WO2021057991A1 - 对lif具有特异性的结合分子及其用途 - Google Patents

对lif具有特异性的结合分子及其用途 Download PDF

Info

Publication number
WO2021057991A1
WO2021057991A1 PCT/CN2020/118247 CN2020118247W WO2021057991A1 WO 2021057991 A1 WO2021057991 A1 WO 2021057991A1 CN 2020118247 W CN2020118247 W CN 2020118247W WO 2021057991 A1 WO2021057991 A1 WO 2021057991A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
seq
light chain
variable region
Prior art date
Application number
PCT/CN2020/118247
Other languages
English (en)
French (fr)
Inventor
刘庆浩
陶俊
周文来
贺珊珊
杨海艳
王红玲
杨贵群
Original Assignee
北京加科思新药研发有限公司
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
Priority to EP20870311.6A priority Critical patent/EP4036114A4/en
Priority to BR112022005909A priority patent/BR112022005909A2/pt
Priority to CN202080046803.4A priority patent/CN114040924A/zh
Priority to AU2020354255A priority patent/AU2020354255A1/en
Priority to KR1020227014353A priority patent/KR20220087457A/ko
Priority to CA3156080A priority patent/CA3156080A1/en
Application filed by 北京加科思新药研发有限公司 filed Critical 北京加科思新药研发有限公司
Priority to MX2022003762A priority patent/MX2022003762A/es
Priority to JP2022519435A priority patent/JP2022550121A/ja
Publication of WO2021057991A1 publication Critical patent/WO2021057991A1/zh
Priority to US17/407,264 priority patent/US20210403582A1/en
Priority to IL291710A priority patent/IL291710A/en
Priority to ZA2022/03636A priority patent/ZA202203636B/en

Links

Images

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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • G01N33/563Immunoassay; Biospecific binding assay; Materials therefor involving antibody fragments
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • 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
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans

Definitions

  • the present invention relates to an isolated antibody or antigen-binding portion thereof that specifically binds to LIF, the use of the isolated antibody or antigen-binding portion thereof of the present invention, and a treatment method using the isolated antibody or antigen-binding portion thereof of the present invention.
  • LIF Leukemia Inhibitory Factor
  • the human LIF protein contains 202 amino acids. It has two receptors on the cell membrane surface, GP130 and LIFR. The LIF protein binds to these two receptors, causing the two receptors to form a heterodimer, thereby activating downstream Signal pathways, such as MAPK signal pathway and JAK/STAT signal pathway [2].
  • the high expression and high serum concentration of LIF protein has been shown to be associated with the poor prognosis of a variety of tumors [3,4].
  • LIF is an important regulator of cancer stem cells, which plays an important role in stem cell maintenance, self-renewal and pluripotency, and is related to chemotherapy resistance [5, 6]. In addition, LIF can also promote tumor growth and metastasis [7]. Recent evidence indicates that LIF upregulates the JAK-STAT3 signaling pathway through autocrine and paracrine methods in tumors, thereby promoting tumor growth and suppressing immune responses [8, 9, 10]. Therefore, LIF is a potential therapeutic target, but the current treatment methods developed for LIF targets are not optimistic.
  • RNA interference technology has disadvantages such as poor targeting, short half-life, and poor transmembrane performance. It is difficult to prepare medicines; EC359 is targeted
  • the small molecule inhibitors of LIFR can not only inhibit the binding of LIFR and LIF, but also inhibit the binding of OSM, CTF1, CNTF and LIFR at the same time [13]. Whether these additional inhibitions will cause additional toxicity is still unknown, but specific targets Small molecule inhibitors of LIF protein have not been reported; currently only one antibody against LIF protein is in clinical development, and relevant safety and efficacy data have not yet been disclosed.
  • the present invention provides isolated antibodies or antigen-binding portions that specifically bind to LIF and their use in treating diseases.
  • the present invention provides an isolated antibody or antigen-binding portion thereof, wherein the isolated antibody or antigen-binding portion thereof binds to the amino acid sequence TYGPDTSGKDVFQKK (SEQ ID NO: 61) of the human LIF protein. Or epitopes in the corresponding amino acid sequence of different mammalian species.
  • the present invention provides an isolated antibody or antigen-binding portion thereof, wherein the isolated antibody or antigen-binding portion thereof comprises:
  • the LCDR1 includes an amino acid sequence selected from the group consisting of SEQ ID NO: 1 or SEQ ID NO: 66 and conservatively modified forms thereof;
  • the LCDR2 includes an amino acid sequence selected from the group consisting of SEQ ID NO: 2 or SEQ ID NO: 67 and conservatively modified forms thereof;
  • LCDR3 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 3 or SEQ ID NO: 68 and conservatively modified forms thereof;
  • HCDR1 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 4 or SEQ ID NO: 69 and conservatively modified forms thereof;
  • HCDR2 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 5, SEQ ID NO: 45 or SEQ ID NO: 70 and conservatively modified forms thereof;
  • HCDR3 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 6 or SEQ ID NO: 71 and conservatively modified forms thereof.
  • the LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 or HCDR3 have additions, substitutions, deletions and/or insertions of 17 or fewer amino acids.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the LCDR1 includes an amino acid sequence selected from the group consisting of SEQ ID NO: 1 and conservatively modified forms thereof;
  • LCDR2 includes an amino acid sequence selected from the group consisting of SEQ ID NO: 2 and conservatively modified forms thereof;
  • LCDR3 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 3 and conservatively modified forms thereof;
  • HCDR1 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 4 and conservatively modified forms thereof;
  • HCDR2 which includes an amino acid sequence selected from the group consisting of SEQ ID NO: 5 or SEQ ID NO: 45 and conservatively modified forms thereof;
  • HCDR3 which comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 6 and conservatively modified forms thereof.
  • the LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 or HCDR3 have additions, substitutions, deletions and/or insertions of 17 or fewer amino acids.
  • the LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, or HCDR3 have additions, substitutions, deletions, and/or insertions of 9 or fewer amino acids.
  • the LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, or HCDR3 have 5 or fewer amino acid additions, substitutions, deletions, and/or insertions.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • LCDR1 including SEQ ID NO: 66
  • LCDR2 including SEQ ID NO: 67
  • LCDR3 including SEQ ID NO: 68
  • HCDR1 including SEQ ID NO: 69
  • HCDR2 comprising SEQ ID NO: 70
  • HCDR3 comprising SEQ ID NO: 71.
  • the isolated antibody or antigen-binding fragment thereof is a murine antibody or antigen-binding fragment thereof, a chimeric antibody or an antigen-binding fragment thereof, a fully human antibody or an antigen-binding fragment thereof, or a humanized antibody Or its antigen-binding fragment.
  • the isolated antibody is a humanized antibody, the humanized antibody comprising a framework region derived from a human antibody or a framework region variant thereof
  • the isolated antibody or antigen binding portion thereof comprises:
  • VL light chain variable region
  • VH Heavy chain variable region
  • the isolated antibody or antigen binding portion thereof comprises:
  • VL light chain variable region
  • VH Heavy chain variable region
  • the isolated antibody or antigen binding portion thereof comprises:
  • VL light chain variable region
  • VH Heavy chain variable region
  • the light chain variable region comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least the light chain variable region selected from (i) 99% or 100% homology of the amino acid sequence;
  • the heavy chain variable region comprises at least 90%, at least 95%, at least 96%, at least 97%, at least the heavy chain variable region selected from (ii) Amino acid sequence with 98%, at least 99% or 100% homology.
  • the isolated antibody or antigen binding portion thereof comprises:
  • VL The light chain variable region (VL), which includes an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 7; and the heavy chain variable region (VH), which includes the amino acid sequence with SEQ ID NO: 23 An amino acid sequence with at least 85% homology to the amino acid sequence;
  • VL The light chain variable region (VL), which includes an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 7; and the heavy chain variable region (VH), which includes the amino acid sequence with SEQ ID NO: 27 An amino acid sequence with at least 85% homology to the amino acid sequence;
  • the amino acid sequence is at least 85% homologous to the amino acid sequence.
  • the isolated antibody or antigen binding portion thereof comprises:
  • VL The light chain variable region (VL), which includes an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 7; and the heavy chain variable region (VH), which includes the amino acid sequence with SEQ ID NO: 23 An amino acid sequence with at least 85% homology to the amino acid sequence;
  • VL The light chain variable region (VL), which includes an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 7; and the heavy chain variable region (VH), which includes the amino acid sequence with SEQ ID NO: 27 An amino acid sequence with at least 85% homology to the amino acid sequence;
  • the amino acid sequence is at least 85% homologous to the amino acid sequence.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain variable region (VL), which includes an amino acid sequence that is at least 85% homologous to the amino acid sequence of SEQ ID NO: 74; and the heavy chain variable region (VH), which includes the amino acid sequence of SEQ ID NO: 75 An amino acid sequence with at least 85% sequence homology.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain variable region and the heavy chain variable region comprise at least 90%, at least 95%, and at least 95% of the light chain variable region and the heavy chain variable region selected from 1)-15).
