WO2023165516A1 - Anticorps bispécifique anti-pd-l1 et vegf et son utilisation - Google Patents

Anticorps bispécifique anti-pd-l1 et vegf et son utilisation Download PDF

Info

Publication number
WO2023165516A1
WO2023165516A1 PCT/CN2023/079017 CN2023079017W WO2023165516A1 WO 2023165516 A1 WO2023165516 A1 WO 2023165516A1 CN 2023079017 W CN2023079017 W CN 2023079017W WO 2023165516 A1 WO2023165516 A1 WO 2023165516A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
antigen
vegf
domain
region
Prior art date
Application number
PCT/CN2023/079017
Other languages
English (en)
Chinese (zh)
Inventor
张震
刘婵娟
郎国竣
王立燕
李文
李佳星
苏飞
Original Assignee
三优生物医药(上海)有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 三优生物医药(上海)有限公司 filed Critical 三优生物医药(上海)有限公司
Publication of WO2023165516A1 publication Critical patent/WO2023165516A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the invention belongs to the field of biomedicine, and in particular relates to a bispecific antibody targeting PD-L1 and VEGF and its preparation method and application.
  • PD-L1 belongs to a type of transmembrane protein on the cell membrane, and is expressed on T cells, B cells and other immune cells and tumor cells.
  • Tasuku Honjo et al. discovered and proved that when PD-L1 on the tumor cell membrane combines with PD-1 on immune cells such as T cells, the tumor cells send out inhibitory signals, and then T cells cannot recognize tumor cells and have a killing effect on tumor cells , the immune function of the body is suppressed (Chamoto, K., Al-Habsi, M., & Honjo, T. (2017). Current topics in microbiology and immunology, 410, 75-97).
  • pathological detection indicators such as PD-L1 expression level and tumor mutation burden (TMB) indicate a low response rate of anti-PD-1/PD-L1 antibodies, which currently only have an effect on less than 40% of solid tumors, and 30% of patients in the A certain amount of drug resistance appears after treatment.
  • TMB tumor mutation burden
  • VEGF-A also known as VEGF.
  • VEGF activates VEGFR2 (the main receptor tyrosine kinase receptor that mediates angiogenesis) to promote mitosis of vascular endothelial cells and increased vascular permeability, thereby promoting new vessel sprouting.
  • VEGFR2 the main receptor tyrosine kinase receptor that mediates angiogenesis
  • the high expression of VEGF in the tumor microenvironment can amplify the immunosuppressive effect of PD-(L)1. Therefore, targeting VEGF or VEGFR2 can effectively inhibit abnormal vascular proliferation (Ferrara, N. (2010). Mol Biol Cell 21 (5): 687-690.).
  • Bevacizumab The anti-VEGF monoclonal antibody Bevacizumab (Bevacizumab) has good efficacy and target safety in tumor treatment and eye diseases related to abnormal blood vessel proliferation (Pfisterer, J., et al. (2020). Lancet Oncol 21(5):699-709. Bhandari, S., et al. (2020). Ophthalmology 127(5):608-615.).
  • anti-VEGF monoclonal antibodies can improve blood vessels, promote T cell infiltration into tumors, inhibit DC maturation, promote T cell activation and activation, improve tumor microenvironment, and increase anti-PD-1/PD-L1 Drug response rate, further through T cell-mediated tumor killing, restore tumor immune function (Socinski, M.A., et al.(2018).N Engl J Med 378(24):2288-2301.N Engl J Med.2018Jun 14 ; 378(24):2288-2301. N Engl J Med 2020; 382:1894-1905).
  • bispecific antibodies have more potential in reducing toxic side effects and dosage, and have better safety; at the same time, anti-PD-L1 has better safety than anti-PD-1 It can better target into the tumor microenvironment, and can improve the toxic and side effects on normal tissues.
  • bispecific antibody drugs the biological activity of candidate antibodies is an important factor that needs to be taken into consideration. Although many bispecific antibodies have been proposed, due to the diversity of therapeutic functional and therapeutic behavior requirements of different therapeutic products, there is no theoretical prediction method that can be generally applied to most different desired molecular combinations. Therefore, it is often necessary to develop bispecific antibodies with good biological activity for specific targets and antibody compositions.
  • Anti-PD-L1 and VEGF bispecific antibodies in the prior art are mainly anti-PD-L1 antibody fused to VEGFR1 protein (US20200172623A1, WO2020200210A1) or Mab-VHH anti-PD-L1 and VEGF bispecific antibody (WO2021147829A1), these Bispecific antibodies all contain full-length IgG1 antibodies, which have problems of large molecular weight and druggability.
  • the inventors developed a bispecific nanobody targeting VEGF and PD-L1 at the same time, with a molecular weight of only 107KDa, which has potential With the advantages of strong tumor tissue infiltration and small dosage, it is superior to major competitors in terms of clinical dosage and safety. Further, the in vitro VEGF neutralizing activity of the anti-VEGF nanobody part of the bispecific antibody of the present invention is better than that of the marketed Bevacizumab, and in the animal model of angiogenesis, in the case of equal dose administration, the blood vessels in vivo The production inhibitory activity is also superior to that of Bevacizumab.
  • the anti-PD-L1 nanobody part of the bispecific antibody of the present invention is similar to the marketed Atezolizumab in terms of in vitro blocking activity and cell binding level. Furthermore, compared with monoclonal antibody combination and other bispecific antibodies of the prior art, the bispecific antibody of the present invention can simultaneously block PD-1/PD-L1 and VEGF/VEGFR signaling pathways, and improve blood vessel and At the same time, by restoring immunity, it has a more significant effect of inhibiting tumors, and has a very broad application prospect.
  • the present invention provides a bispecific antibody specifically binding to PD-L1 and VEGF, said antibody comprising one or two VHH domains specifically binding to PD-L1 (VHH PD-L1 ) and one or two VHH domains (VHH VEGF ) specifically binding to VEGF, wherein the VHH VEGF comprises the CDR1-3 sequence of the VHH domain shown in SEQ ID NO: 2, and preferably, wherein the VHH PD -L1 comprises the CDR1-3 sequence of the VHH domain shown in SEQ ID NO:1.
  • the VHH PD-L1 and VHH VEGF domains may be included in a tandem form on the same polypeptide chain (for example, by means of a linker and/or an Fc region in tandem on a polypeptide chain); or Linked to the immunoglobulin Fc region in a Fab-like structure (for example, by means of the immunoglobulin CH1 and CL constant domains to form a Fab-like structure).
  • the bispecific antibody comprises a polypeptide chain comprising a first VHH domain, an Fc region, a linker and a second VHH domain from the N-terminus to the C-terminus, wherein the first VHH domain and the second VHH domain are The two VHH domains specifically bind the first antigen and the second antigen respectively, wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF, preferably the first antigen is PD-L1 And the second antigen is VEGF.
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is linked at the N-terminus to the first VHH domain through an immunoglobulin hinge region sequence.
  • the Fc region polypeptide is connected to the second VHH domain at the C-terminus via a flexible linker peptide, such as (G 4 S) 3 (SEQ ID NO: 19).
  • the polypeptide chain comprises the amino acid sequence shown in SEQ ID NO: 7 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity amino acid sequence.
  • the bispecific antibody comprises a polypeptide chain comprising a first VHH domain, a linker, a second VHH domain and an Fc region from the N-terminus to the C-terminus, wherein the first VHH domain and The second VHH domain specifically binds the first antigen and the second antigen respectively, wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF, preferably the first antigen is PD- L1 and the second antigen is VEGF.
  • said first and second VHH domains are connected in tandem via a linker, eg ( G4S ) 3 (SEQ ID NO: 19).
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is at the N-terminus Linked to the second VHH domain by an immunoglobulin hinge region sequence.
  • the polypeptide chain comprises the amino acid sequence shown in SEQ ID NO: 8 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% thereof % identity amino acid sequence.
  • said bispecific antibody comprises a first and a second polypeptide chain, wherein,
  • the first polypeptide chain comprises a first VHH domain, a CH1 domain and an Fc region from the N-terminus to the C-terminus;
  • the second polypeptide chain comprises a second VHH domain and a CL domain from the N-terminus to the C-terminus;
  • first VHH domain and the CH1 domain in the first polypeptide chain are paired with the second VHH domain and the CL domain in the second polypeptide chain to form a Fab-like structure
  • the first VHH structural domain and the second VHH structural domain specifically bind the first antigen and the second antigen respectively, wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF, preferably the The first antigen is PD-L1 and the second antigen is VEGF.
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is connected to the CH1 domain at the N-terminus through an immunoglobulin hinge region sequence.
  • the first polypeptide chain comprises or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% of the amino acid sequence shown in SEQ ID NO: 9 or 11 Amino acid sequences with % or 99% identity.
  • the second polypeptide chain comprises or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% of the amino acid sequence shown in SEQ ID NO: 10 or 12 Amino acid sequences with % or 99% identity.
  • the present invention also provides nucleic acids encoding the polypeptide chains of the bispecific antibodies of the present invention, vectors and host cells comprising them.
  • the present invention also provides a pharmaceutical composition comprising the bispecific antibody of the present invention, and its in vitro and in vivo uses, especially in the treatment or diagnosis of diseases.
  • FIG. 1A-1D show schematic structures of candidate bispecific antibodies.
  • BsAb10 has the format of FIG. 1A;
  • BsAb11 has the format of FIG. 1B;
  • BsAb12 has the format of FIG. 1C;
  • BsAb13 has the format of FIG. 1D.
  • Figures 2A-2B show gel electrophoresis profiles of candidate bispecific antibodies.
  • the samples in Fig. 2A are BsAb10 and the reference IPI respectively; the samples in Fig. 2B are BsAb11, BsAb12, BsAb13 and the reference IPI respectively.
  • Figures 3A-3D show the SEC-HPLC monomer detection profiles of candidate bispecific antibodies.
  • Figure 3A is the SEC-HPLC monomer detection spectrum of BsAb10;
  • Figure 3B is the SEC-HPLC monomer detection spectrum of BsAb11;
  • Figure 3C is the SEC-HPLC monomer detection spectrum of BsAb12;
  • Figure 3D is the SEC-HPLC monomer detection spectrum of BsAb13 Atlas.
  • Figure 4 shows the binding activity of candidate bispecific antibodies to recombinant protein VEGF-His.
  • Figure 5 shows the binding activity of candidate bispecific antibodies to recombinant protein PD-L1-His.
  • Figure 6 shows the binding activity of candidate bispecific antibodies to huPD-L1-CHO-K cells.
  • Figures 7A-7B show the binding activity of candidate bispecific antibodies to both VEGF and PD-L1.
  • Figure 7A shows the activity of candidate bispecific antibodies that first bind to recombinant protein VEGF-Fc and then bind to recombinant protein PD-L1-Fc;
  • Figure 7B shows that candidate bispecific antibodies first bind to recombinant protein PD-L1-Fc and then bind to recombinant protein PD-L1-Fc Binding activity to recombinant protein VEGF-Fc.
  • Figure 8 shows the detection of the blocking activity of candidate bispecific antibodies against PD-1/PD-L1 by luciferase reporter gene method.
  • Figure 9 shows the detection of the blocking activity of candidate bispecific antibodies against VEGF/VEGFR2 by luciferase reporter gene method.
  • Figure 10 shows the inhibitory effect of candidate bispecific antibodies on tumor growth in the COLO205 mouse transplantation model.
  • Figure 11 shows the inhibitory effect of candidate bispecific antibodies on tumor growth in the A431 mouse transplantation model.
  • the term “comprising” or “comprising” means including stated elements, integers or steps, but not excluding any other elements, integers or steps. Where “comprising” is mentioned, the expression also covers “consisting of” unless the context clearly dictates otherwise. For example, when referring to a polypeptide chain comprising a specific amino acid sequence, it is also contemplated that the polypeptide chain consists of the specific amino acid sequence.
  • VEGF refers to vascular endothelial growth factor A (VEGF-A) protein.
  • VEGF-A produces multiple isoforms through alternative splicing of exons during transcription, isoforms such as VEGF121, VEGF165, VEGF189, and VEGF206.
  • VEGF refers in particular to the isoform VEGF165, for example the human VEGF165 protein described under UniProtKB-P15692 (in particular, the amino acid sequence without amino acids 27-191 of the signal peptide).
