WO2019184909A1 - 新型抗体分子、其制备方法及其用途 - Google Patents
新型抗体分子、其制备方法及其用途 Download PDFInfo
- Publication number
- WO2019184909A1 WO2019184909A1 PCT/CN2019/079671 CN2019079671W WO2019184909A1 WO 2019184909 A1 WO2019184909 A1 WO 2019184909A1 CN 2019079671 W CN2019079671 W CN 2019079671W WO 2019184909 A1 WO2019184909 A1 WO 2019184909A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- domain
- immunoglobulin
- antibody
- polypeptide chain
- terminus
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/46—Hybrid immunoglobulins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
Definitions
- the present invention generally relates to the field of immunology and antibody engineering.
- the invention relates to a novel novel engineered antibody molecule, a polynucleotide encoding the antibody molecule, a vector comprising the polynucleotide, a host cell comprising the polynucleotide or vector, comprising Immunoconjugates and pharmaceutical compositions of the antibody molecules, and uses of the antibody molecules for immunotherapy, prevention, and/or diagnosis of a disease.
- Antibody molecules are capable of targeted and specific binding to their corresponding antigens, and are increasingly becoming important therapeutic and prophylactic agents for various diseases (eg, cancer, autoimmune diseases, inflammatory diseases, infectious diseases, etc.). / or diagnostic agent.
- diseases eg, cancer, autoimmune diseases, inflammatory diseases, infectious diseases, etc.
- diagnostic agent e.g., cancer, autoimmune diseases, inflammatory diseases, infectious diseases, etc.
- monospecific antibodies directed against only one target have some limitations in clinical applications. Patients may develop resistance or no response after receiving monospecific antibody therapy.
- multispecific antibodies are capable of specifically binding to different antigens, when one antigen is located on a particular immune cell and the other antigen is on a disease cell, a multispecific antibody (eg, bispecific) Sexual antibodies) can redirect specific immune cells to diseased cells to enhance the lethality of immune cells to diseased cells.
- multispecific antibodies eg, bispecific antibodies
- Blinatumomab is the first single-chain bispecific antibody with a molecular weight of approximately 55 kDa for the treatment of B-cell non-Hodgkin's lymphoma (NHL) and B precursor acute lymphoblastic leukemia (ALL).
- NHL B-cell non-Hodgkin's lymphoma
- ALL B precursor acute lymphoblastic leukemia
- the two single-chain Fv molecules directed against the CD19 molecule and against the CD3 molecule are fused by a flexible linker peptide, which utilizes CD19 expressed in almost all B lymphocyte tumors and CD3 expressed on T cells, making T
- the cells are tightly linked to the target cells (tumor cells), which release perforin and telomerase into the synaptic cleft, causing a series of chemical reactions in the tumor cells, thereby destroying the tumor cells (Nagorsen D. and Baeuerle PA, Immunomodulatory) Therapy of cancer with T cell-engaging BiTE antibody blinatumomab, Exp Cell Res, 2011, 317: 1255-1260).
- Catumaxomab is a chimera consisting of two half-antibodies derived from the parental mouse IgG2a isotype and the rat IgG2b isotype, each having a light chain and a heavy chain, anti-CD3 rat IgG2b half.
- Antibodies for T cell recognition mouse IgG2a half antibodies against tumor cell surface antigen EpCAM (epithelial adhesion molecule) for tumor cell recognition (Chelius D et al, Structural and functional characterization of the trifunctional antibody catumaxomab, MAbs, 2010, 2:309-319).
- Catumaxomab Approved in Europe in April 2009 for the treatment of malignant ascites caused by EpCAM-positive epithelial-derived metastases.
- Multispecific antibodies can be divided into many classes depending on the components and the manner in which they are constructed. For example, according to the left and right basic symmetry of the multispecific antibody structure, it can be divided into a symmetric structure and an asymmetric structure; according to the Fc region of the multispecific antibody with or without IgG, it can be divided into an antibody pattern having an Fc region and an Fc-free region.
- the antibody pattern according to the number of antigen binding sites in the multispecific antibody, it can be classified into divalent, trivalent, tetravalent or higher valence antibodies and the like.
- Blinatumomab can be produced by large-scale culture of recombinant Chinese hamster ovary (CHO) cells, it is easy to form aggregates and has a short half-life in vivo. In actual use, an additional infusion set is required; the Catumaxomab production process is complicated and the mouse heterologous antibody is more susceptible to immunogenicity in the human body.
- CHO Chinese hamster ovary
- the present invention provides a novel multispecific antibody format by using a single domain antigen binding site with a small molecular weight and high stability as a building block, with the N-terminus or C-terminus of the Fab fragment. After ligation, the resulting adaptor is ligated to the Fc region, and is easily expressed in cultured cells in vitro without complicated production processes; in addition, the presence of the Fc region in the antibody pattern of the present invention allows expression of the present invention in cultured cells.
- Purified antibodies can be obtained using single-step affinity chromatography, and the antibodies of the invention have longer serum half-life in vivo and can elicit effector functions, such as antibody-dependent cell-mediated cells.
- the multispecific antibody pattern of the present invention is capable of maintaining the affinity of each antigen binding site in the multispecific antibody to bind to a corresponding different epitope, and does not cause steric hindrance when binding different epitopes. Interference, with good drug-forming properties. Further, the multispecific antibody format of the invention is physically stable and biologically stable, which allows the antibody to be more productive and developable.
- the antibody molecule is capable of binding to one or more antigens with high affinity and high specificity, preferably to two or more antigens.
- the invention also provides nucleic acid molecules encoding the antibody molecules, expression vectors, host cells and methods for producing the antibody molecules.
- the invention also provides immunoconjugates and pharmaceutical compositions comprising the antibody molecules of the invention.
- the antibody molecules disclosed herein can be used alone or in combination with other drugs or other therapeutic modalities for the treatment, prevention, and/or diagnosis of diseases such as autoimmune diseases, acute and chronic inflammatory diseases, infectious diseases (eg, chronic infectious diseases or sepsis). ), tumors, etc.
- the invention provides an antibody molecule having one or more of the following properties:
- an antibody molecule of the invention comprises (i) a single domain antigen binding site; (ii) an antigen binding Fab fragment; wherein said (i) is located in said (ii) light chain variable structure The N-terminus of the domain (VL) or the C-terminus of the light chain constant region (CL), or the (i) N-terminal or immunoglobulin CH1 structure of the heavy chain variable domain (VH) of (ii) C-terminus of the domain, and said (i) and (ii) respectively bind the same or different antigens, with or without a linker peptide between (i) and (ii); and at (i) and Iii) The C-terminal (iii) immunoglobulin Fc domain.
- the antibody molecule of the invention comprises at least four antigen binding sites, at least two single domain antigen binding sites and an antigen binding site in at least two Fab fragments, respectively, in combination with at least four, Three, two different antigens, or one antigen.
- the antigen binding site in the antibody molecule of the invention binds to the same or different epitopes in the antigen molecule.
- an antibody molecule of the invention comprises four antigen binding sites, wherein two single domain antigen binding sites bind to the same or different epitopes in the first antigen, and two Fab fragments bind to the second antigen The same or different epitopes, the first antigen being different from the second antigen.
- a glycine and/or serine residue used alone or in combination between (i) and (ii) in the antibody molecule of the present invention is used as a linker peptide, for example, the linker peptide comprises an amino acid sequence (Gly 4 Ser)n, where n is a positive integer equal to or greater than 1, for example, n is a positive integer in 1-7, for example, n is 2, 3, 4, 5, 6.
- the single domain antigen binding site in the antibody molecule of the invention is selected from the group consisting of a heavy chain variable domain (VH), a light chain variable domain (VL), and a heavy chain of an antibody that naturally lacks a light chain.
- VH heavy chain variable domain
- VL light chain variable domain
- a variable domain for example, a heavy chain variable domain of a naturally occurring heavy chain antibody in a Camelidae species
- NAR new antigen receptor
- fish eg, VH-like single domains in naturally occurring IgNAR in shark serum
- recombinant single domain antigen binding sites derived from them eg, camelized human VH domain, humanized camelid antibody heavy Chain variable domain.
- the single domain antigen binding site in an antibody molecule of the invention is selected from the heavy chain variable domain of a naturally occurring heavy chain antibody in camelid species, a camelized human VH domain And humanized camelid antibody heavy chain variable domains.
- the heavy chain variable domain derived from a heavy chain antibody that naturally lacks the light chain is also referred to herein simply as VHH to distinguish it from the conventional VH of a four-chain immunoglobulin.
- VHH molecules can be derived from antibodies produced in camelid species such as camels, alpacas, dromedaries, llamas and guanaco. Other species other than camelids can also produce heavy chain antibodies that naturally lack light chains, and such VHHs are also within the scope of the invention.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light chain and is located in the immunization A N-terminal single domain antigen binding site of a globin light chain variable domain (VL), such as VHH; each polypeptide chain in the second polypeptide chain and the fourth polypeptide chain comprises an immunoglobulin heavy chain.
- VL globin light chain variable domain
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light chain and is located in the immunization A single domain antigen binding site at the C-terminus of the globose light chain constant region (CL), such as VHH; each polypeptide chain in the second polypeptide chain and the fourth polypeptide chain comprises an immunoglobulin heavy chain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each polypeptide chain in the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light chain; a second polypeptide Each polypeptide chain in the chain and the fourth polypeptide chain comprises an immunoglobulin heavy chain and a single domain antigen binding site at the N-terminus of the immunoglobulin heavy chain, such as VHH.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each polypeptide chain in the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light chain; a second polypeptide Each polypeptide chain in the chain and the fourth polypeptide chain comprises an immunoglobulin heavy chain variable region, an immunoglobulin CH1 domain, a single domain antigen binding site (eg, VHH), immunization from the N-terminus to the C-terminus. Globulin CH2, CH3 and optionally CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises a single domain antigen from the N-terminus to the C-terminus Binding site (eg VHH), immunoglobulin heavy chain variable domain (VH) and immunoglobulin light chain constant region (CL); each polypeptide chain in the second polypeptide chain and the fourth polypeptide chain
- the immunoglobulin light chain variable domain (VL), immunoglobulin CH1, CH2, CH3 and optionally the CH4 domain are comprised from the N-terminus to the C-terminus.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin heavy from the N-terminus to the C-terminus a chain variable domain (VH), an immunoglobulin light chain constant region (CL), and a single domain antigen binding site (eg, VHH); each polypeptide chain in the second polypeptide chain and the fourth polypeptide chain
- the immunoglobulin light chain variable domain (VL), immunoglobulin CH1, CH2, CH3 and optionally the CH4 domain are comprised from the N-terminus to the C-terminus.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin heavy from the N-terminus to the C-terminus a chain variable domain (VH) and an immunoglobulin light chain constant region (CL); each polypeptide chain in the second polypeptide chain and the fourth polypeptide chain comprises a single domain antigen binding from the N-terminus to the C-terminus Site (eg, VHH), immunoglobulin light chain variable domain (VL), immunoglobulin CH1, CH2, CH3, and optionally CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin heavy from the N-terminus to the C-terminus a chain variable domain (VH) and an immunoglobulin light chain constant region (CL); each of the second polypeptide chain and the fourth polypeptide chain comprises an immunoglobulin light chain from the N-terminus to the C-terminus Variable domain (VL), immunoglobulin CH1 domain, single domain antigen binding site (eg, VHH), immunoglobulin CH2, CH3, and optionally CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises a single domain antigen from the N-terminus to the C-terminus a binding site (eg, VHH), an immunoglobulin light chain variable domain (VL), and an immunoglobulin CH1 domain; each of the second polypeptide chain and the fourth polypeptide chain is from the N-terminus to The C-terminus comprises an immunoglobulin heavy chain variable domain (VH), an immunoglobulin light chain constant region (CL), an immunoglobulin CH2, CH3, and optionally a CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light from the N-terminus to the C-terminus a chain variable domain (VL), an immunoglobulin CH1 domain, and a single domain antigen binding site (eg, VHH); each of the second polypeptide chain and the fourth polypeptide chain is from the N terminus to The C-terminus comprises an immunoglobulin heavy chain variable domain (VH), an immunoglobulin light chain constant region (CL), an immunoglobulin CH2, CH3, and optionally a CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light from the N-terminus to the C-terminus a chain variable domain (VL) and an immunoglobulin CH1 domain; each of the second polypeptide chain and the fourth polypeptide chain comprises a single domain antigen binding site from the N-terminus to the C-terminus (eg VHH), immunoglobulin heavy chain variable domain (VH), immunoglobulin light chain constant region (CL), immunoglobulin CH2, CH3 and optionally CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the invention provides an antibody molecule comprising four polypeptide chains, wherein each of the first polypeptide chain and the third polypeptide chain comprises an immunoglobulin light from the N-terminus to the C-terminus a chain variable domain (VL) and an immunoglobulin CH1 domain; each of the second polypeptide chain and the fourth polypeptide chain comprises an immunoglobulin heavy chain variable domain from the N-terminus to the C-terminus (VH), immunoglobulin light chain constant region (CL), single domain antigen binding site (eg, VHH), immunoglobulin CH2, CH3, and optionally CH4 domain.
- the immunoglobulin is an IgG1, IgG2 or IgG4 immunoglobulin, more preferably the immunoglobulin is a human IgG1 immunoglobulin.
- the inventors have also designed amino acid residues capable of stabilizing the molecular structure of the antibody and facilitating proper coupling or pairing between the individual strands.
- a hinge region having a "CPPC" amino acid residue is included in an Fc domain of a second polypeptide chain and a fourth polypeptide chain of an antibody molecule such that the second polypeptide chain and the fourth polypeptide chain pass each other
- the disulfide bond formed between the amino acid residues at the hinge region is stably associated.
- the second polypeptide chain and the fourth polypeptide chain of the antibody molecule of the invention comprise Y349C and S354C, respectively, in the respective Fc domains or S354C and Y349C, respectively (according to Kabat et al., Sequences of Proteins of Immunological Interest, 5th edition, Public Health Service, National Institutes of Health, Bethesda, MD (1991), the EU index is numbered, hereinafter referred to as "EU number"), whereby the second polypeptide chain and the fourth polypeptide The strand further forms an interchain disulfide bond in the Fc region to stabilize the correct pairing of the second polypeptide chain and the fourth polypeptide chain.
- the second polypeptide chain and/or the fourth polypeptide chain of an antibody molecule of the invention comprises an amino acid mutation in the Fc domain that affects antibody effector function.
- the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).
- the amino acid mutation is present in the CH2 domain of the Fc region, eg, the antibody molecule is comprised at positions 234 and 235 (EU numbering) of the second polypeptide chain and/or the fourth polypeptide chain Amino acid substitution.
- the amino acid substitutions are L234A and L235A (hereinafter referred to as "LALA mutations").
- the Fc domain of each of the second polypeptide chain and the fourth polypeptide chain of the antibody molecule of the invention comprises a bulge ("knob") or a hole ("hole”, respectively). And said bulges or vacancies in the Fc domain of the second polypeptide chain can be placed in said holes or bulges in the Fc domain of the fourth polypeptide chain, respectively, whereby said second The peptide chain and the fourth polypeptide chain form a "knob-in-hole" stable association with each other.
- an amino acid substitution T366W is included in one of the second polypeptide chain and the fourth polypeptide chain, and is included in the other of the second polypeptide chain and the fourth polypeptide chain Amino acid substitutions T366S, L368A and Y407V (EU numbering).
- the protrusions in one strand can be placed in the cavities in the other strand, facilitating the correct pairing of the second polypeptide chain and the fourth polypeptide chain.
- the first polypeptide chain of the antibody molecule of the invention and the immunoglobulin CL domain and the CH1 domain of the second polypeptide chain comprise a bulge or a hole, respectively, and said The protrusions or holes may be placed in the holes or protrusions in the CL domain, respectively, such that the first polypeptide chain and the second polypeptide chain also form a stable association of "binding" to each other.
- the third polypeptide chain of the antibody molecule of the present invention and the immunoglobulin CL domain and the CH1 domain of the fourth polypeptide chain also respectively contain protrusions or holes, and the protrusions in the CH1 domain Or holes may be placed in the holes or bulges in the CL domain, respectively, such that the third polypeptide chain and the fourth polypeptide chain also form a stable association of "binding" to each other.
- two single domain antigen binding sites in an antibody molecule of the invention bind to the same epitope in a first antigen, and two Fab fragments bind to the same epitope in a second antigen, whereby The antibody molecule is a bispecific antibody directed against the first antigen and the second antigen.
- the type of antigen to which the antibody molecule of the present invention specifically binds is not particularly limited, and the antigen may be, for example, a cytokine, a growth factor, a hormone, a signaling protein, an inflammatory mediator, a ligand, a cell surface receptor, or a fragment thereof.
- the antigen to which the antibody molecule of the invention specifically binds is selected from the group consisting of a tumor-associated antigen, an immunological checkpoint molecule, an angiogenesis-inducing factor, a member of the tumor necrosis factor receptor superfamily, and a costimulatory molecule in the immune system, and Ligands and/or receptors for these molecules, for example, OX40, CD47, PD1, PD-L1, PD-L2, LAG-3, 4-1BB (CD137), VEGF, and GITR.
- the present invention exemplifies several bispecific antibodies as described below.
- the antibody molecule of the invention is an anti-OX40/PD-L1 bispecific antibody capable of being at least about 10 7 M -1 , preferably about 10 8 M -1 and more preferably An affinity constant of about 10 9 M -1 or greater binds to OX40, a member of the tumor necrosis factor (TNF) receptor family expressed on the surface of lymphocytes, thereby activating T cells, for example, enhancing the immune stimulation/effector function of effector T cells and / or allowing these cells to proliferate and / or downregulate the immunosuppressive function of regulatory T cells; and at least about 10 7 M -1 , preferably about 10 8 M -1 and more preferably about 10 9 M -1 or stronger
- the affinity constant binds to PD-L1 on the surface of tumor cells, thereby inhibiting the binding of PD-1 on T cells to PD-L1 on the surface of tumor cells, inducing T cell activation and exerting an anti-tumor effect.
- the anti-OX40/PD-L1 bispecific antibody of the invention consists of four polypeptide chains that are substantially symmetric about the left and right, wherein two polypeptide chains in the left half and two polypeptide chains in the right half Each comprising (i) a single domain antigen binding site; (ii) an antigen binding Fab fragment; wherein said (i) is located at the N-terminus of said (ii) light chain variable domain (VL) or The C-terminus of the light chain constant region (CL), or the (i) the N-terminus of the heavy chain variable domain (VH) of the (ii) or the C-terminus of the immunoglobulin CH1 domain, and (i), (ii) binding to OX40 or PD-L1, respectively, with or without a linking peptide between (i) and (ii); and (iii) at the C-terminus of (i) and (ii) Immunoglobulin Fc domain.
- the single domain antigen binding site in the anti-OX40/PD-L1 bispecific antibody of the invention is a VHH that specifically binds to PD-L1, and the Fab fragment is an anti-OX40 antibody Fab that specifically binds OX40 Fragment.
- the VHH that specifically binds to PD-L1 in the anti-OX40/PD-L1 bispecific antibody of the invention comprises CDR1, SEQ ID NO: 4 set forth in SEQ ID NO: CDR2 shown as CDR2 and SEQ ID NO: 5, or one, two, three, four, five, six or more amino acids with one or more of the three CDRs Sequences that vary (eg, amino acid substitutions or deletions);
- the anti-OX40 antibody Fab fragment that specifically binds OX40 in an anti-OX40/PD-L1 bispecific antibody of the invention comprises a SEQ derived from the anti-OX40 antibody ADI-20112 ID NO: all 6 heavy chain complementarity determining regions (CDRs) and light chain CDRs in the paired heavy chain variable region sequence/light chain variable region sequence shown in 11/7, or in the 6 CDRs
- One or more CDRs have a sequence of one, two, three, four, five, six or more amino acid changes (eg, amino acid substitution
- the VHH that specifically binds to PD-L1 in the anti-OX40/PD-L1 bispecific antibody of the invention comprises the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO: 2, Or a sequence substantially identical thereto (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or more identical); the anti-OX40/PD-L1 of the present invention
- the anti-OX40 antibody Fab fragment that specifically binds OX40 in the bispecific antibody comprises the paired heavy chain variable region sequence/light chain shown in SEQ ID NO: 11/7 derived from the anti-OX40 antibody ADI-20112
- the variable region sequence or having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98 with the paired heavy chain variable region sequence/light chain variable region sequence %, 99% or more sequences of sequence identity.
- the anti-OX40/PD-L1 bispecific antibody of the invention consists of four polypeptide chains substantially symmetric about each other, wherein the two polypeptide chains of the left half of the antibody molecule comprise a first polypeptide chain of SEQ ID NO: 6 and a second polypeptide chain of SEQ ID NO: 10; comprising a first polypeptide chain of SEQ ID NO: 14 and SEQ ID NO: 10, respectively a second polypeptide chain; comprising a first polypeptide chain represented by SEQ ID NO: 15 and a second polypeptide chain represented by SEQ ID NO: 16; respectively comprising the first one represented by SEQ ID NO: 15 a polypeptide chain and a second polypeptide chain of SEQ ID NO: 17; or substantially identical to any of said sequences (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, a sequence of 98%, 99% or higher identical; accordingly, wherein the two polypeptide chains of the right half of the antibody molecule comprise a first polypeptide chain
- the antibody molecule of the invention is an anti-VEGF/GITR bispecific antibody capable of being at least about 10 7 M -1 , preferably about 10 8 M -1 and more preferably about 10 9 M -1 or stronger affinity constant binds to Vascular Endothelial Cell Growth Factor (VEGF), thereby blocking the binding of VEGF to its receptor VEGFR, making VEGFR unable to activate, thereby exerting anti-angiogenic effects.