  • the present invention provides an isolated antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region and a light chain variable region combination selected from any one of the following (a) to (c):
  • VL light chain variable region
  • VH Heavy chain variable region
  • the isolated antibody or antigen binding portion thereof comprises a light chain and a heavy chain, wherein:
  • the light chain comprises an amino acid sequence selected from the group consisting of the following amino acid sequences with at least 85% homology: SEQ ID NOs: 9, 13, 17, 21, 37, 39, 50 or 54 and Amino acid sequence in conservatively modified form; and
  • the heavy chain comprises an amino acid sequence selected from the group consisting of the following amino acid sequences with at least 85% homology: SEQ ID NOs: 25, 29, 33, 35, 52, or 56 and conservatively modified forms thereof Amino acid sequence.
  • the isolated antibody or antigen binding portion thereof comprises a light chain and a heavy chain, wherein:
  • the light chain comprises an amino acid sequence selected from the group consisting of the following amino acid sequences with at least 85% homology: SEQ ID NOs: 9, 13, 17, 21, 37, 39 or 50 and conservative modifications thereof The amino acid sequence of the form; and
  • the heavy chain comprises an amino acid sequence with at least 85% homology to an amino acid sequence selected from the group consisting of SEQ ID NOs: 25, 29, 33, 35 or 52 and amino acid sequences of conservatively modified forms thereof .
  • the isolated antibody or antigen binding portion thereof comprises a light chain and a heavy chain, wherein:
  • the light chain comprises an amino acid sequence with at least 85% homology to an amino acid sequence selected from the group consisting of SEQ ID NOs: 9, 13, 17, or 21 and amino acid sequences of conservatively modified forms thereof;
  • the heavy chain comprises an amino acid sequence with at least 85% homology in an amino acid sequence selected from the group consisting of SEQ ID NOs: 25, 29 or 33 and amino acid sequences of conservatively modified forms thereof.
  • the light chain comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a light chain selected from (I).
  • Source amino acid sequence; said heavy chain comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% homology with a heavy chain selected from (II) The amino acid sequence.
  • the isolated antibody or antigen binding portion thereof comprises:
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 9; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 25 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 9; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 29 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 9; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 33 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 13; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 25 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 13; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 29 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 13; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 33 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 17; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 25 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 17; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 29 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 17; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 33 ;
  • a light chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 21; and a heavy chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 25 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 21; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 29 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 21; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 33 ;
  • a light chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 37; and a heavy chain comprising an amino acid sequence with at least 85% homology to the amino acid sequence of SEQ ID NO: 35 ;
  • a light chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 39; and a heavy chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 35 ;
  • a light chain comprising an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 50; and a heavy chain comprising an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 52 ;or
  • a light chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 54; and a heavy chain comprising an amino acid sequence with at least 85% homology with the amino acid sequence of SEQ ID NO: 56 .
  • the light chain and the heavy chain respectively comprise at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, light chain and heavy chain selected from 1)-16). Amino acid sequence with at least 99% or 100% homology.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 9; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 25.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 9; and the heavy chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 29.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 9; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 33.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 13; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 25.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 13; and a heavy chain, which includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 29.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 13; and the heavy chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 33.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 17; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 25.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 17; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 29.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 17; and a heavy chain, which includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 33.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 21; and the heavy chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 25.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 21; and the heavy chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 29.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 21; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 33.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 37; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 35.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 39; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 35.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 50; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 52.
  • the isolated antibody or antigen binding portion thereof comprises:
  • the light chain includes an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 54; and a heavy chain including an amino acid sequence having at least 85% homology with the amino acid sequence of SEQ ID NO: 56.
  • the present invention provides an isolated antibody or antigen-binding portion thereof, wherein the isolated antibody or antigen-binding portion thereof comprises: (a) LCDR1 comprising SEQ ID NO: 1, (b) comprising LCDR2 of SEQ ID NO: 2, (c) LCDR3 of SEQ ID NO: 3, (d) HCDR1 of SEQ ID NO: 4, (e) HCDR2 of SEQ ID NO: 5, and (f) including SEQ ID NO: 6 HCDR3.
  • the present invention provides an isolated antibody or antigen-binding portion thereof, wherein the isolated antibody or antigen-binding portion thereof comprises: (a) LCDR1 comprising SEQ ID NO: 1, (b) comprising LCDR2 of SEQ ID NO: 2, (c) LCDR3 of SEQ ID NO: 3, (d) HCDR1 of SEQ ID NO: 4, (e) HCDR2 of SEQ ID NO: 45, and (f) of SEQ ID NO: 45 ID NO: 6 HCDR3.
  • the present invention provides an isolated antibody or antigen-binding portion thereof, which contains the light chain variable region (VL) shown in SEQ ID NO: 7 and the heavy chain shown in SEQ ID NO: 23. Chain variable region (VH).
  • VL light chain variable region
  • VH Chain variable region
  • the present invention provides an isolated antibody or antigen-binding portion thereof, which contains the light chain variable region (VL) shown in SEQ ID NO: 11 and the heavy chain shown in SEQ ID NO: 31. Chain variable region (VH).
  • VL light chain variable region
  • VH Chain variable region
  • the present invention provides an isolated antibody or antigen-binding portion thereof, which contains the light chain variable region (VL) shown in SEQ ID NO: 19 and the heavy chain shown in SEQ ID NO: 31. Chain variable region (VH).
  • VL light chain variable region
  • VH Chain variable region
  • the isolated antibody is a monoclonal antibody, chimeric antibody, humanized antibody, human engineered antibody, human antibody, bispecific antibody, Fv, single chain antibody (scFv), Fab, Fab ', Fab'-SH or F(ab') 2 .
  • the isolated antibody is IgG.
  • the isolated antibody is IgG1, IgG2, or IgG4.
  • the isolated antibody or antigen-binding portion thereof is a LIF (leukemia inhibitory factor) antagonist.
  • LIF leukemia inhibitory factor
  • the isolated antibody or antigen binding portion thereof can inhibit LIF expression and/or block LIF activity.
  • the isolated antibody or antigen-binding portion thereof can compete or cross-compete for binding to LIF.
  • the present invention provides a nucleotide composition comprising a nucleotide molecule encoding the isolated antibody or antigen-binding portion thereof according to the present invention.
  • the nucleotide molecule is DNA or RNA.
  • the nucleotide molecule is DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NOs: 7, 11, 15, 19, 46 or 82; and
  • VH variable heavy region
  • the nucleotide composition comprises:
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NOs: 7, 11, 15, 19 or 46;
  • VH variable heavy region
  • the nucleotide composition comprises:
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NOs: 7, 11, 15 or 19;
  • VH variable heavy region
  • the first nucleic acid molecule comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the first nucleic acid molecule selected from (i) Source DNA; the second nucleic acid molecule comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% homology to the second nucleic acid molecule selected from (ii) sexual DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 31 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule, which contains the DNA encoding the amino acid sequence shown in SEQ ID NO: 23 DNA of the variable region of the heavy chain (VH);
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule, which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 31 Chain variable region (VH) DNA;
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 27 DNA of the variable region of the heavy chain (VH);
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 31
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 31
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 46; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 48 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 74; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 75 DNA of the variable region of the heavy chain (VH) as shown; or
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 82; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 83
  • the DNA of the variable region of the heavy chain (VH) is shown.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 31 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule, which contains the DNA encoding the amino acid sequence shown in SEQ ID NO: 23 DNA of the variable region of the heavy chain (VH);
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule, which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 31 Chain variable region (VH) DNA;
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid molecule which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 27 DNA of the variable region of the heavy chain (VH);
  • the first nucleic acid molecule which contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule, which contains the encoding amino acid sequence shown in SEQ ID NO: 31
  • the first nucleic acid sequence which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid sequence, which contains the encoding amino acid sequence shown in SEQ ID NO: 23 The DNA of the variable region of the heavy chain (VH) shown;
  • the first nucleic acid sequence which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid sequence, which contains the encoding amino acid sequence shown in SEQ ID NO: 27 DNA of the variable region of the heavy chain (VH) as shown; or
  • the first nucleic acid sequence which contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid sequence, which contains the encoding amino acid sequence shown in SEQ ID NO: 31
  • the DNA of the variable region of the heavy chain (VH) is shown.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 7; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 31 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule contains the heavy chain encoding amino acid sequence shown in SEQ ID NO: 23 Variable region (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 11; and the second nucleic acid molecule contains DNA encoding the heavy chain shown in SEQ ID NO: 31 Variable region (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule contains DNA encoding the amino acid sequence shown in SEQ ID NO: 27.