  • VEGF165 is active in angiogenesis, vessel formation, and endothelial cell growth; can induce endothelial cell proliferation, promote cell migration, inhibit apoptosis, and induce vascular permeability.
  • VEGF165 binds to the receptors FLT1/VEGFR1 and KDR/VEGFR2; and to the protein Neuropilin-1 (NRP1).
  • NRP1 Neuropilin-1
  • mice with NRP1 gene deletion on killer T cells produced better protection against secondary tumors, including relatively "cold” tumors such as melanoma in B16.F10 mice, against PD1
  • the response to immunotherapy was also more positive (Chang Liu et al, Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity, Nature Immunology (2020). DOI: 10.1038/s41590-020-0733-2).
  • antigen-binding specificity for VEGF refers to a binding site or a binding domain in a molecule that specifically binds to VEGF.
  • the term "PD-L1" refers to programmed cell death 1 ligand 1 (Programmed cell death 1 ligand 1). As a ligand for the inhibitory receptor PD-1, this protein regulates T cell activation thresholds and limits T cell effector responses.
  • the PD1/PD-L1-mediated immunosuppressive pathway can be used by tumors to attenuate anti-tumor immunity and evade clearance by the immune system, thereby promoting tumor survival. Blocking this pathway reverses the exhausted T cell phenotype and normalizes antitumor responses, facilitating cancer immunotherapy.
  • the term PD-L1 herein refers especially to human PD-L1 protein, eg human PD-L1 protein under accession number UniProtKB-Q9NZQ7.
  • antigen-binding specificity for PD-L1 refers to a binding site or binding domain in a molecule that specifically binds to PD-L1.
  • the terms "antigen-binding site” and “antigen-binding specificity” are used interchangeably to refer to the region of the antibody that actually binds to the antigen.
  • the PD-L1 antigen-binding site is provided by the VHH domain from anti-PD-L1; the VEGF antigen-binding site is provided by the VHH domain from anti-VEGF.
  • the term "bind” or “specifically bind” 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 a particular antigen can be determined by enzyme-linked immunosorbent assay (ELISA) or conventional binding assays known in the art, for example, by detecting antibodies by ELISA assays described in Examples 4.1, 4.2 or 4.4 The ability to bind to PD-L1 or VEGF protein or both, or to detect the ability of the antibody to bind to PD-L1 expressed on the cell surface by the FACS assay described in Example 4.3.
  • ELISA enzyme-linked immunosorbent assay
  • an antibody having "blocking activity" on PD-L1 means that the antibody blocks the binding of PD-L1 to the receptor PD-1, and/or reduces the function of PD-L1/PD-1 signal transmission.
  • Assays that can be used to determine this blocking activity can be FACS assays known in the art; or reporter gene-based signaling pathway blockade assays, such as the reporter gene-based assay described in Example 5.
  • the PD-L1 blocking activity of the antibody to be tested can be determined with reference to the PD-L1/PD-1 binding or PD-L1/PD-1 signaling level in the absence of the antibody and/or in the presence of a positive antibody.
  • the neutralizing activity of an antibody to VEGF refers to the function of the antibody to block the signaling pathway between VEGF and the receptor VEGFR2.
  • An assay that can be used to measure this activity can be, for example, a reporter gene-based signaling pathway blockade assay, such as the reporter gene-based assay described in Example 6.
  • the neutralizing activity of the antibody to be tested against VEGF can be determined with reference to the VEGF/VEGFR2 signaling level in the absence of the antibody and/or in the presence of a positive antibody.
  • variable region or “variable domain” of an antibody refers to the domains of the heavy or light chain of an antibody that participate in the binding of the antibody to an antigen.
  • the variable regions of antibodies can be further subdivided into hypervariable regions (ie, complementarity determining regions (CDRs)) and more conserved regions interspersed between hypervariable regions (ie, framework regions (FRs)).
  • CDRs complementarity determining regions
  • FRs framework regions
  • the antigen binding site is composed of a single heavy chain variable domain (also referred to herein as a VHH domain). supply.
  • this single heavy chain variable domain VHH comprises four FR regions and three CDR regions, and is arranged in the following order from the amino-terminus to the carboxy-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • CDR region or “CDR” or “hypervariable region”
  • VHH antibody variable domain
  • the CDRs are primarily responsible for binding to antigenic epitopes. Numbering sequentially from the N-terminus of the antibody chain, the CDRs located in the VHH variable region domain are called CDR1, CDR2, and CDR3.
  • CDR sequence Chothia based on the three-dimensional structure of the antibody and the topology of the CDR loop, Kabat ( Kabat et al., Sequences of Proteins of Immunological Interest, 4th Edition, USDepartment of Health and Human Services, National Institutes of Health (1987)), AbM (University of Bath), Contact (University College London), International ImMunoGeneTics database (IMGT ) (International Immunogenetics Information System, World Wide Web imgt.cines.fr/), and the North CDR definition based on affinity propagation clustering using a large number of crystal structures (North et al., "A New Clustering of Antibody CDR Loop Conformations", Journal of Molecular Biology, 406, 228-256 (2011)).
  • CDR or “CDR sequence” covers a CDR sequence determined in any of the above ways and combinations thereof.
  • VHH domain the antigen-binding site that specifically binds to the antigen is provided by the VHH domain.
  • VHH or "VHH domain” is used herein to refer to a heavy chain variable domain derived from a heavy chain antibody (herein sometimes referred to as a Nanobody) lacking a light chain, also referred to as a single Variable domain (sVD). Unlike the conventional VH of four-chain immunoglobulins, the VHH domain does not need to be paired with a light chain variable domain to form an antigen-binding site.
  • VHH molecules may be derived from antibodies produced in Camelidae species such as llamas, alpacas, dromedaries, llamas and guanacos. In some instances, for therapeutic use of a VHH, it is desirable to reduce its immunogenicity. Thus, preferably, in one embodiment, the invention provides antibodies comprising a humanized VHH domain.
  • the VHH domain from anti-PD-L1 and the VHH domain from anti-VEGF preferably via an immunoglobulin hinge region or a linker , separately connected to the opposite sides of the Fc region polypeptide of the antibody; or in the form of both in series (for example, from N-terminal to C-terminal, VHH PD-L1 -linker-VHH VEGF ; or VHH VEGF -linked Sub-VHH PD-L1 ), linked to the N-terminal side of the Fc region polypeptide of the antibody.
  • Fab-like structure is used herein to refer to, as shown in Figure 1C and Figure 1D, similar to that in a conventional four-chain IgG antibody consisting of a heavy chain variable region and a heavy chain constant region CH1 with complementary light chain variable regions and A structure formed by CL pairing of the light chain constant region, but in which the heavy chain variable region is replaced by a VHH domain and the light chain variable region is replaced by a different VHH domain.
  • the Fab-like structure comprises the first chain consisting of VHH PD-L1 and the CH1 domain of the constant region of the immunoglobulin heavy chain and the CL domain of the constant region of the immunoglobulin light chain consisting of VHH VEGF and composed of the second chain.
  • the Fab-like structure comprises a first chain consisting of VHH VEGF and the CH1 domain of the constant region of the immunoglobulin heavy chain and a CL structure composed of VHH PD-L1 and the constant region of the immunoglobulin light chain The domain consists of the second chain.
  • the VHH domain from anti-PD-L1 and the VHH domain from anti-VEGF are in a Fab-like structure, preferably by immunoglobulin
  • the hinge region is connected to the N-terminal side of the immunoglobulin Fc region polypeptide.
  • the Fab-like structure is linked to the Fc region polypeptide through its CH1 domain.
  • immunoglobulin molecule refers to a protein having the structure of a naturally occurring antibody.
  • IgG class immunoglobulins are heterotetrameric glycoproteins of approximately 150,000 Daltons consisting of two light chains and two heavy chains that are disulfide bonded. From N-terminus to C-terminus, each immunoglobulin heavy chain has a heavy chain variable region (VH), also called a heavy chain variable domain, followed by three heavy chain constant domains (CH1, CH2 and CH3 ). Similarly, from N-terminus to C-terminus, each immunoglobulin light chain has a light chain variable region (VL), also called a light chain variable domain, followed by a light chain constant domain (CL).
  • VH heavy chain variable region
  • CL light chain constant domain
  • an IgG immunoglobulin In an IgG molecule, usually the VH-CH1 of the heavy chain is paired with the VL-CL of the light chain to form a Fab fragment that specifically binds the antigen.
  • an IgG immunoglobulin essentially consists of two Fab molecules and two dimerized Fc regions linked by the immunoglobulin hinge region.
  • the heavy chains of immunoglobulins can be assigned to one of five classes, called ⁇ (IgA), ⁇ (IgD), ⁇ (IgE), ⁇ (IgG), or ⁇ (IgM), based on the type of their constant region, one of which is These classes can be further divided into subclasses such as ⁇ 1 (IgG1), ⁇ 2 (IgG2), ⁇ 3 (IgG3), ⁇ 4 (IgG4), ⁇ 1 (IgA1 ) and ⁇ 2 (IgA2).
  • the light chains of immunoglobulins can also be assigned to one of two types, called kappa and lambda, based on the amino acid sequence of their constant domains.
  • Immunoglobulin heavy chain constant region domain means a constant region domain from or obtained from or derived from an immunoglobulin heavy chain, including the heavy chain constant regions CH1, CH2 covalently linked sequentially from the N-terminus to the C-terminus , CH3 and optionally heavy chain constant region CH4. In most cases, heavy The chain constant regions CH1 and CH2 are connected by the hinge region of the heavy chain, but they can also be connected by a flexible linking peptide when appropriate.
  • the antibody of the present invention comprises a polypeptide chain consisting of immunoglobulin heavy chain constant region CH1-hinge region-CH2-CH3.
  • the antibody of the present invention comprises an Fc region polypeptide chain composed of immunoglobulin heavy chain constant regions CH2-CH3.
  • immunoglobulin constant domains can be selected based on the intended function of the antibody.
  • the constant domain may be an IgA, IgD, IgE, IgG or IgM domain, especially an immunoglobulin constant domain of human IgG, for example, a constant domain of human IgG1, IgG2, IgG3 or IgG4, preferably of human IgG1 constant domain.
  • the CH1 and Fc fragments of an antibody can both be from IgG1.
  • Immunoglobulin light chain constant region domain refers to a constant region domain CL from or obtained from or derived from an immunoglobulin light chain.
  • the light chain CL constant region of an immunoglobulin based on its amino acid sequence, may be a kappa light chain CL domain and a lambda light chain CL domain.
  • Fc domain or “Fc region” or “Fc region polypeptide” are used interchangeably to refer to a C-terminal region polypeptide containing at least a part of the constant region derived from an immunoglobulin heavy chain.
  • the term includes native sequence Fc region polypeptides and variant Fc region polypeptides.
  • a native immunoglobulin "Fc domain” comprises two or three constant domains, a CH2 domain, a CH3 domain and optionally a CH4 domain.
  • the immunoglobulin Fc domain comprises the second and third constant domains (CH2 and CH3 domains) of the heavy chain derived from antibodies of the IgG, IgA and IgD classes; and the second, third and fourth constant domains (CH2 domain, CH3 domain and CH4 domain) of the heavy chain of antibodies of the IgE class.
  • 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 described in 1991 (also known as the EU Index) is used for numbering.
  • Fc domain or "Fc region” or “Fc polypeptide” does not include the heavy chain variable region VH and the light chain variable region VL and the heavy chain constant region CH1 and the light chain constant region of an immunoglobulin cl.
  • “Native sequence Fc region” polypeptides encompass naturally occurring Fc region sequences of various immunoglobulins, such as the Fc region sequences of various Ig subtypes and their allotypes (Gestur Vidarsson et al., IgG subclasses and allotypes: from structure to effector functions , 20 October 2014, doi: 10.3389/fimmu.2014.00520.).