- VEGF Vascular Endothelial Cell Growth Factor
- an anti-tumor angiogenesis effect inhibiting tumor growth; and having an affinity constant of at least about 10 7 M -1 , preferably about 10 8 M -1 and more preferably about 10 9 M -1 or more Glucocorticoid-induced tumor necrosis factor receptor (GITR) binding on CD4 + and CD8 + T cells, thereby reversing the inhibitory effects of regulatory T cells (Treg) and co-stimulating and activating effector T cells to exert antitumor effects .
- GITR Glucocorticoid-induced tumor necrosis factor receptor
- the anti-VEGF/GITR bispecific antibody of the invention consists of four polypeptide chains that are substantially symmetric about each other, wherein in the two polypeptide chains in the left half and the two polypeptide chains in the right half, Each comprising (i) a single domain antigen binding site; (ii) an antigen binding Fab fragment; wherein said (i) is located at the N-terminus or light chain of said light chain variable domain (VL) of (ii) The C-terminus of the constant region (CL), or the (i) the N-terminus of the heavy chain variable domain (VH) of the (ii) or the C-terminus of the immunoglobulin CH1 domain, and the (i) And (ii) binding to VEGF or GITR, respectively, with or without a linker peptide between (i) and (ii); and (iii) immunoglobulin Fc at the C-terminus of (i) and (ii) Domain.
- the single domain antigen binding site in the anti-VEGF/GITR bispecific antibody of the invention is a VHH that specifically binds to GITR, and the Fab fragment is an anti-VEGF antibody Fab fragment that specifically binds to VEGF.
- said VHH that specifically binds to GITR in an anti-VEGF/GITR bispecific antibody of the invention comprises CDR1, SGGGFGD (SEQ ID NO: 26) as shown by GFAGFSS (SEQ ID NO: 25) CDR3 shown in CDR2 and ATDWRKP (SEQ ID NO: 27), or one, two, three, four, five, six with one or more of the three CDRs a sequence of more than one amino acid change (eg, an amino acid substitution or deletion);
- the anti-VEGF antibody Fab fragment that specifically binds to VEGF in an anti-VEGF/GITR bispecific antibody of the invention comprises a derivative derived from the anti-VEGF antibody Avastin All 6 heavy chain complementarity determining regions (CDRs) and light chain CDRs in the paired heavy chain variable region sequence/light chain variable region sequence set forth in SEQ ID NO: 22/20, or with the 6 CDRs
- One or more of the CDRs have a sequence of one, two, three, four, five
- the VHH that specifically binds to GITR in an anti-VEGF/GITR bispecific antibody of the invention comprises, or is substantially derived from, an anti-GITR VHH amino acid sequence set forth in SEQ ID NO: Sequences of the same (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or more identical); of the anti-VEGF/GITR bispecific antibodies of the invention
- the anti-VEGF antibody Fab fragment that specifically binds to VEGF comprises the paired heavy chain variable region sequence/light chain variable region sequence set forth in SEQ ID NO: 22/20 derived from the anti-VEGF antibody Avastin, or
- the heavy chain variable region sequence/light chain variable region sequence has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity Sexual sequence.
- the anti-VEGF/GITR bispecific antibody of the invention consists of four polypeptide chains substantially symmetric about each other, wherein the two polypeptide chains of the left half of the antibody molecule comprise SEQ ID, respectively. a first polypeptide chain represented by NO: 18 and a second polypeptide chain represented by SEQ ID NO: 21; respectively comprising the first polypeptide chain represented by SEQ ID NO: 18 and the SEQ ID NO: 28 a second polypeptide chain; or a sequence substantially identical to any of said sequences (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher)
- the two polypeptide chains of the right half of the antibody molecule comprise a third polypeptide chain of SEQ ID NO: 18 and a fourth polypeptide chain of SEQ ID NO: 21, respectively; a third polypeptide chain set forth in SEQ ID NO: 18 and a fourth polypeptide chain set forth in SEQ ID NO: 28; or substantially identical to any
- the invention provides a polynucleotide encoding any one or more polypeptide chains in an antibody molecule of the invention.
- the invention provides a vector, preferably an expression vector, comprising a polynucleotide encoding any one or more of the polypeptide chains of an antibody molecule of the invention.
- the invention provides a host cell comprising a polynucleotide or vector of the invention.
- the host cell is a mammalian cell, preferably a CHO cell, a HEK293 cell; the host cell is a prokaryotic cell, preferably an E. coli cell.
- the invention provides a method for producing an antibody molecule of the invention, the method comprising the steps of (i) cultivating a host cell of the invention under conditions suitable for expression of the antibody molecule, and (ii) The host cell or the culture medium recovers the antibody molecule.
- the invention provides an immunoconjugate and a pharmaceutical composition comprising an antibody molecule of the invention.
- the antibody molecules disclosed herein can be used alone or in combination with other drugs or other therapeutic modalities for the treatment, prevention, and/or diagnosis of diseases such as autoimmune diseases, acute and chronic inflammatory diseases, infectious diseases (eg, chronic infectious diseases or sepsis). ), tumors, etc.
- the invention provides the use of an antibody molecule, immunoconjugate and pharmaceutical composition of the invention, as a medicament for the treatment and/or prevention of a disease in an individual or as a diagnostic tool for a disease.
- the individual is a mammal, more preferably a human.
- the disease is an autoimmune disease, an acute and chronic inflammatory disease, an infectious disease (eg, a chronic infectious disease or sepsis), a tumor.
- FIGS 1A-1D illustrate four structures of the bispecific antibodies of the invention.
- 2A-2D show the four structures of the anti-OX40/PD-L1 bispecific antibody Bi-110-112HC, Bi-113- prepared by the present invention by size exclusion chromatography (SEC), respectively. Purity of 112HC, Bi-119-112LC and Bi-122-112LC.
- Figure 3 shows the binding of anti-OX40/PD-L1 bispecific antibody Bi-119-112LC detected by FACS, and anti-PD-L1 humanized Nb-Fc antibody as a control to CHO cells overexpressing PD-L1.
- the horizontal axis represents the antibody concentration
- the vertical axis represents the mean fluorescence intensity (MFI).
- Figure 4 shows the binding of the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC detected by FACS, and the anti-OX40 antibody ADI-20112 as a positive control to CHO cells overexpressing OX40.
- the horizontal axis represents the antibody concentration
- the vertical axis represents the mean fluorescence intensity (MFI).
- Figure 5 shows the simultaneous binding of anti-OX40/PD-L1 bispecific antibodies to CHO cells overexpressing OX40 and CHO cells overexpressing PD-L1.
- Figure 6 shows the effect of the anti-OX40/PD-L1 bispecific antibody of the present invention on the binding of human PD-1 to PD-L1, demonstrating that the bispecific antibody Bi-119-112LC of the present invention blocks human PD-1 and The combination of PD-L1.
- the effect of anti-PD-L1 humanized Nb-Fc and IgG1 as a control was also examined.
- Figure 7 shows that the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention effectively abolishes the blocking effect of the PD1/PD-L1 interaction on the NFAT signaling pathway, thereby obtaining a fluorescent signal.
- Figure 8 shows the effect of the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention on the PD-L1-dependent OX40-mediated signaling pathway.
- the effects of anti-PD-L1 humanized Nb-Fc, ADI-20112, anti-PD-L1 humanized Nb-Fc + ADI-20112 and IgG1 were also examined.
- FIG. 9 shows the results of Tm values of the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention as determined by differential scanning fluorescence (DSF).
- Figure 10 shows the activation of human T cells by the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention.
- the effects of anti-PD-L1 humanized Nb-Fc, ADI-20112 and IgG1 were also examined.
- FIGS 11A-11B illustrate two structures of the bispecific antibodies of the invention.
- Figures 12A-12B show the purity of the anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 prepared by the present invention, respectively, detected by SEC.
- Figure 13 shows the binding of anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 detected by FACS to CHO cells overexpressing GITR.
- the horizontal axis represents the antibody concentration
- the vertical axis represents the mean fluorescence intensity (MFI).
- antibody is used herein in its broadest sense to refer to a protein comprising an antigen binding site, encompassing natural and artificial antibodies of various structures including, but not limited to, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (for example, bispecific antibodies), single chain antibodies, intact antibodies, and antibody fragments.
- full antibody full antibody
- full length antibody complete antibody
- intact antibody are used interchangeably herein to refer to a naturally occurring comprising at least two heavy chains (H) interconnected by disulfide bonds. And two light chain (L) glycoproteins.
- Each heavy chain consists of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
- the heavy chain constant region consists of three domains, CH1, CH2 and CH3.
- Each light chain consists of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
- the light chain constant region consists of one domain CL.
- the VH and VL regions can be further subdivided into hypervariable regions (which are complementarity determining regions (CDRs) with more conserved regions interposed (framework regions (FR)).
- CDRs complementarity determining regions
- FR frame regions
- Each VH and VL consists of three CDRs and four
- the FR composition is arranged from the amino terminus to the carboxy terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
- the constant region is not directly involved in the binding of the antibody to the antigen, but exhibits multiple effector functions.
- antigen-binding fragment is a portion or portion of an intact or complete antibody that is less than the number of amino acid residues of an intact or fully antibody, which is capable of binding to an antigen or competing with an intact antibody (ie, an intact antibody derived from an antigen-binding fragment). Binding antigen. Antigen-binding fragments can be prepared by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact antibodies. Antigen binding fragments include, but are not limited to, Fab, Fab', F(ab') 2 , Fv, single chain Fv, diabody, single domain antibody (sdAb).
- the Fab fragment is a monovalent fragment consisting of the VL, VH, CL and CH1 domains, for example, a Fab fragment can be obtained by digestion of a complete antibody by papain.
- F(ab') 2 which is a dimer of Fab', is a divalent antibody fragment by digesting a complete antibody under the disulfide bond of the hinge region by pepsin.
- F(ab') 2 can be reduced under neutral conditions by disrupting the disulfide bond in the hinge region, thereby converting the F(ab') 2 dimer to a Fab' monomer.
- the Fab' monomer is essentially a Fab fragment with a hinge region (for a more detailed description of other antibody fragments, see: Fundamental Immunology, WE Paul, ed., Raven Press, NY (1993)).
- the Fv fragment consists of the VL and VH domains of one arm of the antibody.
- the two domains VL and VH of the Fv fragment are encoded by independent genes, they can be joined by a synthetic linker capable of causing the two domains to be produced as a single protein chain using recombinant methods,
- the VL region and the VH region in a single protein chain are paired to form a single chain Fv.
- the antibody fragment can be obtained by chemical methods, recombinant DNA methods or protease digestion.
- single domain antibody or “single variable domain (SVD) antibody” generally refers to an antibody in which a single variable domain (eg, a heavy chain variable domain (VH) or a light chain can be The variable domain (VL), the heavy chain variable domain derived from the camelid heavy chain antibody, and the VH-like single domain (v-NAR) derived from the fish IgNAR confer antigen binding. That is, the single variable domain does not need to interact with another variable domain to recognize the target antigen.
- single domain antibodies include single domain antibodies derived from camelids (llamas and camels) and cartilage fish (eg, nurse sharks) (WO 2005/035572).
- camelized human VH domain means that the transfer of a key element derived from Camelidae VHH to a human VH domain results in the human VH domain no longer needing to be paired with the VL domain to recognize the target antigen, which is camelized.
- the human VH domain alone confers antigen binding specificity.
- binding site or "antigen binding site” as used herein denotes a region of an antibody molecule that actually binds to an antigen, including by an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).
- VL antibody light chain variable domain
- VH antibody heavy chain variable domain
- a VH/VL pair consisting of a heavy chain variable domain derived from a camelid heavy chain antibody, a VH-like single domain (v-NAR) from a shark family IgNAR, a camelized human VH domain, a human A derived camelid antibody heavy chain variable domain.
- the antibody molecule of the invention comprises at least four antigen binding sites, for example, comprising two single domain antigen binding sites (eg, VHH) and a VH/VL pair in two Fab fragments.
- the antigen binding site formed.
- single domain antigen binding site denotes a single variable domain of an antibody molecule (eg, a heavy chain variable domain (VH), a light chain variable domain (VL), derived from a camelid heavy chain antibody Heavy chain variable domain, v-NAR from IgNAR of sharks, camelized human VH domain, humanized camelid antibody heavy chain variable domain, and their recombined single domain ) the area that actually binds to the antigen.
- the antibody molecule of the invention comprises two single domain antigen binding sites, each of which binds to the same or a different antigen.
- the antibody molecule of the invention comprises two single domain antigen binding sites, each binding to the same or a different epitope.
- the term "monospecific" antibody refers to an antibody having one or more binding sites, each of which binds to the same epitope of the same antigen.
- multispecific antibody refers to an antibody having at least two antigen binding sites, each of the at least two antigen binding sites being different or different from the same epitope of the same antigen. Different epitopes of the antigen bind.
- the antibodies provided herein are typically multispecific antibodies, such as bispecific antibodies.
- Multispecific antibodies are antibodies that have binding specificities for at least two different epitopes.
- a bispecific antibody having binding specificity for a first antigen and a second antigen.
- immunoglobulin molecule refers to a protein having the structure of a naturally occurring antibody.
- an IgG-like immunoglobulin is a heterotetrameric glycoprotein of about 150,000 daltons composed of two light chains and two heavy chains that are disulfide-bonded. From the N-terminus to the C-terminus, each immunoglobulin heavy chain has a heavy chain variable region (VH), also known as a heavy chain variable domain, followed by three heavy chain constant domains (CH1, CH2 and CH3) ).
- VH heavy chain variable region
- each immunoglobulin light chain has a light chain variable region (VL), also referred to as a light chain variable domain, followed by a light chain constant domain (CL).
- VL light chain variable region
- the heavy chain of immunoglobulin can belong to one of five categories, called ⁇ (IgA), ⁇ (IgD), ⁇ (IgE), ⁇ (IgG) or ⁇ (IgM), some of which can be further divided into sub- Classes such as ⁇ 1 (IgG1), ⁇ 2 (IgG2), ⁇ 3 (IgG 3 ), ⁇ 4 (IgG 4 ), ⁇ 1 (IgA 1 ), and ⁇ 2 (IgA 2 ).
- the light chain of an immunoglobulin can be divided into one of two types, called kappa and lambda, based on the amino acid sequence of its constant domain.
- Immunoglobulins consist essentially of two Fab molecules and one Fc domain joined by an immunoglobulin hinge region.
- Fc domain or "Fc region” is used herein to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
- the term includes native sequence Fc regions and variant Fc regions.
- a native immunoglobulin "Fc domain” comprises two or three constant domains, a CH2 domain, a CH3 domain, and an optional CH4 domain.
- the immunoglobulin Fc domain comprises second and third constant domains (CH2 domain and CH3 domain) derived from two heavy chains of IgG, IgA and IgD class antibodies; or a source comprising The second, third, and fourth constant domains (CH2 domain, CH3 domain, and CH4 domain) of the two heavy chains of the IgM and IgE class antibodies.
- amino acid residue numbering in the Fc region or heavy chain constant region is according to, for example, Kabat et al., Sequences of Proteins of Immunological Interes, 5th Edition, Public Health Service, National Institutes of Health, Bethesda, MD, The EU numbering system (also known as the EU index) described in 1991 is numbered.
- effector function refers to those biological activities attributed to the immunoglobulin Fc region that vary with the immunoglobulin isotype.
- immunoglobulin effector functions include: C1q binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) Cytokine secretion, immune complex-mediated uptake by antigen-presenting cells, down-regulation of cell surface receptors (eg, B cell receptors), and B cell activation.
- chimeric antibody is an antibody molecule in which (a) changes, replaces or exchanges a constant region or a portion thereof, such that the antigen binding site is different from a different or altered class, effector function and/or species. a region or a completely different molecule (eg, an enzyme, a toxin, a hormone, a growth factor, a drug) that confers new properties to a chimeric antibody; or (b) a variable region or a portion thereof with a different or altered antigen specificity The variable region is changed, replaced or exchanged.
- a mouse antibody can be modified by replacing its constant region with a constant region derived from human immunoglobulin. Due to the replacement into the human constant region, the chimeric antibody retains its specificity in recognizing the antigen while having reduced antigenicity in humans as compared to the original mouse antibody.
- a “humanized antibody” is an antibody which retains antigen-specific reactivity of a non-human antibody (for example, a mouse monoclonal antibody) and which is less immunogenic when administered to a human as a therapeutic drug. This can be achieved, for example, by retaining the non-human antigen binding site and replacing the remainder of the antibody with their human counterpart (ie, the constant region and the portion of the variable region that is not involved in binding is the corresponding portion of the human antibody). See, for example, Padlan, Anatomy of the antibody molecule, Mol. Immun., 1994, 31: 169-217.
- Other examples of human antibody engineering techniques include, but are not limited to, the Xoma technology disclosed in US 5,766,886.
- ...valent antibody refers to the number of antigen binding sites present in an antibody molecule.
- Bivalent, trivalent and tetravalent antibodies refer to the presence of two antigen binding sites, three binding sites and four binding sites, respectively, in the antibody molecule. In one embodiment, the bispecific antibodies reported herein are "tetravalent.”
- antibody consisting of four polypeptide chains substantially symmetric about left and right means that the antibody molecule consists of four polypeptide chains, including two polypeptide chains to the left of the antibody molecule and two polypeptide chains to the right, and the left side of the antibody molecule 2
- the sequence of the polypeptide chain and the sequence of the two polypeptide chains on the right have 100% identity or at least 95% or at least 99% identity.
- flexible linker peptide refers to a linker peptide consisting of amino acids, such as glycine and/or serine residues, used alone or in combination, to link various variable domains in an antibody.
- the flexible linker peptide is a Gly/Ser linker peptide comprising an amino acid sequence (Gly 4 Ser)n, wherein n is a positive integer equal to or greater than 1, eg, n is a positive integer from 1-7.
- the flexible linker peptide is (Gly 4 Ser) 2 (SEQ ID NO: 9).
- binding means that the binding is selective for the antigen and can be distinguished from unwanted or non-specific interactions.
- the ability of an antigen binding site to bind to a particular antigen can be determined by enzyme-linked immunosorbent assay (ELISA) or conventional binding assays known in the art.
- affinity or "binding affinity” refers to the inherent binding affinity that reflects the interaction between members of a binding pair.
- affinity molecule X for its partner Y can generally dissociation constant (K D) is represented by the solution, the dissociation constant is the ratio of the dissociation rate constant and association rate constant (k dis, respectively and k on) of.
- K D dissociation constant
- association rate constant k dis, respectively and k on
- antigen refers to a molecule that elicits an immune response. This immune response may involve antibody production or activation of specific immune cells, or both.
- any macromolecule including substantially all proteins or peptides, can be used as an antigen.
- the antigen can be derived from recombinant or genomic DNA.
- the first antigen, the second antigen, and the third antigen are three different antigens.
- tumor-associated antigen refers interchangeably to a molecule (usually a protein, carbohydrate or lipid) that is expressed completely or as a fragment (eg, MHC/peptide) on the surface of a cancer cell, as compared to normal cells. And the molecule can be used in the preferential targeting of the agent to cancer cells.
- the tumor associated antigen is a cell surface molecule that is overexpressed in tumor cells as compared to normal cells, eg, 1 fold overexpression, 2 fold overexpression, 3 fold overexpression or more than normal cells Overexpression.
- the tumor associated antigen is a cell surface molecule that is improperly synthesized in tumor cells, such as a molecule that contains a deletion, addition, or mutation compared to a molecule expressed on a normal cell.
- the tumor associated antigen is only expressed intact or as a fragment on the cell surface of the tumor cell and is not synthesized or expressed on the surface of normal cells.
- EGFRvIII epidermal growth factor receptor variant III
- TAG72 tumor associated glycoprotein 72
- CEA carcinoembryonic antigen
- EPCAM epithelial cell adhesion molecule
- interleukin 11 receptor alpha IL-11Ra
- VEGFR2 vascular endothelial growth factor receptor 2
- EGFR epidermal growth factor receptor
- NCAM neural cell adhesion molecule
- IGF insulin-like growth factor 1 receptor
- MAGE-A1 melanoma-associated antigen 1
- immunocheckpoint means a class of inhibitory signaling molecules present in the immune system that protects against tissue damage by modulating the persistence and strength of immune responses in peripheral tissues and is involved in maintaining tolerance to autoantigens (Pardoll DM., The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer, 2012, 12(4): 252-264).
- Immunological checkpoint molecules include, but are not limited to, programmed death 1 (PD-1), PD-L1, PD-L2, cytotoxic T lymphocyte antigen 4 (CTLA-4), LAG-3, and TIM-3.
- PD-1 programmed death 1
- PD-L1 PD-L1
- PD-L2 PD-L2
- CTLA-4 cytotoxic T lymphocyte antigen 4
- LAG-3 LAG-3
- TIM-3 TIM-3
- costimulatory molecule refers to a corresponding binding partner on a T cell that specifically binds to a costimulatory ligand to mediate a costimulatory response to T cells, such as, but not limited to, proliferation.
- Costimulatory molecules are cell surface molecules that contribute to an effective immune response in addition to antigen receptors or their ligands.
- Costimulatory molecules include, but are not limited to, MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activating molecules (SLAM proteins), activated NK cell receptors, OX40 , CD40, GITR, 4-1BB (ie CD137), CD27 and CD28.
- the "costimulatory molecule” is OX40, GITR, 4-1BB (ie, CD137), CD27, and/or CD28.
- cytokine is a generic term for a protein that is released by one cell population and acts as an intercellular medium on another cell.
- cytokines are lymphokines, mononuclear factors, interleukins (IL), such as IL-1, IL-1 ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-9, IL-11, IL-12, IL-15; tumor necrosis factor, such as TNF- ⁇ or TNF- ⁇ ; and other polypeptide factors, including LIF and kit ligand (KL) and ⁇ -interferon.
- IL interleukins
- an “immunoconjugate” is an antibody that is conjugated to one or more heterologous molecules, including but not limited to cytotoxic agents.