  • Chain variable region (VH) DNA is
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 15; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 31 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 23 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 27 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 19; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 31 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 46; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 48 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 74; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 75 The variable region of the heavy chain (VH) DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains the DNA encoding the light chain variable region (VL) shown in SEQ ID NO: 82; and the second nucleic acid molecule contains the encoding amino acid sequence shown in SEQ ID NO: 83 The variable region of the heavy chain (VH) DNA.
  • the first nucleic acid sequence comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98% of the first nucleic acid sequence or the second nucleic acid sequence selected from 1)-15). , DNA sequences with at least 99% or 100% homology.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 7 is shown in SEQ ID NO: 8.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 11 is shown in SEQ ID NO: 12.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 15 is shown in SEQ ID NO: 16.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 19 is shown in SEQ ID NO: 20.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 46 is shown in SEQ ID NO: 47.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 74 is shown in SEQ ID NO: 76.
  • the DNA encoding the light chain variable region (VL) whose amino acid sequence is shown in SEQ ID NO: 82 is shown in SEQ ID NO: 72.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 23 is shown in SEQ ID NO: 24.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 27 is shown in SEQ ID NO: 28.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 31 is shown in SEQ ID NO: 32.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 48 is shown in SEQ ID NO: 49.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 75 is shown in SEQ ID NO: 77.
  • the DNA encoding the variable region of the heavy chain (VH) whose amino acid sequence is shown in SEQ ID NO: 83 is shown in SEQ ID NO: 73.
  • the nucleotide composition includes:
  • the first nucleic acid sequence which contains DNA encoding the light chain shown in SEQ ID NOs: 9, 13, 17, 21, 37, 39, 50 or 54;
  • the nucleotide composition includes:
  • the first nucleic acid molecule which contains DNA encoding the light chain shown in SEQ ID NOs: 9, 13, 17, 21, 37, 39 or 50;
  • a second nucleic acid molecule which contains DNA encoding the heavy chain with an amino acid sequence as shown in SEQ ID NOs: 25, 29, 33, 35 or 52.
  • the nucleotide composition includes:
  • the first nucleic acid molecule which contains DNA encoding the light chain with an amino acid sequence as shown in SEQ ID NOs: 9, 13, 17 or 21;
  • a second nucleic acid molecule which contains DNA encoding the heavy chain whose amino acid sequence is as shown in SEQ ID NOs: 25, 29 or 33.
  • the first nucleic acid molecule comprises at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the first nucleic acid molecule selected from (I) Homologous DNA; the second nucleic acid molecule contains at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the second nucleic acid molecule selected from (II) Sourced DNA.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 13; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 13; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29 ;
  • the first nucleic acid molecule which contains the DNA encoding the light chain with the amino acid sequence as shown in SEQ ID NO: 13; and the second nucleic acid molecule, which contains the DNA encoding the heavy chain with the amino acid sequence as shown in SEQ ID NO: 33 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29 ;
  • the first nucleic acid molecule which contains the DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule, which contains the DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33 ;
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence as SEQ ID NO: 21; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence as SEQ ID NO: 29;
  • the first nucleic acid molecule which contains the DNA encoding the light chain with the amino acid sequence as shown in SEQ ID NO: 21; and the second nucleic acid molecule, which contains the DNA encoding the heavy chain with the amino acid sequence as shown in SEQ ID NO: 33 ;
  • the first nucleic acid molecule which contains the DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 37; and the second nucleic acid molecule, which contains the DNA with the heavy chain encoding the amino acid sequence shown in SEQ ID NO: 35 ;
  • the first nucleic acid molecule and the second nucleic acid molecule comprise at least 90%, at least 95%, at least 96%, at least the first nucleic acid molecule or the second nucleic acid molecule selected from 1)-16) DNA with 97%, at least 98%, at least 99%, or 100% homology.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 9; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 13; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 13; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 13; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 29.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 17; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 21; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 25.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence as SEQ ID NO: 21; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence as SEQ ID NO: 29.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 21; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 33.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 37; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 35.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 39; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 35.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 50; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 52.
  • the nucleotide composition comprises:
  • the first nucleic acid molecule contains DNA encoding the light chain with the amino acid sequence shown in SEQ ID NO: 54; and the second nucleic acid molecule contains DNA encoding the heavy chain with the amino acid sequence shown in SEQ ID NO: 56.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 9 is shown in SEQ ID NO: 10.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 13 is shown in SEQ ID NO: 14.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 17 is shown in SEQ ID NO: 18.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 21 is shown in SEQ ID NO: 22.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 37 is shown in SEQ ID NO: 38.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 39 is shown in SEQ ID NO: 40.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 50 is shown in SEQ ID NO: 51.
  • the DNA encoding the light chain whose amino acid sequence is shown in SEQ ID NO: 54 is shown in SEQ ID NO: 55.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 25 is shown in SEQ ID NO: 26.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 29 is shown in SEQ ID NO: 30.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 33 is shown in SEQ ID NO: 34.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 35 is shown in SEQ ID NO: 36.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 52 is shown in SEQ ID NO: 53.
  • the DNA encoding the heavy chain whose amino acid sequence is shown in SEQ ID NO: 56 is shown in SEQ ID NO: 57.
  • the present invention provides a vector containing the nucleotide composition according to the present invention.
  • the vector is a eukaryotic expression vector, a prokaryotic expression vector or a viral vector.
  • the present invention provides a host cell containing the vector of the present invention.
  • the host cell containing the vector is obtained by transformation with the vector.
  • the host cell is a bacteria, yeast, or mammalian cell.
  • the host cell is Escherichia coli, Pichia pastoris, Chinese hamster ovary cells or human embryonic kidney 293 cells.
  • the present invention provides a method for preparing the isolated antibody or antigen-binding portion thereof of the present invention, the method comprising expressing the antibody or antigen-binding fragment thereof in the host cell of the present invention , And separate the antibody or its antigen-binding fragment.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective therapeutic dose of the aforementioned isolated antibody or antigen-binding portion thereof, and a pharmaceutically acceptable excipient.
  • the present invention provides a reagent for measuring LIF in a biological sample, which comprises the aforementioned isolated antibody or antigen-binding portion thereof.
  • the biological sample is blood, serum, urine, biopsy material, tumor, or any tissue suspected of containing abnormal LIF levels.
  • the present invention provides a method for inhibiting LIF expression and/or blocking LIF activity, comprising administering to a patient in need an effective therapeutic dose of the aforementioned isolated antibody or antigen-binding portion thereof, and/or the aforementioned Pharmaceutical composition.
  • the present invention provides the use of the above-mentioned isolated antibody or antigen-binding portion thereof, and/or the above-mentioned pharmaceutical composition in a therapeutic drug for inhibiting LIF expression and/or blocking LIF activity.
  • the present invention provides the aforementioned isolated antibody or antigen binding portion thereof for inhibiting LIF expression and/or blocking LIF activity, and/or the aforementioned pharmaceutical composition.
  • the present invention provides a method for treating diseases or disorders associated with LIF, the method comprising administering to a patient in need an effective therapeutic dose of the aforementioned isolated antibody or antigen-binding portion thereof, and/or the aforementioned Pharmaceutical composition.
  • the disease associated with LIF is tumor.
  • the tumor is a solid tumor.
  • the solid tumor includes glioblastoma, lung cancer, ovarian cancer, colorectal cancer, pancreatic cancer, or prostate cancer.
  • the present invention provides the use of the above-mentioned isolated antibody or antigen-binding portion thereof, and/or the above-mentioned pharmaceutical composition in a medicament for treating diseases or disorders related to LIF.
  • the disease associated with LIF is tumor.
  • the tumor is a solid tumor.
  • the solid tumor includes glioblastoma, lung cancer, ovarian cancer, colorectal cancer, pancreatic cancer, or prostate cancer.
  • the present invention provides the aforementioned isolated antibody or antigen-binding portion thereof, and/or the aforementioned pharmaceutical composition for use in the treatment of diseases or disorders related to LIF.
  • the disease associated with LIF is tumor.
  • the tumor is a solid tumor.
  • the solid tumor includes glioblastoma, lung cancer, ovarian cancer, colorectal cancer, pancreatic cancer, or prostate cancer.
  • a disease or condition related to LIF means that blocking LIF and LIRR and/or GP130 can treat, alleviate, alleviate and/or stabilize the disease or condition.