  • the human IgG heavy chain Fc region has an amino acid sequence extending from Cys226 or from Pro230 to the carboxy-terminus of the heavy chain.
  • the C-terminal terminal lysine (Lys447) of the Fc region may or may not be present.
  • the human IgG heavy chain Fc region bears at the N-terminus a hinge sequence or a partial hinge sequence of a native immunoglobulin, for example a sequence from E216 to T225 or a sequence from D221 to T225 according to EU numbering.
  • variant Fc region or “Fc region variant” polypeptide herein, used interchangeably herein, refers to an Fc region polypeptide comprising a modification relative to a native sequence Fc region polypeptide.
  • Fc region variant polypeptides of the invention are defined by the amino acid modifications that make them up.
  • L234A is an Fc region variant with alanine substituted for leucine at position 234 relative to the parental polypeptide, where numbering is according to the EU index.
  • Modifications can be additions, deletions or substitutions. Substitutions can include naturally occurring amino acids and non-naturally occurring amino acids. Variants may contain unnatural amino acids.
  • the purpose of the modification may be to alter the binding of the Fc region to its receptor and the effector functions elicited thereby.
  • effector functions refers to those biological activities attributable to the Fc region of an immunoglobulin 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 antigen uptake by antigen-presenting cells, downregulation of cell surface receptors (eg, B-cell receptors), and B-cell activation.
  • the antibody of the invention may have altered effector functions relative to an antibody with a wild-type Fc region, such as reduced or eliminated ADCC activity and the like.
  • the term "flexible linker” or “linker” or “linker” is used interchangeably to refer to a short amino acid sequence consisting of amino acids such as glycine (G) and/or serine ( S) and/or threonine residues (T), or from the hinge region of an immunoglobulin.
  • the connecting peptide is 5-50 amino acids in length, eg, 10, 15, 20, 25, 30 amino acids in length.
  • the connecting peptide comprises the amino acid sequence (G 4 S) n (SEQ ID NO: 20), wherein n is an integer equal to or greater than 1, for example, n is 2, 3, 4, 5, 6 or 7 an integer of .
  • the connecting peptide comprises the amino acid sequence TS(G 4 S) n (SEQ ID NO: 21), wherein n is an integer equal to or greater than 1, for example, n is 2, 3, 4, 5, 6 or Integer of 7.
  • the connecting peptide is a hinge region from an immunoglobulin, for example comprising a hinge region amino acid sequence of "CPPC", for example, the amino acid sequence "EPKSCDKTHTCPPCP” (SEQ ID NO: 22) or "EPKSSDKTHTCPPCP” (SEQ ID NO:23).
  • the connecting peptide that can be used to link the various domains in the antibody of the present invention can also be, for example but not limited to, the following amino acid sequence: GGG (SEQ ID NO: 24); DGGGS (SEQ ID NO: 25); TGEKP (SEQ ID NO :26); GGRR (SEQ ID NO:27); EGKSSGSGSESKVD (SEQ ID NO:28); KESGSVSSEQLAQFRSLD (SEQ ID NO:29); GGRRGGGS (SEQ ID NO:30); LRQRDGERP (SEQ ID NO:31); (SEQ ID NO:32) and GSTSGSGKPGSGEGSTKG (SEQ ID NO:33).
  • suitable flexible linker peptides can be rationally designed using computer programs to model the three-dimensional structures of proteins and peptides, or by phage display methods.
  • chimeric antibody is an antibody in which (a) the constant region, or part thereof, has been altered, substituted or exchanged such that the antigen binding site corresponds to a constant region of a different or altered class, effector function and/or species or linking entirely different molecules (e.g., enzymes, toxins, hormones, growth factors, drugs) etc. that confer new properties on the chimeric antibody; Variable regions are altered, substituted or swapped.
  • a “humanized antibody” is an antibody that retains the antigen-specific reactivity of a non-human antibody (eg, an alpaca heavy chain antibody), while being less immunogenic when administered to humans as a therapeutic. This can be achieved, for example, by retaining the non-human antigen binding sites and replacing the remainder of the antibodies with their human counterparts (ie, the constant regions and the parts of the variable regions that do not participate in binding are those of human antibodies).
  • a non-human antibody eg, an alpaca heavy chain antibody
  • Percent identity (%) of an amino acid sequence refers to after aligning a candidate sequence with the specific amino acid sequence shown in this specification and introducing gaps, if necessary, to achieve the maximum percent sequence identity, without taking into account any When conservative substitutions are taken as part of sequence identity, the percentage of amino acid residues in a candidate sequence that are identical to the amino acid residues of a particular amino acid sequence shown in this specification.
  • the invention contemplates variants of the antibodies of the invention that have a substantial degree of identity, for example at least 80%, 85% identity, relative to the antibodies and sequences thereof specifically disclosed herein. %, 90%, 95%, 97%, 98%, or 99% or higher. Such variants may contain conservative modifications.
  • conservative modifications include substitutions, deletions or additions to a polypeptide sequence that result in the substitution of an amino acid for a chemically similar amino acid.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologues and alleles of the invention.
  • the following 8 groups contain amino acids that are mutually conservative substitutions: 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), valine 6) phenylalanine (F), tyrosine (Y), tryptophan (W); 7) serine (S), threonine (T); and 8) cysteine Acid (C), Methionine (M) (See eg, 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 the amino acid sequence.
  • host cell refers to a cell into which an exogenous polynucleotide has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom.
  • a host cell is any type of cellular system that can be used to produce antibodies 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 cells within transgenic animals, transgenic plants, or cultured plant or animal tissues.
  • expression vector refers to a vector comprising a recombinant polynucleotide comprising expression control sequences operably linked to a nucleotide sequence to be expressed.
  • Expression vectors contain 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, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses) that incorporate recombinant polynucleotides. virus and adeno-associated virus).
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rodents). mouse).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rodents.
  • rodents e.g., mice and rodents.
  • an individual is a human being.
  • treatment refers to clinical intervention intended to alter the natural course of disease in the individual being treated. Desirable therapeutic effects include, but are not limited to, prevention of disease onset or recurrence, alleviation of symptoms, reduction of any direct or indirect pathological consequences of disease, prevention of metastasis, reduction of the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • the antibodies of the invention are used to delay the development of a disease or to slow the progression of a disease.
  • treatment encompasses anti-tumor biological effects that can be induced by human intervention (e.g., administration of drugs such as antibodies of the invention), including, but not limited to, for example, reduction in tumor volume, tumor Decreased cell number, decreased tumor cell proliferation, or decreased tumor cell survival.
  • human intervention e.g., administration of drugs such as antibodies of the invention
  • cancer and “tumor” are used interchangeably to refer to or describe a physiological disorder in mammals that is often characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, carcinoma, solid tumors, and liquid tumors.
  • the present invention provides a VHH domain-based bispecific antibody specifically binding to PD-L1 and VEGF, said antibody comprising a VHH domain specifically binding to PD-L1 (also abbreviated herein as VHH PD-L1 ) and a VHH domain specifically binding to VEGF (also abbreviated herein as VHH VEGF ).
  • the VHH PD-L1 and VHH VEGF domains may be in a tandem form contained on the same polypeptide chain (for example, by means of a linker and/or or Fc regions in series on one polypeptide chain); or can form a Fab-like structure (for example, by means of immunoglobulin CH1 and CL constant domains to form a Fab-like structure).
  • the bispecific antibody according to the invention further comprises an Fc region.
  • said VHH PD-L1 and VHH VEGF domains may be in a tandem form comprising both VHH PD-L1 and VHH VEGF domains (for example, as shown in Figure 1A and Figure 1B ), or in a Fab-like structure comprising both VHH PD-L1 and VHH VEGF domains (eg, as shown in Figure 1C and Figure 1D ), linked to the Fc region.
  • the bispecific antibody according to the present invention can be a tetravalent antibody that specifically binds PD-L1 and VEGF.
  • bispecific antibody of the present invention and the bispecific antibody of the present invention composed of them are described in detail below. Those skilled in the art can understand that unless the context clearly indicates otherwise, any combination of any technical features of these components is within the scope of the present invention. And, those skilled in the art will understand that unless the context clearly indicates otherwise, the antibodies of the invention (including antibodies in any form, such as diabodies and tetrabodies) may comprise any such combination of features.
  • VHH VEGF domain and VHH PD-L1 domain according to the invention
  • the antigen-binding site that specifically binds to PD-L1 is provided by the VHH PD-L1 according to the present invention; and
  • the antigen binding site that specifically binds VEGF is provided by the VHH VEGF according to the invention.
  • a VHH PD-L1 domain according to the invention comprises a VHH domain from an anti-PD-L1 Nanobody.
  • the VHH comprises CDR1, CDR2 and CDR3 sequences having the variable region shown in SEQ ID NO:1.
  • the CDR sequence range of the variable region SEQ ID NO: 1 amino acid sequence can be defined according to the Kabat, AbM, Chothia, Contact or IMGT schemes, or can be defined according to any two or more or all of these definition schemes. Those skilled in the art can easily know the CDR sequences defined by these definitions through http://www.abysis.org/abysis/.
  • the VHH PD-L1 domain according to the invention comprises the CDR1 sequence, the CDR2 sequence and the CDR3 sequence defined according to AbM in the variable region having SEQ ID NO:1.
  • the VHH PD-L1 domain according to the invention comprises
  • the VHH PD-L1 domain comprises the amino acid sequence shown in SEQ ID NO:1. In yet another embodiment, the VHH PD-L1 domain comprises at least 80%, 85%, 90%, 95% or 99% identity to SEQ ID NO: 1 and retains the ability to specifically bind PD-L1 amino acid sequence. In yet another preferred embodiment, the VHH PD-L1 domain comprises one or more (preferably 1-10, more preferably 1-5) amino acid additions compared to SEQ ID NO: 1, Amino acid sequences that are deleted and/or substituted (eg, conservatively substituted) and retain the ability to specifically bind PD-L1. Preferably, said amino acid additions, deletions and/or substitutions do not occur in the CDR regions. Most preferably, in the bispecific antibody according to the present invention, the VHH PD-L1 domain according to the present invention comprises the amino acid sequence of SEQ ID NO: 1, or consists of the amino acid sequence shown in SEQ ID NO: 1.
  • a VHH VEGF domain according to the invention comprises a VHH domain from an anti-VEGF Nanobody.
  • the VHH comprises CDR1, CDR2 and CDR3 sequences of the variable region shown in SEQ ID NO:2.
  • the CDR sequence range of the variable region SEQ ID NO: 2 amino acid sequence can be defined according to the Kabat, AbM, Chothia, Contact or IMGT schemes, or can be defined according to any two or more or all of these definition schemes. Those skilled in the art can easily know the CDR sequences defined by these definitions through http://www.abysis.org/abysis/.
  • the VHH VEGF domain according to the invention comprises the CDR1 sequence, the CDR2 sequence and the CDR3 sequence defined according to AbM in the variable region having SEQ ID NO:2.
  • the VHH VEGF domain according to the invention comprises
  • the VHH VEGF domain comprises the amino acid sequence shown in SEQ ID NO:2. In yet another embodiment, the VHH VEGF domain comprises an amino acid sequence that is at least 80%, 85%, 90%, 95% or 99% identical to SEQ ID NO: 2 and retains the ability to specifically bind VEGF. In yet another preferred embodiment, the VHH VEGF domain comprises one or more (preferably 1-10, more preferably 1-5) amino acid additions, deletions and and/or amino acid sequences that are substituted (eg, conservatively substituted) and retain the ability to specifically bind VEGF. Preferably, said amino acid additions, deletions and/or substitutions do not occur in the CDR regions. Most preferably, in the bispecific antibody according to the present invention, the VHH VEGF domain according to the present invention comprises or consists of the amino acid sequence shown in SEQ ID NO:2.
