth in any one of SEQ ID Nos. 38, 39 or 44, and/or the amino acid sequence of the heavy chain variable region is as set forth in any one of SEQ ID Nos. 30-37, 40-43 or 48-53; or is a variant sequence having at least 70% identity to one of the amino acid sequences set forth SEQ ID Nos. 30-44 or 48-53 and retaining the biological activity of the corresponding parent sequence, such as at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or higher identity; or is a variant sequence of one of the sequences set forth in SEQ ID Nos. 30-44 or 48-53 obtained after deletion, substitution and/or addition of one or more amino acid residues which retains the biological activity of the corresponding parent sequence, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more amino acid residues;

    • preferably, the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 38, and/or the heavy chain variable region has the amino acid sequence selected from the group consisting of those set forth in SEQ ID Nos. 30-37; or
    • the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 39, and/or the heavy chain variable region has the amino acid sequence selected from the group consisting of those set forth in SEQ ID Nos. 30-37 or 48-50; or
    • the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 44, and/or the heavy chain variable region has the amino acid sequence selected from the group consisting of those set forth in SEQ ID Nos. 40-43 or 51-53;
    • more preferably, the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 39, and/or the heavy chain variable region has the amino acid sequence selected from the group consisting of those set forth in SEQ ID Nos. 36 or 48-50; or
    • the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 44, and/or the heavy chain variable region has the amino acid sequence selected from the group consisting of those set forth in SEQ ID Nos. 41 or 51-53;
    • preferably, the amino acid sequences of the light chain constant region and the heavy chain constant region are as set forth in SEQ ID NO. 26 and SEQ ID NO. 27 respectively.


It is well known in the art that when referring to an identity, since the length of an amino acid sequence can only be a natural number, the actual identity value obtained by calculation may not be a finite percentage like 95%, but a number close to a percentage such as 95%. For example, for the variable region sequence of 117 amino acid residues, when only one amino acid residue is changed, the corresponding percentage of identity is a percentage close to 99.15% actually, but for convenience, such a figure would only be recorded as 99% in the context.


In some embodiments, the antigen-binding fragment is one or more selected from the group consisting of F(ab′)2, Fab′, Fab, Fd, Fv, scFv, diabody, camelid antibody, CDR, and antibody minimal recognition unit (dAb), preferably, the antigen-binding fragment is Fab, F(ab′)2 or scFv.


A second aspect of the invention relates to an isolated nucleic acid molecule selected from the group consisting of

    • (1) DNA or RNA encoding the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect;
    • (2) a nucleic acid that is completely complementary to the DNA or RNA defined in (1).


A third aspect of the invention relates to a vector comprising the nucleic acid molecule operably linked as described in the second aspect, preferably, the vector is an expression vector.


A fourth aspect of the invention relates to a host cell comprising the nucleic acid molecule as described in the second aspect or the vector as described in the third aspect.


A fifth aspect of the invention relates to a composition comprising the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect, the nucleic acid molecule as described in the second aspect, the vector as described in the third aspect or the host cell as described in the fourth aspect, and a pharmaceutically acceptable carrier, diluent, or excipient.


A sixth aspect of the invention relates to a method of producing the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect, comprising the steps of culturing the host cells as described in the fourth aspect under culture conditions suitable for the expression of the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof, and optionally, isolating and purifying the obtained products.


A seventh aspect of the invention relates to use of the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect, the nucleic acid molecule as described in the second aspect, the vector as described in the third aspect or the host cell as described in the fourth aspect in the manufacture of a medicament for the prevention and/or the treatment of PD-L1-mediated diseases or disorders such as autoimmune diseases, immune responses against grafts, allergies, infectious diseases, neurodegenerative diseases and tumors.


An eighth aspect of the invention relates to the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect, the nucleic acid molecule as described in the second aspect, the vector as described in the third aspect or the host cell as described in the fourth aspect, for use in the prevention and/or treatment of PD-L1-mediated diseases or disorders such as autoimmune diseases, immune responses to grafts, allergies, infectious diseases, neurodegenerative diseases, and tumors.


A ninth aspect of the invention relates to a method of preventing and/or treating PD-L1-mediated diseases or disorders such as autoimmune diseases, immune responses to grafts, allergies, infectious diseases, neurodegenerative diseases and tumors, comprising administering to a subject in need thereof the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof as described in the first aspect, the nucleic acid molecule as described in the second aspect, the vector as described in the third aspect or the host cell as described in the fourth aspect.


In some embodiments, the autoimmune diseases are one or more selected from the group consisting of arthritis, rheumatoid arthritis, psoriasis, multiple sclerosis, ulcerative colitis, Crohn's disease, systemic lupus erythematosus, glomerulonephritis, dilated cardiomyopathy-like disease, Sjogren's syndrome, allergic contact dermatitis, polymyositis, scleroderma, periarteritis polyarteritis, rheumatic fever, vitiligo, insulin-dependent diabetes mellitus, Behcet's syndrome and chronic thyroiditis; preferably, the autoimmune diseases are one or more selected from the group consisting of arthritis, rheumatoid arthritis, psoriasis, multiple sclerosis, ulcerative colitis, Crohn's disease, systemic lupus erythematosus, glomerulonephritis, rheumatic fever, vitiligo, insulin-dependent diabetes and chronic thyroiditis; more preferably, the autoimmune diseases are one or more selected from the group consisting of rheumatoid arthritis, psoriasis, multiple sclerosis, ulcerative colitis, Crohn's disease, systemic lupus erythematosus, insulin-dependent diabetes mellitus, and chronic thyroiditis.


In some embodiments, the immune response against the graft includes, for example, graft versus host disease.


In some embodiments, the allergies are one of more selected from the group consisting of urticaria, eczema, angioedema, allergic rhinitis, bronchial asthma, laryngeal edema, food allergic gastroenteritis, and anaphylactic shock; preferably, the allergies are one or more selected from the group consisting of urticaria, eczema, allergic rhinitis, bronchial asthma, and anaphylactic shock; more preferably, the allergies are one or more selected from the group consisting of allergic rhinitis, bronchial asthma, and anaphylactic shock.


In some embodiments, the infectious disease refers to the local and systemic inflammatory response caused by the invasion of viruses, bacteria, fungi, parasites and other pathogens and specific toxins into the human body. Examples of pathogenic viruses include, for example, HIV, hepatitis viruses (types A, B, and C), herpes viruses (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein-Barr virus), adenoviruses, influenza virus, flavivirus, echovirus, rhinovirus, coxsackievirus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, Dengue virus, papilloma virus, molluscum virus, polio virus, rabies virus, JC virus, and arboencephalitis virus. Pathogenic bacteria include, for example, Treponema pallidum, Chlamydia, Rickettsia, Mycobacteria, Staphylococci, Streptococci, Pneumococci, Meningococci and Gonorrhoeae (conococci), Klebsiella, Proteus, Serratia, Pseudomonas, Legionella, Diphtheria bacillus, Salmonella, Bacillus, Cholera bacteira, Clostridium tetani, Bacillus botulinus, Bacillus anthracis, Yersinia pestis, Leptospirosis bacteria and Borrelia burgdorferi causing Lyme disease. Pathogenic fungi include, for example, Candida (Candida albicans, Candida krusei, Candida glabrata, Candida tropicalis, etc.), Cryptococcus neoformans, Aspergillus (Aspergillus fumigatus, Aspergillus niger, etc.), Mucorales (mucor, bsidia, rhizophus)), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis, and Histoplasma capsulatum. Pathogenic parasites include, for example, Entamoeba histolytica, Balantidium coli, Naegleria fowleri, Acanthamoeba sp., Giardialambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosomabrucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondii and Nippostrongylus brasiliensis.


In some embodiments, the neurodegenerative diseases are one or more selected from the group consisting of Parkinson's disease, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis, Creutzfeldt-Jakob disease. Preferably, the neurodegenerative diseases are one or more selected from the group consisting of Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis; more preferably, the neurodegenerative diseases are one or more selected from consisting of Parkinson's disease and Huntington's disease.


In some embodiments, the tumors are one or more selected from the group consisting of leukemia, lymphoma, myeloma, brain tumor, head and neck squamous cell carcinoma, non-small cell lung cancer, small cell lung cancer, nasopharyngeal cancer, esophageal cancer, gastric cancer, pancreatic cancer, gallbladder cancer, liver cancer, colorectal cancer, breast cancer, ovarian cancer, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, bladder cancer, urothelial cancer, renal cell carcinoma, osteosarcoma, melanoma, and merkel cell carcinoma; preferably, the tumors are one or more selected from the group consisting of lymphoma, myeloma, head and neck squamous cell carcinoma, non-small cell lung cancer, small cell lung cancer, nasopharyngeal cancer, esophageal cancer, gastric cancer, liver cancer, colorectal cancer, breast cancer, cervical cancer, endometrial cancer, prostate cancer, urothelial cancer, renal cell carcinoma, osteosarcoma, melanoma, and merkel cell carcinoma; more preferably, the tumors are one or more selected from the group consisting of lymphoma, head and neck squamous cell carcinoma, non-small cell lung cancer, gastric cancer, liver cancer, colorectal cancer, cervical cancer, urothelial carcinoma, renal cell carcinoma, and melanoma, and merkel cell carcinoma.


In some embodiments, the subject is selected from mammals, including but not limited to human and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice and/or rats.


In some embodiments, the anti-human PD-L1 antibody, antigen-binding fragment or variant thereof, the nucleic acid molecule, the vector or the host cell of the invention is utilized by conventional administration methods in the art, such as parenteral route, such as intravenous injection.


The anti-PD-L1 monoclonal antibodies of the invention has strong specificity, good stability, strong T cell function regulation activity and good pharmacokinetic properties, and can significantly inhibit tumor growth in the body. Moreover, the anti-PD-L1 monoclonal antibodies of the invention has an activity of blocking the binding to PD-L1/PD-1 stronger than that of Atezolizumab (see FIG. 1B).





BRIEF DESCRIPTION OF THE FIGURES

To illustrate the embodiments of the invention or the technical solutions in the prior art more clearly, the following will briefly introduce the accompanying drawings that need to be used in the embodiments or the prior art. Apparently, the accompanying drawings in the following description show some embodiments of the invention, and for those skilled in the art, they can obtain other accompanying drawings based on these accompanying drawings without any creative work.



FIG. 1 shows the in vitro activity of the anti-human PD-L1 murine monoclonal antibodies secreted by the clone 34-35 in Example 1 of the invention, wherein panel A shows the binding activity of the murine monoclonal antibodies to PD-L1; panel B shows the activity of the murine monoclonal antibodies of blocking the binding to PD-L1/PD-1.



FIG. 2 shows the binding activity of the chimeric anti-human PD-L1 monoclonal antibody to human PD-L1 in Example 3 of the invention.



FIG. 3 shows the binding specificity of the chimeric anti-human PD-L1 monoclonal antibody in Example 4 of the invention, wherein panel A shows the species specificity of the binding; panel B shows the target binding specificity.



FIG. 4 shows the activity of the chimeric anti-human PD-L1 monoclonal antibody of blocking the binding to PD-L1/PD-1 in Example 5 of the invention.



FIG. 5 shows the T cell function regulation activity of the chimeric anti-human PD-L1 monoclonal antibody in Example 6 of the invention.



FIG. 6 shows the drug-time curve of the humanized anti-human PD-L1 monoclonal antibody after a single intraperitoneal injection into mice of Example 9 of the invention.



FIG. 7 shows the in vivo anti-tumor efficacy of the humanized anti-human PD-L1 monoclonal antibody of Example 10 of the invention.





DETAILED DESCRIPTION OF THE EMBODIMENTS
Definitions

The term human “PD-L1” refers to programmed death ligand-1, also known as CD274 and B 7H1, and refers to any native PD-L1 from any vertebrate source, including mammals such as primates (eg, humans) and rodents (eg, mice and rats).