  • the present invention provides a method for detecting LIF in a biological sample, comprising: (i) obtaining a tissue or fluid sample of a subject, (ii) exposing the tissue or fluid sample to the aforementioned isolated antibody or The antigen-binding portion or the above-mentioned reagent; and (iii) detecting the LIF bound to the tissue or fluid sample obtained from (ii) and comparing the LIF bound to the control sample, wherein the amount of LIF bound to the control is increased Shows abnormal levels of LIF production, expression, or activation.
  • tissue or fluid sample includes blood, serum, urine, biopsy material, tumor, or any tissue suspected of containing abnormal LIF levels.
  • Figure 1 shows the ability of the LIF antibody of the present invention to bind to human LIF protein
  • Figure 2 shows the binding ability of the LIF antibody of the present invention to the mouse LIF protein
  • Figure 3 shows the binding ability of the LIF antibody of the present invention with the cynomolgus monkey LIF protein
  • Figure 4 shows the binding affinity of the 38E10E1C11 antibody of the present invention to the antigen
  • Figure 5 shows the ability of the 38E10E1C11 antibody of the present invention to compete with LIFR for binding to human LIF protein
  • Figure 6 shows the ability of the P36-333 antibody of the present invention to compete with the receptor GP130 to bind to human LIF protein
  • Figure 7 shows the cross-reaction of the 38E10E1C11 antibody and P36-333 antibody of the present invention with IL-6 family proteins
  • Figure 8 shows that the present invention shows that the 38E10E1C11R antibody of the present invention recognizes denatured human LIF protein.
  • Panel A in Figure 9 shows that the 38E10E1C11 antibody of the present invention inhibits human LIF protein-induced phosphorylation of colon cancer cells (HCT116);
  • Panel B in Figure 9 shows that the 38E10E1C11 antibody of the present invention inhibits human LIF protein-induced pancreatic cancer cells ( KP4) Phosphorylation of STAT3;
  • FIG. 10 shows that the 38E10E1C11 antibody of the present invention blocks the activation of STAT3 of pancreatic cancer cell KP4 by human LIF secreted by CT26-hLIF cells;
  • Figure 11 shows that the P36-333 antibody of the present invention inhibits the phosphorylation of STAT3 in colon cancer cells (HCT116) induced by human LIF protein;
  • Figure 12 shows that the 38E1E1C11 and P36-033 antibodies of the present invention reverse the inhibition of LIF on the proliferation of M1 cells;
  • Figure 13 shows that the 38E10E1C11 antibody of the present invention inhibits the growth of CT26-hLIF cells in BABL/c mice;
  • Figure 14 shows the sensitivity of three human pancreatic cancer cell lines to human LIF protein stimulation
  • Figure 15 shows that the 38E10E1C11R antibody of the present invention inhibits the phosphorylation of STAT3 in KP4 cells caused by LIF protein stimulation;
  • Panel A in Figure 16 shows the experimental results of the 38E10E1C11 antibody of the present invention recognizing full-length human LIF protein and hybrid LIF protein.
  • Panel B in Figure 16 shows that the 38E10E1C11 antibody of the present invention can reverse the full-length human LIF protein and hybrid LIF.
  • Figures 17A-17B show the experimental results of the binding properties of the humanized anti-LIF antibody of the present invention to the antigen
  • Figure 18 shows the experimental results of the non-specific binding affinity of the humanized anti-LIF antibody of the present invention
  • Figure 19 shows the experimental results of the humanized anti-LIF antibody of the present invention competing with LIFR for binding to human LIF protein
  • Figure 20 shows the experimental results of the humanized anti-LIF antibody of the present invention competing with GP130 for binding to human LIF protein
  • Figure 21 shows the experimental results of the humanized anti-LIF antibody of the present invention in recognizing antigen specificity
  • Panel A in Figure 22 shows the experimental results of the present invention showing that the humanized anti-LIF antibody of the present invention recognizes full-length human LIF protein and hybrid LIF protein
  • panel B in Figure 22 shows the humanized anti-LIF antibody of the present invention The experimental results of blocking the proliferation inhibition of M1 cells by full-length LIF protein and hybrid protein;
  • Figure 23 shows the experimental results of the humanized anti-LIF antibody of the present invention inhibiting the phosphorylation of STAT3 induced by LIF protein
  • Figure 24 shows the results of the experiment that the humanized anti-LIF antibody of the present invention can reverse the inhibition of human LIF protein on the proliferation of M1 cells.
  • Figure 25 shows that the humanized anti-LIF antibody detected by the HTFR method inhibits the phosphorylation level of STAT3 and the total STAT3 level induced by LIF protein.
  • Figure 26 shows the ADCC effect of humanized anti-LIF antibodies.
  • LIF Leukemia Inhibitory Factor
  • the amino acid sequence of LIF is publicly available (Ref Seq NM_001257135).
  • the LIF can be human, mouse (Ref Seq NM_001039537.2), or cynomolgus LIF (XM_015457518.1).
  • LIF can be recombinant and/or glycosylated or non-glycosylated.
  • antibody as used herein may include whole antibodies and any antigen-binding fragments thereof (ie, “antigen-binding portions") or single chains.
  • antibody refers to a glycoprotein or an antigen binding portion thereof comprising at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds.
  • Each heavy chain includes a heavy chain variable region (abbreviated as VH herein) and a heavy chain constant region.
  • VH heavy chain variable region
  • the heavy chain constant region contains three domains, CH1, CH2, and CH3.
  • each light chain includes a light chain variable region (abbreviated as VL herein) and a light chain constant region.
  • the light chain constant region contains a domain CL.
  • the VH and VL regions can be further subdivided into hypervariable regions (called complementarity determining regions (CDR)) and more conservative regions (called framework regions (FR)), which are arranged in a hybrid manner.
  • CDR complementarity determining regions
  • FR framework regions
  • the CDR in the VH region is abbreviated as HCDR, that is, the three CDRs in the VH region can be abbreviated as HCDR1, HCDR2, and HCDR3
  • the CDR in the VL region is abbreviated as LCDR, that is, the three CDRs in the VL region can be abbreviated as LCDR1, LCDR2. LCDR3.
  • Each VH and VL is composed of three CDRs and four FRs, which are arranged in the following order from the amino terminus to the carboxy terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain binding domains that interact with antigens.
  • the heavy chain of the antibody may or may not contain terminal lysine (K) or terminal glycine and lysine (GK). Therefore, any heavy chain sequence and heavy chain constant region sequence provided herein can end in GK or G, or lack K or GK, regardless of the last amino acid of the sequence. This is because the terminal lysine, sometimes glycine and lysine, is cut off during antibody expression.
  • Antibodies usually specifically bind to their cognate antigens with high affinity, which is reflected by the dissociation constant (K D ) of 10 -7 M to 10 -11 M or less. It is generally considered greater than about 10 -6 M K D indicating any non-specific binding.
  • K D dissociation constant
  • an antibody that "specifically binds" to an antigen refers to an antibody that binds to an antigen with high affinity and is substantially the same antigen, but does not bind to an unrelated antigen with high affinity, and high affinity means that it has 10-7 M or less, preferably 10 -8 M or less, even more preferably 5 ⁇ 10 -9 M or less, and most preferably a K D between 10 -8 M and 10 -10 M or less.
  • the antigen exhibits a high degree of sequence identity with a given antigen, for example if it exhibits at least 80%, at least 90%, at least 95%, at least 97%, or at least 99% or greater sequence identity with the sequence of the given antigen, Then the antigen is "substantially the same" as the given antigen.
  • the immunoglobulin can be from any of the generally known isotypes, including but not limited to IgA, secretory IgA, IgG, and IgM.
  • IgG isotypes are divided into multiple subclasses in some species: IgG1, IgG2, IgG3, and IgG4 in humans, and IgG1, IgG2a, IgG2b, and IgG3 in mice.
  • the anti-LIF antibodies described herein have a human IgG1 or mouse IgG1 subtype.
  • Immunoglobulins (such as human IgG1) exist in several isotypes, which differ from each other by at most a few amino acids.
  • "antibody” can include naturally-occurring and non-naturally-occurring antibodies; monoclonal and multi-strain antibodies; chimeric and humanized antibodies; human and non-human antibodies; fully synthetic antibodies; and single-chain antibodies.
  • antigen-binding portion of an antibody refers to one or more fragments of the antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of antibodies can be performed by fragments of full-length antibodies.
  • binding fragments covered by the term "antigen-binding portion" of antibodies include (i) Fab fragments, which are monovalently composed of VL, VH, CL, and CH1 domains Fragment; (ii) F(ab') 2 fragment, which is a bivalent fragment containing two Fab fragments connected at the hinge region by a disulfide bridge; (iii) Fd fragment, which consists of VH and CH1 domains (Iv) Fv fragment, which is composed of a one-arm VL and VH domain of an antibody, (v) dAb fragment (Ward et al.