- cytotoxic agent refers to a substance that inhibits or prevents cellular function and/or causes cell death or destruction.
- Cytotoxic agents include, but are not limited to, radioisotopes (eg, radioisotopes of At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 , and Lu); Or drugs (eg, methotrexate, doxorubicin, vinblastine alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, phenylbutyric acid Nitrogen mustard, Zoorubicin or other intercalating agents; growth inhibitors; enzymes and fragments thereof such as lysozyme; antibiotics; small toxins or enzymatically active toxins such as toxins such as bacterial, fungal, plant or animal sources, Included are fragment
- the “percent identity (%)" of the amino acid sequence means that the candidate sequence is aligned with the specific amino acid sequence shown in the present specification and, if necessary, the vacancy is introduced to achieve the maximum percent sequence identity, and no consideration is given.
- conservative modifications include substitutions, deletions or additions to polypeptide sequences which result in the replacement of an amino acid with a chemically similar amino acid.
- Conservative substitution tables that provide functionally similar amino acids are well known in the art.
- conservatively modified variants are additive relative to the polymorphic variants, interspecies homologs and alleles of the invention and do not exclude them.
- the following 8 groups contain amino acids that are conservatively substituted: 1) alanine (A), glycine (G); 2) aspartic acid (D), glutamic acid (E); 3) asparagine (N) , glutamine (Q); 4) arginine (R), lysine (K); 5) isoleucine (I), leucine (L), methionine (M), guanidine (V); 6) phenylalanine (F), tyrosine (Y), tryptophan (W); 7) serine (S), threonine (T); and 8) cysteine Acid (C), methionine (M) (see, for example, Creighton, Proteins (1984)).
- the term "conservative sequence modification” is used to refer to an amino acid modification that does not significantly affect or alter the binding characteristics of an antibody comprising an amino acid sequence.
- N-terminus refers to the last amino acid at the N-terminus
- C-terminus refers to the last amino acid at the C-terminus
- host cell refers to a cell into which an exogenous polynucleotide has been introduced, including progeny of such a cell.
- Host cells include “transformants” and “transformed cells,” which include primary transformed cells and progeny derived therefrom.
- a host cell is any type of cellular system that can be used to produce an antibody molecule of the invention, including eukaryotic cells, eg, mammalian cells, insect cells, yeast cells; and prokaryotic cells, eg, E. coli cells.
- Host cells include cultured cells, as well as transgenic animals, transgenic plants, or cultured plant tissues or cells within animal tissues.
- expression vector refers to a vector comprising a recombinant polynucleotide comprising an expression control sequence operably linked to a nucleotide sequence to be expressed.
- the expression vector contains sufficient cis-acting elements for expression; other elements for expression may be provided by the host cell or in an in vitro expression system.
- Expression vectors include all those known in the art, including cosmids incorporated into recombinant polynucleotides, plasmids (eg, naked or contained in liposomes), and viruses (eg, lentiviruses, retroviruses, glands) Virus and adeno-associated virus).
- mammals include, but are not limited to, domesticated animals (eg, cows, sheep, cats, dogs, and horses), primates (eg, humans and non-human primates such as monkeys), rabbits, and rodents (eg, mice and large mouse).
- domesticated animals eg, cows, sheep, cats, dogs, and horses
- primates eg, humans and non-human primates such as monkeys
- rabbits eg, mice and large mouse.
- rodents eg, mice and large mouse.
- the individual is a human.
- treatment refers to the clinical intervention intended to alter the natural course of the disease in an individual being treated. Desirable therapeutic effects include, but are not limited to, preventing the onset or recurrence of the disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of progression of the disease, ameliorating or mitigating the disease state, and alleviating or improving the prognosis.
- the antibody molecules of the invention are used to delay the progression of the disease or to slow the progression of the disease.
- anti-tumor effect refers to a biological effect that can be exhibited by a variety of means including, but not limited to, for example, a reduction in tumor volume, a decrease in the number of tumor cells, a decrease in tumor cell proliferation, or a decrease in tumor cell survival.
- tumor and cancer are used interchangeably herein to encompass both solid tumors and liquid tumors.
- cancer refers to or describe a physiological condition in a mammal that is typically characterized by unregulated cell growth.
- cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma and leukemia or lymphoid malignancies. More specific examples of such cancers include, but are not limited to, squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma, and lung squamous cell carcinoma), peritoneal cancer.
- squamous cell carcinoma e.g., epithelial squamous cell carcinoma
- lung cancer including small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma, and lung squamous cell carcinoma
- peritoneal cancer e.g., peritoneal cancer.
- hepatocellular carcinoma gastric cancer (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urinary tract cancer, liver tumor, breast cancer, colon cancer, Rectal cancer, colorectal cancer, endometrial cancer or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, liver cancer, anal cancer, penile cancer, melanoma, superficial diffuse melanoma, Malignant freckle-like melanoma, acral melanoma, nodular melanoma, multiple myeloma and B-cell lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), Hairy cell leukemia, chronic myeloblastic leukemia, and post-transplant lymphoproliferative disorders (PTLD), as well as with phagomatoses, edema (such as those associated with brain
- tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
- cancer refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
- cancer refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
- infectious disease refers to a disease caused by a pathogen, including, for example, a viral infection, a bacterial infection, a parasitic infection, or a fungal infection.
- the present invention provides a novel antibody molecule that can be used for immunotherapy, prevention, and/or diagnosis of a variety of diseases.
- the antibody molecule of the invention comprises at least four antigen binding sites which are capable of functioning as monospecific antibodies or multispecific (eg bispecific) antibodies, preferably capable of acting as multispecific (eg bispecific) Antibodies work.
- the antibody molecule platform constructed herein comprises (i) a single domain antigen binding site; (ii) an antigen binding Fab fragment; wherein said (i) is located in said (ii) light chain variable domain (VL) The C-terminus of the N-terminus or the light chain constant region (CL), or the (i) N-terminus of the heavy chain variable domain (VH) of the (ii) or the C of the immunoglobulin CH1 domain And (i) and (ii) respectively bind the same or different antigens, with or without a linker peptide between (i) and (ii); and at (i) and (ii) (iii) Immunoglobulin Fc domain at the C-terminus.
- an antibody molecule of the invention has four polypeptide chains comprising two single domain antigen binding sites and two Fab fragments and an Fc region.
- the single domain antigen binding site of the antibody molecule of the invention and the Fab fragment do not have a linker peptide.
- a single domain antigen binding site of the antibody molecule of the invention and a Fab fragment have a linker peptide.
- the type of the linker peptide is not particularly limited.
- the linker peptide is a peptide having an amino acid sequence of from 1 to 100, in particular from 1 to 50, more particularly from 1 to 20 amino acids in length.
- the linker peptide is (G 4 S) 2 (SEQ ID NO: 9).
- a single domain antigen binding site in an antibody molecule of the invention is a single variable domain capable of specifically binding a target antigen epitope with higher affinity, for example, a heavy chain variable domain (VH), a light chain variable structure Domain (VL), heavy chain variable domain derived from camelid heavy chain antibody, v-NAR from IgNAR of sharks, camelized human VH domain, humanized camelid antibody heavy chain variable Domains, and their recombined single domains.
- the single domain antigen binding site in an antibody molecule of the invention is a heavy chain variable domain derived from a camelid heavy chain antibody, a camelized human VH domain, and/or a humanized camel Family antibody heavy chain variable domain.
- camelid species such as camelids, alpaca, dromedary, llama and guanaco have been characterized in the prior art.
- a heavy chain variable domain of a camelid heavy chain antibody having high affinity for a target antigen (this region is also referred to as VHH) can be obtained by a genetic engineering method. See U.S. Patent No. 5,759,808, issued June 2, 1998.
- the amino acid sequence of Camelidae VHH can be recombinantly altered to obtain sequences that more realistically mimic human sequences, i.e., "humanized,” thereby reducing the antigenicity of Camelidae VHH to humans.
- key elements derived from Camelidae VHH can be transferred to the human VH domain to obtain a camelized human VH domain.
- the single domain antigen binding site in the antibody molecule of the invention is a humanized VHH directed against PD-L1 having the sequence set forth in SEQ ID NO: 1 and/or SEQ ID NO: 2. Amino acid sequence.
- the single domain antigen binding site in the antibody molecule of the invention is a VHH directed against GITR having the amino acid sequence set forth in SEQ ID NO:24.
- the molecular weight of VHH is one tenth of the molecular weight of a human IgG molecule and has a physical diameter of only a few nanometers.
- VHH itself has extremely high thermal stability, is stable to extreme pH and proteolytic digestion, and has low antigenicity. Therefore, in one embodiment of the antibody molecule of the present invention, VHH is included as a building block for stability of the antibody molecule of the present invention, The low antigenicity of human subjects contributes.
- the Fab fragments in the antibody molecules of the invention are capable of specifically binding to a target antigen epitope with higher affinity.
- the Fab fragment is a Fab fragment of an immunoglobulin comprising a peptide consisting of an immunoglobulin light chain variable region (VL) and an immunoglobulin light chain constant region (CL); a peptide consisting of an immunoglobulin heavy chain variable region (VH) and an immunoglobulin heavy chain constant region 1 (CH1); wherein the CL region and the CH1 region are optionally covalently linked by disulfide bonds and heterodimerized Fab fragments.
- VL immunoglobulin light chain variable region
- CL immunoglobulin light chain constant region
- CH1 immunoglobulin heavy chain constant region 1
- the Fab fragment is a light chain variable region (VL) and a light chain variable region (VL) exchanged Fab fragment of an immunoglobulin Fab fragment, comprising a variable immunoglobulin light chain a peptide consisting of a region (VL) and a heavy chain constant region (CH1); and comprising a peptide consisting of an immunoglobulin heavy chain variable region (VH) and a light chain constant region (CL); wherein the CL region and the CH1 region
- the Fab fragment is heterodimericly heterologously covalently linked by a disulfide bond.
- the Fab fragment is a light chain constant region (CL) and a heavy chain constant region (CH1) exchanged Fab fragment of an immunoglobulin Fab fragment comprising an immunoglobulin heavy chain variable region a peptide consisting of (VH) and a light chain constant region (CL); and comprising a peptide consisting of an immunoglobulin light chain variable region (VL) and a heavy chain constant region (CH1); wherein the CL region and the CH1 region are The Fab fragment is heterodimericly heterologously covalently linked by a disulfide bond.
- Fab fragment in an antibody molecule of the invention does not comprise a disulfide bond.
- the two strands of the Fab fragment can be engineered in such a way as to stably interact without the need for a disulfide bond.
- both strands of a Fab fragment can be engineered to remove a cysteine residue, and the two strands of the Fab fragment still stably interact and function as a Fab.
- the two strands of the Fab fragment are mutated to promote a stable interaction between the two strands.
- a genetic modification strategy can be used (see, for example, John BBRidgway et al., 'Knobs-into-holes' engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Engineering, 1996.9(7): p.
- a Fab fragment in an antibody molecule of the invention is derived from a monoclonal antibody and can be derived from any type of antibody, including IgA, IgM, IgD, IgG, IgE, and subtypes thereof, eg, IgGl, IgG2, IgG3, and IgG4.
- the light chain domain can be derived from a kappa chain or a lambda chain.
- the Fab fragments used herein can also be prepared by recombinant.
- the CH1 domain, CL domain in a Fab fragment of an antibody molecule of the invention is derived from or substantially identical to a corresponding portion of a human immunoglobulin (eg, at least 80%, 85%, 90%) , 92%, 95%, 97%, 98%, 99% or more of the same sequence.
- the immunoglobulin Fc domain in the antibody molecule of the invention is capable of extending the in vivo half-life of the antibody of the invention and providing effector function. See, for example, International Publication No. WO 98/23289; International Publication No. WO 97/34631; and U.S. Patent No. 6,277,375.
- the hinge region having a "CPPC" amino acid residue is contained in the Fc domain of the second polypeptide chain and the fourth polypeptide chain of the antibody molecule of the invention, respectively, and/or comprises Y349C and S354C, respectively. (According to Kabat's "EU No.”), whereby the second polypeptide chain and the fourth polypeptide chain of the antibody molecule of the present invention form an interchain disulfide bond in the Fc region, which also contributes to the antibody molecule of the present invention. Correct pairing of the two polypeptide chains and the fourth polypeptide chain.
- the immunoglobulin Fc domain of an antibody molecule of the invention also employs a "binding" technique that modifies the interface between different strands of an antibody molecule of the invention to facilitate the antibody molecule of the invention
- a "binding" technique that modifies the interface between different strands of an antibody molecule of the invention to facilitate the antibody molecule of the invention
- This technique involves introducing a "bump" at the interface of one strand, introducing a corresponding "hole” at the interface of the other strand to be paired with, such that the projection can be placed in the void.
- the first preferred interface comprises the CH3 domain of the heavy chain constant domain of one strand and the CH3 domain of the heavy chain constant domain of the other strand to be paired with.
- the bulges can be constructed by replacing small amino acid side chains from the interface of the CH3 domain of the heavy chain constant domain of one strand with a larger side chain (eg, tyrosine or tryptophan).
- a larger side chain eg, tyrosine or tryptophan
- the interface of the CH3 domain of the heavy chain constant domain of another strand to be paired is identical or similar to the bulge Compensatory holes of size.
- a second preferred interface is the CL domain comprising the light chain and the CH1 domain of the heavy chain of the Fab fragment described above, which facilitates the correct formation between the two strands of the Fab fragment by constructing a bulge-hole interaction. Heterodimerization.
- the Fc region of an antibody molecule of the invention comprises a modification to the binding affinity of an Fc receptor.
- the Fc receptor is an Fc gamma receptor, in particular a human Fc gamma receptor.
- the Fc receptor is an activating Fc receptor.
- the modification reduces the effector function of an antibody molecule of the invention.
- the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).
- the modification is in the Fc region of the immunoglobulin molecule, particularly in its CH2 region.
- the immunoglobulin molecule comprises an amino acid substitution at position 329 (EU numbering) of the immunoglobulin heavy chain.
- an antibody molecule of the invention comprises an amino acid substitution at positions 234 and 235 (EU numbering) of the immunoglobulin heavy chain.
- the amino acid substitutions are L234A and L235A (LALA mutations) (Armour KL et al, Recombinant human IgG molecules lacking Fcgamma receptor I binding and monocyte triggering activities. Eur J Immunol, 1999. 29(8): 2613 twenty four).
- an antibody molecule of the invention comprises an amino acid substitution at positions 234, 235 and 329 of the immunoglobulin heavy chain (EU numbering).
- the immunoglobulin molecule comprises amino acid substitutions L234A, L235A and P329G (EU numbering) in the immunoglobulin heavy chain.
- At least one single domain antigen binding site eg, two single domain antigen binding sites
- at least one Fab fragment in an antibody molecule of the invention are capable of specifically binding at least one antigen.
- the antibody molecule of the present invention binds two or more antigens, whereby the antibody molecule of the present invention is a multispecific antibody molecule, for example, a bispecific antibody molecule.
- antigens include, but are not limited to, cytokines, growth factors, hormones, signaling proteins, inflammatory mediators, ligands, cell surface receptors or fragments thereof.
- an antibody molecule of the invention inhibits signaling pathways of a plurality (eg, two) of immunological checkpoint molecules, eg, the antibody molecule of the invention has a first binding specificity for PD-L1 and for TIM -3.
- the antibody molecule of the invention inhibits signaling pathways of immunological checkpoint molecules and signaling pathways of agonistic costimulatory molecules, eg, the antibody molecules of the invention are directed against PD-L1, TIM-3, LAG- 3.
- the signaling pathway functions by signaling the signaling pathway of the costimulatory molecule.
- the antibody molecules of the invention inhibit signaling pathways of immunological checkpoint molecules and inhibit abnormal angiogenesis
- the antibody molecules of the invention are directed against PD-L1, TIM-3, LAG-3, PD- 1 or a bispecific antibody molecule of PD-L2 with a first binding specificity and a second binding specificity for VEGF or VEGF receptor, by inhibiting the signaling pathway of the immunological checkpoint molecule and by inhibiting VEGF, VEGF The signal transduction pathway of the body functions.
- the antibody molecules of the invention agonize signaling pathways of multiple (eg, two) costimulatory molecules, eg, the antibody molecules of the invention have a first binding specificity for OX40 and for GITR, 4- A second binding specific bispecific antibody molecule of 1BB, CD27 or CD28 acts by agonizing the signaling pathway of the costimulatory molecule.
- an antibody molecule of the invention agonizes a signaling pathway of a costimulatory molecule and inhibits abnormal angiogenesis
- the antibody molecule of the invention has a first binding to OX40, GITR, 4-1BB, CD27 or CD28 Bispecific antibody molecules that are specific and specific for the second binding specificity of VEGF or VEGF receptors act by agonizing the signaling pathway of the costimulatory molecule and by inhibiting the signaling pathways of VEGF, VEGF receptors.
- the antibody molecule of the invention inhibits abnormal angiogenesis, inhibits signaling pathways of immunological checkpoint molecules, and signaling pathways of agonistic costimulatory molecules, eg, the antibody molecules of the invention are directed against VEGF or VEGF receptors First binding specificity, second binding specificity for PD-L1, TIM-3, LAG-3, PD-1 or PD-L2 and third binding to OX40, GITR, 4-1BB, CD27 or CD28
- a specific trispecific antibody molecule functions by inhibiting a signaling pathway of VEGF, a VEGF receptor, a signal transduction pathway that inhibits the immunological checkpoint molecule, and a signaling pathway that agonizes the costimulatory molecule.
- an antibody molecule of the invention has any of the structures illustrated in Figures 1A-1D of the specification.
- an exemplary antibody molecule of the invention is a four-chain antibody molecule comprising two Fab fragments, a single domain antigen binding site located at the C-terminus of the light chain constant region (CL) of each Fab fragment, respectively. And an immunoglobulin Fc domain which is the C-terminus of the antibody molecule of the present invention, wherein there is or does not have a linker peptide between the C-terminus of the light chain constant region (CL) of the Fab fragment and the single domain antigen binding site.
- an exemplary antibody molecule of the invention is a four-chain antibody molecule comprising two Fab fragments, a single domain antigen binding at the N-terminus of the light chain variable domain (VL) of each Fab fragment, respectively.
- an exemplary antibody molecule of the invention is a four-chain antibody molecule comprising two Fab fragments, a single domain antigen binding site located at the C-terminus of the CH1 domain of each Fab fragment, and as a native An immunoglobulin Fc domain of the C-terminus of an antibody molecule, wherein there is or no linker peptide between the C-terminus of the Fab fragment CH1 domain and the single domain antigen binding site.
- an exemplary antibody molecule of the invention is a four-chain antibody molecule comprising two Fab fragments, single domain antigen binding at the N-terminus of the heavy chain variable domain (VH) of each Fab fragment, respectively.
- the antibody molecule of the invention is an anti-OX40/PD-L1 bispecific antibody or a multispecific antibody.
- OX40 (also known as CD134, TNFRSF4, and ACT35) is a member of the cell surface glycoprotein and tumor necrosis factor (TNF) receptor superfamily, expresses on T lymphocytes and provides a costimulatory signal for the proliferation and survival of activated T cells.
- TNF tumor necrosis factor
- OX40 was originally described as a T cell activation marker on rat CD4 T cells (Paterson DJ et al, Antigens of activated rat T lymphocytes including a molecule of 50,000 Mr detected only on CD4 positive T blasts. Mol Immunol. 1987; 24:1281 -1290) and subsequently shown to be up-regulated in TCR recruitment (Mallett S.
- OX40 signaling promotes costimulatory signals to T cells, resulting in enhanced cell proliferation, survival, effector function, and migration (Gramaglia I et al., Ox-40ligand: a potent costimulatory molecule for sustaining primary CD4T cell responses. J Immunol. 1998; 161:6510–6517; Gramaglia I et al, The OX40 costimulatory receptor determines the development of CD4 memory by regulating primary clonal expansion. J Immunol. 2000; 165:3043–3050).
- Anti-OX40 antibodies that are OX40 agonists are disclosed in the prior art.
- the amino acid sequences of the heavy chain variable region and the light chain variable region of the anti-OX40 antibody mAb 106-222 and humanized 106-222 (Hu106) are disclosed in WO 2012/027328; anti-OX40 antibody mAb 119-122 and The amino acid sequences of the heavy chain variable region and the light chain variable region of humanized 119-122 (Hu119).
- Anti-OX40 antibodies that are OX40 agonists are also disclosed in U.S. Patent No. 7,959,925, PCT Publication No. WO 2006/121810, and Chinese Patent Application No. 201710185399.9.
- the anti-OX40 antibody is capable of activating OX40, thereby inducing proliferation of effector T lymphocytes and promoting an immune response against tumor cells expressing tumor associated antigen (TAA).
- TAA tumor associated antigen
- PD-L1 (also known as differentiation antigen cluster 274 (CD274) or B7 homolog 1 (B7-H1)) is a 40 kDa type I transmembrane protein. PD-L1 binds to its receptor PD-1 present on activated T cells, downregulating T cell activation (Latchman et al, 2001 Nat Immunol 2: 261-8; Carter et al, 2002 Eur J Immunol 32: 634-43 ). PD-L1 expression has been found in many cancers, including human lung cancer, ovarian cancer, colon cancer, and various myeloma, and PD-L1 expression is often associated with poor prognosis of cancer (Iwai et al.
- the anti-OX40/PD-L1 bispecific antibody or multispecific antibody of the invention simultaneously targets at least OX40 and PD-L1 at the same time, and the Fab fragment and the single domain antigen binding site respectively bind to the OX40 or PD-L1 molecule, and can block
- the inhibitory PD-1/PD-L1 signaling pathway is disrupted and the OX40/OX40 ligand signaling pathway in T cells and natural killer (NK) cells is activated to promote an immune response against the disease.