As used herein, the terms “anti-PD-L1 antibody”, “anti-PD-L1”, “PD-L1 antibody” or “antibody binding to PD-L1” refer to an antibody which is capable of binding to the PD-L1 protein or a fragment thereof with sufficient affinity. In some embodiments, the anti-PD-L1 antibody binds to an epitope of PD-L1 that is conserved among PD-L1s from different species.


The term “antibody” refers to an immunoglobulin molecule or a fragment of an immunoglobulin molecule which can bind to an epitope of an antigen. Naturally occurring antibodies typically comprise a tetramer which is usually composed of at least two heavy (H) chains and at least two light (L) chains. Immunoglobulins include the following isotypes: IgG, IgA, IgM, IgD, and IgE, with their corresponding heavy chains being the γ, α, β, δ, and ε chains, respectively. The Igs of the same type can be divided into different subtypes according to the amino acid compositions of the hinge regions and the number and positions of the heavy chain disulfide bonds. For example, IgG can be divided into IgG1, IgG2, IgG3, and IgG4 subtypes, and IgA can be divided into IgA1 and IgA2 subtypes. Light chains can be divided into κ and λ chains according to the constant regions.


Herein, the term “antibody” is used in the broadest sense thereof, and refers to a protein comprising an antigen-binding site, encompassing natural and artificial antibodies of various structures, including but not limited to intact antibodies and antigen-binding fragments of antibodies.


A “variable region” or “variable domain” is a domain of a heavy or light chain of an antibody involving in the binding of the antibody to its antigen. Each heavy chain of an antibody consists of a heavy chain variable region (herein referred to as VH) and a heavy chain constant region (herein referred to as CH), wherein the heavy chain constant region usually consists of 3 domains (CH1, CH2 and CH3). Each light chain consists of a light chain variable region (herein referred to as VL) and a light chain constant region (herein referred to as CL). The variable regions of the heavy and light chains are typically responsible for antigen recognition, while the constant domains of the heavy and light chains can mediate the binding of the immunoglobulin with host tissues or factors (including various cells of the immune system (e.g., effector cells), Fc receptors and the first component of the classical complement system (C1q)). The heavy and light chain variable regions comprise binding regions interacting with an antigen. The VH and VL regions can be further subdivided into hypervariable regions (HVRs) called “complementarity determining regions (CDRs)”, which are interspersed with more conserved regions called “framework regions (FRs)”. Each VH or VL consists of three CDR domains and four FR domains, arranged from the amino-terminus to the carboxy-terminus in the following order: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4.


The terms “complementarity determining region” or “CDR region” or “CDR” (used interchangeably herein with the hypervariable region “HVR”) refer to a region in the antibody variable domains which is hypervariable in sequence and forms a loop structurally determined (“a hypervariable loop”) and/or comprises antigen-contacting residues (“antigen-contacting points”). The CDRs are primarily responsible for binding to an epitope. Herein, the three CDRs of the heavy chain are referred to as HCDR1, HCDR2 and HCDR3, and the three CDRs of the light chain are referred to as LCDR1, LCDR2 and LCDR3.


It should be noted that the boundaries of the CDRs of the variable regions of the same antibody obtained based on different numbering systems (such as IMGT®, Kabat, or Chothia) may be different from each other. That is, the CDR sequences of the variable regions of the same antibody defined under different numbering systems are somewhat different from each other. Thus, when defining an antibody with the particular CDR sequences as defined in the invention, the scope of said antibody also covers antibodies of which the variable region sequences comprise said particular CDR sequences, but due to the different numbering system(s) used (such as different numbering system rules or combinations thereof), the CDR boundaries claimed by them might be different from the specific CDR boundaries defined in the invention.


The terms “mAbs”, “monoclonal antibodies “or “monoclonal antibody composition” refer to a preparation of antibody molecules of single molecular composition, and refer to antibodies obtained from a substantially homogeneous population of antibodies, i.e., the population comprising individual antibodies are identical except for the naturally occurring mutations that may be present in minor amounts. A conventional monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. In certain embodiments, a monoclonal antibody can be composed of more than one Fab domain thereby increasing the specificity to more than one target. The term “monoclonal antibody” or “monoclonal antibody composition” is not limited by any particular method of production (eg, recombinant, transgenic, hybridoma, etc.).


The terms “double antibody”, “bifunctional antibody”, “bispecific antibody” or “BsAb” refers to an antibody which has two different antigen binding sites, so that which can simultaneously bind to two target antigens, alternatively which plays a role in mediating another special function simultaneously besides the targeting effect of the antibody. The special function effector molecules mediated can also be toxins, enzymes, cytokines, radionuclides, etc. The two arms of the bispecific antibody binding to an antigen can be from Fab, Fv, ScFv or dSFv, etc.


The term “polyclonal antibody” refers to a preparation comprising different antibodies raised against different determinants (“epitopes”).


The term “antigen-binding fragment of antibody” refers to a fragment, a part, a region or a domain of an antibody capable of binding to an epitope (e.g., obtainable by cleavage, recombination, synthesis, etc.). An antigen-binding fragment may comprise 1, 2, 3, 4, 5 or all 6 CDR domains of such antibodies and, while capable of binding to the epitope, it may exhibit different specificities, affinities or selectivities. Preferably, an antigen-binding fragment comprises all 6 CDR domains of said antibody. An antigen-binding fragment of an antibody may be a part of a single polypeptide chain (e.g., scFv) or comprise a single polypeptide chain, or may be a part of two or more polypeptide chains (each having an amino- and carboxy-terminus) (e.g., a bispecific antibody, a Fab fragment, a F (ab′)2 fragment, etc.) or comprise two or more polypeptide chains.


Examples of an antigen-binding fragment of the invention include (a) a Fab′ or Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (b) F(ab′)2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bond at the hinge domain; (c) an Fd fragment consisting of the VH and CH1 domains; (d) a Fv fragment consisting of VL and VH domains of a single arm of an antibody; (e) a single chain Fv (scFv), a recombinant protein formed by linking VH and VL domains of an antibody with a peptide linker by genetic engineering; (f) a dAb fragment (Ward et al., Nature, 341, 544-546 (1989)), which consists essentially of a VH domain and also called domain antibodies (Holt et al., Trends Biotechnol., 2i(11): 484-90); (g) camelid or nanobodies (Revets et al., Expert Opin Biol Ther., 5(1): 111-24) and (h) an isolated complementarity determining region (CDR).


The term “chimeric antibody” means that a portion of the heavy and/or light chains is identical or homologous to the corresponding sequences of an antibody from a particular species or belonging to a particular antibody class or subclass, while the rest of the chains is identical or homologous to the corresponding sequences of an antibody from another species or belonging to another antibody class or subclass, providing that they exhibit the desired biological activity. The invention provides the variable region antigen binding sequences from human antibodies. Thus, chimeric antibodies primarily focused herein include antibodies which comprise one or more human antigen-binding sequences (e.g., CDRs) and comprise one or more sequences from non-human antibodies, such as FR or C region sequences. Furthermore, chimeric antibodies described herein are antibodies comprising human variable region antigen-binding sequence from one antibody class or subclass and other sequences from another antibody class or subclass, such as FR or C region sequences.


The term “humanized antibody” refers to an antibody in which CDR sequences from the germline of another mammalian species, such as a mouse, have been grafted among the human framework region sequences. Additional framework region modifications can be made within the human framework region sequences.


The term “human antibody” or “fully human antibody” (“humAb “or “HuMab”) refers to an antibody comprising variable and constant domains derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or during gene rearrangement or by somatic mutation in vivo).


Variant antibodies are also included within the scope of the invention. Accordingly, variants of the sequences recited in the invention are also included within the scope of the invention. Other variants of antibody sequences with improved affinity can be obtained by using methods known in the art and these variants are also included within the scope of the invention. For example, amino acid substitutions can be used to obtain antibodies with further improved affinity. Alternatively, codon optimization of nucleotide sequences can be used to improve the translation efficiency of expression systems in antibody production.


Such variant antibody sequences have 70% or more (e.g., 80%, 85%, 90%, 95%, 97%, 98%, 99% or more) sequence homology to the sequences recited in the invention. Such a sequence homology is obtained by calculation relative to the full length of the reference sequence (i.e., the sequence recited in the invention).


The numbering of amino acid residues in the invention is according to IMGT®, the international ImMunoGeneTics Information System® or Kabat, E. A., Wu, T. T., Perry, H. M., Gottesmann, K. S. & Foeller, C. (1991). Sequences of Proteins of Immunological Interest, 5th edit., NIH Publication no. 91-3242 U.S. Department of Health and Human Services; Chothia, C. & Lesk, A. M. (1987). Canonical structures For The Hypervariable domains Of Immunoglobulins. J. Mol. Biol. 196, 901-917. Unless otherwise specified, the numbering of amino acid residues in the invention is according to the Kabat numbering system.


An antibody or an antigen-binding fragment thereof is said to “specifically” bind a region of another molecule (i.e., an epitope) if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity or avidity with that epitope relative to alternative epitopes. In some embodiments, the antibody or antigen-binding fragment thereof of the invention binds human PD-L1 with an affinity of at least 10−7 M, such as 10−8 M, 10−9 M, 10−0 M, 10−11 M or higher. Preferably, the antibody or antigen-binding fragment thereof binds under physiological conditions (e.g., in vivo). Therefore, “specifically binding to PD-L1” refers to the ability of the antibody or antigen-binding fragment thereof to bind to PD-L1 with the above-mentioned specificity and/or under such conditions. Methods suitable for determining such a binding are known in the art.


The term “binding” in the context of the binding of an antibody to a predetermined antigen typically refers to binding with an affinity corresponding to a KD of about 10−6 M or less, which is at least ten-fold lower, such as at least 100-fold lower, for instance at least 1,000-fold lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen.


As used herein, the term “kd” (sec-1 or 1/s) refers to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred as the koff value.


As used herein, the term “ka” (M-1×sec-1 or 1/Msec) refers to the association rate constant of a particular antibody-antigen interaction.


As used herein, the term “KD” (M) refers to the dissociation equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the kd by the ka.


As used herein, the term “KA” (M-1 or 1/M) refers to the association equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the ka by the kd.


In some embodiments, only part of a CDR, namely the subset of CDR residues required for binding, termed the SDRs, are needed to retain binding in a humanized antibody. CDR residues not contacting the relevant epitope and not in the SDRs can be identified based on previous studies (for example residues H60-H65 in CDR H2 are often not required), from regions of Kabat CDRs lying outside Chothia hypervariable loops (see, Kabat et al. (1992), “Sequences of Proteins of Immunological Interest”, National Institutes of Health Publication No. 91-3242; Chothia, C. et al. (1987), “Canonical Structures For The Hypervariable Regions Of Immunoglobulins”, J. Mol. Biol. 196:901-917), by molecular modeling and/or empirically, or as described in Gonzales, N. R. et al. (2004), “SDR Grafting Of A Murine Antibody Using Multiple Human Germline Templates To Minimize Its Immunogenicity”, Mol. Immunol. 41:863-872. In such humanized antibodies at positions in which one or more donor CDR residues is absent or in which an entire donor CDR is omitted, the amino acid occupying the position can be an amino acid occupying the corresponding position (by Kabat numbering) in the acceptor antibody sequence. The number of such substitutions of acceptor for donor amino acids in the CDRs to include reflects a balance of competing considerations. Such substitutions are potentially advantageous in decreasing the number of mouse amino acids in a humanized antibody and consequently decreasing potential immunogenicity. However, substitutions can also cause changes of affinity, and significant reductions in affinity are preferably avoided. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically.