  • conservatively modified form of amino acid sequence refers to an amino acid modification that does not significantly affect or change the binding properties of an antibody comprising the amino acid sequence, and the modification includes amino acid substitutions, additions, and deletions. Modifications can be introduced into the antibodies of the invention by standard techniques such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are those in which amino acid residues are replaced by amino acid residues with similar side chains. A family of amino acid residues with similar side chains has been determined in the art.
  • amino acids with basic side chains such as lysine, arginine, histidine
  • amino acids with acidic side chains such as aspartic acid, glutamic acid
  • uncharged polar sides Chain amino acids such as glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • amino acids with non-polar side chains such as alanine, va Acid, leucine, isoleucine, proline, phenylalanine, methionine
  • amino acids with ⁇ -branched side chains such as threonine, valine, isoleucine
  • amino acids with aromatic side chains such as tyrosine, phenylalanine, tryptophan, histidine
  • one or more amino acid residues in the CDR region of the antibody of the present invention can be replaced by other amino acid residues from the same side chain family, and the function measurement method described herein can be used to retain the function of the modified antibody. carry out testing.
  • the number of conservative modifications does not exceed one or two.
  • amino acid sequences described herein are desirable in the present invention, especially those human heavy chain constant regions to adapt the sequence to the desired allotype, such as those found in Asian populations.
  • one or more CDRs or CDR groups of an antibody can be grafted into a framework (such as a human framework) to provide an antibody molecule.
  • the framework regions can be human germline gene sequences or non-germline gene sequences.
  • the framework can be reproductive, where one or more residues within the framework can be exchanged to match the residues at a comparable position within the most similar human germline framework.
  • the binding member of the present invention may be an isolated VH domain having a group of HCDRs within a human germline framework, for example, a human germline IgG VH framework.
  • the binding member may also have a VL domain containing the LCDR group, such as in the human germline IgG VL framework.
  • VH and/or VL scaffold residues can be modified as discussed, as exemplified herein, such as using site-directed mutagenesis.
  • the VH or VL domains, or binding members of the present invention include such VL domains.
  • Changes can be made in one or more framework regions and/or one or more CDRs, and the changes usually do not result in loss of function, so the binding member comprising such changes in amino acid sequence should keep binding and/or neutralizing LIF ability. It can maintain the same number of binding and/or neutralizing capabilities as the binding members that have not undergone changes, as measured by the analytical method described herein.
  • a binding member comprising such a changed amino acid sequence may have an improved ability to bind and/or neutralize LIF.
  • Variations can include replacing one or more amino acid residues with non-naturally occurring or non-standard amino acids, modifying one or more amino acid residues into non-naturally occurring or non-standard forms, or replacing one or more non-natural Existing or non-standard amino acids are inserted into this sequence. Examples of the location and number of changes in the sequence of the invention are described elsewhere herein.
  • Naturally occurring amino acids include G, A, V, L, I, M, P, F, W, S, T, N, Q, Y, C, K, R, H, 20 "standard” L-amino acids of D and E.
  • Non-standard amino acids include any other residues that can be incorporated into the polypeptide backbone or modified from existing amino acid residues.
  • Non-standard amino acids can be naturally occurring or non-naturally occurring.
  • Several naturally occurring non-standard amino acids are known in the art, 4-hydroxyproline, 5-hydroxylysine, 3-methylhistidine, N-ethylserine, etc. (Voet & Voet, 1995, Biochemistry, 2nd Edition, (Wiley)).
  • Those amino acid residues derivatized at their N- ⁇ position will only be positioned at the N-terminus of the amino acid sequence.
  • the amino acid in the present invention is an L-amino acid, but it may be a D-amino acid. Therefore changes may include modification with L-amino acids or replacement of L-amino acids with D-amino acids.
  • the formylated, acetylated and/or phosphorylated forms of amino acids are known, and the amino acids of the present invention can be modified as such.
  • amino acid sequences in the binding members and antibody domains of the present invention may include the unnatural or non-standard amino acids described above.
  • Non-standard amino acids such as D-amino acids
  • D-amino acids can be incorporated into the amino acid sequence during synthesis, or by modification or substitution of "original" standard amino acids after amino acid synthesis.
  • non-standard and/or non-naturally occurring amino acids improves the diversity of structure and function, and can increase the potential to achieve the desired LIF binding and neutralizing properties in the binding members of the present invention.
  • D-amino acids and analogs have been shown to have better pharmacokinetic properties due to the degradation of polypeptides with L-amino acids in vivo after administration to animals such as humans.
  • the generation of the new VH or VL region with CDR-derived sequences of the present invention can use one or more random mutagenesis selected from VH and/or VL genes to generate mutants in all the variant regions.
  • a technique is described in Gram et al. (Gram et al., 1992, Proc. Natl. Acad. Sci., USA, 89: 3576-3580), which uses error-prone PCR.
  • one or more amino acid substitutions are made in all variant regions or CDR groups.
  • Another method that can be used is targeted mutagenesis of the CDR regions of VH or VL genes.
  • Such methods are published in Barbas et al. (Barbas et al., 1994, Proc. Natl Acad. Sci., USA, 91: 3809-3813) and Schier et al. (Schier et al., 1996, J. Mol. Biol. 263: 551-567).
  • VH and VL domain amino acid sequence variants of the specific sequences disclosed herein can be used in accordance with the present invention, as discussed.
  • Specific variants may include one or more amino acid sequence changes (additions, deletions, substitutions and/or insertions of amino acid residues).
  • the variant has less than about 17, less than 9, or less than 5 such changes.
  • the CDR amino acid sequence substantially as described herein can be carried as a CDR in a human antibody variant structure region or most of it.
  • the HCDR3 sequence substantially as described herein represents an embodiment of the present invention, and each of these can be carried as a CDR in a human antibody variant region or most of it, optionally in combination with the HCDR1, HCDR2, LCDR1, and LCDR1 of the present invention. LCDR2 and LCDR3.
  • the term “monoclonal antibody” refers to an antibody that exhibits a single binding specificity and affinity for a specific epitope, or an antibody composition in which all antibodies in the antibody composition are displayed for a specific epitope The single binding specificity and affinity.
  • these monoclonal antibodies will be derived from a single antibody-encoding cell or nucleic acid, and will proliferate without intentionally introducing any sequence changes. Therefore, the term “human monoclonal antibody” refers to a monoclonal antibody having variable regions derived from human germline immunoglobulin sequences, and optionally constant regions.
  • the human monoclonal antibody is obtained by, for example, fusing B cells and immortal cells obtained from a transgenic or transchromosomal non-human animal (e.g., a transgenic mouse having a genome containing a human heavy chain transgene and a light chain transgene) Of hybridomas.
  • a transgenic or transchromosomal non-human animal e.g., a transgenic mouse having a genome containing a human heavy chain transgene and a light chain transgene
  • mAb refers to a monoclonal antibody.
  • the term "recombinant human antibody” includes all human antibodies prepared, expressed, produced or isolated by recombinant means, such as (a) from an animal (such as a mouse) that has been transgenic or transchromosome to a human immunoglobulin gene Or an antibody isolated from a hybridoma prepared therefrom, (b) an antibody isolated from a host cell transformed to express the antibody, such as an antibody isolated from a transfectoma, (c) an antibody isolated from a recombinant, combinatorial human antibody library, and ( d) Antibodies prepared, expressed, produced or isolated by any other means involving the splicing of human immunoglobulin gene sequences to other DNA sequences.
  • variable and constant regions that utilize specific human germline immunoglobulin sequences and are encoded by germline genes, but include subsequent rearrangements and mutations that occur, for example, during antibody maturation.
  • the variable region contains antigen bindings encoded by different genes rearranged to form antibodies specific to foreign antigens Structural domain.
  • the variable region can be further modified with multiple single amino acid changes (called somatic mutations or hypermutations) to increase the affinity of the antibody to the foreign antigen.
  • the constant region will further change in response to the antigen (ie, isotype switching).
  • the rearranged and somatically mutated nucleic acid sequences encoding light chain and heavy chain immunoglobulin polypeptides in response to the antigen may be different from the original germline sequence, but will be substantially the same or similar (ie, have at least 80% identity) ).
  • a “human” antibody refers to an antibody having a variable region: the framework and CDR regions in the variable region are all derived from human germline immunoglobulin sequences. In addition, if the antibody contains a constant region, the constant region is also derived from human germline immunoglobulin sequences.
  • the antibodies described herein may include amino acid residues that are not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random mutagenesis in vitro or site-specific mutagenesis or by somatic mutation in vivo).