- an antibody molecule of the invention comprises a single domain antigen binding site that specifically binds to PD-L1 and a Fab fragment that specifically binds OX40. In one embodiment, an antibody molecule of the invention comprises a single domain antigen binding site that specifically binds OX40 and a Fab fragment that specifically binds to PD-L1.
- the Fab fragment that specifically binds to PD-L1 or OX40 it comprises an anti-PD-L1 antibody (for example, the anti-PD-L1 antibody exemplified above) which is derived from any of the prior art and which is developed in the future.
- an anti-PD-L1 antibody for example, the anti-PD-L1 antibody exemplified above
- an anti-PD-L1 antibody for example, the anti-PD-L1 antibody exemplified above
- the anti-OX40 antibody is ADI-20112 having the heavy chain amino acid sequence set forth in SEQ ID NO: 10 and the light chain amino acid sequence set forth in SEQ ID NO: 15.
- the single domain antigen binding site that specifically binds to PD-L1 or OX40, it comprises a heavy chain variable domain (VH), a light chain variable domain (VL) that specifically binds to PD-L1 or OX40 a heavy chain variable domain in camelid antibodies consisting of only two heavy chains, which are naturally free of light chains from camelid serum, a VH-like single domain of IgNAR from sharks, and a camelized human VH structure. Domain, humanized camelid antibody heavy chain variable domain.
- an anti-OX40/PD-L1 bispecific antibody of the invention comprises two Fab fragments that specifically bind OX40 and two single domain antigen binding sites that specifically bind to PD-L1 (eg, VHH) ) has any of the structures illustrated in Figures 1A-1D, respectively.
- the two Fab fragments that specifically bind to OX40 specifically bind to the same epitope or different epitopes on the OX40 molecule; the two single domain antigen binding sites that specifically bind to PD-L1 specifically bind to PD-L1 The same epitope or different epitopes on the molecule.
- the Fab fragment that specifically binds OX40 in an anti-OX40/PD-L1 bispecific antibody of the invention comprises the paired weight of SEQ ID NO: 11/7 derived from the anti-OX40 antibody ADI-20112 All 6 heavy chain complementarity determining regions (CDRs) and light chain CDRs contained in the chain variable region sequence/light chain variable region sequence, or one or two with one or more of the 6 CDRs Sequences of one, three, four, five, six or more amino acid changes (eg, amino acid substitutions or deletions).
- CDRs heavy chain complementarity determining regions
- the Fab fragment that specifically binds OX40 in an anti-OX40/PD-L1 bispecific antibody of the invention comprises the paired weight of SEQ ID NO: 11/7 derived from the anti-OX40 antibody ADI-20112 a chain variable region sequence/light chain variable region sequence, or at least 90%, 91%, 92%, 93%, 94%, 95 with the paired heavy chain variable region sequence/light chain variable region sequence %, 96%, 97%, 98%, 99% or more sequences of sequence identity.
- the single domain antigen binding site that specifically binds to PD-L1 in an anti-OX40/PD-L1 bispecific antibody of the invention comprises CDR1, SEQ ID set forth in SEQ ID NO: CDR2 represented by NO: 4 and CDR3 represented by SEQ ID NO: 5, or one, two, three, four, five, six or one or more of the three CDRs More sequences of amino acid changes (eg, amino acid substitutions or deletions).
- the single domain antigen binding site that specifically binds to PD-L1 in an anti-OX40/PD-L1 bispecific antibody of the invention comprises SEQ ID NO: 1 and/or SEQ ID NO The amino acid sequence shown in 2, or a sequence substantially identical thereto (for example, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or more identical).
- the type of the heavy chain constant region CH1 domain and the Fc region (including the CH2 domain, the CH3 domain, and the optional CH4 domain) in the anti-OX40/PD-L1 bispecific antibody of the present invention is not particularly limited, and is preferably
- the corresponding domain derived from the IgG1, IgG2 or IgG4 immunoglobulin heavy chain constant region is substantially identical (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or more of the same sequence.
- the heavy chain constant region CH1 domain and Fc region are derived from, or substantially identical to, the CH1 domain and the Fc region of the heavy chain constant region of a human IgG1 immunoglobulin (eg, at least 80%, 85) Sequence of %, 90%, 92%, 95%, 97%, 98%, 99% or more of the same).
- an anti-OX40/PD-L1 bispecific antibody of the invention comprises a CH1 domain and an Fc region of an IgG4 (eg, human IgG4) heavy chain constant region.
- an anti-OX40/PD-L1 bispecific antibody of the invention comprises a CH1 domain and an Fc region of an IgG1 (eg, human IgG1) heavy chain constant region.
- an anti-OX40/PD-L1 bispecific antibody of the invention comprises a CH1 domain of an IgG4 (eg, human IgG4) heavy chain constant region and an Fc of an IgG1 (eg, human IgG1) heavy chain constant region a region; or a Fc region comprising a IgG1 (eg, human IgG1) heavy chain constant region and an IgG4 (eg, human IgG4) heavy chain constant region.
- the Fc domain of the second polypeptide chain and the fourth polypeptide chain of the anti-OX40/PD-L1 bispecific antibody of the invention comprises a hinge region having a "CPPC" amino acid residue, respectively, and / Or separately comprising Y349C and S354C (according to Kabat's "EU numbering"), whereby the second polypeptide chain and the fourth polypeptide chain of the anti-OX40/PD-L1 bispecific antibody of the invention form an interchain disulfide bond in the Fc region. Thereby, the correct pairing of the second polypeptide chain and the fourth polypeptide chain is stabilized.
- the second polypeptide chain and/or the fourth polypeptide chain of an anti-OX40/PD-L1 bispecific antibody of the invention comprises an amino acid mutation in the Fc domain that affects antibody effector function.
- the amino acid substitution is a LALA mutation.
- the anti-OX40/PD-L1 bispecific antibody of the invention comprises a kappa light chain constant region and/or a lambda light chain constant region, eg, a human kappa light chain constant region and/or a human lambda light chain is constant Area.
- the light chain constant region comprises or is substantially identical to the amino acid sequence set forth in SEQ ID NO: 8 (eg, at least 80%, 85%, 90%, 92%, 95%, 97%) , 98%, 99% or more of the same sequence.
- the second polypeptide chain and the fourth polypeptide chain of the anti-OX40/PD-L1 bispecific antibody of the invention comprise a stable association of "binding" in the respective Fc domains, respectively.
- an amino acid substitution T366W is included in one of the second polypeptide chain and the fourth polypeptide chain, and is included in the other of the second polypeptide chain and the fourth polypeptide chain Amino acid substitutions T366S, L368A and Y407V (EU numbering).
- the immunoglobulin CHI1 domain and the CL domain of the anti-OX40/PD-L1 bispecific antibody of the invention comprise a bulge or a hole, respectively, and the bulge or empty in the CH1 domain
- the wells can be placed in the holes or bulges in the CL domain, respectively, such that the first polypeptide chain and the second polypeptide chain of the anti-OX40/PD-L1 bispecific antibody of the invention also form an "inclusion" with each other.
- the stable association of the buckle is described in one embodiment, the immunoglobulin CHI1 domain and the CL domain of the anti-OX40/PD-L1 bispecific antibody of the invention.
- the anti-OX40/PD-L1 bispecific antibody of the invention consists of four polypeptide chains that are substantially symmetric about each other, wherein the two polypeptide chains of the left half of the antibody molecule comprise SEQ ID, respectively.
- the anti-OX40/PD-L1 bispecific antibody of the present invention is capable of binding to both PD-L1 and OX40 proteins simultaneously, and maintains the affinity constant of the parent antibody, thereby being able to block the PD-1/PD-L1 signaling pathway and Activate the OX40/OX40 ligand signaling pathway in T cells and natural killer (NK) cells.
- the anti-OX40/PD-L1 bispecific antibodies of the invention can be used in the treatment, prevention or diagnosis of diseases associated with such signaling pathways.
- the antibody molecule of the invention is an anti-VEGF/GITR bispecific antibody or a multispecific antibody.
- VEGF Vascular endothelial growth factor
- VPF vascular permeability factor
- Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid, Science, 1983, 219 (4587): 983-985
- VEGF is a signaling protein produced by cells that stimulate blood vessels.
- VEGF is a subfamily of growth factors that are a major class of signaling proteins involved in angiogenesis.
- Vascular endothelial growth factor and vascular endothelial growth inhibitory factor are simultaneously present in normal tissues and remain relatively balanced, which allows human blood vessels to be normally produced and differentiated.
- VEGF family molecules are proliferating, and the imbalance between regulation and angiogenesis inhibitors is greatly promoted, thereby greatly promoting the proliferation and migration of vascular endothelial cells, and improving vascular permeability.
- Inhibition of tumor cell apoptosis provides a good microenvironment for tumor growth and metastasis (Lapeyre-Prost A et al, Immunomodulatory Activity of VEGF in Cancer, Int Rev Cell Mol Biol. 2017; 330:295-342).
- the VEGF family contains six closely related polypeptides, each of which is a highly conserved homodimeric glycoprotein, with six subtypes: VEGF-A, -B, -C, -D, -E, and placental growth factor ( Placental growth factor (PLGF)), with molecular weights ranging from 35 to 44 kDa.
- VEGF-A including its splices such as VEGF 165
- VEGF-A is associated with microvessel density in some solid tumors
- the concentration of VEGF-A in tissues is associated with the prognosis of solid tumors such as breast, lung, prostate and colon cancers. .
- each VEGF family member is mediated by one or more of the cell surface VEGF receptor (VEGFR) family, including VEGFR1 (also known as Flt-1), VEGFR2 (also known as KDR). Flk-1), VEGFR3 (also known as Flt-4), etc., wherein VEGFR1 and VEGFR2 are closely related to angiogenesis, and VEGF-C/D/VEGFR3 is closely related to lymphangiogenesis.
- VEGFR1 also known as Flt-1
- VEGFR2 also known as KDR
- Flk-1 Flk-1
- VEGFR3 also known as Flt-4
- Bevacizumab (trade name: Avastin), developed by Genentech, is a recombinant human-mouse chimeric anti-VEGF antibody that blocks VEGFR from inactivation by blocking the binding of VEGF-A to VEGFR. This exerts an anti-angiogenic effect.
- Bevacizumab is currently used for first-line treatment of metastatic colorectal cancer, and may be used for the treatment of metastatic lung cancer, breast cancer, pancreatic cancer, and kidney cancer in the future. Bevacizumab is also one of the more successful antibody drugs developed.
- Glucocorticoid-induced tumor necrosis factor receptor (GITR, also known as TNFRSF18, activation-inducible TNFR family members (AITR), CD357 and GITR-D), is a tumor necrosis factor (tumor necrosis factor) , the 18th member of the TNF) receptor superfamily. Initially identified in a murine T cell line treated with dexamethasone (Nocentini G et al, A new member of the tumor necrosis factor/nerve growth factor receptor family inhibits T cell receptor-induced apoptosis, Proc Natl Acad Sci U S A. 1997; 94(12): 6216-21).
- TNF receptor superfamily include CD40, CD27, 4-1BB, and OX40.
- GITR expression is lower in naive CD4+ and CD8+ cells, it is constitutively expressed in regulatory T cells (Tone M et al., Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells, Proc Natl Acad Sci U S A. 2003; 100 (25): 15059-64).
- GITR expression is induced on effector T cells, effector T cell activation, proliferation, and cytokine production are promoted.
- CD4+CD25+ regulatory T cells Shimizu used mixed culture repression assays to report that GITR activation inhibits Treg function (Shimizu J et al., Stimulation of CD25(+)CD4(+) regulatory T cells through GITR Breaks immunological self-tolerance, Nature Immunology 2002; 3: 135-42).
- Anti-GITR antibody DTA-1 mediated GITR stimulation promotes anti-tumor immunity in a variety of tumor models (Cohen AD, Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor Accumulation, PLoS One. 2010; 5(5): e10436; Coe D et al, Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for cancer immunotherapy, Cancer Immunol Immunother, 2010; 59(9): 1367- 77).
- GITR is activated by binding to GITR ligand (GITRL), which is mainly expressed on APC. After activation, GITR can increase resistance to tumor and viral infection, participate in autoimmune processes/inflammatory processes, and regulate leukocyte extravasation.
- GITRL GITR ligand
- Anti-GITR antibodies are described in U.S. Patent No. 7,025,962, European Patent No. 1,947,183 B1, U.S. Patent No. 7,812,135, U.S. Patent No. 8,388,967, U.S. Patent No. 8,591,886, European Patent No. EP 1866339, PCT Publication No. WO 2011/028683, U.S. Patent No. 8,709,424, PCT Publication No. WO 2013/039954, International Publication No. WO 2013/039954, US Publication No. US 2014/0072566, International Publication No. WO 2015/026684, PCT Publication No. WO 2005/007190, PCT Publication No. WO 2007/133822 PCT Publication No.
- the anti-VEGF/GITR bispecific antibody or multispecific antibody of the present invention targets at least VEGF and GITR at the same time, and the Fab fragment and the single domain antigen binding site respectively bind to the VEGF or GITR molecule, and can block the VEGF family signaling pathway. And the function of activating effector T cells and suppressing Treg.
- an antibody molecule of the invention comprises a single domain antigen binding site that specifically binds to GITR and a Fab fragment that specifically binds to VEGF. In one embodiment, an antibody molecule of the invention comprises a single domain antigen binding site that specifically binds to VEGF and a Fab fragment that specifically binds to GITR.
- the Fab fragment that specifically binds GITR or VEGF comprises an anti-GITR antibody (e.g., an anti-GITR antibody exemplified above) derived from any of the prior art and an anti-GITR antibody VH/VL developed in the future.
- an anti-GITR antibody e.g., an anti-GITR antibody exemplified above
- the anti-VEGF antibody is Avastin having the heavy chain amino acid sequence set forth in SEQ ID NO: 19 and the light chain amino acid sequence set forth in SEQ ID NO: 18.
- the single domain antigen binding site that specifically binds GITR or VEGF, it comprises a heavy chain variable domain (VH), a light chain variable domain (VL), which is specifically binding to GITR or VEGF, from Camelidae
- VH heavy chain variable domain
- VL light chain variable domain
- the heavy chain variable domain of a camel antibody consisting of only two heavy chains, which is naturally free of light chains, the VH-like single domain of IgNAR from sharks, the camelized human VH domain, human origin Camelid antibody heavy chain variable domain.
- an anti-VEGF/GITR bispecific antibody of the invention comprises two Fab fragments that specifically bind to VEGF and two single domain antigen binding sites (eg, VHH) that specifically bind to GITR, each having Any of the structures illustrated in FIGS. 1A, 1B, 1D, 11A, and 11B.
- the two Fab fragments that specifically bind to VEGF specifically bind to the same epitope or different epitopes on the VEGF molecule; the two single domain antigen binding sites that specifically bind to GITR specifically bind to the same on the GITR molecule Epitope or different epitopes.
- the Fab fragment that specifically binds to VEGF in an anti-VEGF/GITR bispecific antibody of the invention comprises a pair of heavy chain variable regions of SEQ ID NO: 22/20 derived from the anti-VEGF antibody Avastin All six heavy chain complementarity determining regions (CDRs) and light chain CDRs contained in the sequence/light chain variable region sequence, or one, two, three with one or more of the six CDRs Sequence of four, five, six or more amino acid changes (eg, amino acid substitutions or deletions).
- CDRs heavy chain complementarity determining regions
- the Fab fragment that specifically binds to VEGF in an anti-VEGF/GITR bispecific antibody of the invention comprises a pair of heavy chain variable regions of SEQ ID NO: 22/20 derived from the anti-VEGF antibody Avastin a sequence/light chain variable region sequence, or at least 90%, 91%, 92%, 93%, 94%, 95%, 96% with the paired heavy chain variable region sequence/light chain variable region sequence , 97%, 98%, 99% or more sequences of sequence identity.
- the single domain antigen binding site that specifically binds to GITR in an anti-VEGF/GITR bispecific antibody of the invention comprises CDR1, SGGGFGD (SEQ ID NO: 25) ID NO: 26) CDR3 shown by CDR2 and ATDWRKP (SEQ ID NO: 27), or one, two, three, four, five with one or more of the three CDRs Sequence of one, six or more amino acid changes (eg, amino acid substitutions or deletions).
- the single domain antigen binding site that specifically binds to GITR in an anti-VEGF/GITR bispecific antibody of the invention comprises, or is essential to, the amino acid sequence set forth in SEQ ID NO: Sequences that are identical (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or more identical).
- the type of the heavy chain constant region CH1 domain and the Fc region (including the CH2 domain, the CH3 domain, and the optional CH4 domain) in the anti-VEGF/GITR bispecific antibody of the present invention is not particularly limited, and is preferably derived from
- the corresponding domain of the IgG1, IgG2 or IgG4 immunoglobulin heavy chain constant region is substantially identical (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99%) Or more of the same sequence.
- the heavy chain constant region CH1 domain and Fc region are derived from or substantially identical to the CH1 domain and the Fc region of the heavy chain constant region of a human IgG1 immunoglobulin (eg, at least 80%, 85) Sequence of %, 90%, 92%, 95%, 97%, 98%, 99% or more of the same).
- an anti-VEGF/GITR bispecific antibody of the invention comprises a CH1 domain and an Fc region of an IgG4 (eg, human IgG4) heavy chain constant region. In one embodiment, an anti-VEGF/GITR bispecific antibody of the invention comprises a CH1 domain and an Fc region of an IgG1 (eg, human IgG1) heavy chain constant region.
- an anti-VEGF/GITR bispecific antibody of the invention comprises a CH1 domain of an IgG4 (eg, human IgG4) heavy chain constant region and an Fc region of an IgG1 (eg, human IgG1) heavy chain constant region;
- the Fc region comprising the IgG1 (eg, human IgG1) heavy chain constant region and the IgG4 (eg, human IgG4) heavy chain constant region.
- the Fc domains of the second polypeptide chain and the fourth polypeptide chain of the anti-VEGF/GITR bispecific antibody of the invention comprise a hinge region having a "CPPC" amino acid residue, respectively, and/or respectively Containing Y349C and S354C ("EU number" according to Kabat), whereby the second polypeptide chain and the fourth polypeptide chain of the anti-VEGF/GITR bispecific antibody of the invention form an interchain disulfide bond in the Fc region, Thus, the correct pairing of the second polypeptide chain and the fourth polypeptide chain is stabilized.
- the second polypeptide chain and/or the fourth polypeptide chain of an anti-VEGF/GITR bispecific antibody of the invention comprises an amino acid mutation in the Fc domain that affects antibody effector function.
- the amino acid substitution is a LALA mutation.
- an anti-VEGF/GITR bispecific antibody of the invention comprises a kappa light chain constant region and/or a lambda light chain constant region, eg, a human kappa light chain constant region and/or a human lambda light chain constant region.
- the light chain constant region comprises or is substantially identical to the amino acid sequence set forth in SEQ ID NO: 8 (eg, at least 80%, 85%, 90%, 92%, 95%, 97%) , 98%, 99% or more of the same sequence.
- the second polypeptide chain and the fourth polypeptide chain of the anti-VEGF/GITR bispecific antibody of the invention comprise a stable association of "binding" in the respective Fc domains, respectively.
- an amino acid substitution T366W is included in one of the second polypeptide chain and the fourth polypeptide chain, and is included in the other of the second polypeptide chain and the fourth polypeptide chain Amino acid substitutions T366S, L368A and Y407V (EU numbering).
- the immunoglobulin CHI1 domain and the CL domain of the anti-VEGF/GITR bispecific antibody of the invention comprise a bulge or a hole, respectively, and the bulge or cavity in the CH1 domain may Separately placed in the holes or bulges in the CL domain, such that the first polypeptide chain and the second polypeptide chain of the anti-VEGF/GITR bispecific antibody of the invention also form a "binding" stability Association.
- the anti-VEGF/GITR bispecific antibody of the invention consists of four polypeptide chains that are substantially symmetric about each other, wherein the two polypeptide chains of the left half of the antibody molecule comprise SEQ ID NO: a first polypeptide chain represented by 18 and a second polypeptide chain of SEQ ID NO: 21; comprising a first polypeptide chain of SEQ ID NO: 18 and a second of SEQ ID NO: 28, respectively a polypeptide chain; or a sequence substantially identical (eg, at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher) to any of said sequences; Wherein the two polypeptide chains of the right half of the antibody molecule comprise a third polypeptide chain of SEQ ID NO: 18 and a fourth polypeptide chain of SEQ ID NO: 21, respectively; a third polypeptide chain represented by NO: 18 and a fourth polypeptide chain represented by SEQ ID NO: 28; or substantially identical to any of the sequences (eg.,
- the anti-VEGF/GITR bispecific antibody of the present invention is capable of binding to both GITR and VEGF proteins, and maintains the affinity constant of the parent antibody, thereby blocking the VEGF family signaling pathway and activating effector T cells and natural killing (NK) GITR/GITR ligand signaling pathway in cells.
- the anti-VEGF/GITR bispecific antibodies of the invention can be used in the treatment, prevention or diagnosis of diseases associated with such signaling pathways.
- amino acid sequence variants of the bispecific antibodies exemplified herein are contemplated.
- Amino acid sequence variants of bispecific antibodies can be made by introducing appropriate modifications to the nucleotide sequence encoding the bispecific antibody or by peptide synthesis. Such modifications include, for example, deletion of residues from within the amino acid sequence of the antibody and/or insertion of residues into and/or substitution of residues in the amino acid sequence. Any combination of deletions, insertions, and substitutions can be made to obtain the final construct, so long as the final construct possesses a desired characteristic, such as antigen binding.
- Amino acids can be grouped according to common side chain properties:
- Non-conservative substitutions will cause members of one of these categories to be exchanged for members of another classification.
- the antibody molecules of the invention are capable of recombinant fusion or chemical conjugation (including covalent and non-covalent conjugation) to a heterologous protein or polypeptide to produce a fusion protein.