  • the bispecific antibody of the present invention in addition to the aforementioned VHH VEGF domain and VHH PD-L1 domain, in some embodiments, it may also include an immunoglobulin constant domain, for example, the immunoglobulin constant regions CH2 and CH3 Constituent Fc region, and/or CH1 and CL constant domains in a Fab-like structure.
  • an immunoglobulin constant domain for example, the immunoglobulin constant regions CH2 and CH3 Constituent Fc region, and/or CH1 and CL constant domains in a Fab-like structure.
  • the invention provides a bispecific antibody comprising an Fc region polypeptide chain consisting of immunoglobulin heavy chain constant regions CH2-CH3.
  • the present invention provides a bispecific antibody comprising an Fc region and further comprising an immunoglobulin CH1 domain and a CL domain, preferably, wherein the CH1 domain passes through an immunoglobulin hinge region connects the Fc region polypeptide, and more preferably, the antibody comprises a polypeptide chain consisting of an immunoglobulin heavy chain constant region CH1-hinge region-CH2-CH3.
  • the immunoglobulin constant domains comprised in the bispecific antibody according to the invention including the CH2 and CH3 constant domains in the Fc region and the CH1 and CL constant domains in the Fab-like structure (if present), May be, independently of each other, an immunoglobulin constant domain from human IgG, eg, a constant domain of human IgGl, IgG2, IgG3 or IgG4, preferably a constant domain of human IgGl.
  • immunoglobulin constant domains comprised in the bispecific antibodies according to the invention may independently of each other be native sequence constant domains (for example, Human native sequence constant domain) or an amino acid sequence variant thereof, preferably a native sequence constant domain.
  • the Fc region comprised in the bispecific antibody according to the invention is an Fc region sequence from human IgG1.
  • the Fc region has the amino acid sequence shown in SEQ ID NO: 3, or an amino acid sequence at least 90%, 95% or 99% identical thereto, or no more than Amino acid sequence with 1-10 (preferably 1-5) amino acid residues altered.
  • the bispecific antibody according to the present invention may comprise two polypeptide chains having an Fc region, for example, the di-chain antibody and the tetra-chain antibody of the present invention as shown in FIG. 1 , wherein the two polypeptide chains The Fc region of is capable of pairing and dimerization.
  • both the first Fc region polypeptide and the second Fc region polypeptide constituting the dimerized Fc region of the antibody may be Fc domains from IgG, such as human IgG1, IgG2, IgG3 or IgG4, preferably human IgG1 and may independently of each other be a native sequence Fc region and a variant Fc region as defined herein, provided that said first Fc region polypeptide and said second Fc region polypeptide can pair and dimerize.
  • the first Fc region and the second Fc region are native sequence Fc regions, especially human IgG1 native sequence Fc regions.
  • the first Fc region and the second Fc region have the amino acid sequence shown in SEQ ID NO: 3, or an amino acid sequence at least 90%, 95% or 99% identical thereto, or identical thereto An amino acid sequence that has no more than 1-10 (preferably 1-5) amino acid residue changes.
  • the antibody according to the present invention may comprise a Fab-like structure having a heavy chain constant region CH1 domain and a light chain constant region CL domain, wherein the CH1 structure The domain is preferably linked C-terminally to the first VHH domain and the light chain constant region CL domain is linked C-terminally to the second VHH domain.
  • the heavy chain constant regions CH1 and CL constituting the antibody may be constant domains from IgG, such as human IgG1, IgG2, IgG3 or IgG4, preferably human IgG1.
  • the CL domain of the antibody may be a Kappa light chain CL domain or a Lamda light chain CL domain, preferably a Kappa light chain CL domain.
  • the CH1 and CL domains may independently of each other be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variants thereof, provided that the CH1 structure is included Domain and the antibody polypeptide chain of the first VHH domain connected at its N-terminus, and the antibody polypeptide chain comprising said CL domain and the second VHH domain connected at its N-terminus can be paired with each other to form a Fab according to the invention like structure.
  • the CH1 and CL domains are native sequence constant domains, especially human IgG1 native sequence CH1 domains and CL domains. More preferably, the CH1 domain has the amino acid sequence shown in SEQ ID NO:4. More preferably, the CL domain has the amino acid sequence shown in SEQ ID NO:5 or SEQ ID NO:6.
  • linkers can be used to connect antibody components.
  • a VHH domain according to the present invention may be linked to the N-terminal or C-terminal of the Fc region via a linker, or may be linked to another VHH domain according to the present invention via a linker.
  • Linkers that can be used in the antibodies of the present invention are not particularly limited. Those skilled in the art can easily determine the available linker sequences according to the components to be linked and the position of the link.
  • the VHH domains according to the invention are in tandem form (eg, as shown in Figures 1A and 1B ) or in a Fab-like structure (eg, as shown in Figure 1C and 1D), connected to the Fc region polypeptide chain through a linker.
  • linker e.g., as shown in Figures 1A and 1B
  • linker e.g., as shown in Figure 1C and 1D
  • the polypeptide chains of the Fc region are connected at the N-terminus to a single VHH domain or two VHH domains in tandem, or a Fab-like structure via a linker, wherein the linker preferably comprises (one or two) cysteine Amino acid residues, which are capable of forming (one or two) disulfide bonds when the two antibody polypeptide chains comprising the Fc region pair to stabilize the structure of the bispecific antibody thus formed.
  • the linker preferably comprises (one or two) cysteine Amino acid residues, which are capable of forming (one or two) disulfide bonds when the two antibody polypeptide chains comprising the Fc region pair to stabilize the structure of the bispecific antibody thus formed.
  • the two-chain antibody structure and the four-chain antibody structure shown in FIG. 1 shown in FIG. 1 . Therefore, in a preferred embodiment, the Fc region polypeptide chain contained in the bispecific antibody of the present invention has a linker sequence comprising "CPPC" at the N-terminus.
  • the Fc region polypeptide chain contained in the bispecific antibody of the present invention has a hinge region derived from an immunoglobulin at the N-terminus, for example, the amino acid sequence of the hinge region comprising "CPPC", for example, amino acid The sequence "EPKSCDKTHTCPPCP” (SEQ ID NO:22) or "EPKSSDKTHTCPPCP” (SEQ ID NO:23); more preferably, the amino acid sequence "EPKSCDKTHTCPPCP" (SEQ ID NO:22).
  • the Fc region polypeptide chain is connected to the VHH domain at the C-terminus by a linker, wherein the linker is preferably a flexible connecting peptide of 5-50 amino acids, preferably comprising glycine (G) and/or serine (S) and/or a linking peptide of a threonine residue (T).
  • the linker is 5-50 amino acids in length, eg, 10, 15, 20, 25 or 30 amino acids in length, or has an amino acid length falling between any two integers.
  • the linker comprises the amino acid sequence (G 4 S) n , wherein n is an integer equal to or greater than 1, eg, n is an integer of 2, 3, 4, 5, 6 or 7.
  • the linker consists of the amino acid sequence (G 4 S) 3 .
  • the VHH VEGF domain and the VHH PD-L1 domain according to the invention form a tandem VHH domain via a linker.
  • the first and second domains are connected in series through a linker, wherein the linker is a flexible connecting peptide of 5-50 amino acids.
  • the linker is 5-50 amino acids in length, eg, 10, 15, 20, 25 or 30 amino acids in length, or has an amino acid length falling between any two integers.
  • the linker is a linker peptide comprising glycine (G) and/or serine (S) and/or threonine residues (T).
  • the linker comprises the amino acid sequence (G 4 S) n (SEQ ID NO: 20), wherein n is an integer equal to or greater than 1, for example, n is 2, 3, 4, 5, 6 or Integer of 7.
  • the linker consists of the amino acid sequence (G 4 S) 3 (SEQ ID NO: 19).
  • the present invention provides a bispecific antibody specifically binding to PD-L1 and VEGF, wherein the antibody comprises a VHH domain specifically binding to a first antigen and a VHH domain specifically binding to a second antigen , wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF.
  • the VHH domain specifically binding to VEGF comprises the CDR1-3 sequence of the VHH domain shown in SEQ ID NO:2.
  • the VHH domain specifically binding to PD-L1 comprises the CDR1-3 sequence of the VHH domain shown in SEQ ID NO:1.
  • the VHH domain specifically binding to VEGF comprises the amino acid sequence shown in SEQ ID NO: 2, or has at least 80%, 85%, 90%, 95%, or An amino acid sequence with 99% identity, or an amino acid sequence with one or more (preferably 1-10, more preferably 1-5) amino acid additions, deletions and/or substitutions compared to SEQ ID NO:2 , most preferably, the VHH domain comprises the amino acid sequence of SEQ ID NO: 2, or consists of the amino acid sequence shown in SEQ ID NO: 2.
  • the VHH domain specifically binding to PD-L1 comprises the amino acid sequence shown in SEQ ID NO: 1, or has at least 80%, 85%, 90%, 95% with SEQ ID NO: 1 Amino acid sequence with % or 99% identity, or with one or more (preferably 1-10, more preferably 1-5) amino acid additions, deletions and/or substitutions compared to SEQ ID NO: 1 Amino acid sequence, most preferably, the VHH domain comprises the amino acid sequence of SEQ ID NO: 1, or consists of the amino acid sequence shown in SEQ ID NO: 1.
  • the bispecific antibody according to the present invention further comprises a linker comprising 10-20 amino acids in length, preferably comprising the amino acid sequence (G 4 S) 3 (SEQ ID NO: 19) .
  • the bispecific antibody according to the present invention further comprises an Fc region of an immunoglobulin, wherein the Fc region is an IgG Fc region, for example, the Fc domain of human IgG1, IgG2, IgG3 or IgG4, preferably human The Fc domain of IgG1, more preferably the Fc region carries an immunoglobulin hinge region sequence at the N-terminus.
  • the bispecific antibody according to the invention comprises a polypeptide chain comprising VHH PD-L1 and VHH VEGF domains connected in series by means of an Fc region and a linker.
  • the bispecific antibody comprises a polypeptide chain comprising a first VHH domain, an Fc region, a linker and a second VHH domain from the N-terminus to the C-terminus, wherein the first VHH domain and the second VHH domain
  • the two VHH domains specifically bind the first antigen and the second antigen respectively, wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF, preferably, the first antigen is PD-L1 and The second antigen is VEGF.
  • the bispecific antibody comprises the amino acid sequence shown in SEQ ID NO: 7 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% thereof or amino acid sequences with 99% identity.
  • the bispecific antibody according to the invention comprises a polypeptide chain comprising VHH PD-L1 and VHH VEGF domains connected in series by means of a linker.
  • the bispecific antibody comprises a polypeptide chain comprising from the N-terminus to the C-terminus Containing a first VHH domain, a linker, a second VHH domain and an Fc region, wherein the first VHH domain and the second VHH domain specifically bind to a first antigen and a second antigen, respectively, wherein the first antigen and The second antigens are different from each other and are independently selected from PD-L1 and VEGF, preferably, the first antigen is PD-L1 and the second antigen is VEGF.
  • the bispecific antibody comprises the amino acid sequence shown in SEQ ID NO: 8 or has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% thereof or amino acid sequences with 99% identity.
  • the bispecific antibody according to the invention comprises a first and a second polypeptide chain forming a Fab-like structure.