  • human antibody is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species (e.g., mouse) have been grafted onto human framework sequences.
  • mammalian species e.g., mouse
  • human antibody and “fully human” antibody are used synonymously.
  • a “humanized” antibody refers to an antibody in which some, most, or all of the amino acids other than the CDR domains of the non-human antibody are replaced by corresponding amino acids derived from human immunoglobulins.
  • some, most, or all of the amino acids outside the CDR domain have been replaced by amino acids from human immunoglobulin, while some, most, or all of the amino acids in one or more CDR regions have been replaced by amino acids from human immunoglobulin. All amino acids have not changed. Small additions, deletions, insertions, substitutions or modifications of amino acids can be tolerated, as long as they do not eliminate the ability of the antibody to bind to a specific antigen.
  • "Humanized” antibodies retain antigen specificity similar to the original antibody.
  • a “chimeric antibody” refers to an antibody in which the variable region is derived from one species and the constant region is derived from another species, for example, an antibody in which the variable region is derived from a mouse antibody and the constant region is derived from a human antibody.
  • the functional antibody fragments of the present invention include any functional fragments whose half-life is increased by chemical modification such as by PEGylation or incorporated into liposomes.
  • the antibodies of the invention include bispecific antibodies.
  • Bispecific or bifunctional antibodies form the second generation of monoclonal antibodies, in which two different variant regions are combined into the same molecule (Holliger and Bohlen, 1999 Cancer and metastasis rev. 18: 411-419). From their ability to recruit new effector functions or target some molecules to the surface of tumor cells, their applications in the field of diagnosis and treatment have been elucidated.
  • bispecific antibodies can be conventional bispecific antibodies, which can be manufactured in various ways (Holliger P. & Winter G. Current Opinion Biotechnol. 4, 446-449: 1993), such as chemical preparations , Or from a hybrid hybridoma, or can be any of the bispecific antibody fragments mentioned above.
  • bispecific antibodies include those of the BiTETM method, in which the binding domains of two antibodies with different specificities can be used and linked directly by a short flexible peptide. This combines two antibodies on a short single polypeptide chain.
  • the diabody and scFc are constructed without the Fc region, and only the variant region is used, potentially reducing the effect of the anti-idiotypic response.
  • Bispecific antibodies can be constructed as full IgG, bispecific (Fab')2, (Fab')PEG, diabody or other bispecific scFv. Furthermore, two bispecific antibodies can be linked to form a tetravalent antibody using conventional methods known in the art.
  • bispecific diabodies are also particularly useful because they can be easily constructed and expressed in E. coli.
  • diabodies with appropriate binding specificity and many other polypeptides, such as antibody fragments
  • Bispecific full antibodies can be prepared by different engineering methods, which are described in Ridgeway et al. (Ridgeway, JBB et al., Protein Eng. 9, 616-621 , 1996) or WO1996/27011, WO1998/50431 and WO2006/028936.
  • Modified heavy chain constant region refers to a heavy chain constant region comprising constant domains CH1, hinge, CH2, and CH3, wherein one or more constant domains are from different isotypes (eg, IgG1, IgG2, IgG3, IgG4).
  • the modified constant region includes a human IgG2 CH1 domain and a human IgG2 hinge and a human IgG1 CH3 domain fused to the human IgG1 CH2 domain.
  • these modified constant regions also include amino acid modifications in one or more domains relative to the wild-type amino acid sequence.
  • isotype refers to the antibody class (eg, IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE antibodies) encoded by heavy chain constant region genes.
  • “Alterotype” refers to naturally-occurring variants within a specific homotypic group that differ by a few amino acids (for example, see Jefferis et al. (2009) mAbs 1:1).
  • the antibodies described herein can have any isotype.
  • Antibody that recognizes an antigen and "antibody that specifically targets an antigen” are used interchangeably with the term “antibody that specifically binds to an antigen” herein.
  • isolated antibody is intended to refer to an antibody that is substantially free from other antibodies with different antigen specificities (for example, an isolated antibody that specifically binds to LIF is substantially free from specific binding other than LIF. Antibodies to other antigens). However, an isolated antibody that specifically binds to an epitope of LIF may have cross-reactivity against other LIF proteins of different species.
  • effector function refers to the interaction of the Fc region of an antibody with an Fc receptor or ligand, or a biochemical event derived therefrom.
  • exemplary “effector functions” include Clq binding, complement-dependent cytotoxicity (CDC), Fc receptor binding, Fc ⁇ R-mediated effector functions (e.g. ADCC and antibody-dependent cell-mediated phagocytosis (ADCP)) and Down-regulate cell surface receptors (e.g., B cell receptor; BCR). These effector functions usually require a combination of an Fc region and a binding domain (such as an antibody variable domain).
  • Fc receptor or “FcR” is a receptor that binds to the Fc region of an immunoglobulin.
  • the FcR that binds to an IgG antibody includes receptors of the Fc ⁇ R family, including allelic variants and alternatively spliced forms of these receptors.
  • the Fc ⁇ R family consists of three activating receptors (Fc ⁇ RI, Fc ⁇ RIII and Fc ⁇ RIV in mice; Fc ⁇ RIA, Fc ⁇ RIIA and Fc ⁇ RIIIA in humans) and one inhibitory receptor (Fc ⁇ RIIB).
  • Table A The various properties of human Fc ⁇ R are summarized in Table A.
  • Hinge refers to the domain of the constant region of the heavy chain that connects the CH1 domain and the CH2 domain and includes the upper, middle and lower parts of the hinge ( Roux et al., J. Immunol. 1998 161:4083).
  • the hinge provides the different flexibility between the binding region of the antibody and the effector region and also provides a site for the intermolecular disulfide bond connection between the constant regions of the two heavy chains.
  • hinge includes wild-type hinges and variants thereof (for example, non-naturally occurring hinges or modified hinges).
  • IgG2 hinge includes wild-type IgG2 hinges and having 1, 2, 3, 4, 5, 1-3, 1-5, 3-5, and/or at most 5, Variants with 4, 3, 2 or 1 mutations (such as substitutions, deletions or additions).
  • CH1 domain refers to the constant region of the heavy chain that connects the variable domain with the hinge in the constant domain of the heavy chain.
  • CH1 domain includes wild-type CH1 domains and variants thereof (for example, non-naturally occurring CH1 domains or modified CH1 domains).
  • CH1 domain includes wild-type CH1 domains and those having 1, 2, 3, 4, 5, 1-3, 1-5, 3-5, and/or at most Variants with 5, 4, 3, 2 or 1 mutations (e.g. substitutions, deletions or additions).
  • Exemplary CH1 domains include CH1 domains with mutations that alter the biological activity of the antibody (e.g., ADCC, CDC, or half-life).
  • CH2 domain refers to the heavy chain constant region that connects the hinge in the heavy chain constant domain with the CH3 domain.
  • CH2 domain includes wild-type CH2 domains and variants thereof (for example, non-naturally occurring CH2 domains or modified CH2 domains).
  • CH2 domain includes wild-type CH2 domains and those having 1, 2, 3, 4, 5, 1-3, 1-5, 3-5, and/or at most Variants with 5, 4, 3, 2 or 1 mutations (such as substitutions, deletions or additions).
  • Exemplary CH2 domains include CH2 domains with mutations that alter the biological activity of the antibody (e.g., ADCC, CDC, or half-life).
  • CH3 domain refers to the heavy chain constant region that is the C-terminus of the CH2 domain in the heavy chain constant domain.
  • CH3 domain includes wild-type CH3 domains and variants thereof (for example, non-naturally occurring CH3 domains or modified CH3 domains).
  • CH3 domain includes wild-type CH3 domains and those having 1, 2, 3, 4, 5, 1-3, 1-5, 3-5, and/or at most Variants with 5, 4, 3, 2 or 1 mutations (such as substitutions, deletions or additions).
  • Exemplary CH3 domains include CH3 domains with mutations that alter the biological activity of the antibody (e.g., ADCC, CDC, or half-life).
  • CL domain refers to the constant domain of the light chain.
  • CL domain includes wild-type CL domains and variants thereof.
  • the “native sequence Fc region” or “native sequence Fc” includes the same amino acid sequence as the amino acid sequence of the Fc region found in nature.
  • the natural sequence human Fc region includes the natural sequence human IgG1 Fc region; the natural sequence human IgG2 Fc region; the natural sequence human IgG3 Fc region; and the natural sequence human IgG4 Fc region and its natural variants.
  • the native sequence Fc includes multiple allotypes of Fc (for example, see Jefferis et al. (2009) mAbs 1:1).