- Methods of fusing or conjugating a protein, polypeptide or peptide to an antibody are known in the art. See, for example, U.S. Patent Nos. 5,336,603, 5,622,929 and EP 367,166.
- antibody molecules of the invention can be fused to a labeling sequence (e.g., a peptide) to facilitate purification.
- the labeled amino acid sequence is a hexahistidine peptide, such as the one provided in the pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), etc., many of which are commercially available. of. As described in Gentz et al., 1989, Proc. Natl. Acad. Sci. USA 86:821-824, for example, hexahistidine provides convenient purification of the fusion protein.
- peptide tags for purification include, but are not limited to, hemagglutinin ("HA") tags, which correspond to epitopes derived from influenza hemagglutinin proteins (Wilson et al., 1984, Cell 37: 767) and "flag" label.
- HA hemagglutinin
- an antibody molecule of the invention is conjugated to a diagnostic or detectable agent.
- a diagnostic or detectable agent e.g., to determine the efficacy of a particular therapy
- Such antibodies can be used as part of a clinical test (eg, to determine the efficacy of a particular therapy) for monitoring or predicting the onset, formation, progression, and/or severity of a disease or condition.
- Such diagnosis and detection can be accomplished by coupling an antibody to a detectable substance, including but not limited to a variety of enzymes such as, but not limited to, horseradish peroxidase, alkaline phosphatase, beta-galactose Glycosidase or acetylcholinesterase; prosthetic groups such as, but not limited to, streptavidin/biotin and avidin/biotin; fluorescent substances such as, but not limited to, umbelliferone, fluorescein, isothiocyanate , rhodamine, dichlorotriazinamide fluorescein, dansyl chloride or phycoerythrin; luminescent substances such as, but not limited to, luminol; bioluminescent substances such as, but not limited to, luciferase, luciferin and jellyfish Photoprotein; radioactive material such as, but not limited to, iodine ( 131 I, 125 I, 123 I and 121 I), carbon ( 14 C
- the invention also encompasses the use of antibody molecules conjugated to a therapeutic moiety.
- the antibody molecule can be conjugated to a therapeutic moiety, such as a cytotoxin (eg, a cytostatic or cytotoxic agent), a therapeutic agent, or a radioactive metal ion, such as an alpha emitter.
- a cytotoxin eg, a cytostatic or cytotoxic agent
- a therapeutic agent e.g, a cytostatic or cytotoxic agent
- a radioactive metal ion such as an alpha emitter.
- an antibody molecule of the invention can be conjugated to a therapeutic moiety or moiety that modulates a given biological response.
- the therapeutic or drug moiety should not be construed as being limited to classical chemotherapeutics.
- the drug moiety can be a protein, peptide or polypeptide possessing the desired biological activity.
- Such proteins may, for example, include toxins such as abrin, ricin A, Pseudomonas exotoxin, cholera toxin, or diphtheria toxin; proteins such as tumor necrosis factor, alpha interferon, beta interferon, nerve Growth factors, platelet-derived growth factors, tissue plasminogen activators, apoptotic agents, anti-angiogenic agents, or biological response modifiers, such as lymphokines.
- toxins such as abrin, ricin A, Pseudomonas exotoxin, cholera toxin, or diphtheria toxin
- proteins such as tumor necrosis factor, alpha interferon, beta interferon, nerve Growth factors, platelet-derived growth factors, tissue plasminogen activators, apoptotic agents, anti-angiogenic agents, or biological response modifiers, such as lymphokines.
- the antibody molecules of the invention can be conjugated to a therapeutic moiety such as a radioactive metal ion, such as an alpha-emitter such as 213 Bi or can be used to catalyze the emission of metal ions (including but not limited to 131 In, 131 LU, 131 Y, 131 Ho , 131 Sm) a macrocyclic chelating agent conjugated to the polypeptide.
- a macrocyclic chelating agent is 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA), which can be passed through a linker The molecule attaches to the antibody.
- linker molecules are well known in the art and are described in Denardo et al., 1998, Clin Cancer Res. 4(10):2483-90, each of which is incorporated by reference in its entirety.
- the antibody may also be linked to a solid support, which is particularly useful for immunoassays or purification of target antigens.
- solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
- the antibody molecules of the invention can be obtained, for example, by solid peptide synthesis (e.g., Merrifield solid phase synthesis) or recombinant production.
- a polynucleotide encoding any one of the polypeptide chains and/or a plurality of polypeptide chains of the antibody molecule is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
- the polynucleotide can be easily isolated and sequenced using conventional methods.
- a vector, preferably an expression vector, comprising one or more polynucleotides of the invention is provided.
- Expression vectors can be constructed using methods well known to those of skill in the art.
- Expression vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YAC).
- the expression vector can be transfected or introduced into a suitable host cell.
- a variety of techniques can be used to accomplish this, for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene guns, liposome-based transfection, or other conventional techniques.
- a host cell comprising one or more polynucleotides of the invention.
- a host cell comprising an expression vector of the invention.
- the term "host cell” refers to any type of cellular system that can be engineered to produce an antibody molecule of the invention.
- Host cells suitable for replicating and supporting expression of an antibody molecule of the invention are well known in the art. Such cells can be transfected or transduced with a specific expression vector, as desired, and a large number of cells containing the vector can be cultured for inoculating a large scale fermenter to obtain a sufficient amount of the antibody molecule of the present invention for clinical use.
- Suitable host cells include prokaryotic microorganisms such as E.
- coli eukaryotic microorganisms such as filamentous fungi or yeast, or various eukaryotic cells such as Chinese hamster ovary cells (CHO), insect cells, and the like.
- CHO Chinese hamster ovary cells
- Mammalian cell lines suitable for suspension culture can be used.
- Examples of useful mammalian host cell lines include SV40 transformed monkey kidney CV1 line (COS-7); human embryonic kidney line (HEK 293 or 293F cells), baby hamster kidney cells (BHK), monkey kidney cells (CV1), African green monkey kidney cells (VERO-76), human cervical cancer cells (HELA), canine kidney cells (MDCK), Buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (Hep G2), CHO cells, NSO cells, myeloma cell lines such as YO, NS0, P3X63, and Sp2/0.
- the host cell is a CHO, HEK293 or NSO cell.
- a method of producing an antibody molecule of the invention comprising culturing a host cell as provided herein under conditions suitable for expression of the antibody molecule, the host cell comprising the encoding A polynucleotide of an antibody molecule, and the antibody molecule is recovered from a host cell (or host cell culture medium).
- Antibody molecules prepared as described herein can be purified by known prior art techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like.
- the actual conditions used to purify a particular protein also depend on factors such as net charge, hydrophobicity, hydrophilicity, and the like, and these will be apparent to those skilled in the art.
- the purity of the antibody molecules of the present invention can be determined by any of a variety of well known analytical methods, including size exclusion chromatography, gel electrophoresis, high performance liquid chromatography, and the like.
- the physical/chemical properties and/or biological activities of the antibody molecules provided herein can be identified, screened or characterized by a variety of assays known in the art.
- compositions for example, pharmaceutical compositions comprising an antibody molecule described herein formulated together with a pharmaceutically acceptable carrier.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible.
- the pharmaceutical compositions of the invention are suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g., by injection or infusion).
- compositions of the invention may be in a variety of forms. These forms include, for example, liquid, semi-solid, and solid dosage forms, such as liquid solutions (for example, injectable solutions and infusible solutions), dispersions or suspensions, liposomes, and suppositories.
- liquid solutions for example, injectable solutions and infusible solutions
- dispersions or suspensions for example, liposomes, and suppositories.
- the preferred form depends on the intended mode of administration and therapeutic use.
- a common preferred composition is in the form of an injectable solution or an infusible solution.
- a preferred mode of administration is parenteral (eg, intravenous, subcutaneous, intraperitoneal (i.p.), intramuscular) injection.
- the antibody molecule is administered by intravenous infusion or injection.
- the antibody molecule is administered by intramuscular, intraperitoneal or subcutaneous injection.
- parenteral administration and “parenteral administration” as used herein mean modes of administration other than enteral administration and topical administration, usually by injection, and include, but are not limited to, intravenous, intramuscular, intraarterial, Intradermal, intraperitoneal, transtracheal, subcutaneous injection and infusion.
- compositions should generally be sterile and stable under the conditions of manufacture and storage.
- the compositions can be formulated as solutions, microemulsions, dispersions, liposomes or lyophilized forms.
- Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody molecule) in a suitable amount in a suitable solvent, followed by filter sterilization.
- dispersions are prepared by incorporating the active compound into a sterile vehicle containing base dispersion medium and other ingredients.
- a coating agent such as lecithin or the like can be used.
- the proper fluidity of the solution can be maintained by the use of surfactants.
- Prolonged absorption of the injectable compositions can be brought about by the inclusion in the compositions of the compositions which delay the absorption, such as the monostearate and gelatin.
- an antibody molecule of the invention can be administered orally, for example, orally with an inert diluent or an edible carrier.
- the antibody molecules of the invention may also be enclosed in hard or soft shell gelatin capsules, compressed into tablets or incorporated directly into the subject's diet.
- the compound can be incorporated with excipients and in ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, glutinous rice papers It is used in the form of a wafer or the like.
- Therapeutic compositions can also be administered using medical devices known in the art.
- compositions of the invention may comprise a "therapeutically effective amount” or a “prophylactically effective amount” of an antibody molecule of the invention.
- “Therapeutically effective amount” means an amount effective to achieve the desired therapeutic result at the desired dosage and for the period of time required.
- the therapeutically effective amount can vary depending on various factors such as the disease state, the age, sex, and weight of the individual.
- a therapeutically effective amount is any amount that is toxic or detrimental to a therapeutically beneficial effect.
- a "therapeutically effective amount” preferably inhibits a measurable parameter (eg, a tumor growth rate) of at least about 20%, more preferably at least about 40%, even more preferably at least about 60%, and still more, relative to an untreated subject. Preferably at least about 80%.
- the ability of an antibody molecule of the invention to inhibit measurable parameters e.g., tumor volume
- prophylactically effective amount is meant an amount effective to achieve the desired prophylactic result at the desired dosage and for the period of time required. Generally, the prophylactic amount is less than the therapeutically effective amount since the prophylactic dose is administered to the subject prior to the earlier stage of the disease or at an earlier stage of the disease.
- Kits comprising the antibody molecules described herein are also within the scope of the invention.
- the kit may contain one or more additional elements including, for example, instructions for use; other reagents, such as labels or reagents for coupling; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
- the antibody molecules disclosed herein have diagnostic and therapeutic and prophylactic uses in vitro and in vivo.
- these molecules can be administered to cultured cells in vitro or ex vivo or to a subject, eg, a human subject, to treat, prevent, and/or diagnose a variety of antigen-related diseases, such as cancer, autoimmune diseases. , acute and chronic inflammatory diseases, infectious diseases (for example, chronic infectious diseases or sepsis).
- the invention provides a diagnostic method for detecting a biological sample, such as serum, semen, or a urine or tissue biopsy sample (eg, from a hyperproliferative or cancerous lesion) in vitro or in vivo.
- the diagnostic method comprises: (i) contacting a sample (and optionally a control sample) with an antibody molecule as described herein or administering the antibody molecule to a subject, and (ii) allowing the interaction to occur.
- the formation of a complex between the antibody molecule and the sample (and optionally, the control sample) is detected. Formation of the complex indicates the presence of a relevant antigen and may indicate the suitability or need for the treatment and/or prevention described herein.
- the relevant antigen is detected prior to treatment, for example, prior to initiation of treatment or prior to treatment after the treatment interval.
- Detection methods that can be used include immunohistochemistry, immunocytochemistry, FACS, ELISA assays, PCR techniques (eg, RT-PCR), or in vivo imaging techniques.
- antibody molecules used in in vivo and in vitro assays are labeled, directly or indirectly, with a detectable substance to facilitate detection of bound or unbound conjugates.
- Suitable detectable materials include a variety of biologically active enzymes, prosthetic groups, fluorescent materials, luminescent materials, paramagnetic (eg, nuclear magnetic resonance) materials, and radioactive materials.
- the level and/or distribution of the relevant antigen is determined in vivo, eg, in a non-invasive manner (eg, by detecting using a suitable imaging technique (eg, positron emission tomography (PET) scan)) Labeled antibody molecules of the invention.
- a suitable imaging technique eg, positron emission tomography (PET) scan
- PET positron emission tomography
- the relevant antigen is determined in vivo, for example, by detecting an antibody molecule of the invention that is detectably labeled with a PET reagent (eg, 18 F-fluorodeoxyglucose (FDG)).
- FDG F-fluorodeoxyglucose
- the invention provides a diagnostic kit comprising the antibody molecule described herein and instructions for use.
- the invention relates to the use of an antibody molecule of the invention in vivo for the treatment or prevention of a condition in which an immune response is modulated in a subject, thereby inhibiting or reducing related diseases such as cancerous tumors, autoimmune diseases, acute and chronic The onset or recurrence of inflammatory diseases, infectious diseases (eg, chronic infectious diseases or sepsis).
- the antibody molecule of the present invention can be used alone.
- the antibody molecule can be administered in combination with other cancer therapeutic/preventive agents.
- the antibody molecule of the invention is administered in combination with one or more other drugs, such combinations can be administered in any order or simultaneously.
- the invention provides a method of modulating an immune response in a subject, the method comprising administering to the subject a therapeutically effective amount of an antibody molecule described herein.
- the invention provides a method of preventing the onset or recurrence of a disease in a subject, the method comprising administering to the subject a prophylactically effective amount of an antibody molecule described herein.
- cancers treated and/or prevented with antibody molecules include, but are not limited to, solid tumors, hematological cancers (eg, leukemias, lymphomas, myeloma, eg, multiple myeloma), and metastatic lesions.
- the cancer is a solid tumor.
- solid tumors include malignant tumors, for example, sarcomas and carcinomas of multiple organ systems, such as invasive lungs, breasts, ovaries, lymphoid, gastrointestinal (eg, colon), anal, genital, and genitourinary tract (eg, Kidney, bladder epithelium, bladder cells, prostate), pharynx, CNS (eg, brain, nerve or glial cells), head and neck, skin (eg, melanoma), nasopharynx (eg, differentiated or undifferentiated) Metastatic or locally recurrent nasopharyngeal carcinoma) and those of the pancreas, as well as adenocarcinomas, including malignant tumors such as colon cancer, rectal cancer, renal cell carcinoma, liver cancer, non-small cell lung cancer, small bowel cancer, and esophageal cancer. Cancer can be in early, intermediate or advanced stages or metastatic cancer.
- the cancer is selected from the group consisting of melanoma, breast cancer, colon cancer, esophageal cancer, gastrointestinal stromal tumor (GIST), renal cancer (eg, renal cell carcinoma), liver cancer, non-small cell lung cancer (NSCLC) ), ovarian cancer, pancreatic cancer, prostate cancer, head and neck cancer, stomach cancer, hematological malignancies (eg, lymphoma).
- GIST gastrointestinal stromal tumor
- renal cancer eg, renal cell carcinoma
- liver cancer eg, non-small cell lung cancer (NSCLC)
- ovarian cancer pancreatic cancer
- prostate cancer head and neck cancer
- stomach cancer hematological malignancies
- an infectious disease that is treated and/or prevented with an antibody molecule includes a pathogen that is currently in the absence of an effective vaccine or a pathogen to which a conventional vaccine is not fully effective.
- pathogens include, but are not limited to, HIV, (A, B, and C) hepatitis, flu, herpes, Giardia, malaria, Leishmania, Staphylococcus aureus, Pseudomonas aeruginosa.
- the blocking effect of the exemplified antibody molecules of the present invention on PD-L1 is particularly useful for combating infections established by pathogens (e.g., HIV) in which a variant antigen occurs as the infection progresses.
- variant antigens can be regarded as foreign antigens upon administration of an anti-human PD-L1 antibody, whereby the antibody molecules exemplified in the present invention are capable of eliciting a strong T cell response which is not inhibited by a negative signal by PD-L1.
- the immune system is downregulated by treatment and/or prevention of inflammatory and autoimmune diseases and graft versus host disease (GvHD) with an antibody molecule of the invention.
- autoimmune diseases that can be treated and/or prevented by administration of an antibody molecule of the invention include, but are not limited to, alopecia areata, ankylosing spondylitis, autoimmune hepatitis, Crohn's disease, lupus erythematosus, ulcerative colitis, uveitis Wait.
- inflammatory diseases that can be treated and/or prevented by administration of the antibody molecules of the invention include, but are not limited to, asthma, encephalitis, inflammatory bowel disease, allergic diseases, septic shock, pulmonary fibrosis, arthritis, and chronic virality. Or chronic inflammation caused by bacterial infections.
- Example 1 Construction, expression, purification and characterization of anti-OX40/PD-L1 bispecific antibody
- bispecific antibody Bi-110-112HC and its structure is shown in FIG. 1A
- Bispecific antibody Bi-113-112HC its structure is shown in Figure 1B
- bispecific antibody Bi-119-112LC its structure is shown in Figure 1C
- bispecific antibody Bi-122-112LC its structure is shown in Figure 1D.
- the four anti-OX40/PD-L1 bispecific antibodies are described separately below.
- the bispecific antibody Bi-110-112HC is composed of four polypeptide chains symmetrically bilaterally, wherein two polypeptide chains in the left half (ie, peptide chain #1 and peptide chain#) 2)
- the VL amino acid sequence represented by SEQ ID NO: 7 derived from the anti-OX40 antibody ADI-20112 is contained in the peptide chain #1 shown in SEQ ID NO: 6 from the N-terminus to the C-terminus, in the VL a human kappa light chain constant region (CL) amino acid sequence represented by SEQ ID NO: 8 at the C-terminus of the amino acid sequence, and a linker peptide represented by SEQ ID NO: 9 at the C-terminus of the human kappa light chain constant region (CL) amino acid sequence
- VH amino acid sequence represented by SEQ ID NO: 11 derived from the anti-OX40 monoclonal antibody ADI-20112, and the human IgG1 derived from the C-terminus of the VH amino acid sequence are contained in the peptide chain #2 shown in SEQ ID NO:
- the bispecific antibody Bi-113-112HC is composed of four polypeptide chains symmetrically bilaterally, wherein two polypeptide chains in the left half (ie, peptide chain #1 and peptide chain#) 2)
- the anti-PD-L1 VHH amino acid sequence represented by SEQ ID NO: 2 and the linker peptide represented by SEQ ID NO: 9 are included in the peptide chain #1 shown in SEQ ID NO: 14 from the N-terminus to the C-terminus.
- amino acid sequence The amino acid sequence, the VL amino acid sequence derived from the anti-OX40 antibody ADI-20112 shown in SEQ ID NO: 7, and the human kappa light chain constant region (CL) amino acid sequence set forth in SEQ ID NO: 8.
- Peptide chain #2 has the amino acid sequence shown in SEQ ID NO: 10.
- the bispecific antibody Bi-119-112LC is composed of four polypeptide chains symmetrically bilaterally, wherein two polypeptide chains in the left half (ie, peptide chain #1 and peptide chain#) 2)
- the amino acid sequence shown by SEQ ID NO: 15 and SEQ ID NO: 16 from the N-terminus to the C-terminus, respectively.
- the VL amino acid sequence derived from the anti-OX40 antibody ADI-20112 represented by SEQ ID NO: 7 and the SEQ ID NO: are included from the N-terminus to the C-terminus in the peptide chain #1 shown in SEQ ID NO: 15.
- the N-terminal to C-terminus of the peptide chain #2 shown in SEQ ID NO: 16 comprises the VH amino acid sequence derived from the anti-OX40 monoclonal antibody ADI-20112 represented by SEQ ID NO: 11, and the CH1 derived from human IgG1.
- the bispecific antibody Bi-122-112LC is composed of four polypeptide chains symmetrically left and right, and two polypeptide chains in the left half (ie, peptide chain #1 and peptide chain#) 2) having an amino acid sequence of SEQ ID NO: 15 and SEQ ID NO: 17 from the N-terminus to the C-terminus, respectively, wherein the peptide chain #2 comprises the SEQ ID NO: 2 from the N-terminus to the C-terminus.
- the anti-PD-L1 VHH amino acid sequence, the linker peptide amino acid sequence shown in SEQ ID NO: 9, the VH amino acid sequence derived from the anti-OX40 monoclonal antibody ADI-20112 shown in SEQ ID NO: 11, and the SEQ ID NO: 12 The CH1 amino acid sequence derived from human IgG1 and the amino acid sequence derived from the human IgG1 Fc region set forth in SEQ ID NO: 13.
- Example 1.2 Expression, purification and analysis of anti-OX40/PD-L1 bispecific antibodies
- nucleotide sequences encoding the peptide chain #1 and peptide chain #2 of the anti-OX40/PD-L1 bispecific antibody constructed in Example 1.1 were respectively ligated into the commercially available true
- the nuclear expression vector pTT5 was expressed and purified in eukaryotic cells, and anti-OX40/PD-L1 bispecific antibodies Bi-110-112HC, Bi-113-112HC, Bi-119-112LC and Bi-122-112LC were obtained. .
- the specific operation is as follows.
- the gene encoding the above-mentioned peptides of the bispecific antibodies Bi-110-112HC, Bi-113-112HC, Bi-119-112LC and Bi-122-112LC was synthesized by Genewiz. sequence.
- the nucleotide sequence encoding the synthesized peptide chain was ligated into the vector pTT5 using a suitable restriction enzyme and ligase, respectively, to obtain a recombinant vector containing the nucleotide sequence encoding the peptide chain, respectively.
- the recombinant vector was verified by sequencing and used for subsequent expression.