The fact that a single amino acid alteration of a CDR residue can result in loss of functional binding (Rudikoff, S. et al. (1982), “Single Amino Acid Substitution Altering Antigen-Binding Specificity”, Proc. Natl. Acad. Sci. (USA) 79(6):1979-1983) provides a means for systematically identifying alternative functional CDR sequences. In one preferred method for obtaining such variant CDRs, a polynucleotide encoding the CDR is mutagenized (for example via random mutagenesis or by a site-directed method (e.g., polymerase chain-mediated amplification with primers that encode the mutated locus)) to produce a CDR having a substituted amino acid residue. By comparing the identity of the relevant residue in the original (functional) CDR sequence to the identity of the substituted (non-functional) variant CDR sequence, the BLOSUM62.iij substitution score for that substitution can be identified. The BLOSUM system provides a matrix of amino acid substitutions created by analyzing a database of sequences for trusted alignments (Eddy, S. R. (2004), “Where Did The BLOSUM62 Alignment Score Matrix Come From?”, Nature Biotech. 22(8):1035-1036; Henikoff, J. G. (1992), “Amino acid substitution matrices from protein blocks”, Proc. Natl. Acad. Sci. (USA) 89:10915-10919; Karlin, S. et al. (1990), “Methods For Assessing The Statistical Significance Of Molecular Sequence Features By Using General Scoring Schemes”, Proc. Natl. Acad. Sci. (USA) 87:2264-2268; Altschul, S. F. (1991), “Amino Acid Substitution Matrices From An Information Theoretic Perspective”, J. Mol. Biol. 219, 555-565. Currently, the most advanced BLOSUM database is the BLOSUM62 database (BLOSUM62.iij). Table 1 presents the BLOSUM62.iij substitution scores (the higher the score the more conservative the substitution and thus the more likely the substitution will not affect function). If an epitope-binding fragment comprising the resultant CDR fails to bind to PD-L1, for example, then the BLOSUM62.iij substitution score is deemed to be insufficiently conservative, and a new candidate substitution is selected and produced having a higher substitution score. Thus, for example, if the original residue was glutamate (E), and the non-functional substitute residue was histidine (H), then the BLOSUM62.iij substitution score will be 0, and more conservative changes (such as to aspartate, asparagine, glutamine, or lysine) are preferred.





























TABLE 1






A
R
N
D
C
Q
E
G
H
I
L
K
M
F
P
S
T
W
Y
V



































A
+4
−1
−2
−2
0
−1
−1
0
−2
−1
−1
−1
−1
−2
−1
+1
0
−3
−2
0


R
−1
+5
0
−2
−3
+1
0
−2
0
−3
−2
+2
−1
−3
−2
−1
−1
−3
−2
−3


N
−2
0
+6
+1
−3
0
0
0
+1
−3
−3
0
−2
−3
−2
+1
0
−4
−2
−3


D
−2
−2
+1
+6
−3
0
+2
−1
−1
−3
−4
−1
−3
−3
−1
0
−1
−4
−3
−3


C
0
−3
−3
−3
+9
−3
−4
−3
−3
−1
−1
−3
−1
−2
−3
−1
−1
−2
−2
−1


Q
−1
+1
0
0
−3
+5
+2
−2
0
−3
−2
+1
0
−3
−1
0
−1
−2
−1
−2


E
−1
0
0
+2
−4
+2
+5
−2
0
−3
−3
+1
−2
−3
−1
0
−1
−3
−2
−2


G
0
−2
0
−1
−3
−2
−2
+6
−2
−4
−4
−2
−3
−3
−2
0
−2
−2
−3
−3


H
−2
0
+1
−1
−3
0
0
−2
+8
−3
−3
−1
−2
−1
−2
−1
−2
−2
+2
−3


I
−1
−3
−3
−3
−1
−3
−3
−4
−3
+4
+2
−3
+1
0
−3
−2
−1
−3
−1
+3


L
−1
−2
−3
−4
−1
−2
−3
−4
−3
+2
+4
−2
+2
0
−3
−2
−1
−2
−1
+1


K
−1
+2
0
−1
−3
+1
+1
−2
−1
−3
−2
+5
−1
−3
−1
0
−1
−3
−2
−2


M
−1
−1
−2
−3
−1
0
−2
−3
−2
+1
+2
−1
+5
0
−2
−1
−1
−1
−1
+1


F
−2
−3
−3
−3
−2
−3
−3
−3
−1
0
0
−3
0
+6
−4
−2
−2
+1
+3
−1


P
−1
−2
−2
−1
−3
−1
−1
−2
−2
−3
−3
−1
−2
−4
+7
−1
−1
−4
−3
−2


S
+1
−1
+1
0
−1
0
0
0
−1
−2
−2
0
−1
−2
−1
+4
+1
−3
−2
−2


T
0
−1
0
−1
−1
−1
−1
−2
−2
−1
−1
−1
−1
−2
−1
+1
+5
−2
−2
0


W
−3
−3
−4
−4
−2
−2
−3
−2
−2
−3
−2
−3
−1
+1
−4
−3
−2
+11
+2
−3


Y
−2
−2
−2
−3
−2
−1
−2
−3
+2
−1
−1
−2
−1
+3
−3
−2
−2
+2
+7
−1


V
0
−3
−3
−3
−1
−2
−2
−3
−3
+3
+1
−2
+1
−1
−2
−2
0
−3
−1
+4









The invention thus contemplates the use of random mutagenesis to identify improved CDRs. In the context of the present invention, conservative substitutions may be defined by substitutions within the classes of amino acids reflected in one or more of Tables 2, 3, or 4:


Amino Acid Residue Classes for Conservative Substitutions:










TABLE 2







Acidic Residues
Asp (D) and Glu (E)


Basic Residues
Lys (K), Arg (R), and His (H)


Hydrophilic Uncharged Residues
Ser (S), Thr (T), Asn (N), and Gln (Q)


Aliphatic Uncharged Residues
Cly (G), Ala (A), Val (V), Leu (L),



and Ile (I)


Non-polar Uncharged Residues
Cys (C), Met (M), and Pro (P)


Aromatic Residues
Phe (F), Tyr (Y), and Trp (W)









Alternative Conservative Amino Acid Residue Substitution Classes:














TABLE 3









1
A
S
T



2
D
E



3
N
Q



4
R
K



5
I
L
M



6
F
Y
W










Alternative Physical and Functional Classifications of Amino Acid Residues










TABLE 4







Alcohol Group-Containing Residues
S and T


Aliphatic Residues
I, L, V and M


Cycloalkenyl-Associated Residues
F, H, W and Y


Hydrophobic Residues
A, C, F, G, H, I, L, M, R, T, V,



W and Y


Negatively Charged Residues
D and E


Polar Residues
C, D, E, H, K, N, Q, R, S and T


Positively Charged Residues
H, K and R


Small Residues
A, C, D, G, N, P, S, T and V


Very Small Residues
A, G and S


Residues Involved In Turn Formation
A, C, D, E, G, H, K, N, Q, R, S,



P and T


Flexible Residues
Q, T, K, S, G, P, D, E and R









More conservative substitutions groupings include: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, and asparagine-glutamine.


In some embodiments, the hydrophilic amino acid is selected from the group consisting of Arg, Asn, Asp, Gln, Glu, His, Tyr, and Lys.


Additional groups of amino acids may also be formulated using the principles described in, e.g., Creighton (1984) Proteins: Structure and Molecular Properties (2d Ed. 1993), W. H. Freeman and Company.


Thus, the sequences of the CDR variants comprised in antibodies or antigen-binding fragments thereof may differ from those of the CDRs of the parent antibody by substitutions, such as substitutions of 4, 3, 2 or 1 amino acid residues. According to an embodiment of the invention, amino acid(s) in the CDR regions may be substituted with conservative substitutions, as defined in 3 Tables above.


“Homology” or “sequence identity” refers to the percentage of residues in a variant polynucleotide or polypeptide sequence that are identical to those of the non-variant sequence, after alignment of the sequences and introduction of gaps. In specific embodiments, polynucleotide and polypeptide variants have at least 70%, at least 75%, at least 80%, at least 90%, at least 95%, at least 98%, or at least 99% polynucleotide or polypeptide homology.


Such variant polypeptide sequences have 70% or higher (i.e., 80%, 85%, 90%, 95%, 97%, 98%, 99% or higher) sequence identity to the sequences recited in the invention. In other embodiments, the invention provides polypeptide fragments comprising contiguous stretches of various lengths of the amino acid sequences disclosed herein. For example, where applicable, the peptide sequences provided herein comprise at least about 5, 10, 15, 20, 30, 40, 50, 75, 100, 150 or more, and all intermediate lengths therebetween, contiguous amino acid residues of one or more sequences disclosed herein.


The term “treatment” or “treating” as used herein means ameliorating, slowing, attenuating, or reversing the progress or severity of a disease or disorder, or ameliorating, slowing, attenuating, or reversing one or more symptoms or side effects of such disease or disorder. In this invention, “treatment” or “treating” further means an approach for obtaining beneficial or desired clinical results, where “beneficial or desired clinical results” include, without limitation, alleviation of a symptom, diminishment of the extent of a disorder or disease, stabilized (i.e., not worsening) disease or disorder state, delay or slowing of the progression a disease or disorder state, amelioration or palliation of a disease or disorder state, and remission of a disease or disorder, whether partial or total, detectable or undetectable.


The term “prevention” or “preventing” refers to the administration of antibodies and functional fragments thereof of the invention, thereby preventing or retarding the development of at least one symptom of a disease or condition. The term also includes treating a subject in remission to prevent or hinder relapse.


The antibody of the invention may be of any isotype. The choice of isotype typically will be guided by the desired effector functions, such as ADCC induction. Exemplary isotypes are IgG1, IgG2, IgG3, and IgG4. Either of the human light chain constant domains, κ or λ, may be used. If desired, the class of an anti-PD-L1 antibody of the present invention may be switched by known methods. For example, an antibody of the present invention that was originally IgG may be class switched to an IgM antibody of the present invention. Further, class switching techniques may be used to convert one IgG subclass to another, for instance from IgG1 to IgG2. Thus, the effector function of the antibodies of the present invention may be changed by isotype switching to, e.g., an IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM antibody for various therapeutic uses. In one embodiment an antibody of the present invention is an IgG2 antibody, such as IgG2a. An antibody is said to be of a particular isotype if its amino acid sequence is most homologous to that isotype, relative to other isotypes.


In some embodiments, the antibodies of the invention are full-length antibodies, preferably IgG antibodies. In other embodiments, the antibodies of the invention are antibody antigen-binding fragments or single chain antibodies.


In some embodiments, the anti-PD-L1 antibody is a monovalent antibody, preferably a monovalent antibody as described in WO2007059782 (which is incorporated herein by reference in its entirety) having a deletion of the hinge region. Accordingly, in some embodiments, the antibody is a monovalent antibody, wherein said anti-PD-L1 antibody is constructed by a method comprising: i) providing a nucleic acid construct encoding the light chain of said monovalent antibody, said construct comprising a nucleotide sequence encoding the VL region of a selected antigen specific anti-PD-L1 antibody and a nucleotide sequence encoding the constant CL region of an Ig, wherein said nucleotide sequence encoding the VL region of a selected antigen specific antibody and said nucleotide sequence encoding the CL region of an Ig are operably linked together, and wherein, in case of an IgG1 subtype, the nucleotide sequence encoding the CL region has been modified such that the CL region does not contain any amino acids capable of forming disulfide bonds or covalent bonds with other peptides comprising an identical amino acid sequence of the CL region in the presence of polyclonal human IgG or when administered to an animal or human being; ii) providing a nucleic acid construct encoding the heavy chain of said monovalent antibody, said construct comprising a nucleotide sequence encoding the VH region of a selected antigen specific antibody and a nucleotide sequence encoding a constant CH region of a human Ig, wherein the nucleotide sequence encoding the CH region has been modified such that the region corresponding to the hinge region and, as required by the Ig subtype, other regions of the CH region, such as the CH3 region, does not comprise any amino acid residues which participate in the formation of disulphide bonds or covalent or stable non-covalent inter-heavy chain bonds with other peptides comprising an identical amino acid sequence of the CH region of the human Ig in the presence of polyclonal human IgG or when administered to an animal human being, wherein said nucleotide sequence encoding the VH region of a selected antigen specific antibody and said nucleotide sequence encoding the CH region of said Ig are operably linked together; iii) providing a cell expression system for producing said monovalent antibody; iv) producing said monovalent antibody by co-expressing the nucleic acid constructs of (i) and (ii) in cells of the cell expression system of (iii).