  • epitopes within protein antigens can be formed from both continuous amino acids (usually linear epitopes) or non-contiguous amino acids juxtaposed by tertiary folding of the protein (usually conformational epitopes). When exposed to a denaturing solvent, epitopes formed from consecutive amino acids are usually but not always retained, and epitopes formed by tertiary folding are usually lost when treated with a denaturing solvent. In a unique spatial conformation, an epitope usually includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids .
  • epitope mapping Methods of determining the epitope bound by a given antibody (ie epitope mapping) are well known in the art and include, for example, immunoblotting and immunoprecipitation analysis, in which overlapping or continuous peptides (e.g. from LIF) are tested against a given antibody (e.g., anti- LIF antibody) reactivity.
  • Methods for determining the spatial conformation of epitopes include techniques in the art and those described herein, such as x-ray crystallography, 2-dimensional nuclear magnetic resonance, and HDX-MS (see, for example, Epitope Mapping Protocols in Molecular Biology, Vol. 66 , Edited by GEMorris (1996)).
  • An antibody that "competes with another antibody for binding to a target” refers to an antibody that inhibits (partially inhibits or completely inhibits) the binding of another antibody to the target.
  • Known competition experiments can be used to determine whether two antibodies compete with each other for binding to the target, that is, whether one antibody inhibits the binding of the other antibody to the target and the degree of inhibition.
  • the antibody competes with another antibody for binding to the target and inhibits at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% Of the combination.
  • the degree of inhibition or competition can vary depending on which antibody is a "blocking antibody” (ie, a cold antibody that is first incubated with the target).
  • Competition assays can be implemented as described in, for example, the following documents: Ed Harlow and David Lane, Cold Spring Harb Protoc; 2006; doi:10.1101/pdb.prot4277 or "Using Antibodies", Ed Harlow and David Lane, Cold Spring Harbor Laboratory Press, Chapter 11 of ColdSpring Harbor, NY, USA 1999. Competing antibodies bind to the same epitope, overlapping epitopes, or adjacent epitopes (e.g., as evidenced by steric hindrance).
  • Kassoc or “Ka” is intended to refer to the association rate constant of a specific antibody-antigen interaction
  • Kdis or “Kd” is intended to refer to a specific The dissociation rate constant of the antibody-antigen interaction
  • K D is intended to refer to the equilibrium dissociation constant, which is obtained from the ratio of Kd to Ka (ie Kd/Ka) and expressed in molar concentration (M).
  • the K D value of an antibody can be determined using methods well established in the art.
  • the preferred method for determining the K D of an antibody is by using surface plasmon resonance, preferably using a biosensor system, such as Surface plasmon resonance system or flow cytometry or Scatchard analysis.
  • EC50 in the context of in vitro or in vivo analysis using antibodies or antigen-binding fragments thereof refers to the concentration of the antibody or its antigen-binding portion that induces 50% of the maximum response, that is, the reaction halfway between the maximum response and the baseline.
  • IC50 in the functional analysis, IC50 is the concentration of the binding member that can reduce the biological response to 50% of its maximum value, in nM. In ligand-binding studies, IC50 is the concentration that reduces receptor binding to 50% of the maximum specific binding level.
  • the IC50 can be calculated by plotting the percentage of the maximum biological activity response as a function of the log of the binding member concentration, and using a software program such as Origin (OriginLab Software Company, Northampton, Massachusetts, USA) to fit the S function to the data to generate the IC50 value.
  • Origin OpinLab Software Company, Northampton, Massachusetts, USA
  • the potency is determined or measured using one or more analytical methods known to those skilled in the art and/or described or referenced herein.
  • the neutralizing potency of the binding members can be expressed as the geometric mean (Geomean).
  • naturally occurring refers to the fact that the target can be found in nature when applied to a target.
  • polypeptide or polynucleotide sequences that exist in organisms (including viruses) that can be isolated from natural sources and have not been intentionally modified by humans in the laboratory are naturally occurring.
  • Polypeptide refers to a chain comprising at least two consecutively linked amino acid residues, and the length of the chain has no upper limit.
  • One or more amino acid residues in the protein may contain modifications, such as (but not limited to) glycosylation, phosphorylation, or disulfide bonds.
  • a “protein” may comprise one or more polypeptides.
  • nucleic acid molecule is intended to include DNA molecules and RNA molecules.
  • the nucleic acid molecule may be single-stranded or double-stranded, and may be cDNA.
  • the “conservative sequence modification” of the sequence described in the SEQ ID NOs described herein is also provided, that is, the nucleotide and amino acid sequence modifications that do not eliminate the binding of the antibody encoded by the nucleotide sequence or containing the amino acid sequence to the antigen. These conservative sequence modifications include conservative nucleotide and amino acid substitutions, and nucleotide and amino acid additions and deletions.
  • the modification can be introduced into the SEQ ID NOs described herein by standard techniques known in the art (such as site-directed mutagenesis and PCR-mediated mutagenesis).
  • Conservative sequence modifications include conservative amino acid substitutions in which amino acid residues are replaced with amino acid residues with similar side chains. Families of amino acid residues with similar side chains are already defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), amino acids with acidic side chains (e.g., aspartic acid, glutamic acid), and side chains with no electrical polarity.
  • Amino acids e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • amino acids with non-polar side chains e.g., alanine, valine
  • amino acids with ⁇ -branched side chains e.g. threonine, valine, isoleucine
  • Amino acids with aromatic side chains for example, tyrosine, phenylalanine, tryptophan, histidine).
  • nucleic acids For nucleic acids, the term "substantial homology" means that two nucleic acids or their designated sequences have at least about 80% of the nucleotides in the best alignment and comparison (where appropriate insertion or deletion of nucleotides), usually At least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides are identical. Alternatively, when the segment hybridizes to the complement of the strand under selective hybridization conditions, substantial homology exists.
  • polypeptides for polypeptides, the term "substantial homology" means that two polypeptides or their designated sequences have at least about 80% amino acids, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the amino acids are identical.
  • the percent identity between two nucleotide sequences can use the GAP program in the GCG software package (available on http://www.gcg.com), use the NWSgapdna.CMP matrix and gap weights 40, 50, 60 , 70 or 80 and length weight 1, 2, 3, 4, 5 or 6.
  • the percent identity between two nucleotide or amino acid sequences can also use E. Meyers and W. Miller (CABIOS, 4:11-17 (1989)) included in the alignment program (ALIGN program, version 2.0) Algorithm, use PAM120 weight residue table, gap length penalty of 12 and gap penalty of 4 to determine.
  • percent identity between two amino acid sequences can be used in the gap program included in the GCG software package (available on http://www.gcg.com) by Needleman and Wunsch (J.Mol.Biol. (48):444-453 (1970)) algorithm, using Blossum 62 matrix or PAM250 matrix and slot weight 16, 14, 12, 10, 8, 6 or 4 and length weight 1, 2, 3, 4, 5 or 6 To determine.
  • nucleic acid and protein sequences described herein can be further used as "query sequences" to perform searches against public databases, for example to identify related sequences. These searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul et al. (1990) J. Mol. Biol. 215:403-10.
  • gapped BLAST can be used as described in Altschul et al. (1997) Nucleic Acids Res. 25(17): 3389-3402.
  • the preset parameters of each program such as XBLAST and NBLAST can be used. See www.ncbi.nlm.nih.gov.
  • nucleic acids may be present in intact cells, in cell lysates, or in a partially purified or substantially pure form.
  • purification is performed by standard techniques to remove other cellular components or other contaminants such as other cellular nucleic acids (such as other parts of chromosomes or proteins), the nucleic acid is "isolated” or “substantially pure”.
  • These standard techniques include Alkali/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and other methods well known in the art. See F. Ausubel et al. Edit. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987).
  • Nucleic acids can be mutated according to standard techniques to provide gene sequences. For coding sequences, these mutations can optionally affect the amino acid sequence. Specifically, it encompasses DNA sequences that are substantially homologous or derived from natural V sequences, D sequences, J sequences, constant sequences, switching sequences, and other sequences described herein.
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid linked to it.
  • plasmid refers to a circular double-stranded DNA loop into which other DNA segments can be ligated.
  • viral vector in which other DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in the host cell into which they have been introduced (for example, bacterial vectors with a bacterial origin of replication and episomal mammalian vectors). Other vectors (such as non-episomal mammalian vectors) can integrate into the host cell's genome when introduced into the host cell, and thereby replicate with the host genome.
  • vectors can direct the expression of genes operably linked to them. These vectors are referred to herein as “recombinant expression vectors” (or simply “expression vectors”).
  • expression vectors used in recombinant DNA technology are usually in the form of plasmids.
  • plasmid and “vector” are used interchangeably, because plasmid is the most commonly used form of vector.