- HEK293 cells (purchased from Invitrogen) were subcultured in Expi293 cell culture medium (purchased from Invitrogen). The cell culture was centrifuged one day before the transfection to obtain a cell pellet, and the cells were suspended with fresh Expi293 cell culture medium to adjust the cell density to 1 ⁇ 10 6 cells/ml. The HEK293 cells were further cultured so that the cell density in the culture on the day of transfection was about 2 x 10 6 cells/ml. A final volume of HEK293 cell suspension of 1/10 F17 medium (purchased from Gibco, Cat. No. A13835-01) was used as a transfection buffer.
- the culture flask was supplemented with FEED (Sigma, catalog number: H6784-100G) at a concentration of 1/50 of the culture volume after transfection and a concentration of 1/50 of the culture volume after transfection. 200 g / L of glucose solution, gently mixed, placed in 8% CO 2 , 36.5 ° C continue to culture. After 20 hours, VPA (Gibco, catalog number: 11140-050) was added to a final concentration of 2 mM/L.
- FEED Sigma, catalog number: H6784-100G
- the specific affinity chromatography purification step is: using MabSelect SuRe (GE Healthcare, catalog number: 17-5438-03) affinity chromatography column, and placed in the AKTApure system.
- the AKTApure system equipped with a MabSelect SuRe affinity chromatography column was detoxified overnight with 0.1 M NaOH, and then the system was washed with 5 column volumes of binding buffer (Tris 20 mM, NaCl 150 mM, pH 7.2) and the column was equilibrated. The supernatant of the above filtered cells was passed through a column. The cells were re-equilibrated with 5 to 10 column volumes of binding buffer and monitored for UV-leveling using an ultraviolet detection device equipped with an AKTApure system.
- the antibody was eluted with an elution buffer (citric acid + sodium citrate 100 mM, pH 3.5), and samples were collected according to the ultraviolet absorption value. Each 1 ml of the collection solution was neutralized by adding 80 ⁇ l of a neutralization buffer (Tris-HCl 2M).
- the purity of the samples in the collected fractions was measured by size exclusion chromatography (SEC).
- SEC results are shown in Fig. 2A, Fig. 2B, Fig. 2C and Fig. 2D, respectively.
- the purity of the bispecific antibody Bi-110-112HC is 71.40%
- the purity of Bi-113-112HC is 84.54%
- the purity of Bi-119-112LC is 99.43. %
- Bi-122-112LC purity was 94.79%.
- the purified bispecific antibody solution was centrifuged at 4500 rpm for 30 minutes using a 15 ml ultrafiltration centrifuge tube.
- the protein was diluted with PBS, centrifugation was continued, and centrifugation was performed at 4500 rpm for 30 minutes, and the operation was repeated twice to replace the buffer.
- the antibodies after buffer exchange were combined and the antibody concentration was measured.
- the equilibrium dissociation constant (K D ) of the above-described exemplary anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention in combination with OX40 and PD-L1 was determined by a kinetic binding assay using an Octet system (manufactured by ForteBio) . ).
- the ForteBio affinity assay was performed according to the method reported in the literature (Estep, P et al, High throughput solution Based measurement of antibody-antigen affinity and epitope binning. MAbs, 2013, 5(2): p. 270-278). Briefly, AHC sensor (Pall, Cat. No.
- the anti-human IgG Fc biosensor AHC was immersed in wells containing the antibody solution, respectively, and immersed at room temperature for 600 seconds. The sensor was then washed in SD buffer until baseline was reached and then immersed in wells containing 100 ⁇ l of antigen solution to monitor binding of the antibody to the antigen. The sensor was then transferred to a well containing 100 ⁇ l of SD buffer to monitor antibody dissociation. The speed was 400 rpm and the temperature was 30 °C. The background corrected binding curves and dissociation curves were fitted by Octet analysis software (ForteBio) to generate binding (k on ) and dissociation (k dis ) rate constants which were subsequently used to calculate the equilibrium dissociation constant (K D ). The on , k dis and K D data of the bispecific antibody Bi-119-112LC and the antigen OX40 or PD-L1 are shown in Tables 1 and 2.
- the bispecific antibody Bi-119-112LC of the present invention is capable of simultaneously binding to PD-L1 and OX40 proteins in solution, and maintains the parent antibody ADI-20112 and humanized Nb-Fc and each corresponding antigen. Affinity constant.
- Example 1.4 Binding analysis of anti-OX40/PD-L1 bispecific antibody of the present invention to CHO cells overexpressing OX40 or PD-L1
- Binding of the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention to CHO cells overexpressing OX40 or PD-L1 was measured by FACS.
- ExpiCHO TM Expression System Kit (Invitrogen, catalog number: A29133), according to the manufacturer's instructions embodiment operates as follows: carrying cloned into the multiple cloning site MCS human PD-L1 cDNA (Sino Biological Inc. ) of The pCHO1.0 vector (Invitrogen) was transfected into Chinese hamster ovarian cancer cells (CHO) (Invitrogen) to produce CHO cells (CHO-PD-L1 cells) overexpressing human PD-L1. CHO-PD-L1 cells were counted, diluted to 1 ⁇ 10 6 cells/ml with a cell culture medium, and added to a U-bottom 96-well plate at 100 ⁇ l/well.
- the cell culture medium was removed by centrifugation at 400 g for 5 minutes on a centrifuge.
- 100 ⁇ l of the serially diluted bispecific antibody Bi-119-112LC of the present invention and the humanized Nb-Fc as a control were separately added to the U-shaped plate and the cells were resuspended and allowed to stand on ice for 30 minutes.
- the supernatant was removed, and unbound antibody was removed by washing the cells with PBS.
- 100 ⁇ l of 1:200 diluted PE-conjugated anti-human Fc antibody (SOUTHERN BIOTECH) was added to each well and incubated on ice for 30 minutes in the dark.
- the bispecific antibody Bi-119-112LC of the present invention is capable of binding to PD-L1 expressed on the cell surface, and has an EC50 of 2.654 nM, and PD-L1 expressed on the cell surface with the parent anti-PD-L1 antibody.
- the binding capacity (EC50 is 1.940nM) is similar.
- overexpression of human OX40 was generated by transfecting the human OX40 cDNA (Invitrogen) carrying human OX40 cDNA (Sino Biological Inc.) cloned into the multiple cloning site MCS into Chinese hamster ovarian cancer cells (CHO) (Invitrogen). CHO cells (CHO-OX40 cells).
- FACS detection was performed on CHO-OX40, except that the cells used were different and the ADI-20112 antibody was used as the control antibody, and the other experimental procedures were the same as those of the CHO-PD-L1 cells described above.
- the bispecific antibody Bi-119-112LC of the present invention is capable of binding to OX40 expressed on the cell surface, and has an EC50 of 3.195 nM, and the binding ability of the parent anti-OX40 antibody to OX40 expressed on the cell surface (EC50 is 2.193nM) is similar.
- Example 1.5 Analysis of the anti-OX40/PD-L1 bispecific antibody of the present invention simultaneously binding to CHO cells overexpressing OX40 and CHO cells overexpressing PD-L1 -
- the present embodiment detects the bispecific antibody by flow cytometry. Induction of different cell cross-linking conditions.
- the specific experimental process is as follows.
- CHO-PD-L1 cells and CHO-OX40 cells were obtained and cultured as described in Example 1.4. Cultures containing CHO-PD-L1 cells and CHO-OX40 cells were separately centrifuged at 400 g for 5 minutes on a centrifuge to remove the cell culture medium. After washing once with PBS, the cells were resuspended in PBS. The cells were counted and the cell density was adjusted to 2 x 10 6 cells/ml. The CHO-PD-L1 cells, and CHO-OX40 cells were 1: 5000 was added CellTracker TM Deep Red (Thermo) and Cell Trace CFSE (Invitrogen) dye, placed in 37 °C 30 minutes. The cells were centrifuged at 400 g for 5 minutes on a centrifuge, the supernatant was removed, and the cells were washed once with PBS.
- TM Deep Red Thermo
- Cell Trace CFSE Invitrogen
- the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC was able to induce cross-linking of CHO-PD-L1 cells and CHO-OX40 cells, thereby indicating the bispecificity of the present invention.
- Antibodies are capable of binding to target antigens from different cell surfaces simultaneously.
- the heavy chain (HC) amino acid sequence of the IgG1 negative control used in this example is shown in SEQ ID NO: 29; the light chain (LC) amino acid sequence of the IgG1 negative control is shown in SEQ ID NO:30.
- Anti-OX40/PD-L1 bispecific antibody of the invention blocks binding of PD-1 to CHO cells overexpressing PD-L1
- this example detects the anti-OX40/PD of the present invention by flow cytometry.
- the -L1 bispecific antibody blocks the binding of PD-1 protein to CHO cells overexpressing PD-L1.
- CHO-PD-L1 cells were obtained and cultured as described in Example 1.4.
- the culture containing 2.4 ⁇ 10 7 CHO-PD-L1 cells was centrifuged at 400 g for 5 minutes on a centrifuge to remove the cell culture medium. After washing once with PBS, the cells were resuspended in 5 ml of PBS.
- the IgG1 negative control used in this example was the same as the IgG1 negative control used in Example 1.5 above.
- Experimental Results As shown in Fig. 6 , the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention can effectively block the binding of PD-1 to CHO cells overexpressing PD-L1, and block the activity and
- the anti-PD-L1 humanized Nb-Fc antibody was similar (the IC50 of the anti-OX40/PD-L1 bispecific antibody was 3.522 nM, and the IC50 of the anti-PD-L1 humanized Nb-Fc antibody was 4.906 nM).
- Example 1.7 Detection of anti-PD-L1 activity of anti-OX40/PD-L1 bispecific antibody based on luciferase reporter gene
- this example uses a luciferase reporter gene detection cell line (Promega, CS187109) By detecting the expression of luciferase, the bispecific antibody inhibits the interaction of PD-1/PD-L1 interaction.
- a luciferase reporter gene detection cell line Promega, CS187109
- this example uses PD-1/PD-L1 Blockade Bioassay, Cell Propagation Model (Promega) The anti-PD-L1 biological activity of the bispecific antibody of the present invention was investigated.
- Promega's PD-1/PD-L1 Blockade Bioassay is a biologically relevant MOA-based assay for determining the potency and stability of antibodies that block PD-1/PD-L1 interaction.
- the assay consists of two genetically engineered cell lines:
- PD-1 effector cells Stable expression of human PD-1 and Jurkat T cells expressing luciferase by a nuclear factor of activated T cells (NFAT).
- PD-L1 aAPC/CHO-K1 cells CHO-K1 cells stably expressing human PD-L1 and cell surface proteins that activate the corresponding TCR in an antigen-independent manner.
- PD-1 binds to PD-L1 to block the transduction of NFAT downstream signals, thereby inhibiting the expression of luciferase.
- PD-1 antibody or PD-L1 antibody is added, this blocking effect is reversed.
- the photozyme is expressed to thereby detect a fluorescent signal.
- the detection method has good sensitivity, specificity and accuracy, and the stability is very good.
- PD-L1 aAPC/CHO-K1 cells were plated one day before the activity test: the culture supernatant was discarded, washed once with PBS, trypsin (Gibco, 25200072), incubated at 37 ° C for 3-5 min, using four volumes of 10
- the RPMI1640 (Gibco, 22400-071) medium of %FBS (HyClone, SH30084.03) was digested, the cells were collected, and a small amount of cell mixture was taken to determine the cell concentration. The required volume of cell fluid was taken, 400 g, centrifuged for 10 min, discarded.
- the cells were resuspended in RPMI 1640 (Gibco, 22400-071) medium containing 10% FBS (HyClone, SH30084.03) as an assay buffer so that the cell density was 4 ⁇ 10 5 cells/ml.
- the cell suspension was added to a 96-well white cell culture plate (Nunclon, 136101) at 100 ⁇ L/well, and a side hole of a 96-well white cell culture plate was added to PBS at 200 ⁇ l/well.
- the cells were cultured overnight in a carbon dioxide incubator at 37 ° C in a 5% CO 2 incubator.
- PD-1 effector cells were taken, counted, 400 g, centrifuged for 5 min, and the cells were resuspended in an assay buffer so that the cell concentration was 1.25 ⁇ 10 6 cells/ml.
- the IgG1 negative control used in this example was the same as the IgG1 negative control used in Example 1.5 above.
- Experimental Results As shown in Figure 7, the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention can effectively abolish the blocking effect of PD1/PD-L1 interaction on the NFAT signaling pathway, and the activity and anti-PD
- the -L1 humanized Nb-Fc antibody was similar (the EC50 of the anti-OX40/PD-L1 bispecific antibody was 0.4585 nM, and the EC50 of the anti-PD-L1 humanized Nb-Fc antibody was 0.3283 nM).
- the OX40-mediated signaling pathway biological activity was activated in the presence of CHO-PD-L1 cells as described in Example 1.4.
- This example uses the Jurkat-OX40-NFkB-Luc-Rep stable cell line of Cinda Biopharmaceutical (Suzhou) Co., Ltd. to measure OX40-mediated transcriptional activation to evaluate whether the anti-OX40/PD-L1 bispecific antibody of the present invention is Has activator activity against the OX40 antibody.
- Human OX40 construct purchased from Sino
- NFkB Human OX40 construct (purchased from Sino) and NFkB were introduced by anti-human CD3 (BD Biosciences, catalog number: 555329), anti-human CD28 (BD Biosciences, catalog number: 555725) plus the antibody of the present invention in solution.
- - luciferase construct promoter of NFkB -luc, Promega
- human OX40 overexpressing Jurkat cells obtained from the American ATCC for 16 hours, followed by addition of Bio-Glo TM reagent color.
- the specific experimental process is as follows:
- Solution preparation Analytical buffer: RPIM-1640 (90%) (Gibco, 22400-071), FBS (10%) (HyClone, SH30084.03), anti-human CD3 (2 ⁇ g/ml) (BD Biosciences, catalog number: 555329), anti-human CD28 (2 ⁇ g/ml) (BD Biosciences, catalog number: 555725), now available.
- the IgG1 negative control used in this example was the same as the IgG1 negative control used in Example 1.5 above.
- the results of the experiment are shown in Figure 8.
- the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention has a significant activation of the NFkB signaling pathway, while the anti-OX40 Antibody ADI-20112 detected a lower NFkB signaling pathway activation effect, and anti-PD-L1 humanized Nb-Fc antibody did not have an NFkB signaling pathway activation effect.
- the anti-OX40/PD-L1 bispecific antibody of the present invention shows that the NFkB signaling pathway downstream of OX40 can be better activated in the presence of PD-L1 expressing cells.
- Differential scanning fluorimetry provides information about the structural stability of a protein based on the fluorescence changes in the protein profile, detects changes in the conformation of the protein, and obtains the melting temperature (T m ) of the protein.
- T m melting temperature
- the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC antibody of the present invention was diluted to 1 mg/ml with a PBS solution, respectively.
- SYPRO Orange Protein Gel Stain (Gibco, catalog number: S6650)
- 196 ⁇ l of PBS was added, and SYPRO Orange Protein Gel Stain was diluted 50-fold.
- the bispecific antibody of the present invention has a T m of >60 ° C and, therefore, has good thermal stability.
- the present example evaluated the antibody by measuring the change in purity of the prepared antibody at 40 ° C for 0, 1, 3, 7, 10, 20, and 30 days. Long-term thermal stability.
- the initial purity of the prepared batch of Bi-119-112LC antibody was 92.91% as determined by SEC.
- the experimental method was as follows: The antibody sample was concentrated to 5 mg/ml (dissolved in PBS), dispensed in an EP tube, 200 ⁇ l/tube, and placed at 40 ° C in the dark. One tube was taken on days 0, 1, 3, 7, 10, 20, and 30, and the purity of the main peak of the monomer was measured by SEC-HPLC.
- the experimental results are shown in Table 4.
- the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention was allowed to stand at 40 ° C for 30 days, and the ratio of the main peak of the monomer was reduced by only 3.69%.
- the results indicate that the anti-OX40/PD-L1 bispecific antibody of the present invention has good thermal stability.
- Example 1.11 Detection of activation of human CD4 + T cells by anti-OX40/PD-L1 bispecific antibody of the present invention
- This example detects the activation of human CD4 + T cells by anti-OX40/PD-L1 bispecific antibody in vitro.
- the detailed experimental procedure is as follows:
- PBMC cells Resuscitate human PBMC cells (ALLCELLS, PB005F), the cells after standing for 3 hours are mononuclear cells, add 10ml AIM Medium CTS (GIBCO, A3021002) medium, adding IL4 (20ng/ml) (R&D, 204-IL), GM-CSF (10ng/ml) (R&D, 215-GM) to induce monocyte differentiation into dendritic cells (ie, DC cells), cultured until day 5, adding cytokine TNF ⁇ (1000 U/ml) that induces DC maturation (R&D, catalog number: 210-TA), RhIL-1 ⁇ (5 ng/ml) (R&D, catalog number :201-LB), RhIL-6 (10 ng/ml) (R&D, catalog number: 206-IL), 1 ⁇ M PGE (Tocris, catalog number: 2296), in a carbon dioxide incubator at 37 ° C, 5% CO 2 culture conditions Continue to culture for 2 days as mature DC cells (mo
- CD4 + T cells were cultured in Medium CTS medium.
- Add CD4 + T cells: anti-CD3/CD28 beads 1:1 to Dynabeads Human T-Activator CD3/CD28 (INVITROGEN, catalog number: 11131D), culture in a carbon dioxide incubator at 37 ° C, 5% CO 2 culture conditions 3 Days, performing bead stimulation on CD4 + T cells;
- the above isolated DC cells were mixed with bead-stimulated CD4 + T cells, and added to the Staphylococcal enterotoxin E superantigen (Toxin technology, catalog number: ET404) at a final concentration of 1 ng/ml, 200 ⁇ l per well, and 12,000 DC cells.
- 120,000 CD4 + T cells were added to the diluted antibody for 3 days.
- the expression of IL2 in each sample was detected by Cisbio IL2 detection kit (CISBIO, catalog number: 62HIL02PEG), and the expression of IL2 in different antibodies was reflected. The ability of the antibody to activate T cells.
- the anti-OX40/PD-L1 bispecific antibody Bi-119-112LC of the present invention can effectively activate human CD4 + T cells in vitro, and its activation effect is stronger than that of anti-PD-L1 humanized Nb-Fc.
- the antibody, anti-OX40 antibody ADI-20112 is stronger.
- bispecific antibody Bi-2-50 the structure of which is shown in Figure 11A
- the bispecific antibody Bi-2-51 has a structural schematic diagram as shown in Fig. 11B.
- the two anti-VEGF/GITR bispecific antibodies are described separately below.
- the bispecific antibody Bi-2-50 is composed of four polypeptide chains symmetrically left and right, and two polypeptide chains in the left half (i.e., peptide chain #1 and peptide chain#) 2)
- the VL amino acid sequence represented by SEQ ID NO: 20 derived from the anti-VEGF antibody Avastin and the SEQ ID NO at the C-terminus of the VL amino acid sequence are contained in the peptide chain #1 shown in SEQ ID NO: 18.
- the human kappa light chain constant region (CL) amino acid sequence shown in Figure 8 comprising the VH amino acid set forth in SEQ ID NO: 22 derived from the anti-VEGF monoclonal antibody Avastin in the peptide chain #2 set forth in SEQ ID NO:21. a sequence, a CH1 amino acid sequence represented by SEQ ID NO: 23 derived from human IgG1 at the C-terminus of the VH amino acid sequence, a linker peptide amino acid sequence represented by SEQ ID NO: 9 at the C-terminus of the CH1 amino acid sequence, and SEQ ID The anti-GITR VHH amino acid sequence shown by NO: 24, and the Fc region amino acid sequence derived from human IgG1 shown by SEQ ID NO: 13.
- the bispecific antibody Bi-2-51 is composed of four polypeptide chains symmetrically left and right, and two polypeptide chains in the left half (i.e., peptide chain #1 and peptide chain#) 2) The amino acid sequences shown in SEQ ID NO: 18 and SEQ ID NO: 28, respectively, from the N-terminus to the C-terminus.
- the N-terminal to C-terminus of the peptide chain #2 shown in SEQ ID NO: 28 comprises the VH amino acid sequence derived from the anti-VEGF monoclonal antibody Avastin represented by SEQ ID NO: 22, and the SEQ ID NO: 23
- Example 2.2 Expression, purification and analysis of anti-VEGF/GITR bispecific antibodies
- nucleotide sequences encoding the anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 constructed in Example 2.1 were respectively passed through the peptide sequences #1 and # ⁇ #2.
- the multiple cloning site was ligated into the commercially available eukaryotic expression vector pTT5, and expressed and purified in eukaryotic cells to obtain anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51.
- the anti-VEGF/GITR bispecific antibody After purification, the anti-VEGF/GITR bispecific antibody has good purity, and the main peak purity of the bispecific antibodies Bi-2-50 and Bi-2-51 are 99.57% and 99.48%, respectively.
- an antibody having the amino acid sequence of SEQ ID NO: 31 named "hcIgG-10", which comprises the SEQ ID NO: 24 from the N-terminus to the C-terminus is used.
- the anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 of the present invention were able to simultaneously bind VEGF165 (R&D, 293-VE-500) in solution. Binding to the GITR (AcroBiosystems, GIR-H5228-1MG) protein and maintaining the affinity constant of the parental antibody Avastin or hcIgG-10.
- Example 2.4 Binding analysis of anti-VEGF/GITR bispecific antibodies of the invention with CHO cells overexpressing VEGF or GITR
- Binding of the anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 of the present invention to CHO cells overexpressing VEGF or GITR was measured by FACS.
- the other specific experimental procedures were the same as in the above Example 1.4 except that the antibody and antigen used were different.
- the IgG1 negative control used in this example was the same as the IgG1 negative control used in Example 1.5 above. The result is shown in FIG.
- the anti-VEGF/GITR bispecific antibodies Bi-2-50 and Bi-2-51 of the present invention were both able to bind to GITR expressed on the cell surface with binding EC50 of 2.990 nM and 3.168 nM, respectively.