Similarly, in some embodiments, the anti-PD-L1 antibody of the invention is a monovalent antibody, which comprises:

    • (i) a variable domain of an antibody of the invention as described herein or an epitope-binding part of the said domain, and
    • (ii) a CH domain of an immunoglobulin or a domain thereof comprising the CH2 and CH3 domains, wherein the CH domain or domain thereof has been modified such that the domain corresponding to the hinge domain and, if the immunoglobulin is not an IgG4 subtype, other domains of the CH domain, such as the CH3 domain, do not comprise any amino acid residues, which are capable of forming disulfide bonds with an identical CH domain or other covalent or stable non-covalent inter-heavy chain bonds with an identical CH domain in the presence of polyclonal human IgG.


In some other embodiments, the heavy chain of the monovalent antibody of the invention has been modified such that the entire hinge region has been deleted.


In other embodiments, the sequence of the monovalent antibody has been modified so that it does not comprise any acceptor sites for N-linked glycosylation.


The invention also includes “Bispecific Antibodies,” wherein an anti-PD-L1 binding region (e.g., a PD-L1-binding region of an anti-PD-L1 monoclonal antibody) is part of a bivalent or polyvalent bispecific scaffold that targets more than one epitope, (for example a second epitope could comprise an epitope of an active transport receptor, such that the bispecific antibody would exhibit improved transcytosis across a biological barrier, such as the Blood Brain Barrier, or the second epitope is an epitope of another target protein). Thus, in other embodiments, the monovalent Fab of an anti-PD-L1 antibody may be joined to an additional Fab or scfv that targets a different protein to generate a bispecific antibody. A bispecific antibody can have a dual function, for example a therapeutic function imparted by an anti-PD-L1 binding domain and a transport function that can bind to a receptor molecule to enhance transfer cross a biological barrier, such as the blood brain barrier.


Antibodies and epitope-binding fragments thereof of the invention also include single chain antibodies. Single chain antibodies are peptides in which the heavy and light chain Fv domains are connected. In some embodiment, the present invention provides a single-chain Fv (scFv) wherein the heavy and light chains in the Fv of an anti-PD-L1 antibody of the present invention are joined with a flexible peptide linker (typically of about 10, 12, 15 or more amino acid residues) in a single peptide chain. Methods of producing such antibodies are described in for instance U.S. Pat. No. 4,946,778, Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds. Springer-Verlag, New York, pp. 269-315 (1994), Bird et al., Science 242, 423-426 (1988), Huston et al., PNAS USA 85, 5879-5883 (1988) and McCafferty et al., Nature 348, 552-554 (1990). The single chain antibody may be monovalent, if only a single VH and VL are used, bivalent, if two VH and VL are used, or polyvalent, if more than two VH and VL are used.


Antibodies of the invention are produced by any technique known in the art, such as, but not limited to, any chemical, biological, genetic, or enzymatic technique, alone or in combination. In general, if knowing the amino acid sequence of a desired sequence, one skilled in the art can readily generate such an antibody by standard techniques used to generate polypeptides. For example, these antibodies can be synthesized using well-known solid-phase methods, preferably using a commercially available peptide synthesis apparatus (such as that manufactured by Applied Biosystems, Foster City, California) and following the manufacturer's instructions. Alternatively, antibodies of the invention can be synthesized by recombinant DNA techniques well known in the art. For example, after incorporating a DNA sequence encoding an antibody into an expression vector and introducing these vectors into eukaryotic or prokaryotic host cells suitable for expressing the desired antibodies, antibodies as DNA expression products can be obtained, which can then be isolated from the host cells using known techniques.


The antibodies and epitope-binding fragments thereof described herein may be modified by inclusion of any suitable number of modified amino acids and/or associations with such conjugated substituents. Suitability in this context is generally determined by the ability to retain the PD-L1 selectivity and/or the PD-L1 specificity at least substantially associated with the non-derivatized parent anti-PD-L1 antibody. The inclusion of one or more modified amino acids may be advantageous in, for example, increasing polypeptide serum half-life, reducing polypeptide antigenicity, or increasing polypeptide storage stability. Amino acid(s) are modified, for example, co-translationally or post-translationally during recombinant production (e.g., N-linked glycosylation at N-X-S/T motifs during expression in mammalian cells) or modified by synthetic means. Non-limiting examples of a modified amino acid include a glycosylated amino acid, a sulfated amino acid, a prenylated (e.g., farnesylated, geranyl-geranylated) amino acid, an acetylated amino acid, an acylated amino acid, a PEGylated amino acid, a biotinylated amino acid, a carboxylated amino acid, a phosphorylated amino acid, and the like. References adequate to guide one of skill in the modification of amino acids are replete throughout the literature, see e.g., Walker (1998) Protein Protocols On CD-Rom, Humana Press, Totowa, NJ. The modified amino acid may, for instance, be selected from a glycosylated amino acid, a PEGylated amino acid, a farnesylated amino acid, an acetylated amino acid, a biotinylated amino acid, an amino acid conjugated to a lipid moiety, or an amino acid conjugated to an organic derivatizing agent.


The antibodies and antigen-binding fragments thereof of the invention may also be chemically modified by covalent conjugation to a polymer to for instance increase their circulating half-life. Exemplary polymers and methods to attach them to peptides are illustrated in for instance U.S. Pat. Nos. 4,766,106; 4,179,337; 4,495,285 and 4,609,546. Additional illustrative polymers include polyoxyethylated polyols and polyethylene glycol (PEG) (e.g., a PEG with a molecular weight of between about 1,000 and about 40,000 D, such as between about 2,000 and about 20,000 D, e.g., about 3,000-12,000 D).


The term “subject” refers to a warm-blooded animal, preferably a mammal (including humans, domestic and farm animals, zoo animals, sport animals, or pet animals such as dogs, cats, cows, horses, sheep, pigs, goats, rabbits, etc.), more preferably humans. In one embodiment, a subject may be a “patient”, i.e., a warm-blooded animal, more preferably a human, who is waiting to receive or is receiving medical care or will be a subject of a medical procedure, or monitoring for the development of a disease. In one embodiment, the subject is an adult (e.g., a subject over 18 years old). In another embodiment, the subject is a child (e.g., a subject under 18 years old). In one embodiment, the subject is a male. In another embodiment, the subject is a female.


In one embodiment of the invention, a sample is a biological sample. Examples of biological samples include, but are not limited to, tissue lysates and extracts prepared from diseased tissues, body fluids, preferably blood, more preferably serum, plasma, synovial fluid, bronchoalveolar lavage fluid, sputum, lymph fluid, ascites, urine, amniotic fluid, peritoneal fluid, cerebrospinal fluid, pleural fluid, pericardial fluid, and alveolar macrophages.


In one embodiment of the invention, the term “sample” is intended to mean a sample taken from an individual prior to any analysis.


Accordingly, in some embodiments, the anti-PD-L1 antibodies and antigen-binding fragments thereof of the invention include intact antibodies, such as IgG (IgG1, IgG2, IgG3 and IgG4 subtypes), IgA (IgA1 and IgA2 subtypes), IgD, IgM and IgE; antigen-binding fragments such as SDR, CDR, Fv, dAb, Fab, Fab2, Fab′, F(ab′)2, Fd, scFv, camelid antibodies or nanobodies; variant sequences of antibodies or antigen-binding fragments thereof, such as variant sequences having at least 80% sequence identity to the above antibodies or antigen-binding fragments thereof. In some embodiments, the invention also includes derivatives comprising the anti-PD-L1 antibody or antigen-binding fragment thereof, such as chimeric antibodies, humanized antibodies, fully human antibodies, recombinant antibodies, and bispecific antibodies derived from an intact antibody.


In a further aspect, the invention relates to an expression vector comprising nucleotide sequences encoding one or more polypeptide chains of the antibody or antigen-binding fragment thereof of the invention. Such expression vectors can be used to recombinantly produce the antibodies and antigen-binding fragments thereof of the invention.


An expression vector in the context of the present invention may be any suitable DNA or RNA vector, including chromosomal, non-chromosomal, and synthetic nucleic acid vectors (a nucleic acid sequence comprising a suitable set of expression control elements). Examples of such vectors include derivatives of SV40, bacterial plasmids, phage DNA, baculovirus, yeast plasmids, vectors derived from combinations of plasmids and phage DNA, and viral nucleic acid (RNA or DNA) vectors. In some embodiments, an anti-PD-L1 antibody-encoding nucleic acid is comprised in a naked DNA or RNA vector, including, for example, a linear expression element (as described in, for instance, Sykes and Johnston, Nat Biotech 12, 355-59 (1997)), a compacted nucleic acid vector (as described in for instance U.S. Pat. No. 6,077,835 and/or WO 00/70087), a plasmid vector such as pBR322, pUC 19/18, or pUC 118/119, a minimally-sized nucleic acid vector (as described in, for instance, Schakowski et al., MoI Ther 3, 793-800 (2001)), or as a precipitated nucleic acid vector construct, such as a CaPO4-precipitated construct (as described in, for instance, WO 00/46147, Benvenisty and Reshef, PNAS USA 83, 9551-55 (1986), Wigler et al., Cell 14, 725 (1978), and Coraro and Pearson, Somatic Cell Genetics 2, 603 (1981)). Such nucleic acid vectors and the usage thereof are well known in the art (see for instance U.S. Pat. Nos. 5,589,466 and 5,973,972). In some embodiments, the expression vector is X0GC (from the PCT publication WO2008/048037), pCDNA3.1 (ThermoFisher, catalog number V79520), pCHO1.0 (ThermoFisher, catalog number R80007).


In some embodiments, the vector is suitable for expression of anti-PD-L1 antibodies or epitope-binding fragments thereof of the invention in a bacterial cell. Examples of such vectors include expression vectors such as BlueScript (Stratagene), pIN vectors (Van Heeke & Schuster, J. Biol. Chem. 264, 5503-5509 (1989), pET vectors (Novagen, Madison, Wisconsin), and the like.


An expression vector may also be a vector suitable for expression in a yeast system. Any vector suitable for expression in a yeast system may be employed. Suitable vectors include, for example, vectors comprising constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH (reviewed in: F. Ausubel et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley InterScience New York (1987), Grant et al., Methods in Enzymol 153, 516-544 (1987), Mattanovich, D. et al. Methods Mol. Biol. 824, 329-358 (2012), Celik, E. et al. Biotechnol. Adv. 30(5), 1108-1118 (2012), Li, P. et al. Appl. Biochem. Biotechnol. 142(2), 105-124 (2007), Boer, E. et al. Appl. Microbiol. Biotechnol. 77(3), 513-523 (2007), van der Vaart, J. M. Methods Mol. Biol. 178, 359-366 (2002), and Holliger, P. Methods Mol. Biol. 178, 349-357 (2002)).