  • the bispecific antibody comprises a first polypeptide chain and a second polypeptide chain, wherein the first polypeptide chain comprises a first VHH domain, a CH1 domain and an Fc region from the N-terminus to the C-terminus; the The second polypeptide chain comprises a second VHH domain and a CL domain from the N-terminus to the C-terminus; wherein the first VHH domain and the CH1 domain in the first polypeptide chain are the same as the second VHH in the second polypeptide chain domain and CL domain pair to form a Fab-like structure; wherein the first VHH domain and the second VHH domain specifically bind a first antigen and a second antigen, respectively, wherein the first and second antigens are different and independent of each other is selected from PD-L1 and VEGF, preferably, the first antigen is PD-L1 and the
  • the bispecific antibody comprises first and second polypeptide chains, wherein the first antigen is PD-L1 and the second antigen is VEGF, wherein the first polypeptide chain comprises SEQ The amino acid sequence shown in ID NO: 9 or an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto; and
  • the two polypeptide chains comprise an amino acid sequence shown in SEQ ID NO: 10 having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity therewith amino acid sequence.
  • the antibody may comprise two polypeptide chains comprising an Fc region, wherein the two polypeptide chains Dimers can be formed by dimerizing association of the Fc region.
  • the two polypeptide chains that associate to form a dimer need not be identical.
  • asymmetric modifications can be introduced in one or both polypeptide chains (eg, in the Fc region of the polypeptide chains) without affecting the desired target activity.
  • the two polypeptide chains are identical. For example, as shown in Figures 1A and 1B.
  • the present invention provides a bispecific antibody that specifically binds PD-L1 and VEGF, wherein said antibody comprises two polypeptide chains, said two polypeptide chains may be the same or different, wherein:
  • Each polypeptide chain comprises a first VHH domain, an Fc region, a linker, and a second VHH domain from N-terminus to C-terminus (as shown in Figure 1A); or each polypeptide chain comprises from N-terminus to C-terminus A first VHH domain, a linker, a second VHH domain and an Fc region (as shown in Figure 1B);
  • the Fc region polypeptides of the two polypeptide chains are paired to form a dimerized Fc region, wherein the first VHH domain and the second VHH domain specifically bind the first antigen and the second antigen respectively, wherein the The first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF.
  • the first antigen is PD-L1 and the second antigen is VEGF; in another embodiment, the first antigen is VEGF and the second antigen is PD-L1.
  • the present invention provides a bispecific antibody that specifically binds PD-L1 and VEGF, wherein the antibody comprises two polypeptide chains, which may be the same or different, wherein:
  • Each polypeptide chain comprises a first VHH domain, an Fc region, a linker and a second VHH domain from the N-terminus to the C-terminus,
  • the Fc region polypeptides of the two polypeptide chains are paired and form a dimerized Fc region, wherein the first VHH domain and the second VHH domain specifically bind the first antigen and the second antigen respectively, wherein the first antigen and the second antigen
  • the two antigens are different from each other and are independently selected from PD-L1 and VEGF, preferably the first antigen is PD-L1 and the second antigen is VEGF.
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is linked at the N-terminus to the first VHH domain through an immunoglobulin hinge region sequence. Still preferably, the Fc region polypeptide is connected to the second VHH domain at the C-terminus through a flexible linking peptide with a length of 10-20 amino acids, such as (G 4 S) 3 .
  • the first VHH domain is a VHH PD-L1 domain according to the present invention, preferably, comprising the CDR1-3 sequence of the variable region shown in SEQ ID NO: 1, more preferably comprising SEQ ID NOs: 13- 15 CDR1-3 sequence; most preferably comprising the variable region sequence shown in SEQ ID NO: 1; and the second VHH domain is a VHH VEGF domain according to the present invention, preferably comprising SEQ ID NO: 2
  • the CDR1-3 sequence of the variable region more preferably comprising the CDR1-3 sequence of SEQ ID NOs: 16-18; most preferably comprising the variable region sequence shown in SEQ ID NO:2.
  • the two polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 7 or have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, Amino acid sequences of 98% or 99% identity.
  • the two polypeptide chains are identical and each comprise the amino acid sequence of SEQ ID NO:7.
  • the present invention provides a bispecific antibody specifically binding to PD-L1 and VEGF, wherein the antibody comprises two polypeptide chains, and the two polypeptide chains may be the same or different, wherein :
  • Each polypeptide chain comprises a first VHH domain, a linker, a second VHH domain and an Fc region from the N-terminus to the C-terminus,
  • the Fc region polypeptides of the two polypeptides pair and form a dimerized Fc region, wherein the first VHH domain and the second VHH domain specifically bind the first antigen and the second antigen respectively, wherein the first antigen and The second antigens are different from each other and are independently selected from PD-L1 and VEGF, preferably the first antigen is PD-L1 and the second antigen is VEGF.
  • the linker is a flexible connecting peptide with a length of 10-20 amino acids, such as (G 4 S) 3 (SEQ ID NO: 19) .
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is linked at the N-terminus to a tandem VHH domain through an immunoglobulin hinge region sequence.
  • the first VHH domain is a VHH PD-L1 domain according to the present invention, preferably, comprising the CDR1-3 sequence of the variable region shown in SEQ ID NO: 1, more preferably comprising SEQ ID NOs: 13- 15 CDR1-3 sequence; most preferably comprising the variable region sequence shown in SEQ ID NO: 1; and the second VHH domain is a VHH VEGF domain according to the present invention, preferably comprising SEQ ID NO: 2
  • the CDR1-3 sequence of the variable region more preferably comprising the CDR1-3 sequence of SEQ ID NOs: 16-18; most preferably comprising the variable region sequence shown in SEQ ID NO:2.
  • the two polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 8 or have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, Amino acid sequences of 98% or 99% identity.
  • the two polypeptide chains are identical and each comprise the amino acid sequence of SEQ ID NO:8.
  • said antibody may comprise two first polypeptide chains comprising an Fc region and two The second polypeptide chain, wherein the two first polypeptide chains can form a dimer through dimerization association of the Fc region.
  • the two first polypeptide chains comprised in the bispecific antibody need not be identical; and similarly, the two second polypeptide chains need not be identical either.
  • the two first polypeptide chains are identical and the two second polypeptide chains are also identical.
  • the present invention provides a bispecific antibody specifically binding to PD-L1 and VEGF, wherein the antibody comprises two first polypeptide chains and two second polypeptide chains, wherein the two The first polypeptide chains may be the same or different, and the two second polypeptide chains may be the same or different, wherein:
  • the two first polypeptide chains each comprise a first VHH domain, a CH1 domain and an Fc region from the N-terminus to the C-terminus;
  • the two second polypeptide chains each comprise a second VHH domain and a CL domain from the N-terminus to the C-terminus;
  • first VHH structural domain and the second VHH structural domain specifically bind the first antigen and the second antigen respectively, wherein the first antigen and the second antigen are different from each other and are independently selected from PD-L1 and VEGF.
  • first antigen is PD-L1 and the second antigen is VEGF; or in another embodiment, the first antigen is VEGF and the second antigen is PD-L1.
  • the Fc region is from IgG, especially IgG1, and includes CH2 and CH3 domain sequences from N-terminus to C-terminus.
  • the Fc region may be a native sequence Fc region or a variant Fc region, preferably a native sequence Fc region.
  • the Fc region polypeptide is connected to the CH1 domain at the N-terminus through an immunoglobulin hinge region sequence.
  • the first VHH domain is a VHH PD-L1 domain according to the present invention, preferably, comprising the CDR1-3 sequence of the variable region shown in SEQ ID NO: 1, more preferably comprising SEQ ID NOs: 13- 15 CDR1-3 sequence; most preferably comprising the variable region sequence shown in SEQ ID NO: 1; and the second VHH domain is a VHH VEGF domain according to the present invention, preferably comprising SEQ ID NO: 2
  • the CDR1-3 sequence of the variable region more preferably comprising the CDR1-3 sequence of SEQ ID NOs: 16-18; most preferably comprising the variable region sequence shown in SEQ ID NO:2.
  • the bispecific antibody of the invention comprises two first polypeptide chains and two second polypeptide chains, wherein the first antigen is PD-L1 and the second antigen is VEGF, wherein:
  • the two first polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 9 or have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequences;
  • the two second polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 10 and have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% % identity amino acid sequence.
  • the two first polypeptide chains are identical, respectively comprising the amino acid sequence of SEQ ID NO:9; and the two second polypeptide chains are identical, respectively comprising the amino acid sequence of SEQ ID NO:10.
  • the bispecific antibody of the invention comprises two first polypeptide chains and two second polypeptide chains, wherein the first antigen is VEGF and the second antigen is PD-L1, wherein:
  • the two first polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 11 or have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequences;
  • the two second polypeptide chains respectively comprise the amino acid sequence shown in SEQ ID NO: 12 and have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% % identity amino acid sequence.
  • the two first polypeptide chains are identical, respectively comprising the amino acid sequence of SEQ ID NO: 11; and the two second polypeptide chains are identical, respectively comprising the amino acid sequence of SEQ ID NO: 12.
  • the invention also contemplates an embodiment in which the CH1 and CL domains in the first and second polypeptide chains are exchanged.
  • the two Fc regions are linked by the CL domain of the Fab-like structure through an immunoglobulin hinge region.
  • the invention provides methods for producing antibodies of the invention.
  • the polypeptide chains of the antibodies of the invention can be obtained, eg, by solid-state peptide synthesis (eg, Merrifield solid-phase synthesis) or recombinant production, and assembled under suitable conditions.
  • polynucleotides encoding any polypeptide chain and/or multiple polypeptide chains of the antibody can be isolated and inserted into one or more vectors for further cloning and/or expression in host cells.
  • the polynucleotides can be readily isolated and sequenced using conventional methods.
  • polynucleotides encoding one or more polypeptide chains of an antibody of the invention are provided.
  • the invention provides a vector, preferably an expression vector, comprising one or more polynucleotides of the invention.
  • the invention provides a method for producing an antibody of the invention, said method comprising: culturing a host cell comprising a polypeptide chain encoding said antibody under conditions suitable for expressing said polypeptide chain; and The antibody is produced by assembling the polypeptide chains under conditions suitable for assembly of the polypeptide chains into the antibody.
  • Expression vectors can be constructed using methods well known to those skilled in the art.
  • Expression vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YACs).
  • the expression vector can be transfected or introduced into a suitable host cell.
  • a suitable host cell for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, liposome-based transfection or other conventional techniques.
  • the invention also provides host cells comprising one or more polynucleotides of the invention.
  • host cells comprising an expression vector of the invention are provided.
  • the term "host cell” refers to any kind of cellular system that can be engineered to produce the antibodies of the invention.
  • Host cells suitable for replicating and supporting expression of the antibodies of the invention are well known in the art. Such cells can be transfected or transduced with specific expression vectors as needed, and large numbers of cells containing the vector can be grown for inoculation of large-scale fermenters to obtain sufficient quantities of the antibodies of the invention for clinical use.