- the EC50 of the parental antibody hcIgG-10 binding to the cell surface GITR was 0.6061 nM.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Animal Behavior & Ethology (AREA)
- Wood Science & Technology (AREA)
- Biomedical Technology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Engineering & Computer Science (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Microbiology (AREA)
- General Chemical & Material Sciences (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Cell Biology (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Mycology (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
| 放置于40℃(天) | Bi-119-112LC |
| 0 | 92.91% |
| 1 | 91.51% |
| 3 | 91.10% |
| 7 | 90.93% |
| 10 | 90.45% |
| 20 | 89.72% |
| 30 | 89.22% |
Claims (24)
- 一种抗体分子,其包含(i)单结构域抗原结合位点;(ii)结合抗原的Fab片段;其中所述(i)位于所述(ii)的轻链可变结构域(VL)的N端或轻链恒定区(CL)的C端,或者所述(i)位于所述(ii)的重链可变结构域(VH)的N端或免疫球蛋白CH1结构域的C端,且所述(i)和(ii)分别结合相同或者不同的抗原,所述(i)和(ii)之间具有或者不具有连接肽;以及位于所述(i)和(ii)的C端的(iii)免疫球蛋白Fc结构域。
- 根据权利要求1所述的抗体分子,其中所述单结构域抗原结合位点选自重链可变结构域(VH)、轻链可变结构域(VL)、天然缺乏轻链的抗体的重链可变结构域(例如,骆驼科物种中天然存在的重链抗体的重链可变结构域)、鱼类中称为新型抗原受体(NAR)的免疫球蛋白(如鲨鱼血清中天然存在的IgNAR)中的VH样单结构域、和衍生自它们的经重组的单结构域抗原结合位点(例如,骆驼化的人VH结构域、人源化的骆驼科抗体重链可变结构域),例如,所述单结构域抗原结合位点选自骆驼科物种中天然存在的重链抗体的重链可变结构域、骆驼化的人VH结构域和人源化的骆驼科抗体重链可变结构域(它们简称为“VHH”)。
- 根据权利要求1或2所述的抗体分子,其中所述免疫球蛋白是IgG1、IgG2或IgG4免疫球蛋白,优选地,所述免疫球蛋白是IgG1免疫球蛋白,更优选地,所述免疫球蛋白是人IgG1免疫球蛋白;所述免疫球蛋白的轻链型别是κ型或λ型,优选地为κ型。
- 根据权利要求1-3中任一项所述的抗体分子,其中所述Fc结构域包含免疫球蛋白恒定部分的铰链区,并且所述抗体分子的重链彼此通过所述铰链区处的二硫键稳定缔合,例如,在所述抗体分子的重链的Fc结构域中包含具有“CPPC”氨基酸残基的铰链区,从而所述重链彼此通过所述铰链区处氨基酸残基之间形成的二硫键稳定缔合;优选地,所述抗体分子的重链在各自的Fc结构域中还分别包含Y349C和S354C或者S354C和Y349C(根据Kabat的“EU编号”),从而所述抗体分子的重链彼此在Fc区进一步形成链间二硫键。
- 根据权利要求1-4中任一项所述的抗体分子,其中所述Fc结构域中还包含影响抗体效应子功能的突变,例如,LALA突变。
- 根据权利要求1-5中任一项所述的抗体分子,其中所述抗体分子的重链在各自的Fc结构域中分别包含凸起或空穴,并且一条重链Fc结构域中的所述凸起或空穴可分别置于另一条重链Fc结构域中的所述空穴或凸起中,由此所述抗体分子的重链彼此形成“结入扣”的稳定缔合。
- 根据权利要求1-6中任一项所述的抗体分子,其中所述免疫球蛋白CH1结构域和轻链恒定结构域(CL)中分别包含凸起或空穴,并且CH1结构域中的所述凸起或空穴可分别置于CL结构域中的所述空穴或凸起中,由此所述抗体分子的重链和轻链彼此形成“结入扣”的稳定缔合。
- 根据权利要求1-7中任一项所述的抗体分子,其中所述(i)和(ii)分别结合相同抗原上的表位或者不同抗原上的表位,例如,所述(i)结合第一抗原的表位,所述(ii)结合第二抗原上的表位,由此,所述抗体分子是针对第一抗原和第二抗原的双特异性抗体。
- 根据权利要求1-8中任一项所述的抗体分子,其中所述连接肽是单独或组合使用的甘氨酸和/或丝氨酸残基,例如,所述连接肽包含氨基酸序列(Gly 4Ser)n,其中n是等于或大于1的正整数,例如,n是1-7中的正整数,例如,n是2。
- 根据权利要求1-9中任一项所述的抗体分子,其中所述抗原是细胞因子、生长因子、激素、信号传导蛋白、炎性介质、配体、细胞表面受体或其片段。
- 根据权利要求10所述的抗体分子,其中所述抗原选自肿瘤相关抗原、免疫检查点分子、血管新生诱导因子、肿瘤坏死因子受体超家族成员和免疫系统中的共刺激分子,以及这些分子的配体和/或受体。
- 根据权利要求11所述的抗体分子,其中所述抗原选自OX40、CD47、PD1、PD-L1、PD-L2、LAG-3、4-1BB(CD137)、VEGF和GITR。
- 根据权利要求1-12中任一项所述的抗体分子,其是包含四条多肽链的抗体分子,其中第一多肽链和第三多肽链中的每一多肽链包含免疫球蛋白轻链以及位于所述免疫球蛋白轻链可变结构域(VL)的N端的单结构域抗原结合位点,例如VHH;第二多肽链和第四多肽链中的每一多肽链包含免疫球蛋白重链;或者其中第一多肽链和第三多肽链中的每一多肽链包含免疫球蛋白轻链以及位于所述免疫球蛋白轻链恒定区(CL)的C端的单结构域抗原结合位点,例如VHH;第二多肽链和第四多肽链中的每一多肽链包含免疫球蛋白重链;或者其中第一多肽链和第三多肽链中的每一多肽链包含免疫球蛋白轻链;第二多肽链和第四多肽链中的每一多肽链包含免疫球蛋白重链以及位于所述免疫球蛋白重链N端的单结构域抗原结合位点,例如VHH;或者其中第一多肽链和第三多肽链中的每一多肽链包含免疫球蛋白轻链;第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变区、免疫球蛋白CH1结构域、单结构域抗原结合位点(例如VHH)、免疫球蛋白CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含单结构域抗原结合位点(例如VHH)、免疫球蛋白重链可变结构域(VH)和免疫球蛋白轻链恒定区(CL);第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)、免疫球蛋白CH1、CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)、免疫球蛋白轻链恒定区(CL)和单结构域抗原结合位点(例如VHH);第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)、免疫球蛋白CH1、CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)和免疫球蛋白轻链恒定区(CL);第二多肽链和第四多肽链中的每一多肽链从N端至C端包含单结构域抗原结合位点(例如VHH)、免疫球蛋白轻链可变结构域(VL)、免疫球蛋白CH1、CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)和免疫球蛋白轻链恒定区(CL);第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)、免疫球蛋白CH1结构域、单结构域抗原结合位点(例如VHH)、免疫球蛋白CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含单结构域抗原结合位点(例如VHH)、免疫球蛋白轻链可变结构域(VL)和免疫球蛋白CH1结构域;第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)、免疫球蛋白轻链恒定区(CL)、免疫球蛋白CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)、免疫球蛋白CH1结构域和单结构域抗原结合位点(例如VHH);第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)、免疫球蛋白轻链恒定区(CL)、免疫球蛋白CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)和免疫球蛋白CH1结构域;第二多肽链和第四多肽链中的每一多肽链从N端至C端包含单结构域抗原结合位点(例如VHH)、免疫球蛋白重链可变结构域(VH)、免疫球蛋白轻链恒定区(CL)、免疫球蛋白CH2、CH3和任选地CH4结构域;或者其中第一多肽链和第三多肽链中的每一多肽链从N端至C端包含免疫球蛋白轻链可变结构域(VL)和免疫球蛋白CH1结构域;第二多肽链和第四多肽链中的每一多肽链从N端至C端包含免疫球蛋白重链可变结构域(VH)、免疫球蛋白轻链恒定区(CL)、单结构域抗原结合位点(例如VHH)、免疫球蛋白CH2、CH3和任选地CH4结构域。
- 根据权利要求1-13中任一项所述的抗体分子,其是抗OX40/PD-L1双特异性抗体,且(i)单结构域抗原结合位点和(ii)结合抗原的Fab片段分别结合OX40或PD-L1分子,例如,所述抗体分子由左右基本上对称的4条多肽链组成,其中在左半部分的2条多肽链和右半部分的2条多肽链中,均包含(i)特异性结合PD-L1的单结构域抗原结合位点;(ii)特异性结合OX40的Fab片段;优选地,所述(i)单结构域抗原结合位点包含SEQ ID NO:3所示的CDR1、SEQ ID NO:4所示的CDR2和SEQ ID NO:5所示的CDR3,或者与所述3个CDR中的一个或多个CDR具有一个、两个、三个、四个、五个、六个或更多个氨基酸变化(例如,氨基酸置换或缺失)的序列;所述(ii)结合抗原的Fab片段包含衍生自抗OX40抗体的SEQ ID NO:11/7所示的成对重链可变区序列/轻链可变区序列中的全部6个重链互补决定区(CDR)与轻链CDR,或者与所述6个CDR中的一个或多个CDR具有一个、两个、三个、四个、五个、六个或更多个氨基酸变化(例如,氨基酸置换或缺失)的序列;更优选地,所述(i)单结构域抗原结合位点包含衍生自SEQ ID NO:1或SEQ ID NO:2所示的抗PD-L1VHH氨基酸序列,或与之基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更多同一)的序列;所述(ii)结合抗原的Fab片段包含衍生自抗OX40抗体的SEQ ID NO:11/7所示的成对重链可变区序列/轻链可变区序列,或与所述成对重链可变区序列/轻链可变区序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%或更多序列同一性的序列;最优选地,所述抗体分子的左半部分的2条多肽链分别包含SEQ ID NO:6所示的第一多肽链和SEQ ID NO:10所示的第二多肽链;分别包含SEQ ID NO:14所示的第一多肽链和SEQ ID NO:10所示的第二多肽链;分别包含SEQ ID NO:15所示的第一多肽链和SEQ ID NO:16所示的第二多肽链;分别包含SEQ ID NO:15所示的第一多肽链和SEQ ID NO:17所示的第二多肽链;或与任一所述序列基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更高同一)的序列;相应地,其中所述抗体分子的右半部分的2条多肽链分别包含SEQ ID NO:6所示的第三多肽链和SEQ ID NO:10所示的第四多肽链;分别包含SEQ ID NO:14所示的第三多肽链和SEQ ID NO:10所示的第四多肽链;分别包含SEQ ID NO:15所示的第三多肽链和SEQ ID NO:16所示的第四多肽链;分别包含SEQ ID NO:15所示的第三多肽链和SEQ ID NO:17所示的第四多肽链;或与任一所述序列基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更高同一)的序列。
- 根据权利要求1-13中任一项所述的抗体分子,其是抗VEGF/GITR双特异性抗体,且(i)单结构域抗原结合位点和(ii)结合抗原的Fab片段分别结合VEGF或GITR分子,例如,所述抗体分子由左右基本上对称的4条多肽链组成,其中在左半部分的2条多肽链和右半部分的2条多肽链中,均包含(i)特异性结合GITR的单结构域抗原结合位点;(ii)特异性结合VEGF的Fab片段;优选地,所述(i)单结构域抗原结合位点包含SEQ ID NO:25所示的CDR1、SEQ ID NO:26所示的CDR2和SEQ ID NO:27所示的CDR3,或者与所述3个CDR中的一个或多个CDR具有一个、两个、三个、四个、五个、六个或更多个氨基酸变化(例如,氨基酸置换或缺失)的序列;所述(ii)结合抗原的Fab片段包含衍生自抗VEGF抗体的SEQ ID NO:22/20所示的成对重链可变区序列/轻链可变区序列中的全部6个重链互补决定区(CDR)与轻链CDR,或者与所述6个CDR中的一个或多个CDR具有一个、两个、三个、四个、五个、六个或更多个氨基酸变化(例如,氨基酸置换或缺失)的序列;更优选地,所述(i)单结构域抗原结合位点包含衍生自SEQ ID NO:24所示的抗GITR VHH氨基酸序列,或与之基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更多同一)的序列;所述(ii)结合抗原的Fab片段包含衍生自抗VEGF抗体的SEQ ID NO:22/20所示的成对重链可变区序列/轻链可变区序列,或与所述成对重链可变区序列/轻链可变区序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%或更多序列同一性的序列;最优选地,所述抗体分子的左半部分的2条多肽链分别包含SEQ ID NO:18所示的第一多肽链和SEQ ID NO:21所示的第二多肽链;分别包含SEQ ID NO:18所示的第一多肽链和SEQ ID NO:28所示的第二多肽链;或与任一所述序列基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更高同一)的序列;相应地,其中所述抗体分子的右半部分的2条多肽链分别包含SEQ ID NO:18所示的第三多肽链和SEQ ID NO:21所示的第四多肽链;分别包含SEQ ID NO:18所示的第三多肽链和SEQ ID NO:28所示的第四多肽链;或与任一所述序列基本上同一(例如,至少80%、85%、90%、92%、95%、97%、98%、99%或更高同一)的序列。
- 多核苷酸,其编码权利要求1-15中任一项所述的抗体分子中的任意一条或者多条多肽链。
- 载体,优选地表达载体,其包含权利要求16的多核苷酸。
- 宿主细胞,其包含权利要求16所述的多核苷酸或权利要求17所述的载体,例如,所述宿主细胞是哺乳动物细胞,优选地是CHO细胞、HEK293细胞;所述宿主细胞是原核细胞,优选地是大肠杆菌细胞。
- 用于生产权利要求1-15中任一项所述的抗体分子的方法,所述方法包括步骤(i)在适于表达所述抗体分子的条件下培养权利要求18所述的宿主细胞,和(ii)从所述宿主细胞或所述培养基回收所述抗体分子。
- 药物组合物,其包含权利要求1-15中任一项所述的抗体分子和可药用载体。
- 根据权利要求20所述的药物组合物,其还包含至少一种其他有效成分。
- 免疫缀合物,其包含权利要求1-15中任一项所述的抗体分子和与所述抗体分子缀合的一个或多个异源分子,优选地,所述一个或多个异源分子是细胞毒性剂。
- 根据权利要求1-15中任一项所述的抗体分子、权利要求20-21所述的药物组合物、和权利要求22所述的免疫缀合物的用途,用作在个体中治疗和/或预防疾病的药物或用作疾病的诊断工具,优选地,所述个体是哺乳动物,更优选地是人。
- 根据权利要求23所述的用途,用于自身免疫病、急性和慢性炎性疾病、感染性疾病(例如,慢性传染病或败血症)、肿瘤的治疗和/或预防或诊断。
Priority Applications (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US16/965,229 US11746148B2 (en) | 2018-03-27 | 2019-03-26 | Antibody molecules comprising a single-domain antigen-binding site and Fab fragments |
| EP19775977.2A EP3778648A4 (en) | 2018-03-27 | 2019-03-26 | NOVEL ANTIBODY MOLECULE, METHOD OF PRODUCTION AND USE THEREOF |
| JP2020545187A JP7438958B2 (ja) | 2018-03-27 | 2019-03-26 | 新規な抗体分子、その調製方法及びその使用 |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN201810259102 | 2018-03-27 | ||
| CN201810259102.3 | 2018-03-27 | ||
| CN201910196438.4A CN110305210B (zh) | 2018-03-27 | 2019-03-15 | 新型抗体分子、其制备方法及其用途 |
| CN201910196438.4 | 2019-03-15 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2019184909A1 true WO2019184909A1 (zh) | 2019-10-03 |
Family
ID=68062524
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2019/079671 Ceased WO2019184909A1 (zh) | 2018-03-27 | 2019-03-26 | 新型抗体分子、其制备方法及其用途 |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2019184909A1 (zh) |
Cited By (12)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110531089A (zh) * | 2019-10-10 | 2019-12-03 | 首都医科大学附属北京同仁医院 | 鼻分泌物总IgE胶体金检测卡、试剂盒和应用 |
| US11066465B2 (en) | 2015-12-30 | 2021-07-20 | Kodiak Sciences Inc. | Antibodies and conjugates thereof |
| CN113214400A (zh) * | 2020-01-21 | 2021-08-06 | 甫康(上海)健康科技有限责任公司 | 一种双特异性抗pd-l1/vegf抗体及其用途 |
| US11155610B2 (en) | 2014-06-28 | 2021-10-26 | Kodiak Sciences Inc. | Dual PDGF/VEGF antagonists |
| EP3731875A4 (en) * | 2017-12-29 | 2021-12-08 | AP Biosciences, Inc. | MONOSPECIFIC AND BISPECIFIC PROTEINS AS REGULATORY OF IMMUNE CONTROL POINTS INTENDED FOR THE TREATMENT OF CANCER |
| WO2022002038A1 (zh) * | 2020-06-30 | 2022-01-06 | 和铂医药(上海)有限责任公司 | 免疫细胞衔接器多特异性结合蛋白及其制备和应用 |
| CN114106190A (zh) * | 2020-08-31 | 2022-03-01 | 普米斯生物技术(珠海)有限公司 | 一种抗vegf/pd-l1双特异性抗体及其用途 |
| JP2022553176A (ja) * | 2019-10-17 | 2022-12-22 | 江蘇康寧杰瑞生物制薬有限公司 | Ox40/pd-l1二重特異性抗体 |
| EP4093765A4 (en) * | 2020-01-21 | 2023-06-28 | Wuxi Biologics (Shanghai) Co. Ltd. | A bispecific anti-pd-l1/vegf antibody and uses thereof |
| US11912784B2 (en) | 2019-10-10 | 2024-02-27 | Kodiak Sciences Inc. | Methods of treating an eye disorder |
| US12071476B2 (en) | 2018-03-02 | 2024-08-27 | Kodiak Sciences Inc. | IL-6 antibodies and fusion constructs and conjugates thereof |
| US12409233B2 (en) | 2019-09-12 | 2025-09-09 | Biotheus Inc. | Anti-PD-L1 single-domain antibody and derivatives and use thereof |
Citations (38)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0367166A1 (en) | 1988-10-31 | 1990-05-09 | Takeda Chemical Industries, Ltd. | Modified interleukin-2 and production thereof |
| WO1994004678A1 (en) | 1992-08-21 | 1994-03-03 | Casterman Cecile | Immunoglobulins devoid of light chains |
| US5336603A (en) | 1987-10-02 | 1994-08-09 | Genentech, Inc. | CD4 adheson variants |
| US5622929A (en) | 1992-01-23 | 1997-04-22 | Bristol-Myers Squibb Company | Thioether conjugates |
| WO1997034631A1 (en) | 1996-03-18 | 1997-09-25 | Board Of Regents, The University Of Texas System | Immunoglobin-like domains with increased half lives |
| WO1998023289A1 (en) | 1996-11-27 | 1998-06-04 | The General Hospital Corporation | MODULATION OF IgG BINDING TO FcRn |
| US5766886A (en) | 1991-12-13 | 1998-06-16 | Xoma Corporation | Modified antibody variable domains |
| WO1999020758A1 (en) | 1997-10-21 | 1999-04-29 | Human Genome Sciences, Inc. | Human tumor necrosis factor receptor-like proteins tr11, tr11sv1, and tr11sv2 |
| WO1999040196A1 (en) | 1998-02-09 | 1999-08-12 | Genentech, Inc. | Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same |
| WO2001003720A2 (en) | 1999-07-12 | 2001-01-18 | Genentech, Inc. | Promotion or inhibition of angiogenesis and cardiovascularization by tumor necrosis factor ligand/receptor homologs |
| US6277375B1 (en) | 1997-03-03 | 2001-08-21 | Board Of Regents, The University Of Texas System | Immunoglobulin-like domains with increased half-lives |
| US6689607B2 (en) | 1997-10-21 | 2004-02-10 | Human Genome Sciences, Inc. | Human tumor, necrosis factor receptor-like proteins TR11, TR11SV1 and TR11SV2 |
| WO2004060319A2 (en) | 2002-12-30 | 2004-07-22 | 3M Innovative Properties Company | Immunostimulatory combinations |
| WO2005007190A1 (en) | 2003-07-11 | 2005-01-27 | Schering Corporation | Agonists or antagonists of the clucocorticoid-induced tumour necrosis factor receptor (gitr) or its ligand for the treatment of immune disorders, infections and cancer |
| WO2005035572A2 (en) | 2003-10-08 | 2005-04-21 | Domantis Limited | Antibody compositions and methods |
| WO2005055808A2 (en) | 2003-12-02 | 2005-06-23 | Genzyme Corporation | Compositions and methods to diagnose and treat lung cancer |
| WO2005115451A2 (en) | 2004-04-30 | 2005-12-08 | Isis Innovation Limited | Methods for generating improved immune response |
| US7025962B1 (en) | 1996-08-16 | 2006-04-11 | Schering Corporation | Mammalian cell surface antigens; related reagents |
| WO2006083289A2 (en) | 2004-06-04 | 2006-08-10 | Duke University | Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity |
| WO2006121810A2 (en) | 2005-05-06 | 2006-11-16 | Providence Health System | Trimeric ox40-immunoglobulin fusion protein and methods of use |
| WO2007005874A2 (en) | 2005-07-01 | 2007-01-11 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (pd-l1) |
| WO2007133822A1 (en) | 2006-01-19 | 2007-11-22 | Genzyme Corporation | Gitr antibodies for the treatment of cancer |
| EP1866339A2 (en) | 2005-03-25 | 2007-12-19 | TolerRx, Inc | Gitr binding molecules and uses therefor |
| US7618632B2 (en) | 2003-05-23 | 2009-11-17 | Wyeth | Method of treating or ameliorating an immune cell associated pathology using GITR ligand antibodies |
| WO2010077634A1 (en) | 2008-12-09 | 2010-07-08 | Genentech, Inc. | Anti-pd-l1 antibodies and their use to enhance t-cell function |
| WO2011028683A1 (en) | 2009-09-03 | 2011-03-10 | Schering Corporation | Anti-gitr antibodies |
| WO2011051726A2 (en) | 2009-10-30 | 2011-05-05 | Isis Innovation Ltd | Treatment of obesity |
| WO2012027328A2 (en) | 2010-08-23 | 2012-03-01 | Board Of Regents, The University Of Texas System | Anti-ox40 antibodies and methods of using the same |
| WO2012138475A1 (en) | 2011-04-08 | 2012-10-11 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Anti-epidermal growth factor receptor variant iii chimeric antigen receptors and use of same for the treatment of cancer |
| WO2013039954A1 (en) | 2011-09-14 | 2013-03-21 | Sanofi | Anti-gitr antibodies |
| EP1947183B1 (en) | 1996-08-16 | 2013-07-17 | Merck Sharp & Dohme Corp. | Mammalian cell surface antigens; related reagents |
| US8591886B2 (en) | 2007-07-12 | 2013-11-26 | Gitr, Inc. | Combination therapies employing GITR binding molecules |
| WO2014012479A1 (en) | 2012-07-18 | 2014-01-23 | Shanghai Birdie Biotech, Inc. | Compounds for targeted immunotherapy |
| US20140072566A1 (en) | 2012-06-08 | 2014-03-13 | National Cancer Center | Novel epitope for switching to th2 cell and use thereof |
| WO2015026684A1 (en) | 2013-08-20 | 2015-02-26 | Merck Sharp & Dohme Corp. | Modulation of tumor immunity |
| CN105143270A (zh) * | 2013-02-26 | 2015-12-09 | 罗切格利卡特公司 | 双特异性t细胞活化抗原结合分子 |
| WO2016065038A1 (en) * | 2014-10-23 | 2016-04-28 | Five Prime Therapeutics, Inc. | Slamf1 antagonists and uses thereof |
| CN107750164A (zh) * | 2015-06-08 | 2018-03-02 | 豪夫迈·罗氏有限公司 | 使用抗ox40抗体和pd‑1轴结合拮抗剂治疗癌症的方法 |
-
2019
- 2019-03-26 WO PCT/CN2019/079671 patent/WO2019184909A1/zh not_active Ceased
Patent Citations (43)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5336603A (en) | 1987-10-02 | 1994-08-09 | Genentech, Inc. | CD4 adheson variants |