In an expression vector of the invention, the anti-PD-L1 antibody-encoding nucleic acids may comprise or be associated with any suitable promoter, enhancer, and other expression-facilitating elements. Examples of such elements include strong expression promoters (e.g., human CMV IE promoter/enhancer as well as RSV, SV40, SL3-3, MMTV, and HIV LTR promoters), effective poly (A) termination sequences, an origin of replication for plasmid product in E. coli, an antibiotic resistance gene as selectable marker, and/or a convenient cloning site (e.g., a polylinker). Nucleic acids may also comprise an inducible promoter as opposed to a constitutive promoter such as CMV IE (the skilled artisan will recognize that such terms are actual descriptors of a degree of gene expression under certain conditions).


In an even further aspect, the invention relates to a recombinant eukaryotic or prokaryotic host cell, such as a transfectoma, which produces an antibody or epitope-binding fragment thereof of the invention as defined herein or a bispecific molecule of the invention as defined herein. Examples of host cells include yeast, bacteria, and mammalian cells, such as CHO or HEK cells. For example, in some embodiments, the present invention provides a cell comprising a nucleic acid stably integrated into the cellular genome that comprises a sequence coding for expression of an anti-PD-L1 antibody of the present invention or an epitope-binding fragment thereof. In other embodiments, the present invention provides a cell comprising a non-integrated nucleic acid, such as a plasmid, cosmid, phagemid, or linear expression element, which comprises a sequence coding for expression of an anti-PD-L1 antibody or epitope-binding fragment thereof of the invention.


Antibodies and antigen-binding fragments thereof of the invention can be produced in different cell lines, such as human cell lines, non-human mammalian cell lines, and insect cell lines, such as CHO cell lines, HEK cell lines, BHK-21 cell lines, murine cell lines (such as myeloma cell line), fibrosarcoma cell line, PER.C6 cell line, HKB-11 cell line, CAP cell line, and HuH-7 human cell line (Dumont et al., 2015, Crit Rev Biotechnol., September 18, 1-13., the contents of which are incorporated herein by reference).


The antibodies of the invention are suitably separated from the culture medium by conventional immunoglobulin purification methods, such as Protein A-Sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.


The present invention further relates to a composition comprising, consisting of, or consisting essentially of the antibodies of the invention.


As used herein, the term “consisting essentially of” with respect to a composition means that at least one antibody of the invention as described above is the only biologically active therapeutic agent or reagent in the composition.


In one embodiment, the composition of the invention is a pharmaceutical composition and further comprises a pharmaceutically acceptable excipient, diluent, or carrier.


The term “pharmaceutically acceptable carrier” refers to an excipient that does not produce adverse, allergic, or other adverse reactions when administered to animals, preferably humans, including any, and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. For human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by regulatory agencies (e.g., FDA or EMA).


The invention further relates to a medicament comprising, consisting of, or consisting essentially of the antibodies of the invention.


In some embodiments, the glycosylation of the antibodies of the invention is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for an antigen or change the ADCC activity of the antibody. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for an antigen. Such an approach is described in further detail in U.S. Pat. Nos. 5,714,350 and 6,350,861 by Co et al. Additionally, or alternatively, an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated or an afucosylated antibody having reduced amounts of fucosyl residues or having no fucosyl residues, or an antibody having increased bisecting GlcNac structures. Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. For example, EP 1,176,195 by Hang et al. describes a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation. PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Lecl3 cells, with reduced ability to attach fucose to Asn (297)-linked carbohydrates, also resulting in hypofocosylation of antibodies expressed in that host cell (see also Shields, R. L. et al., (2002), J. Biol Chem. 277: 26733-26740). PCT Publication WO 99/54342 by Umana et al., describes cell lines engineered to express glycoprotein-modifying glycosyl transferases (e.g., beta(1,4)-N-acetylglucosammyltransferase III (GnTIII)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (see also Umana et al., (1999), Nat. Biotech. 17:176-180). Eureka Therapeutics further describes genetically engineered CHO mammalian cells capable of producing antibodies with altered mammalian glycosylation patterns lacking fucosyl residues (http://www.eurekainc.com/a&boutus/companyoverview.html). Alternatively, the human antibodies (preferably monoclonal antibodies) of the invention can be produced in yeast or filamentous fungi which are used in the production of antibodies having mammalian-like glycosylation patterns and capable of producing a glycosylation model lacking fucose (see e.g., EP1297172B1).


The antibody of the present invention acts on PD-L1, is involved in the activation and regulation of T cells, and can regulate the strength and duration of an immune response. Under normal conditions, PD-L1 can mediate and maintain the autoimmune tolerance of body tissues, prevent the excessive activation of the immune system from damaging self-tissues during inflammatory responses, and play a positive role in avoiding the occurrence of autoimmune diseases; under pathological conditions, it is involved in tumor immunity and the occurrence and development of various autoimmune diseases. Clinical evidence shows that PD-L1 is highly expressed on the surface of a variety of tumor cells, including melanoma, lung cancer, kidney cancer, breast cancer, ovarian cancer, cervical cancer, bladder cancer, esophageal cancer, gastric cancer, pancreatic cancer, intestinal cancer, etc. Tumor cells bind to PD-1 or CD80 on T cells through highly expressed PD-L1, and transmit immunosuppressive signals, causing the body to develop immune tolerance to tumor cells and thereby, facilitating the growth and metastasis of tumor cells. High expression of PD-L1 is closely related to poor prognosis and drug resistance of tumor patients. The PD-L1 antibody of the invention enhances the anti-tumor immune response by blocking the PD-L1/PD-1 and PD-L1/CD80 signaling pathways, thereby exerting a broad-spectrum anti-tumor effect, including non-small cell lung cancer, urothelial carcinoma, Merkel cell carcinoma, melanoma, renal cell carcinoma, lymphoma, head and neck cancer, colorectal cancer, liver cancer, gastric cancer, etc.


Where a range of values is provided, it is understood that unless the context clearly dictates otherwise, each intervening value, to the tenth of the unit of the lower limit, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both the limits, ranges excluding either or both of those included limits are also included in the invention.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference.


Embodiments of the invention will be illustrated in detail by following examples, but those skilled in the art will understand that the following examples are only for illustrating the invention, and should not be construed as limiting the scope of the invention. On the one hand, the examples provided by the invention are used to describe the preparation processes of the antibodies of the invention. The preparation processes are only to illustrate the relevant methods and is not limiting. It is well known to those skilled in the art that the invention can be modified without departing from the spirit of the invention, and such modifications also fall within the scope of the invention. On the other hand, the invention provides examples to illustrate the characteristics and advantages of the antibodies of the invention, but the invention is not limited to these characteristics and advantages.


The following experimental methods are all conventional methods unless otherwise specified. Those not indicating the specific conditions in the examples are carried out according to the conventional conditions or the conditions suggested by the manufacturer. The reagents or instruments used of which the manufacturers are not specified are all conventional products that could be commercially purchased. The various antibodies used in the following examples of the present invention are all standard antibodies commercially available.


EXAMPLES
Example 1 Preparation of the Murine Anti-Human PD-L1 Monoclonal Antibodies
1.1. Animal Immunizations

The 6-8-week-old female Balb/c mice (Beijing HFK Bioscience Co., Ltd) were selected as the experimental animals. After the mice acclimated to the environment for one week, the immunization began. For the first immunization, 50 μg of the recombinant human PD-L1-Fc protein (prepared by Beijing Hanmi Pharmaceutical Co., LTD., with the amino acid sequence of human PD-L1 from genebank #: NP_001254635.1) and complete Freund's adjuvant (Sigma-Aldrich, catalog number F5881) were thoroughly mixed to form an emulsion and injected intraperitoneally into the mice. Two weeks later, a booster immunization was given. For the booster immunization, 25 μg of the recombinant human PD-L1-Fc protein and incomplete Freund's adjuvant (Sigma-Aldrich, catalog number F5806) were thoroughly mixed to form an emulsion and injected intraperitoneally into the mice. The immunization was boosted with the same process every 2 weeks, a total of 3 times. On day 10 after the last immunization, blood was collected from the posterior orbital venous plexus of the mice, centrifuged to isolate serum, and the antibody titer was determined with ELISA. Mice with high titers were selected for fusion to make hybridomas. Three days before fusion, 50 μg of the recombinant human PD-L1-Fc protein was injected intraperitoneally without an adjuvant. On the day of fusion, the spleen was taken aseptically, prepared into individual splenocytes suspension for fusion.


1.2. Preparation of Hybridomas

The myeloma cells SP2/0 grew to logarithmic phase, centrifuged at 1000 rpm for 5 min, the supernatant was discarded, the cells were suspended with incomplete DMEM culture medium (Gibco, catalog number 11965) and counted, the required number of cells were taken, washed twice with the incomplete culture medium above. At the same time, the immunized spleen cell suspension was prepared and washed twice with the incomplete culture medium. The myeloma cells and splenocytes were mixed together at a ratio of 1:10 or 1:5, washed once with the incomplete culture medium in a 50 ml plastic centrifuge tube, centrifuged at 1200 rpm for 8 min. The supernatant was discarded and the residual liquid was removed with a dropper and tapping the bottom of the centrifuge tubes with your hand to loosen the precipitated cells evenly; preheated in a 40° C. water bath. 1 ml of 45% PEG-4000 (pH 8.0, Sigma, catalog number P7181) preheated to 40° C. were added with a 1 ml pipette within about 1 minute (the best time is 45 seconds), stirring gently while adding (stirring with a pipette), and there should be visible particles when observed with the naked eye. 20-30 ml of the incomplete medium preheated to 37° C. were added with a 10 ml pipette within 90 seconds to terminate the action of PEG, let stand at 20-37° C. for 10 minutes, centrifuged at 1000 rpm for 5 minutes, the supernatant was discarded. 5 ml of HAT medium (DMEM+HAT, Sigma, catalog number: H0262-10VL) was added, the pelleted cells were gently blew and aspirated (note: not to blow and aspirate hard, so as not to disperse the fused cells), to suspend and mix the cells well, and then HAT medium were supplemented to 80-100 ml (to make the concentration of the splenocytes to 1˜2×106/ml). The cells were aliquoted into a 96-well cell culture plate, 0.1 ml per well; were aliquoted into a 24-well plate, 1.0-1.5 ml per well, and the plates were cultured at 37° C. in a 6% CO2 incubator. Totally six 96-well cell culture plates were plated. After 5 days, 1/2 medium was replaced with HAT medium. After 7˜10 days, HAT medium was replaced with HT medium (DMEM+HT, Sigma, catalog number H0137-10VL). The growth of the hybridomas was observed frequently, and the supernatant was pipetted when the hybridomas grew to more than 1/10 of the area of the bottom of the well for antibody detection. The cells of the positive clones were expanded and freezed.


1.3. Screening and Identification of Clones

ELISA was used to screen the anti-human PD-L1 antibodies in the hybridoma culture supernatants. A 96-well high-adsorption ELISA plate (Corning, catalog number 42592) was coated with 1 μg/mL of the recombinant human PD-L1 (Sino Biological Inc.) at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The culture supernatant samples and positive serum control (positive serum from immunized mice (immunized with human PD-L1-Fc protein)) were added, 100 μL/well, incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The horseradish peroxidase-labeled anti-mouse IgG antibody (Abcam, catalog number Ab7068) diluted 1:10000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader. Positive clones capable of secreting the anti-human PD-L1 binding antibodies were selected according to the magnitude of OD450 nm.


ELISA was used to determine whether the anti-human PD-L1 antibodies secreted by the positive clones can block the binding of PD-L1/PD-1. A 96-well high-adsorption ELISA plate was coated with 1 μg/mL of the recombinant human PD-L1-Fc at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The anti-human PD-L1 antibody samples and the positive control Atezolizumab were added, 50 μL/well, the biotin-labelled PD-1-Fc (Beijing Hanmi Pharmaceutical) was added at a concentration of 40 nM (a final concentration of 20 nM), 50 μL/well, incubated at 25° C. for 90 minutes, and then washed 5 times with PBST. The Streptavidin-HRP (BD Pharmingen, catalog number 554066) diluted 1:1000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader. The anti-human PD-L1 antibodies capable of inhibiting the binding of the human PD-L1-Fc/biotin-labelled PD-1-Fc have a neutralizing activity. The positive clones capable of secreting the anti-human PD-L1 neutralizing antibodies were selected according to the magnitude of the blocking capability.