  • Suitable host cells include prokaryotic microorganisms such as Escherichia 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. 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 (HEK293 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 (HepG2) , CHO cells, NSO cells, myeloma cell lines such as YO, NSO, P3X63 and Sp2/0, etc.
  • the host cell is a CHO, HEK293 or NSO cell.
  • Antibodies prepared by the methods described herein can be purified by known art techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like. After purification, the antibodies of the invention can be determined by any of a variety of well known assays.
  • the well-known analytical methods include size exclusion chromatography, gel electrophoresis, high performance liquid chromatography, etc.
  • the physical/chemical properties and/or biological activities of the antibodies provided herein can be identified, screened or characterized by a variety of assays known in the art.
  • the bispecific antibody of the present invention exhibits good production properties in recombinant production in mammalian host cells such as CHO cells, in particular, good expression yield and good by-product profile.
  • the transient expression level of the bispecific antibody of the present invention reaches above 50 ⁇ g/mL, preferably above 150 ⁇ g/mL; After purification, the purity of the product is determined by SEC-HPLC, showing a purity of more than 95%, preferably more than 96%, 97%, 98% or 99%.
  • the determination of the transient expression level is determined according to the method described in Example 3; the determination of the purity of the product after one-step protein A purification is performed according to the SEC-HPLC monomer purity identification method described in Example 3.4.
  • the invention provides compositions, eg, pharmaceutical compositions, comprising an antibody 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 (eg, by injection or infusion).
  • an antibody of the invention is the sole active ingredient in a pharmaceutical composition.
  • a pharmaceutical composition may comprise an antibody described herein and more than one therapeutic agent.
  • the invention also provides pharmaceutical combinations comprising an antibody described herein and more than one therapeutic agent.
  • Therapeutic agents suitable for use in the pharmaceutical compositions and drug combinations of the invention may be therapeutic agents selected from any of the following categories (i)-(iv): (i) agents that enhance antigen presentation (e.g., tumor antigen presentation) Drugs; (ii) drugs that enhance effector cell responses (eg, B cell and/or T cell activation and/or mobilization); (iii) drugs that reduce immunosuppression; (iv) drugs that have tumor suppressive effects.
  • agents that enhance antigen presentation e.g., tumor antigen presentation
  • Drugs e.g., tumor antigen presentation
  • drugs that enhance effector cell responses eg, B cell and/or T cell activation and/or mobilization
  • drugs that reduce immunosuppression e.g., B cell and/or T cell activation and/or mobilization
  • drugs that reduce immunosuppression e.g., B cell and/or T cell activation and/or mobilization
  • drugs that reduce immunosuppression e.g., B cell and/or T cell activ
  • compositions of the invention can be in a variety of forms. These forms include, for example, liquid, semisolid, and solid dosage forms, such as liquid solutions (eg, injectable solutions and infusible solutions), dispersions or suspensions, liposomes, and suppositories.
  • liquid solutions eg, injectable solutions and infusible solutions
  • dispersions or suspensions e.g., liposomes, and suppositories.
  • the preferred form depends on the intended mode of administration and therapeutic use. Commonly preferred compositions are in the form of injectable solutions or infusible solutions.
  • the preferred mode of administration is parenteral (eg, intravenous, subcutaneous, intraperitoneal (i.p.), intramuscular) injection.
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular, intraperitoneal or subcutaneous injection.
  • parenteral administration and “parenteral administration” as used herein mean modes of administration other than enteral 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.
  • Compositions can be formulated as solutions, microemulsions, dispersions, liposomes or in lyophilized form.
  • Sterile injectable solutions can be prepared by incorporating the active compound (ie, antibody) in the required amount in an appropriate solvent, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and other ingredients. Coating agents such as lecithin and the like can be used.
  • proper fluidity of the solution can be maintained by using a surfactant.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition substances which delay absorption, for example, monostearate salts and gelatin.
  • the pharmaceutical composition of the present invention may contain "therapeutically effective amount” or “prophylactically effective amount” of the antibody of the present invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages required, and for periods of time required, to achieve the desired therapeutic result. can be based on a variety of factors such as disease status, individual age, A therapeutically effective amount for such changes as sex and weight. A therapeutically effective amount is one in which any noxious or detrimental effects are outweighed by the therapeutically beneficial effects.
  • a “therapeutically effective amount” preferably inhibits a measurable parameter (e.g. tumor growth rate) by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60% and still more relative to an untreated subject. Preferably at least about 80%.
  • the ability of the antibodies of the invention to inhibit a measurable parameter eg, tumor volume
  • prophylactically effective amount refers to an amount effective, at dosages required, and for periods of time required, to achieve the desired prophylactic result. Typically, the prophylactically effective amount is less than the therapeutically effective amount because the prophylactic dose is used in the subject before or at an earlier stage of the disease.
  • Kits comprising the antibodies described herein are also within the scope of the invention.
  • a kit may comprise one or more other elements including, for example: instructions for use; other reagents, such as labels or reagents for conjugation; a pharmaceutically acceptable carrier; and devices or other materials for administration to a subject.
  • the present invention provides in vivo and in vitro uses and methods of application of the antibodies of the present invention.
  • the uses and methods of the invention involve the in vivo and/or in vitro application of the antibodies of the invention to:
  • the antibody of the present invention or a pharmaceutical composition comprising the antibody of the present invention is used as a drug for treating and/or preventing a disease or as a diagnostic tool for a disease in an individual, preferably, the individual is a mammal, more preferably Preferably a human.
  • the present invention provides methods and uses of using the antibodies of the present invention, especially bispecific antibodies, to treat cancer, wherein the cancer can be PD-L1 positive solid tumors or blood cancers, for example selected from head and neck squamous cell carcinoma, Melanoma, renal cell carcinoma, non-small cell lung cancer, bladder cancer, urothelial carcinoma, gastric cancer, colon cancer, colorectal cancer, ovarian cancer, breast cancer, lung cancer, cervical cancer, glioblastoma, pancreatic cancer, prostate cancer Carcinoma, esophageal cancer, lymphoma, liver cancer, microsatellite unstable solid tumors.
  • said cancer is colorectal cancer, liver cancer, ovarian cancer, breast cancer, lung cancer.
  • the invention provides in vitro or in vivo diagnostic methods for detecting the presence of relevant antigens in biological samples such as serum, semen or urine or tissue biopsy samples (eg, from hyperproliferative or cancerous lesions).
  • the diagnostic method comprises: (i) contacting a sample (and optionally, a control sample) with an antibody as described herein or administering the antibody to a subject under conditions that allow the interaction to occur and (ii) detecting the detected Complex formation between the antibody and the sample (and optionally, a control sample) is performed. Formation of the complex indicates the presence of the relevant antigen and may indicate suitability or need for treatment and/or prophylaxis as described herein.
  • the invention provides a diagnostic kit comprising an antibody described herein and instructions for use.
  • Embodiment 1 raw material preparation
  • the extracellular region of human PD-L1 (UniProtKB-Q9NZQ7), the extracellular region of human PD-1 (UniProtKB-Q15116) and human VEGF (UniProtKB-P15692) were synthesized by General Biosystems (Anhui) Co., Ltd. for the target fragment gene.
  • Each target fragment was amplified by PCR, and a His tag or hIgG1Fc (shown in the amino acid sequence SEQ ID NO: 3) was introduced at the C-terminus by primers, and then respectively constructed into the eukaryotic expression vector pcDNA3.4 (Invitrogen) by homologous recombination .
  • the constructed recombinant protein expression vectors were respectively transformed into Escherichia coli DH5 ⁇ , cultured overnight at 37°C, and then plasmids were extracted using an endotoxin-free plasmid extraction kit (OMEGA, D6950-01) to obtain endotoxin-free plasmids and For eukaryotic expression.
  • OEGA endotoxin-free plasmid extraction kit
  • Recombinant protein VEGF-His, recombinant protein VEGF-Fc, recombinant protein PD-L1-His, recombinant protein PD-L1-Fc, recombinant protein PD-1-His and recombinant protein PD-1-Fc were all passed through the Expi293 transient expression system (ThermoFisher, A14635) expression, see Expi293 TM Expression System Kit Manual for the transient method. After 5-7 days of transfection, the cell expression supernatant was centrifuged at a high speed of 15000 g for 10 min.
  • the resulting His-tagged recombinant protein expression supernatant was affinity-purified with Ni Smart Beads 6FF (Changzhou Tiandi Renhe Biotechnology Co., Ltd., SA036050), and then the target protein was eluted with gradient concentrations of imidazole.
  • the filter concentration tube (Millipore, UFC901096) was replaced with PBS buffer; the resulting Fc-tagged recombinant protein expression supernatant was filtered through a 0.22 ⁇ m filter membrane, and then purified by Protein A/G affinity chromatography column affinity method.
  • the target protein was eluted with 100mM glycine salt (pH3.0), then concentrated and replaced, and then frozen at -80°C after being identified by SDS-PAGE and qualified for activity identification.
  • anti-human VEGF antibody P30-10-26 (VHH VEGF -hIgG1Fc, VHH VEGF amino acid sequence shown in SEQ ID NO: 2) is derived from patent application CN202110995278.7; anti-human PD-L1 antibody D21-4 (VHH PD-L1 -hIgG1Fc, VHH PD-L1 amino acid sequence shown in SEQ ID NO: 1) is derived from the patent application PCT/CN2020/125301; the anti-VEGF control antibody Bevacizumab (Bevacizumab, abbreviated as Beva) sequence is derived from Drug Bank (Drug Bank No: DB00112); anti-PD-L1 control antibody Atezolizumab (Ate) sequence is derived from Drug Bank (Drug Bank No: DB11595); anti-PD-L1 and VEGF control bispecific
  • Bevacizumab Bevacizumab, abbreviated as Beva
  • Beva anti-PD-L1 control
  • the antibodies were all expressed using the transient system (ExpiCHO).
  • ExpiCHO transient system
  • the expressed cell suspension was subjected to high-speed centrifugation and the supernatant was taken, and the obtained supernatant was filtered through a 0.22 ⁇ m filter membrane, and then purified by using a Protein A/G affinity chromatography column affinity method. After purification, the target protein was eluted with 100 mM glycine salt (pH 3.0), then concentrated and replaced, and then frozen at -80°C after being identified by SDS-PAGE and qualified for activity identification.
  • the CHO-K cells (Thermo, A1461801) were passaged to 5 ⁇ 10 6 cells/mL, and the next day, the constructed cells containing full-length human PD-L1 Plasmid introduction of (UniProtKB-Q9NZQ7) gene sequence in CHO-K cells.
  • the cells after electroporation were transferred to CD-CHO medium (Gibco, 10743029), and placed in a cell culture incubator at 37°C for 48 hours. Then, 2000 cells/well were plated into a 96-well plate, and MSX (Millipore, GSS-1015-F) and GS supplement (Sigma, 58672C) were added to a final concentration of 30 ⁇ M for pressurized screening.
  • MSX Micropore, GSS-1015-F
  • GS supplement Sigma, 58672C
  • the pGL4.30 plasmid (promega, #E8481) containing the NF-AT-re nucleic acid sequence was electroporated into HEK293 cells ( CRL-1573 TM ) to obtain HEK293-NFAT cell line.