| EP0367166A1 (en) | 1988-10-31 | 1990-05-09 | Takeda Chemical Industries, Ltd. | Modified interleukin-2 and production thereof |
| US5766886A (en) | 1991-12-13 | 1998-06-16 | Xoma Corporation | Modified antibody variable domains |
| US5622929A (en) | 1992-01-23 | 1997-04-22 | Bristol-Myers Squibb Company | Thioether conjugates |
| WO1994004678A1 (en) | 1992-08-21 | 1994-03-03 | Casterman Cecile | Immunoglobulins devoid of light chains |
| US5759808A (en) | 1992-08-21 | 1998-06-02 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
| WO1997034631A1 (en) | 1996-03-18 | 1997-09-25 | Board Of Regents, The University Of Texas System | Immunoglobin-like domains with increased half lives |
| US7025962B1 (en) | 1996-08-16 | 2006-04-11 | Schering Corporation | Mammalian cell surface antigens; related reagents |
| EP1947183B1 (en) | 1996-08-16 | 2013-07-17 | Merck Sharp & Dohme Corp. | Mammalian cell surface antigens; related reagents |
| WO1998023289A1 (en) | 1996-11-27 | 1998-06-04 | The General Hospital Corporation | MODULATION OF IgG BINDING TO FcRn |
| US6277375B1 (en) | 1997-03-03 | 2001-08-21 | Board Of Regents, The University Of Texas System | Immunoglobulin-like domains with increased half-lives |
| WO1999020758A1 (en) | 1997-10-21 | 1999-04-29 | Human Genome Sciences, Inc. | Human tumor necrosis factor receptor-like proteins tr11, tr11sv1, and tr11sv2 |
| US6689607B2 (en) | 1997-10-21 | 2004-02-10 | Human Genome Sciences, Inc. | Human tumor, necrosis factor receptor-like proteins TR11, TR11SV1 and TR11SV2 |
| WO1999040196A1 (en) | 1998-02-09 | 1999-08-12 | Genentech, Inc. | Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same |
| WO2001003720A2 (en) | 1999-07-12 | 2001-01-18 | Genentech, Inc. | Promotion or inhibition of angiogenesis and cardiovascularization by tumor necrosis factor ligand/receptor homologs |
| WO2004060319A2 (en) | 2002-12-30 | 2004-07-22 | 3M Innovative Properties Company | Immunostimulatory combinations |
| US7618632B2 (en) | 2003-05-23 | 2009-11-17 | Wyeth | Method of treating or ameliorating an immune cell associated pathology using GITR ligand antibodies |
| WO2005007190A1 (en) | 2003-07-11 | 2005-01-27 | Schering Corporation | Agonists or antagonists of the clucocorticoid-induced tumour necrosis factor receptor (gitr) or its ligand for the treatment of immune disorders, infections and cancer |
| WO2005035572A2 (en) | 2003-10-08 | 2005-04-21 | Domantis Limited | Antibody compositions and methods |
| WO2005055808A2 (en) | 2003-12-02 | 2005-06-23 | Genzyme Corporation | Compositions and methods to diagnose and treat lung cancer |
| WO2005115451A2 (en) | 2004-04-30 | 2005-12-08 | Isis Innovation Limited | Methods for generating improved immune response |
| WO2006083289A2 (en) | 2004-06-04 | 2006-08-10 | Duke University | Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity |
| US7812135B2 (en) | 2005-03-25 | 2010-10-12 | Tolerrx, Inc. | GITR-binding antibodies |
| US8388967B2 (en) | 2005-03-25 | 2013-03-05 | Gitr, Inc. | Methods for inducing or enhancing an immune response by administering agonistic GITR-binding antibodies |
| EP1866339A2 (en) | 2005-03-25 | 2007-12-19 | TolerRx, Inc | Gitr binding molecules and uses therefor |
| WO2006121810A2 (en) | 2005-05-06 | 2006-11-16 | Providence Health System | Trimeric ox40-immunoglobulin fusion protein and methods of use |
| US7959925B2 (en) | 2005-05-06 | 2011-06-14 | Providence Health System | Trimeric OX40-immunoglobulin fusion protein and methods of use |
| WO2007005874A2 (en) | 2005-07-01 | 2007-01-11 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (pd-l1) |
| WO2007133822A1 (en) | 2006-01-19 | 2007-11-22 | Genzyme Corporation | Gitr antibodies for the treatment of cancer |
| US8591886B2 (en) | 2007-07-12 | 2013-11-26 | Gitr, Inc. | Combination therapies employing GITR binding molecules |
| WO2010077634A1 (en) | 2008-12-09 | 2010-07-08 | Genentech, Inc. | Anti-pd-l1 antibodies and their use to enhance t-cell function |
| US8709424B2 (en) | 2009-09-03 | 2014-04-29 | Merck Sharp & Dohme Corp. | Anti-GITR antibodies |
| WO2011028683A1 (en) | 2009-09-03 | 2011-03-10 | Schering Corporation | Anti-gitr antibodies |
| WO2011051726A2 (en) | 2009-10-30 | 2011-05-05 | Isis Innovation Ltd | Treatment of obesity |
| WO2012027328A2 (en) | 2010-08-23 | 2012-03-01 | Board Of Regents, The University Of Texas System | Anti-ox40 antibodies and methods of using the same |
| WO2012138475A1 (en) | 2011-04-08 | 2012-10-11 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Anti-epidermal growth factor receptor variant iii chimeric antigen receptors and use of same for the treatment of cancer |
| WO2013039954A1 (en) | 2011-09-14 | 2013-03-21 | Sanofi | Anti-gitr antibodies |
| US20140072566A1 (en) | 2012-06-08 | 2014-03-13 | National Cancer Center | Novel epitope for switching to th2 cell and use thereof |
| WO2014012479A1 (en) | 2012-07-18 | 2014-01-23 | Shanghai Birdie Biotech, Inc. | Compounds for targeted immunotherapy |
| CN105143270A (zh) * | 2013-02-26 | 2015-12-09 | 罗切格利卡特公司 | 双特异性t细胞活化抗原结合分子 |
| WO2015026684A1 (en) | 2013-08-20 | 2015-02-26 | Merck Sharp & Dohme Corp. | Modulation of tumor immunity |
| WO2016065038A1 (en) * | 2014-10-23 | 2016-04-28 | Five Prime Therapeutics, Inc. | Slamf1 antagonists and uses thereof |
| CN107750164A (zh) * | 2015-06-08 | 2018-03-02 | 豪夫迈·罗氏有限公司 | 使用抗ox40抗体和pd‑1轴结合拮抗剂治疗癌症的方法 |
Non-Patent Citations (38)
| Title |
|---|
| "Fundamental Immunology", 1993, RAVEN PRESS |
| ARMOUR KL ET AL.: "Recombinant human IgG molecules lacking Fcgamma receptor I binding and monocyte triggering activities", EUR. J. IMMUNOL., vol. 29, no. 8, 1999, pages 2613 - 24, XP002501617, DOI: 10.1002/(SICI)1521-4141(199908)29:08<2613::AID-IMMU2613>3.0.CO;2-J |
| ARNON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", pages: 243 - 256 |
| BRINKMANN U.KONTERMANN R. E.: "The making of bispecific antibodies", MABS, vol. 9, no. 2, 2017, pages 182 - 212, XP055531122, DOI: 10.1080/19420862.2016.1268307 |
| CARTER ET AL., EUR. J. IMMUNOL., vol. 32, 2002, pages 634 - 43 |
| CHELIUS D ET AL.: "Structural andfunctional characterization of the trifunctional antibody Catumaxomab", MABS, vol. 2, 2010, pages 309 - 319, XP009137730, DOI: 10.4161/mabs.2.3.11791 |
| COE D ET AL.: "Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for cancer immunotherapy", CANCER IMMUNOL IMMUNOTHER, vol. 59, no. 9, 2010, pages 1367 - 77, XP019842210 |
| COHEN AD ET AL., CANCER IMMUNOL IMMUNOTHER |
| COHEN AD ET AL.: "Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation", PLOS ONE, vol. 5, no. 5, 2010, pages e10436, XP055150996, DOI: 10.1371/journal.pone.0010436 |
| CUZZOCREA S ET AL.: "Genetic and pharmacological inhibition of GITR-GITRL interaction reduces chronic lung injury induced by bleomycin instillation", FASEB J., vol. 21, no. 1, 2007, pages 117 - 129 |
| DENARDO ET AL., CLIN CANCER RES., vol. 4, no. 10, 1998, pages 2483 - 90 |
| ESTEP, P ET AL.: "High throughput solution Based measurement of antibody-antigen affinity and epitope binning", MABS, vol. 5, no. 2, 2013, pages 270 - 278, XP055105281, DOI: 10.4161/mabs.23049 |
| GENTZ ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 821 - 824 |
| GRAMAGLIA I ET AL.: "OX40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses", J IMMUNOL., vol. 161, 1998, pages 6510 - 6517, XP002109418 |
| GRAMAGLIA I ET AL.: "The OX40 costimulatory receptor determines the development of CD4 memory by regulating primary clonal expansion", J IMMUNOL., vol. 165, 2000, pages 3043 - 3050 |
| IWAI ET AL., PNAS, vol. 99, 2002, pages 12293 - 7 |
| JOHN B.B.RIDGWAY ET AL.: "Knobs-into-holes' engineering of antibody CH3 domains for heavy chain heterodimerization", PROTEIN ENGINEERING, vol. 9, no. 7, 1996, pages 617 - 21 |
| KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE |
| LAPEYRE-PROST A ET AL.: "Immunomodulatory Activity of VEGF in Cancer", INT REV CELL MOL BIOL., vol. 330, 2017, pages 295 - 342 |
| LATCHMAN ET AL., NAT. IMMUNOL., vol. 2, 2001, pages 261 - 8 |
| MALLETT S. ET AL.: "Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes-a molecule related to nerve growth factor receptor", EMBO J., vol. 9, 1990, pages 1063 - 1068 |
| NAGORSEN D.BAEUERLE P.A.: "Immunomodulatory therapy of cancer with T cell-engaging BiTE antibody blinatumomab", EXP. CELL RES., vol. 317, 2011, pages 1255 - 1260, XP028205663, DOI: 10.1016/j.yexcr.2011.03.010 |
| NOCENTINI G ET AL.: "A new member of the tumor necrosis factor/nerve growth factor receptor family inhibits T cell receptor-induced apoptosis", PROC NATL ACAD SCI USA., vol. 94, no. 12, 1997, pages 6216 - 21, XP002061244, DOI: 10.1073/pnas.94.12.6216 |
| OHIGASHI ET AL., CLIN CANCER RES., vol. 11, 2005, pages 2947 - 53 |
| OKAZAKI ET AL., INTERN. IMMUN., vol. 19, 2007, pages 813 - 24 |
| ORCUTT KD ET AL.: "A modular IgG-scFv bispecific antibody topology", PROTEIN ENG DES SEL., vol. 23, no. 4, 2010, pages 221 - 228 |
| PADLAN: "Anatomy of the antibody molecule", MOL. IMMUN., vol. 31, 1994, pages 169 - 217, XP023681629, DOI: 10.1016/0161-5890(94)90001-9 |
| PARDOLL DM.: "The Blockade of Immune Checkpoints in Cancer Immunotherapy", NAT REV CANCER, vol. 12, no. 4, 2012, pages 252 - 264, XP037114946, DOI: 10.1038/nrc3239 |
| PATERSON D. J. ET AL.: "Antigens of activated Rat T lymphocytes including a molecule of 50,000 M(r) detected only on CD4 positive T blasts", MOLECULAR IMMUNOLOGY, vol. 24, no. 12, 1987, pages 1281 - 1290, XP023682271, DOI: 10.1016/0161-5890(87)90122-2 |
| PATERSON DJ ET AL.: "Antigens of activated rat T lymphocytes including a molecule of 50, 000 Mr detected only on CD4 positive T blasts", MOLLMMUNOI., vol. 24, 1987, pages 1281 - 1290, XP023682271, DOI: 10.1016/0161-5890(87)90122-2 |
| SENGER, DR ET AL.: "Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid", SCIENCE, vol. 219, no. 4587, 1983, pages 983 - 985 |
| SHANE ATWELL ET AL.: "Stable heterodimers for remodeling the domain interface of a homodimer using a phage display library", J.MOL.BIOL., vol. 270, 1997, pages 26 - 35 |
| SHIMIZU J ET AL.: "Stimulation of CD25 (+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance", NATURE IMMUNOLOGY, vol. 3, 2002, pages 135 - 42, XP002247979, DOI: 10.1038/ni759 |
| THOMPSON ET AL., CANCER RES., vol. 66, 2006, pages 3381 - 5 |
| TONE M ET AL.: "Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells", PROC NATL ACAD SCI USA., vol. 100, no. 25, 2003, pages 15059 - 64, XP002307883, DOI: 10.1073/pnas.2334901100 |
| WILSON ET AL., CELL, vol. 37, 1984, pages 767 |
| YAO SZHU YCHEN L.: "Advances in Targeting Cell Surface Signaling Molecules for Immune Modulation", NAT REV DRUGDISCOV, vol. 12, no. 2, 2013, pages 130 - 146 |
| YAZAKIWU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268 |
Cited By (18)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US11155610B2 (en) | 2014-06-28 | 2021-10-26 | Kodiak Sciences Inc. | Dual PDGF/VEGF antagonists |
| US11066465B2 (en) | 2015-12-30 | 2021-07-20 | Kodiak Sciences Inc. | Antibodies and conjugates thereof |
| EP4268848A3 (en) * | 2017-12-29 | 2024-01-10 | AP Biosciences, Inc. | Monospecific and bispecific proteins with immune checkpoint regulation for cancer therapy |
| US11643470B2 (en) | 2017-12-29 | 2023-05-09 | Ap Biosciences, Inc. | PD-L1 and OX40 binding proteins for cancer Regulation |
| EP3731875A4 (en) * | 2017-12-29 | 2021-12-08 | AP Biosciences, Inc. | MONOSPECIFIC AND BISPECIFIC PROTEINS AS REGULATORY OF IMMUNE CONTROL POINTS INTENDED FOR THE TREATMENT OF CANCER |
| US12304961B2 (en) | 2017-12-29 | 2025-05-20 | Ap Biosciences, Inc. | PD-L1 and OX40 binding proteins for cancer regulation |
| US12071476B2 (en) | 2018-03-02 | 2024-08-27 | Kodiak Sciences Inc. | IL-6 antibodies and fusion constructs and conjugates thereof |
| US12409233B2 (en) | 2019-09-12 | 2025-09-09 | Biotheus Inc. | Anti-PD-L1 single-domain antibody and derivatives and use thereof |
| CN110531089A (zh) * | 2019-10-10 | 2019-12-03 | 首都医科大学附属北京同仁医院 | 鼻分泌物总IgE胶体金检测卡、试剂盒和应用 |
| US11912784B2 (en) | 2019-10-10 | 2024-02-27 | Kodiak Sciences Inc. | Methods of treating an eye disorder |
| JP2022553176A (ja) * | 2019-10-17 | 2022-12-22 | 江蘇康寧杰瑞生物制薬有限公司 | Ox40/pd-l1二重特異性抗体 |
| EP4047021A4 (en) * | 2019-10-17 | 2023-11-22 | Jiangsu Alphamab Biopharmaceuticals Co., Ltd. | BISPECIFIC OX40/PD1-L1 ANTIBODIES |
| EP4093765A4 (en) * | 2020-01-21 | 2023-06-28 | Wuxi Biologics (Shanghai) Co. Ltd. | A bispecific anti-pd-l1/vegf antibody and uses thereof |
| CN113214400A (zh) * | 2020-01-21 | 2021-08-06 | 甫康(上海)健康科技有限责任公司 | 一种双特异性抗pd-l1/vegf抗体及其用途 |
| CN113214400B (zh) * | 2020-01-21 | 2022-11-08 | 甫康(上海)健康科技有限责任公司 | 一种双特异性抗pd-l1/vegf抗体及其用途 |
| WO2022002038A1 (zh) * | 2020-06-30 | 2022-01-06 | 和铂医药(上海)有限责任公司 | 免疫细胞衔接器多特异性结合蛋白及其制备和应用 |
| CN114106190A (zh) * | 2020-08-31 | 2022-03-01 | 普米斯生物技术(珠海)有限公司 | 一种抗vegf/pd-l1双特异性抗体及其用途 |
| US12325759B2 (en) | 2020-08-31 | 2025-06-10 | Biotheus Inc. | Anti-VEGF-anti-PD-L1 bispecific antibody, pharmaceutical composition of same, and uses thereof |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| TWI754800B (zh) | 新型抗ox40/pd-l1雙特異性抗體分子、新型抗vegf/gitr雙特異性抗體分子及其用途 | |
| AU2018393424B2 (en) | Triabody, preparation method and use thereof | |
| WO2019184909A1 (zh) | 新型抗体分子、其制备方法及其用途 | |
| TWI793129B (zh) | 細胞毒性t淋巴球相關蛋白4 (ctla-4) 之新穎單株抗體 | |
| US20210009693A1 (en) | Novel anti-pd-l1 antibodies | |
| CA3170025A1 (en) | Pvrig binding protein and its medical uses | |
| WO2019129054A1 (zh) | 三链抗体、其制备方法及其用途 | |
| HK1215035A1 (zh) | 人源化抗cd134(ox40)抗體及其應用 | |
| CN110678484B (zh) | 抗pd-l1/抗lag3双特异性抗体及其用途 | |
| CN115943161A (zh) | 结合mait和肿瘤细胞两者的多特异性抗体 | |
| TWI858393B (zh) | 介白素2突變體以及其融合蛋白 | |
| CA3227854A1 (en) | Novel anti-sirpa antibodies | |
| WO2021143914A1 (zh) | 一种激活型抗ox40抗体、生产方法及应用 | |
| US20230174670A1 (en) | Anti-cd25 antibodies, antigen-binding fragments thereof, and medical uses thereof | |
| WO2019192493A1 (zh) | 抗人lag-3单克隆抗体及其应用 | |
| CN118339179A (zh) | 经修饰的蛋白或多肽 | |
| TWI835166B (zh) | 靶向pd-1和/或ox40的特異性結合蛋白及其應用 | |
| WO2025113640A1 (zh) | 结合lilrb1/2或pd1-lilrb1/2的抗体及其用途 | |
| WO2023236991A1 (zh) | 靶向her2,pd-l1和vegf的三特异性抗体 | |
| WO2024175030A1 (zh) | 抗Nectin-4抗体和包含其的多特异性抗体 | |
| WO2024235317A1 (zh) | 结合lilrb4和cd3的抗体及其用途 | |
| WO2025092742A1 (zh) | 结合bcma的抗体以及包含其的多特异性抗体 | |
| WO2024199294A1 (zh) | 一种靶向cd3的抗体或其抗原结合片段及其用途 | |
| CN117736323A (zh) | 抗pd-l1抗体及其用途 | |
| HK40010370A (zh) | 三链抗体、其制备方法及其用途 |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 19775977 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2020545187 Country of ref document: JP Kind code of ref document: A |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2019775977 Country of ref document: EP Effective date: 20201027 |
|
| WWW | Wipo information: withdrawn in national office |
Ref document number: 2019775977 Country of ref document: EP |