As shown in panel A of FIG. 1, the clone 34-35 has strong human PD-L1 binding activity, and as shown in panel B of FIG. 1, the clone 34-35 also has strong activity of blocking the binding of human PD-L1/PD-1, which is slightly stronger than that of Atezolizumab.


1.4. Determination of the Sequences of the Monoclonal Antibodies

The DNA sequences of the antibodies secreted by the clones that have both antigen-binding activity and antigen-neutralizing activity screened above were determined. Cellular mRNAs were first extracted using the RNAprep Pure kit (Tiangen, DP419). Then the QuantScript RT kit (Tiangen, catalog number KR103) was used to synthetize the first strand of the cDNAs and the reversely transcribed first strand of the cDNAs was used for the subsequent PCT reactions.


The primers used in the PCR reactions are shown in Table 5.









TABLE 5





PCR primers















GAGGTGAAGCTGCAGGAGTCAGGACCTAGCCTGGTG (SEQ ID No. 1)





AGGT(C/G)(A/C)AACTGCAG(C/G)AGTC(A/T)GG (SEQ ID No. 2)





AGGT(C/G)(A/C)AGCTGCAG(C/G)AGTC(A/T)GG (SEQ ID No. 3)





AGGT(C/G)CAGCTGCAG(C/G)AGTC(A/T)GG (SEQ ID No. 4)





CCAGGGGCCAGTGGATAGACAAGCTTGGGTGTCGTTTT (SEQ ID No. 5)





ATAGACAGATGGGGGTGTCGTTTTGGC (SEQ ID No. 6)





CTTGACCAGGCATCCTAGAGTCA (SEQ ID No. 7)





AGGGGCCAGTGGATAGACTGATGG (SEQ ID No. 8)





AGGGACCAAGGGATAGACAGATGG (SEQ ID No. 9)





SARGTNMAGCTGSAGSAGTC (SEQ ID No. 10)





SARGTNMAGCTGSAGSAGTCWGG (SEQ ID No. 11)





GGTGATATCGTGAT(A/G)AC(C/A)CA(G/A)GATGAACTCTC (SEQ ID No. 12)





GGTGATATC(A/T)TG(A/C)TGACCCAA(A/T)CTCCACTCTC (SEQ ID No. 13)





GGTGATATCGT(G/T)CTCAC(C/T)CA(A/G)TCTCCAGCAAT (SEQ ID No. 14)





GGGAAGATGGATCCAGTTGGTGCAGCATCAGC (SEQ ID No. 15)





GGATACAGTTGGTGCAGCATC (SEQ ID No. 16)





GAYATTGTGMTSACMCARWCTMCA (SEQ ID NO. 17)









When using the primers, any upstream primer in the heavy chain variable region primers (VH primers) can be used in combination with any downstream primer; similarly, any upstream primer in the light chain variable region primers (VL primers) can also be used in combination with any downstream primer. The bands of interest obtained by PCR amplification were cloned into a pGEM-T vector. Single clones were picked up for DNA sequencing.


Example 2 Preparation of the Chimeric Anti-Human PD-L1 Monoclonal Antibodies

The sequence encoding the antibody light chain variable region amplified by PCR was set forth in SEQ ID NO: 18, and the sequence encoding the antibody heavy chain variable region was set forth in SEQ ID NO: 19, wherein the amino acid sequences of the three complementarity determining regions LCDR1, LCDR2 and LCDR3 of the light chain are set forth in SEQ ID Nos: 20, 21 and 22 respectively, the amino acid sequences of the three complementary determining regions HCDR1, HCDR2 and HCDR3 of the heavy chain are set forth in SEQ ID Nos: 23, 24 and 25 respectively. The nucleotide sequences encoding the sequences of the variable regions above were cloned into the eukaryotic cell expression vector X0GCs comprising the nucleotide sequences encoding the antibody light chain constant region of the amino acid sequence set forth in SEQ ID NO. 26 and the antibody heavy chain constant region of the amino acid sequence set forth in SEQ ID NO. 27, respectively. The full-length light chain sequence of the antibody was obtained by linking the light chain variable region of the antibody to SEQ ID NO. 26 and the full-length heavy chain sequence of the antibody was obtained by linking the heavy chain variable region of the antibody to SEQ ID NO. 27. The expression vectors containing the heavy chain and light chain of the antibody were then transfected into the ExpiCHO cell line (ExpiCHO™ Cells, Catalog number A29127, invitrogen). The cells were inoculated one day before transfection with an inoculation density of 35*105 cells/mL. On the day of transfection, the cells were diluted with fresh ExpiCHO expression medium (ExpiCHO™ Expression Medium, Catalog number A29100-01, Invitrogen) to a density of 60*105 cells/mL. The plasmids were taken out according to the transfection volume, and the final concentration of the plasmids was 0.5 μg/mL. The plasmids were diluted to 4% of the transfection volume with OptiPRO™ SFM medium (OptiPRO™ SFM, Catalog number 12309-019, invitrogen), and mixed by inversion. ExpiFectamine™ transfection reagent (ExpiFectamine™ CHO Transfection Kit, Catalog number A29129, invitrogen) 6.4 times the amount of plasmids was taken out, and the transfection reagent was diluted to 4% of the transfection volume with OptiPRO™ SFM medium, and mixed well by inversion. The diluted transfection reagent was added into the diluted plasmids, mixed gently, let stand at room temperature for 1-5 min, and were slowly added dropwise to the cells. Then they were placed in an incubator (8% CO2) and incubated at 37° C. on a shaker at 120 rpm for 20 hours. ExpiCHO™ Enhancer (ExpiFectamine™ CHO Transfection Kit, catalog number A29129, invitrogen) 0.006 times the transfection volume and ExpiCHO™ Feed (ExpiCHO™ Feed, catalog number A29101-02, invitrogen) 0.24 times the transfection volume were added dropwise into the cells slowly. The cells were incubated at 120 rpm on a shaker at 32° C. Cell culture supernatants 10 days after transfection were collected by centrifugation.


The expression level was determined by ELISA assay. The precipitates were removed through a 0.2 μm filter membrane before chromatography column purification. This step was carried out at 4° C.


Example 3 Binding Activity and Binding Kinetic Constant of the Chimeric Anti-Human PD-L1 Monoclonal Antibodies to Human PD-L1

The binding activity of the chimeric anti-human PD-L1 mAb to its antigen, human PD-L1, was determined by ELISA. A 96-well high-adsorption ELISA plate was coated with 1 μg/mL of the recombinant human PD-L1 (Sino Biological Inc.) at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The chimeric anti-human PD-L1 antibody sample serially diluted in PBST with 1% BSA and the control Atezolizumab were added, 100 μL/well, incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The horseradish peroxidase-labeled anti-human IgG antibody (Chemicon, catalog number AP309P) diluted 1:10000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader.


As shown in FIG. 2, the chimeric anti-human PD-L1 monoclonal antibody has good binding affinity to human PD-L1, similar to that of Atezolizumab.


Biacore X100 instrument was used to detect the kinetic constant of the chimeric anti-human PD-L1 monoclonal antibody binding to its antigen human PD-L1. This instrument utilizes optical surface plasmon resonance technology to detect the association and dissociation between the molecules coated on the biochip and the molecules to be tested. Association constants and dissociation constants were analyzed and calculated by Biacore X100 evaluation software. The association constants, dissociation constants and dissociation equilibrium constants of the chimeric anti-human PD-L1 antibody were shown in Table 6. The data indicated that, compared to Atezolizumab, the chimeric anti-human PD-L1 monoclonal antibody can bind to PD-L1 faster, with a comparable dissociation rate. Table 6. Binding kinetic constants of the chimeric anti-human PD-L1 mAb to human PD-L1















Test sample
Kon (1/Ms)
Koff (1/s)
KD (M)







Atezolizumab
1.82E+05
3.95E−04
2.17E−09


Chimeric anti-human PD-L1
9.46E+05
5.96E−04
0.63E−09


monoclonal antibody









Example 4 Species Binding Specificity and Target Binding Specificity of the Chimeric Anti-Human PD-L1 Monoclonal Antibody

Species binding specificity of the chimeric anti-human PD-L1 mAb was determined by ELISA. A 96-well high-adsorption ELISA plate was coated with 1 μg/mL of the recombinant human PD-L1, cynomolgus PD-L1, rat PD-L1 and mouse PD-L1 (all purchased from Sino Biological Inc.), at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The chimeric anti-human PD-L1 antibody samples serially diluted in PBST with 1% BSA were added, 100 μL/well, incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The horseradish peroxidase-labeled anti-human IgG antibody (Chemicon, catalog number AP309P) diluted 1:10000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader.


Target binding specificity of the chimeric anti-human PD-L1 mAb was determined by ELISA. A 96-well high-adsorption ELISA plate was coated with 1 μg/mL of the recombinant human PD1, CD28, CTLA4, ICOS, BTLA, PD-L1, PD-L2, CD80, CD86, B7-H2 (all purchased from Sino Biological Inc.), at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The chimeric anti-human PD-L1 mAb samples serially diluted in PBST with 1% BSA and the control were added, 100 μL/well, incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The horseradish peroxidase-labeled anti-human IgG antibody (Chemicon, catalog number AP309P) diluted 1:10000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader.


As shown in FIG. 3 (panel A), the chimeric anti-human PD-L1 monoclonal antibody can bind to human PD-L1 and cynomolgus PD-L1 with similar affinities, but do not bind to rat and mouse PD-L1 and has species binding specificity. At the same time, as shown in panel B of FIG. 3, the chimeric anti-human PD-L1 mAb also has very strong target binding specificity, only binding to PD-L1, not to other members of B7 family, and CD28 family members.


Example 5 Activity of the Chimeric Anti-Human PD-L1 Monoclonal Antibody of Blocking the Binding of PD-L1 to PD-1

A 96-well high-adsorption ELISA plate was coated with 1 μg/mL of the recombinant human PD-L1-Fc at 100 μL per well using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST, and then blocked using PBST with 1% BSA, 300 μL/well, and incubated at 25° C. for 1 hour, and then washed 5 times with PBST. The anti-human PD-L1 antibody samples and the positive control were added, 50 μL/well, the biotin-labelled PD-1-Fc was added at a concentration of 40 nM (a final concentration of 20 nM), 50 μL/well, incubated at 25° C. for 90 minutes, and then washed 5 times with PBST. The Streptavidin-HRP (BD Pharmingen, catalog number 554066) diluted 1:1000 in PBST with 1% BSA was then added, 100 μL per well, and incubated at 25° C. for 1 hour. The plate was washed 5 times with PBST. The colorimetric substrate TMB was added, 100 μL/well, and developed at room temperature for 10 minutes. 1M H2SO4 was added, 100 μL/well, to stop the development. The absorbance at 450 nm was read on a microplate reader.


As shown in FIG. 4, the chimeric anti-human PD-L1 monoclonal antibody has an activity of blocking the binding of PD-L1/PD-1 similar to that of Atezolizumab.


Example 6 T Cell Function Regulation Activity of the Chimeric Anti-Human PD-L1 Monoclonal Antibody

PBMCs used in the experiment were purchased from Lonza (catalog number CC-2702).