  • the HEK293-NFAT cells were passaged to 2 ⁇ 105 cells/mL, and the next day, the electroporation kit (Invitrogen, MPK10096) and electroporation instrument (Invitrogen, MP922947) were used to transform the constructed human VEGFR2 gene sequence (NCBI Gene ID: 3791) plasmid into HEK293-NFAT cells.
  • the electroporated cells were transferred to DMEM medium (Gibco, 12634010), and placed in a 37°C cell culture incubator for 48 hours. Then, 1000 cells/well were plated in a 96-well plate, and puromycin at a final concentration of 2 ⁇ g/mL was added for pressure selection. The single-cell clones grown in the 96-well plate were picked, and after expanded culture, the HEK293-VEGFR2-NFAT cell line was obtained by adding a luciferase catalytic substrate to detect the fluorescent signal and identify it.
  • the pGL4.30 plasmid (promega, #E8481) containing the NF-AT-re nucleic acid sequence was electroporated into Jurkat cells ( TIB-152) to obtain Jurkat-NFAT cell line.
  • stably transform the full-length expression gene sequence of PD-1 NCBI Gene ID: 5133
  • Monoclonal cell lines were screened, and Jurkat-PD-1-NFAT cell lines were obtained by FACS identification after expansion.
  • the scFv sequence of OKT-3 (Drug Bank No: DB00075) was electrotransfected into CHO cells to obtain CHO-CD3L stably transfected cell line, on this basis, the full-length expression gene sequence of PD-L1 was stably transfected (NCBI Gene ID: 29126), The method was consistent with 1.3.2. The final concentration of 30 mM methionine iminosulfone and 8 ⁇ g/mL puromycin were added to screen monoclonal cell lines, and the CHO-PD-L1-CD3L cell line was obtained by FACS identification after expansion.
  • This example describes the construction of exemplary anti-PD-L1 and VEGF bispecific antibody (BsAb) structures and expression vectors.
  • BsAb bispecific antibody
  • Four constructs were designed and constructed: the amino acid sequence of VHH PD-L1 is from D21-4, and its variable region amino acid sequence is shown in SEQ ID NO:1; the VHH VEGF amino acid sequence is from P30-10-26, and its variable region The amino acid sequence is shown in SEQ ID NO: 2; the amino acid sequence of the linker is GGGGSGGGGSGGGGS (SEQ ID NO: 19).
  • Exemplary BsAb constructs are shown in Table 1 and the corresponding amino acid sequences are provided in Table 2.
  • BsAb10 Contains two identical polypeptide chains, including anti-PD-L1 VHH domain, IgG1 heavy chain hinge region, IgG1 heavy chain Fc, linker and anti-VEGF VHH domain from N-terminal to C-terminal.
  • BsAb10 has the format of Figure 1A.
  • BsAb11 Contains two identical polypeptide chains, including anti-PD-L1 VHH domain, linker, anti-VEGF VHH domain, IgG1 heavy chain hinge region and IgG1 heavy chain Fc from N-terminal to C-terminal.
  • BsAb11 has the format of Figure 1B.
  • Construct BsAb12 Contains two identical first polypeptide chains, including anti-PD-L1 VHH domain from N-terminal to C-terminal, IgG1 heavy Chain constant region domain (including IgG1 heavy chain CH1, IgG1 heavy chain hinge region, IgG1 heavy chain Fc); contains two identical second polypeptide chains, from N-terminal to C-terminal contains anti-VEGF VHH domain, antibody Kappa light chain CL domain.
  • BsAb12 has the format of Figure 1C.
  • BsAb13 Contains two identical first polypeptide chains, including anti-VEGF VHH domain from N-terminal to C-terminal, IgG1 heavy chain constant region domain (including IgG1 heavy chain CH1, IgG1 heavy chain hinge region, IgG1 Heavy chain Fc); Contains two identical second polypeptide chains, including VHH domain of anti-PD-L1 and CL domain of antibody kappa light chain from N-terminus to C-terminus.
  • BsAb13 has the format of Figure 1D.
  • fragments of the variable region and constant region of each antibody are amplified by PCR method, and the fragments are connected by overlap extension PCR method, and then constructed into modified eukaryotic expression vectors by homologous recombination method
  • the full-length gene of the polypeptide chain constituting the complete construct.
  • the constructed vectors containing the full-length gene of the polypeptide chain of the construct were respectively transformed into Escherichia coli DH5 ⁇ , and cultured overnight at 37°C.
  • the endotoxin-free plasmid extraction kit (OMEGA, D6950-01) was used for plasmid extraction to obtain an endotoxin-free construct polypeptide chain plasmid for eukaryotic expression.
  • Example 3 Expression, purification, and analysis of physicochemical properties of anti-PD-L1 and VEGF bispecific antibody
  • Example 2 The construct in Example 2 was expressed through the ExpiCHO transient expression system (Thermo Fisher, A29133), and the specific method was as follows: on the day of transfection, confirm that the cell density was about 7 ⁇ 10 6 to 1 ⁇ 10 7 cells/mL, and the cell survival At this point, adjust the cells to a final concentration of 6 ⁇ 10 6 cells/mL with fresh ExpiCHO expression medium pre-warmed at 37°C.
  • OptiPRO TM SFM Dilute the target plasmid with 4°C pre-cooled OptiPRO TM SFM (add 1 ⁇ g of plasmid to 1 mL of the medium), and at the same time, dilute ExpiFectamine TM CHO with OptiPRO TM SFM, then mix the two in equal volumes and gently blow and mix to prepare Form ExpiFectamine TM CHO/plasmid DNA mixture, incubate at room temperature for 1-5min, slowly add to the prepared cell suspension and shake gently at the same time, and finally place in a cell culture shaker at 37°C, 8% CO 2 conditions under cultivation.
  • the cell culture supernatant expressing the target protein was centrifuged at a high speed of 15000g for 10min, and the resulting supernatant was affinity purified with MabSelect SuRe LX (GE, 17547403), and then purified with 100mM sodium acetate (pH3.0 ) to elute the target protein, then neutralize it with 1M Tris-HCl, and finally replace the obtained protein into PBS buffer through an ultrafiltration concentrator tube (Millipore, UFC901096).
  • the concentration of the purified bispecific antibody in Example 3.1 was measured with a verified ultra-micro spectrophotometer (Hangzhou Aosheng Instrument Co., Ltd., Nano-300), and the measured A280 value was divided by the theoretical extinction coefficient of the antibody. The value is used as the antibody concentration value for follow-up research. After passing the quality inspection, it is aliquoted and stored at -80°C.
  • Non-reducing solution preparation 1 ⁇ g of candidate bispecific antibody and reference product IPI (the IPI is the abbreviation of Ipilimumab (Ipilimumab), prepared by the method in Example 3.1) was added to 5 ⁇ SDS loading buffer and 40mM Iodoacetamide was heated in a dry bath at 75°C for 10 minutes, cooled to room temperature, centrifuged at 12,000 rpm for 5 minutes, and the supernatant was taken.
  • IPI is the abbreviation of Ipilimumab (Ipilimumab)
  • Preparation of reducing solution Add 2 ⁇ g of candidate bispecific antibody and reference IPI to 5 ⁇ SDS loading buffer and 5 mM DTT, heat in a dry bath at 100°C for 10 min, cool to room temperature, centrifuge at 12,000 rpm for 5 min, and take the supernatant.
  • SEC-HPLC results of candidate bispecific antibodies are as follows: the percentages of high molecular polymers, antibody monomers and low molecular substances in the sample were calculated according to the area normalization method, and the results are shown in Figures 3A-3D and Table 3.
  • huPD-L1-CHO-K cells Collect the huPD-L1-CHO-K cells in the exponential growth phase, centrifuge at 300g to remove the supernatant, resuspend the cells in FACS buffer (PBS containing 1% BSA), count and adjust the density of the cell suspension to 2 ⁇ 10 6 individual/mL. Subsequently, huPD-L1-CHO-K cells were added to a 96-well round bottom plate at 100 ⁇ L per well, and centrifuged at 300 g to remove the supernatant. Different concentrations of candidate bispecific antibody and control antibody D21-4 dilutions were added to the corresponding wells, the cells were resuspended and incubated at 4°C for 30 min.
  • a PE-labeled anti-human-IgG-Fc flow antibody (Abcam, 98596) was added, resuspended and incubated at 4°C for 30 min. After the incubated cell mixture was washed three times, 200 ⁇ L of FACS buffer was added to resuspend the cells, and the cells were detected and analyzed by flow cytometry (Beckman, CytoFLEX AOO-1-1102). Data were analyzed using PRISM TM (GraphPad Software, San Diego, CA), and EC50 values were calculated.
  • PRISM TM GraphPad Software, San Diego, CA
  • bispecific antibodies BsAb10 and BsAb12 showed better binding ability to PD-L1 expressed on cells than control antibody D21-4.
  • the recombinant protein PD-L1-Fc was coated on a 96-well ELISA plate overnight at 4°C. On the next day, the well plate was washed 3 times with PBST and then blocked with 5% skim milk for 2 hours. After the plate was washed 3 times with PBST, different concentrations of candidate bispecific antibodies were added and incubated for 1 hour.
  • the luciferase reporter gene system was used to detect the activity of candidate anti-PD-L1 and VEGF bispecific antibodies in blocking the PD-1/PD-L1 signaling pathway, and the combination of PD-1 and PD-L1 can block the downstream signal of CD3 Transduced to inhibit luciferase expression system, when adding anti-PD-L1 and VEGF bispecific antibody, the blocking effect is reversed, luciferase expression, stimulated with antibodies with different concentration gradients, will have antibody concentration-dependent A specific fluorescence readout curve allows the evaluation of the blocking activity of the antibody.
  • the luciferase reporter gene system was used to detect the activity of candidate anti-PD-L1 and VEGF bispecific antibodies in blocking the PD-1/PD-L1 signaling pathway. The specific method is as follows:
  • the HEK293-VEGFR2-NFAT cell line was used (the VEGF recombinant protein was added to the cell line culture system to activate the cell line through the VEGF-VEGFR2 signaling axis). Transcription and expression of the internal NFAT luciferase reporter gene, adding the catalytic substrate of luciferase to generate a fluorescent signal) as a material to detect the binding of the candidate bispecific antibody to VEGF-VEGFR2 to block the expression of the downstream NFAT luciferase reporter gene ability.
  • the specific implementation is as follows:
  • Adjust the HEK293-VEGFR2-NFAT cell line to 4 ⁇ 10 5 cells/mL add 100 ⁇ L per well to a new 96-well cell culture plate, and place it in a 37°C cell culture incubator.
  • the candidate bispecific antibody and control antibody P30-10-26 were serially diluted in DMEM medium, 60 ng/mL VEGF-Fc was added, mixed and incubated at room temperature for 30 min. Subsequently, the co-incubated serially diluted antibody and VEGF-Fc mixture was added to a 96-well cell culture plate, and cultured in a 37° C. incubator for 18 hours. After the incubation, 30 ⁇ L of luciferase substrate Bright-Lite (Vazyme, DD1204-03) was added to each well, and the fluorescence value of the 96-well plate was detected after shaking for 2 minutes.
  • Example 7 In vivo tumor suppression experiment of anti-PD-L1 and VEGF bispecific antibody (COLO205 mouse tumor-bearing model)
  • NOG mice purchased from Shanghai Weitong Lihua, strain: NOD.Cg-Prkdc scid Il2rg tm1Sug /JicCrl
  • the experimental mice were kept in an independent ventilated box with constant temperature and humidity, and the temperature of the feeding room was 21 -24°C, humidity 30-53%.
  • the huPD-L1-COLO205 cells (a stably transfected cell line overexpressing human PD-L1 obtained by stably transfecting PD-L1 (Gene ID: 29126) into COLO205 cells (purchased from Beina Biotech, catalog number: BNCC338682)) were 3 ⁇ 10 6 mice/mouse were subcutaneously injected into the right back (day 0), and then randomly divided into groups (6 mice per group): PBS treatment group, Atezolizumab administration group, Bevacizumab administration group , Atezolizumab+Bevacizumab combined administration group, W3256 administration group, AK112 administration group and bispecific antibody BsAb10 administration group, each administration group has a high-dose group, in which the bispecific antibody BsAb10 administration group and AK112 The administration group also set up a middle and low dose group.
  • PBMC peripheral blood mononuclear cell
  • ip intraperitoneal injection
  • Example 8 In vivo tumor suppression experiment of anti-PD-L1 and VEGF bispecific antibody (A431 mouse tumor-bearing model)
  • mice 6-8 weeks old female NSG mice (purchased from Shanghai Nsweeping Model Organisms, strain: M-NSG) were used, and the experimental mice were kept in an independent ventilation box with constant temperature and humidity. %.
  • A431 cells human epidermal cancer cells
  • PBS treatment group respectively , D21-4 low-dose administration group (0.37mpk (mg/kg)), P30-10-26 low-dose administration group (0.37mpk), bispecific antibody BsAb10 high-dose administration group (2mpk), bispecific Antibody BsAb10 low-dose administration group (0.5mpk), D21-4 and P30-10-26 high-dose combination treatment group (1.48+1.48mpk), D21-4 and P30-10-26 low-dose combination treatment group (0.37 +0.37mpk).
  • PBMC C2106025
  • ip intraperitoneal injection

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)

Abstract

La présente invention concerne un anticorps bispécifique ciblant PD-L1 et VEGF et son procédé de préparation, et concerne également une composition pharmaceutique comprenant l'anticorps bispécifique et son utilisation dans le traitement et le diagnostic.
PCT/CN2023/079017 2022-03-02 2023-03-01 Anticorps bispécifique anti-pd-l1 et vegf et son utilisation WO2023165516A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210198544.8A CN116731188A (zh) 2022-03-02 2022-03-02 抗pd-l1和vegf双特异性抗体及其应用
CN202210198544.8 2022-03-02

Publications (1)

Publication Number Publication Date
WO2023165516A1 true WO2023165516A1 (fr) 2023-09-07

Family

ID=87883048

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/079017 WO2023165516A1 (fr) 2022-03-02 2023-03-01 Anticorps bispécifique anti-pd-l1 et vegf et son utilisation

Country Status (2)

Country Link
CN (1) CN116731188A (fr)
WO (1) WO2023165516A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103965363A (zh) * 2013-02-06 2014-08-06 上海白泽生物科技有限公司 与pd-1和vegf高效结合的融合蛋白、其编码序列及用途
CN109942712A (zh) * 2019-04-01 2019-06-28 华博生物医药技术(上海)有限公司 抗pd-l1/vegf双功能抗体及其用途
WO2020181221A1 (fr) * 2019-03-07 2020-09-10 Children's Medical Center Corporation Administration ciblée de vhh immunomodulateur et de protéine de fusion vhh
CN111848800A (zh) * 2020-07-31 2020-10-30 三优生物医药(上海)有限公司 Pd-l1单结构域抗体及其用途

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR110040A1 (es) * 2016-05-27 2019-02-20 Abbvie Biotherapeutics Inc Proteínas de unión biespecíficas
EP4067387A4 (fr) * 2019-11-25 2023-12-27 Akeso Biopharma, Inc. Anticorps bispécifique anti-pd-1-anti-vegfa, composition pharmaceutique et leur utilisation
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

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103965363A (zh) * 2013-02-06 2014-08-06 上海白泽生物科技有限公司 与pd-1和vegf高效结合的融合蛋白、其编码序列及用途
WO2020181221A1 (fr) * 2019-03-07 2020-09-10 Children's Medical Center Corporation Administration ciblée de vhh immunomodulateur et de protéine de fusion vhh
CN109942712A (zh) * 2019-04-01 2019-06-28 华博生物医药技术(上海)有限公司 抗pd-l1/vegf双功能抗体及其用途
CN111848800A (zh) * 2020-07-31 2020-10-30 三优生物医药(上海)有限公司 Pd-l1单结构域抗体及其用途

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE Protein 14 September 2010 (2010-09-14), ANONYMOUS: "Immunoglobulin Heavy Chain Variable Region, Partial [Vicugna Pacos]", XP093088336, retrieved from NCBI Database accession no. ACS73864.1 *

Also Published As

Publication number Publication date
CN116731188A (zh) 2023-09-12

Similar Documents

Publication Publication Date Title
US11214616B2 (en) Anti-TIGIT antibodies and their use as therapeutics and diagnostics
JP7082620B2 (ja) 抗pd1モノクローナル抗体、その医薬組成物およびその使用
CN110305210B (zh) 新型抗体分子、其制备方法及其用途
KR20210042117A (ko) Cldn18.2 및 cd3에 대한 항체 작제물
JP2021519610A (ja) 多価抗体
JP7462611B2 (ja) Ox40結合性ポリペプチド及びその使用
KR20210142638A (ko) Cd3 항원 결합 단편 및 이의 응용
CN111971298A (zh) 抗体
US20230357389A1 (en) Anti-claudin18.2 and cd3 bispecific antibody and use thereof
WO2022199555A1 (fr) Protéine de liaison à un antigène multispécifique se liant à cd3 et son utilisation
Masuko et al. Towards therapeutic antibodies to membrane oncoproteins by a robust strategy using rats immunized with transfectants expressing target molecules fused to green fluorescent protein
JP2024502758A (ja) Cd73結合タンパク質及びその使用
JP2023554326A (ja) Cd73の抗原結合蛋白及びその使用
CN113527493A (zh) 一种b7-h3抗体及其应用
CN113698492A (zh) 人间皮素嵌合抗原受体及其用途
KR20220044748A (ko) 4가 대칭 이중 특이적 항체
WO2022161454A1 (fr) Protéine de liaison à l'antigène et son utilisation
JP2021525537A (ja) 三量体ポリペプチド複合体およびその使用
US20240043487A1 (en) Interleukin-21 mutant and use thereof
WO2023165516A1 (fr) Anticorps bispécifique anti-pd-l1 et vegf et son utilisation
EP4373854A1 (fr) Polypeptides se liant à cd8 et leurs utilisations
KR20230154235A (ko) NKp46에 대한 항체 및 이의 적용
CN116987196A (zh) 多特异性抗体、其制备方法及其用途
WO2023241480A1 (fr) Anticorps trispécifique anti-pd-l1, vegf et egfr et son utilisation
CN115768482A (zh) 结合siglec-3/cd33的材料和方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23762914

Country of ref document: EP

Kind code of ref document: A1