DC cells were induced with PBMCs firstly: PBMC cells were thawed and collected with a complete medium (RPMI 1640 with 10% FBS), washed once with a corresponding serum-free medium, then re-suspended in serum-free medium and seeded in a cell culture flask, and cultured in a 5% CO2 incubator at 37° C. for 90 minutes. Discarding the culture supernatant and suspension cells, and the adherent cells were incubated for 3 days in the complete medium+100 ng/ml GM-CSF (granulocyte-macrophage colony stimulating factor) and 100 ng/ml IL-4 (interleukin 4) (Sino Biological Inc.), and the medium was replaced, and incubated for another 3 days. Then the medium was replaced with the complete medium+100 ng/ml GM-CSF, 100 ng/ml IL-4 and 20 ng/ml TNF-α (tumor necrosis factor) (Sino Biological Inc.), and incubated for one day to accomplish the induction of the DCs. T cells were separated from PBMCs from another individual: T cells were isolated with the Pan T cell isolation kit (Miltenyi Biotech, catalog number 5150414820) and please refer to the instruction manual for the specific experimental process. The induced mature DC cells were seeded in a 96-well plate, 10,000 cells per well, and isolated T cells were added, 100,000 cells per well, and finally the samples to be tested were added, and incubated together for 120 hours. At the end of the incubation, the supernatant was taken, and the level of IFN-γ was detected with the ELISA kit purchased from RayBiotech.


As shown in FIG. 5, the chimeric anti-human PD-L1 monoclonal antibody can enhance the secretion of IFN-γ in the mixed lymphocyte culture system, and has a T cell function regulation activity slightly stronger than that of Atezolizumab.


Example 7 Preparation of the Humanized Anti-Human PD-L1 Monoclonal Antibodies

The humanized anti-human PD-L1 mAbs were obtained according to the method described by Leung et al. (1995, Molecule Immunol 32:1413-27). The humanized templates that best match the variable region sequences of the murine antibody were selected from the Germline database, wherein the template for the light chain variable region is IGKV1-33*01 of the sequence set forth in SEQ ID NO: 28; the template for the heavy chain variable region is IGHV1-69*01 of the sequence set forth in SEQ ID NO: 29. The CDR regions of the murine antibody were grafted into the selected humanized templates to replace the CDR regions of the human templates, obtaining the grafted, humanized antibody light chain variable region and the grafted, humanized antibody heavy chain variable region. Particular sites were selected to carry out back mutations in the light chain framework regions to obtain the light chain variable region sequences set forth in SEQ ID Nos: 38, 39 and 44, and particular sites were selected to carry out back mutations in the heavy chain framework regions to obtain the heavy chain variable region sequences set forth in SEQ ID Nos: 30-37 and 40-43. Then, the NG site on HCDR2 of the sequence set forth in SEQ ID No. 24 was mutated to remove the possible deamidation site, obtaining the mutated heavy chain HCDR2s of the amino acid sequences set forth in SEQ ID Nos. 45-47. The heavy chain variable region sequences obtained after mutating HCDR2 of the heavy chain variable region as set forth in SEQ ID No. 36 are set forth in SEQ ID Nos. 48-50; the heavy chain variable region sequences obtained after mutating HCDR2 of the heavy chain variable region as set forth in SEQ ID No. 41 are set forth in SEQ ID Nos. 51-53. The light chain variable region was linked to the light chain constant region (SEQ ID NO: 26) to obtain the corresponding full-length sequences of the light chains respectively, and the heavy chain variable region was linked to the heavy chain constant region (SEQ ID NO: 27) to obtain the corresponding full-length sequences of the heavy chains respectively. The humanized sequences are shown in Table 7. Exemplary combinations of the heavy chain variable region VH and the light chain variable region VL in the humanized antibodies are shown in Table 8. The affinity data (dissociation constants) of the humanized antibodies are shown in Table 9.









TABLE 7





Humanized sequences



















SEQ ID No. of the amino acid sequence



VL number
corresponding to the VL number







VLS9
SEQ ID NO. 38



VLS10
SEQ ID NO. 39



VLS15
SEQ ID NO. 44








SEQ ID No. of the amino acid sequence



VH number
corresponding to the VH number







VHS1
SEQ ID NO. 30



VHS2
SEQ ID NO. 31



VHS3
SEQ ID NO. 32



VHS4
SEQ ID NO. 33



VHS5
SEQ ID NO. 34



VHS6
SEQ ID NO. 35



VHS7
SEQ ID NO. 36



VHS8
SEQ ID NO. 37



VHS11
SEQ ID NO. 40



VHS12
SEQ ID NO. 41



VHS13
SEQ ID NO. 42



VHS14
SEQ ID NO. 43



VHS7 P1
SEQ ID NO. 48



VHS7 P2
SEQ ID NO. 49



VHS7 P3
SEQ ID NO. 50



VHS12 P1
SEQ ID NO. 51



VHS12 P2
SEQ ID NO. 52



VHS12 P3
SEQ ID NO .53

















TABLE 8







Exemplary combinations of the heavy chain variable region


VH and the light chain variable region VL in the antibodies










VH comprising the
VL comprising the



amino acid sequence set
amino acid sequence set



forth in the following
forth in the following



SEQ ID NO and
SEQ ID NO and


Sample
composition thereof
composition thereof





VLS9-VHS1
SEQ ID NO. 30
SEQ ID NO. 38


VLS9-VHS2
SEQ ID NO. 31
SEQ ID NO. 38


VLS9-VHS3
SEQ ID NO. 32
SEQ ID NO. 38


VLS9-VHS4
SEQ ID NO. 33
SEQ ID NO. 38


VLS9-VHS5
SEQ ID NO. 34
SEQ ID NO. 38


VLS9-VHS6
SEQ ID NO. 35
SEQ ID NO. 38


VLS9-VHS7
SEQ ID NO. 36
SEQ ID NO. 38


VLS9-VHS8
SEQ ID NO. 37
SEQ ID NO. 38


VLS10-VHS1
SEQ ID NO. 30
SEQ ID NO. 39


VLS10-VHS2
SEQ ID NO. 31
SEQ ID NO. 39


VLS10-VHS3
SEQ ID NO. 32
SEQ ID NO. 39


VLS10-VHS4
SEQ ID NO. 33
SEQ ID NO. 39


VLS10-VHS5
SEQ ID NO. 34
SEQ ID NO. 39


VLS10-VHS6
SEQ ID NO. 35
SEQ ID NO. 39


VLS10-VHS7
SEQ ID NO. 36
SEQ ID NO. 39


VLS10-VHS8
SEQ ID NO. 37
SEQ ID NO. 39


VLS15-VHS11
SEQ ID NO. 40
SEQ ID NO. 44


VLS15-VHS12
SEQ ID NO. 41
SEQ ID NO. 44


VLS15-VHS13
SEQ ID NO. 42
SEQ ID NO. 44


VLS15-VHS14
SEQ ID NO. 43
SEQ ID NO. 44


VLS10-VHS7 P1
SEQ ID NO. 48
SEQ ID NO. 39


VLS10-VHS7 P2
SEQ ID NO. 49
SEQ ID NO. 39


VLS10-VHS7 P3
SEQ ID NO. 50
SEQ ID NO. 39


VLS15-VHS12 P1
SEQ ID NO. 51
SEQ ID NO. 44


VLS15-VHS12 P2
SEQ ID NO. 52
SEQ ID NO. 44


VLS15-VHS12 P3
SEQ ID NO. 53
SEQ ID NO. 44


Chimeric antibody
SEQ ID NO. 19
SEQ ID NO. 18
















TABLE 9







Affinity determination (dissociation


constants) of the humanized sequences










Sample
kd (1/s)







VLS9-VHS1
5.97E−03



VLS9-VHS2
5.99E−03



VLS9-VHS3
5.20E−03



VLS9-VHS4
5.44E−03



VLS9-VHS5
4.38E−03



VLS9-VHS6
6.18E−03



VLS9-VHS7
5.80E−03



VLS9-VHS8
5.32E−03



VLS10-VHS1
4.24E−03



VLS10-VHS2
3.25E−03



VLS10-VHS3
1.21E−03



VLS10-VHS4
1.56E−03



VLS10-VHS5
9.94E−04



VLS10-VHS6
1.21E−03



VLS10-VHS7
6.72E−04



VLS10-VHS8
6.52E−04



VLS15-VHS11
8.41E−04



VLS15-VHS12
4.86E−04



VLS15-VHS13
7.63E−04



VLS15-VHS14
4.52E−04



Chimeric antibody
5.31E−04










Example 8 Thermal Stability of the Humanized Anti-Human PD-L1 Monoclonal Antibodies

Waters Xbridge BEH200 3.5 um, 7.8 mm×30 cm chromatographic column (catalog number: 186007640) was used, with the mobile phase: 0.1 mol/L phosphate buffer (NaH2PO4—Na2HPO4), 0.1 mol/L sodium sulfate buffer, pH 6.7, the flow rate: 0.6 mL/min, the column temperature: 25° C., the sample cell temperature: 4° C., and the detection wavelength: 280 nm. The sample was diluted to 1 mg/mL with the sample buffer, and the injection volume is 10 μL. Agilent High-Performance Liquid Chromatography 1260 system workstation was used to process the data of the experimental results, and the proportion of the main peak (i.e. the purity) was calculated through the area normalization method. To determine the thermal stability of these monoclonal antibodies, the above samples were placed under a high temperature condition of 40° C., samples were taken at the 2nd week and 4th week for SE-HPLC detection to observe the thermal stability, and the results were shown in Table 10 below. All the humanized anti-human PD-L1 antibodies, except for VLS10-VHS7 P2, showed good and considerable stability.









TABLE 10







The thermal stability of the humanized anti-human


PD-L1 mAbs detected by SE-HPLC at 40° C.








Humanized anti-human PD-L1
SE-HPLC purity (%)










monoclonal antibody
T = 0
week 2
week 4













VLS10-VHS7
97.36
96.36
96.04


VLS10-VHS7 P1
97.41
97.17
95.52


VLS10-VHS7 P2
97.61
97.26
93.96


VLS10-VHS7 P3
98.18
96.81
97.07


VLS15-VHS12
97.71
96.44
95.10


VLS15-VHS12 P1
97.82
96.50
95.24


VLS15-VHS12 P2
98.56
97.54
97.50


VLS15-VHS12 P3
98.59
97.48
97.22


Atezolizumab
96.35
93.38
93.31









Example 9 In Vitro Biological Activities of the Humanized Anti-Human PD-L1 Monoclonal Antibodies

In vitro biological activities of the humanized anti-human PD-L1 humanized monoclonal antibodies were determined, including the binding activity to human PD-L1, the activities of blocking the binding of PD-L1/PD-1 and blocking the binding of PD-L1/CD80, the binding kinetic constant to human PD-L1. The humanized sequences to be determined include VLS10-VHS7, VLS10-VHS7 P1, VLS10-VHS7 P2, VLS10-VHS7 P3, VLS15-VHS12, VLS15-VHS12 P1, VLS15-VHS12 P2, VLS15-VHS12 P3 and the control Atezolone, the chimeric anti-human PD-L1 monoclonal antibody. The specific experimental method is the same as the method for measuring the in vitro biological activities of the chimeric anti-human PD-L1 monoclonal antibody.


The experimental results are shown in Table 11 and Table 12. Compared to the chimeric anti-human PD-L1 mAb, the humanized sequences to be tested all maintained their activities well, showing strong PD-L1 binding activity and PD-L1/PD-1 blocking activity and PD-L1/CD80 blocking activity, among which VLS15-VHS12-P3 sequence is the best.









TABLE 11







Activities of the humanized anti-human PD-L1 monoclonal antibodies


of binding to PD-L1 and blocking the binding of PD-L1/receptor












PD-L1/PD-1
PD-L1/CD80



PD-L1 binding
blocking
blocking



activity
activity
activity


Sample
(EC50, nM)
(IC50, nM)
(IC50, nM)













Atezolizumab
0.055
0.892
0.904


Chimeric mAb
0.066
0.596
0.762


VLS10-VHS7
0.067
0.749
0.890


VLS10-VHS7 P1
0.067
0.756
0.865


VLS10-VHS7 P2
0.059
0.725
0.857


VLS10-VHS7 P3
0.063
0.617
0.804


VLS15-VHS12
0.046
0.666
0.802


VLS15-VHS12 P1
0.048
0.567
0.795


VLS15-VHS12 P2
0.060
0.585
0.755


VLS15-VHS12 P3
0.075
0.613
0.813
















TABLE 12







Binding kinetic constants of the humanized anti-


human PD-L1 monoclonal antibodies to human PD-L1










Sample
Kon (1/Ms)
Koff (1/s)
KD (M)





Atezolizumab
1.92E+5
3.86E−4
2.01E−9


Chimeric mAb
5.07E+5
5.94E−4
1.17E−9


VLS10-VHS7
3.41E+5
5.84E−4
1.71E−9


VLS10-VHS7 P1
3.41E+5
 1.08E−03
3.16E−9


VLS10-VHS7 P2
3.07E+5
9.94E−4
3.23E−9


VLS10-VHS7 P3
2.55E+5
7.47E−4
2.93E−9


VLS15-VHS12
3.42E+5
4.77E−4
1.39E−9


VLS15-VHS12 P1
3.26E+5
8.33E−4
2.56E−9


VLS15-VHS12 P2
3.72E+5
7.20E−4
1.94E−9


VLS15-VHS12 P3
3.38E+5
5.28E−4
1.56E−9









Example 10 Pharmacokinetic Study of the Humanized Anti-Human PD-L1 Monoclonal Antibodies in Human PD-1/Human PD-L1 Double Gene Knock-In Mice

The 6-8-week-old female human PD-1/human PD-L1 double gene knock-in mice (C57BL/6 background) (Biocytogen Beijing Gene Biotechnology Co., Ltd.) were selected as the experimental material. After the mice acclimated to the environment for one week, the mice were intraperitoneally injected with the humanized anti-human PD-L1 monoclonal antibody VLS15-VHS12 P3 at a dosage of 70 nmol/kg, single administration. At the time of administration, 6 hours, 24 hours, 72 hours, 120 hours, 168 hours, 240 hours, and 312 hours after administration, orbital blood was collected, 2 mice at each blood collection time point. No anticoagulation was given, and the blood samples were placed at room temperature for 30 minutes to 1 hour. After coagulation, the blood samples were centrifuged at 3000 rpm for 10 minutes, and the obtained serum samples were frozen at −80° C. and stored until testing.


The levels of the humanized anti-human PD-L1 monoclonal antibodies in serum were determined by ELISA. Briefly, a high-adsorption ELISA plate was coated with the recombinant human PD-L1 using carbonate buffer solution, pH=9.6 at 4° C. overnight. The plate was washed 5 times with PBST. To prevent non-specific binding, the plate was blocked using PBST with 5% skim milk powder and washed with PBST. Then, the serum samples to be tested which were diluted with PBST containing 10% mixed mouse serum and 1% BSA were added, and incubated at 25° C. for 1 hour, and the plate was washed with PBST. The horseradish peroxidase-labeled anti-human IgG antibody (Chemicon, catalog number AP309P) diluted in PBST containing 5% skim milk powder, the plate was then incubated at 25° C. for 1 hour, and washed with PBST. Finally, the colorimetric substrate TMB was used for development at room temperature for 10 minutes. 1M H2SO4 was added to stop the development. The absorbance at 450 nm was read on a microplate reader.


As shown in FIG. 6, a single intraperitoneal injection of 70 nmol/kg the humanized anti-human PD-L1 monoclonal antibody showed good drug-time curve and pharmacokinetic characteristics in human PD-1/human PD-L1 double gene knock-in mice. The pharmacokinetic parameters of the humanized anti-human PD-L1 monoclonal antibody were as follows: the half-life t1/2 was 130 hours; the area under the drug-time curve AUC0-312 hr was 77917 nM·hr; the apparent volume of distribution Vd was 147 mL/Kg; the clearance rate CL was 0.78 mL/hr/kg; the mean residence time MRTlast was 90 hours.


Example 11 Anti-Tumor Efficacy Study of the Humanized Anti-Human PD-L1 mAb

This example examines the growth inhibitory effect of the humanized anti-human PD-L1 mAb on MC38/human PD-L1 tumor grafts inoculated in human PD-1/human PD-L1 double gene knock-in mice.


The 6-8-week-old female human PD-1/human PD-L1 double gene knock-in mice (C57BL/6 background) (Biocytogen Beijing Gene Biotechnology Co., Ltd.) were selected as the experimental material. After the mice acclimated to the environment for one week, each mouse was inoculated with 5×105 MC38/human PD-L1 mouse colon cancer cells constructed for expressing human PD-L1. Once the tumors grew to a volume of about 150 mm3, the mice were divided into different groups according to the tumor volumes, 6 mice per group. The vehicle control group, the humanized anti-human PD-L1 mAb VLS15-VHS12 P3 administration group were set up, with a dosage of 70 nmol/kg, twice a week for 2 consecutive weeks, via an intraperitoneal injection. The tumor volumes were measured twice a week beginning at the day of administration, and the long diameter a and the short diameter b were measured and the tumor volume calculation formula is: tumor volume (mm3)=(a×b2)/2.


As shown in FIG. 7, the humanized anti-human PD-L1 monoclonal antibody had an anti-tumor activity, significantly inhibiting the growth of MC38/human PD-L1 tumor grafts inoculated in human PD-1/human PD-L1 double gene knock-in mice.


Finally, it should be noted that the above examples are only used to illustrate the technical solutions of the invention, rather than to limit them. While the inventions have been described in detail with reference to the foregoing examples, those of ordinary skill in the art will recognize that numerous modifications can be made to the technical solutions described in the foregoing embodiments, or various equivalent alternatives can be made for some or all the technical features described herein without making the essence of the corresponding technical solutions depart from the scopes of various embodiments of the invention.

Claims
  • 1. An isolated anti-human PD-L1 antibody or antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof comprises a light chain variable region and/or a heavy chain variable region, wherein, the light chain variable region comprises a LCDR1 of the amino acid sequence set forth in SEQ ID No. 20, a LCDR2 of the amino acid sequence set forth in SEQ ID No. 21, and a LCDR3 of the amino acid sequence set forth in SEQ ID No. 22; and/orthe heavy chain variable region comprises a HCDR1 of the amino acid sequence set forth in SEQ ID No. 23, a HCDR2 of the amino acid sequence at least about 94% identical to SEQ ID Nos. 24, 45, 46 or 47; a HCDR3 of the amino acid sequence set forth in SEQ ID No. 25.
  • 2. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein said anti-human PD-L1 antibody or antigen-binding fragment thereof is a chimeric antibody, a humanized antibody, or a fully human antibody.
  • 3. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein the heavy chain constant region sequence of the antibody is the constant region sequence of one selected from the group consisting of human IgG1, IgG2, IgG3, IgG4, IgA, IgM, IgE, IgD, and/or, the light chain constant region sequence of the antibody is the constant region sequence of κ chain or λ chain; preferably, the heavy chain constant region sequence is the constant region sequence of IgG1 or IgG4, and/or the light chain constant region sequence is the constant region sequence of κ chain.
  • 4. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein the light chain variable region has the amino acid sequence set forth in SEQ ID NO. 18, and/or the heavy chain variable region has the amino acid sequence set forth in SEQ ID NO. 19.
  • 5. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein the framework regions of the light chain variable region of the anti-human PD-L1 antibody or antigen-binding fragment thereof include FR-L1, FR-L2, FR-L3 and FR-L4, and the framework regions of the heavy chain variable include FR-H1, FR-H2, FR-H3 and FR-H4, wherein the FR-L1 has the amino acid sequence set forth in SEQ ID NO. 54;the FR-L2 has the amino acid sequence set forth in SEQ ID NO. 55, or an amino acid sequence as set forth in SEQ ID NO. 55 obtained by one of the following substitutions or any combination thereof:the 2nd amino acid Y is substituted with I,the 3rd amino acid Q is substituted with H,the 9th amino acid A is substituted with S;the FR-L3 has the amino acid sequence set forth in SEQ ID NO. 56;the FR-L4 has the amino acid sequence set forth in SEQ ID NO. 57;the FR-H1 has the amino acid sequence set forth in SEQ ID NO. 58, or an amino acid sequence as set forth in SEQ ID NO. 58 obtained by one of the following substitutions or any combination thereof:the 1st amino acid Q is substituted with E,the 23rd amino acid K is substituted with T;the FR-H2 has the amino acid sequence set forth in SEQ ID NO. 59, or an amino acid sequence as set forth in SEQ ID NO. 59 obtained by the following substitution:the 13th amino acid M is substituted with I;the FR-H3 has the amino acid sequence set forth in SEQ ID NO. 60, or an amino acid sequence as set forth in SEQ ID NO. 60 obtained by one of the following substitutions or any combination thereof:the 2nd amino acid V is substituted with A,the 8th amino acid E is substituted with T,the 11th amino acid S is substituted with N,the 31st amino acid A is substituted with G; and/orthe FR-H4 has the amino acid sequence set forth in SEQ ID NO. 61.
  • 6. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein (a) the light chain variable region has the amino acid sequence set forth in SEQ ID No. 38; and/orthe heavy chain variable region has the amino acid sequence selected from those set forth in SEQ ID Nos. 30-37;(b) the light chain variable region has the amino acid sequence set forth in SEQ ID No. 39; and/orthe heavy chain variable region has the amino acid sequence selected from those set forth in SEQ ID Nos. 30-37 or 48-50; or(c) the light chain variable region has the amino acid sequence set forth in SEQ ID No. 44; and/orthe heavy chain variable region has the amino acid sequence selected from those set forth in SEQ ID Nos. 40-43 or 51-53.
  • 7. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 6, wherein (a) the light chain variable region has the amino acid sequence set forth in SEQ ID No. 39; and/orthe heavy chain variable region has the amino acid sequence selected from those set forth in SEQ ID Nos. 36 or 48-50;(b) the light chain variable region has the amino acid sequence set forth in SEQ ID No. 44; and/orthe heavy chain variable region has the amino acid sequence selected from those set forth in SEQ ID Nos. 41 or 51-53.
  • 8. The anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, wherein the antigen-binding fragment is one or more selected from the group consisting of F(ab′)2, Fab′, Fab, Fd, Fv, scFv, diabody, camelid antibody, CDR and antibody minimal recognition unit (dAb), preferably, the antigen-binding fragment is Fab, F(ab′)2 or scFv.
  • 9. An isolated nucleic acid molecule selected from the group consisting of: (1) DNA or RNA encoding the anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1;(2) a nucleic acid that is completely complementary to the nucleic acid defined in (1).
  • 10-11. (canceled)
  • 12. A composition comprising the anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • 13-14. (canceled)
  • 15. A method of preventing and/or treating PD-L1-mediated diseases or disorders, including administering to a subject in need thereof the anti-human PD-L1 antibody or antigen-binding fragment thereof according to claim 1, preferably, the diseases or disorders are tumors; more preferably, the tumors are one or more selected from the group consisting of leukemia, lymphoma, myeloma, brain tumor, head and neck squamous cell carcinoma, non-small cell lung cancer, nasopharyngeal cancer, esophageal cancer, gastric cancer, pancreatic cancer, gallbladder cancer, liver cancer, colorectal cancer, breast cancer, ovarian cancer, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, bladder cancer, renal cell carcinoma, and melanoma.
Priority Claims (1)
Number Date Country Kind
20210025262.3 Jan 2021 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2022/070628 1/7/2022 WO