  • the present invention also includes other forms of expression vectors that provide equivalent functions, such as viral vectors (such as replication-defective retroviruses, adenoviruses, and adeno-associated viruses).
  • the term "recombinant host cell” (or simply “host cell”) is intended to refer to a cell that contains nucleic acid that is not naturally present in the cell, and may be a cell that has been introduced into a recombinant expression vector. It should be understood that these terms are intended not only to refer to a specific individual cell, but also to refer to the progeny of this cell. Because mutations or environmental influences can cause certain changes in subsequent generations, the offspring may actually be different from the parent cell, but they are still included in the category of the term "host cell” as used herein.
  • the term "antigen" refers to any natural or synthetic immunogenic substance, such as a protein, peptide, or hapten.
  • the antigen may be LIF or a fragment thereof.
  • inhibition or “blocking” (for example, referring to inhibition/blocking of LIF binding or activity) are used interchangeably and encompass both partial and complete inhibition/blocking.
  • cancer refers to a large group of diseases characterized by uncontrolled abnormal cell growth in the body. Due to dysregulated cell division, malignant tumors or cells can be formed, which invade adjacent tissues and can metastasize to distant parts of the body via the lymphatic system or bloodstream.
  • treatment refers to the implementation of any type of intervention or process on the subject, or the administration of an active agent to the subject to reverse, alleviate, ameliorate, inhibit or slow down or prevent disease-related The progress, development, severity, or recurrence of symptoms, complications, conditions, or biochemical markers of the disease.
  • Prevention refers to administration to subjects without a disease to prevent the occurrence of the disease, or to minimize its impact if the disease occurs.
  • the term "effective dose or effective dose” is defined as an amount sufficient to achieve or at least partially achieve the desired effect.
  • the "therapeutically effective dose” or “therapeutically effective dose” of a drug or therapeutic agent is any amount that can promote the regression of the disease when used alone or in combination with another therapeutic agent, and the regression of the disease is reduced by the severity of the symptoms of the disease. , As evidenced by the increase in the frequency and duration of the asymptomatic period, or the prevention of damage or disability due to illness.
  • the "prophylactically effective dose” or “prophylactically effective dose” of a drug means that when the drug is administered alone or in combination with another therapeutic agent, it inhibits the development or recurrence of the disease when it is administered to a subject who is at risk of suffering from the disease or suffering from the recurrence of the disease. ⁇ The amount.
  • the ability of a therapeutic or preventive agent to promote regression of a disease or inhibit the development or recurrence of a disease can be assessed using a variety of methods known to those skilled in the art, such as evaluation in human subjects during clinical trials, and predictability in humans. Efficacy in the animal model system is evaluated, or by measuring the activity of the agent in an in vitro assay.
  • patient and “subject” refer to any human or non-human animal receiving prophylactic or therapeutic treatment.
  • methods and compositions described herein can be used to treat subjects suffering from cancer.
  • non-human animals includes all vertebrates, such as mammals and non-mammals, such as non-human primates, sheep, dogs, cows, chickens, amphibians, reptiles, and the like.
  • mice were immunized with recombinant human LIF protein (purchased from Sino Biological).
  • 50 ⁇ g of recombinant human LIF protein was injected into the abdominal cavity for booster immunization. Three days later, the animals were sacrificed and splenocytes were taken for fusion.
  • the SP2/0 cells of the mouse myeloma cells in the logarithmic growth phase were counted, and the spleen cell suspension of the immune mice was prepared at the same time, and the spleen cells and SP2/0 cells were fused with 50% PEG according to the conventional method.
  • the fused cells were added to a 96-well plate of trophoblast cells (six-week-old BABL/c mouse peritoneal macrophages), which had been selected and cultured in DMEM containing 1% HAT and 20% fetal bovine serum. When the clones grow to 1/3 of the bottom of the plate, collect the culture supernatant.
  • the ELISA plate was coated with recombinant human LIF protein, the anti-LIF antibody in the culture supernatant was detected by the indirect ELISA method, the clones secreting anti-human LIF antibody were screened, and the limiting dilution method was used for monoclonalization to obtain a stable secreting high-affinity antibody.
  • the human LIF monoclonal antibody cell line is labeled as 38E10E1C11. After determination, the full-length gene sequences encoding the light chain and heavy chain of the 38E10E1C11 antibody are shown in SEQ ID NO: 42 and SEQ ID NO: 44, respectively.
  • the full-length amino acid sequences of the light chain and heavy chain of the corresponding 38E10E1C11 antibody are shown in SEQ ID NO: 41 and SEQ ID NO: 43, respectively; the variable region gene sequences encoding the light chain and heavy chain of the 38E10E1C11 antibody are shown in SEQ ID NO, respectively.
  • the amino acid sequences of the variable regions of the light chain and heavy chain of the corresponding 38E10E1C11 antibody are shown in SEQ ID NO: 74 and SEQ ID NO: 75, as shown in SEQ ID NO: 76 and SEQ ID NO: 77; 38E10E1C11 is obtained according to the Kabat coding rules
  • the amino acid sequence of the LCDR1 of the antibody is shown in SEQ ID NO: 1, the amino acid sequence of LCDR2 is shown in SEQ ID NO: 2, the amino acid sequence of LCDR3 is shown in SEQ ID NO: 3, and the amino acid sequence of HCDR1 is shown in SEQ ID NO: As shown in 4, the amino acid sequence of HCDR2 is
  • thermofisher's CD hybridoma serum-free medium After obtaining a hybridoma cell line capable of stably secreting antibodies, use thermofisher's CD hybridoma serum-free medium to domesticate the cells, adapt them to serum-free suspension and shaking culture, and then use serum-free medium to express purified antibodies.
  • Invitrogen s TRIZOL Reagent was used to extract the total RNA of spleen cells, and the invitrogen cDNA reverse transcription kit was used for reverse transcription to synthesize cDNA.
  • the antibody gene was amplified using mouse light and heavy chain variable region degenerate primers and constructed into a phage display vector.
  • the phage antibody library was constructed, and the phage antibody library was panned using thermo's automatic magnetic bead sorting system.
  • the E. coli clones that could bind recombinant human LIF protein were selected by phage ELISA, and the antibody gene sequence was determined. Further through ELISA and cell viability identification, a strain of antibody labeled P36-033 was obtained.
  • the full-length gene sequences of the light chain and heavy chain of P36-033 are shown in SEQ ID NO: 55 and SEQ ID NO: 57, and the corresponding P36
  • the full-length amino acid sequences of the light chain and heavy chain of the -033 antibody are shown in SEQ ID NO: 54 and SEQ ID NO: 56 respectively;
  • the variable region gene sequences encoding the light chain and heavy chain of the P36-033 antibody are shown in SEQ ID, respectively
  • the amino acid sequences of the variable regions of the light chain and heavy chain of the corresponding P36-033 antibody are shown in NO: 72 and SEQ ID NO: 73, respectively, shown in SEQ ID NO: 82 and SEQ ID NO: 83; coded by Kabat
  • the amino acid sequence of LCDR1 of the P36-033 antibody is shown in SEQ ID NO: 66
  • the amino acid sequence of LCDR2 is shown in SEQ ID NO: 67
  • the amino acid sequence of LCDR3 is shown in SEQ ID NO: 68
  • the 5D8 antibody is an antibody that blocks the binding of LIF protein to its receptor GP130.
  • the present invention synthesizes multiple gene sequences to perform different combinations of light and heavy chains. Constructed full-length antibodies in the form of human IgG1, and finally found that one of the pairings has the best LIF protein binding activity, and can block the binding of recombinant human LIF protein to human GP130 protein. After cell activity verification, it can block the recombinant human LIF protein.
  • the present invention named it 5D8, which was used as a positive control antibody in subsequent experiments.
  • the full-length gene sequences encoding the heavy chain and light chain of the 5D8 antibody are as shown in SEQ ID NO: 63 and SEQ ID NO: 65, the corresponding 5D8 antibody heavy chain and light chain full-length amino acid sequences are shown in SEQ ID NO: 62 and SEQ ID NO: 64, respectively; encoding the 5D8 antibody heavy chain and light chain
  • the variable region gene sequences are shown in SEQ ID NO: 80 and SEQ ID NO: 81, respectively, and the amino acid sequences of the variable regions of the heavy chain and light chain of the corresponding 5D8 antibody are shown in SEQ ID NO: 78 and SEQ ID NO: 79 shown.
  • Use KinExA 4000 to determine the affinity of purified monoclonal antibody to recombinant human LIF protein.
  • the buffer composition is 1 ⁇ PBS, pH 7.4, 0.02% NaN3. Make sure that the beads are completely suspended in the solution. Spin the beads for 2 hours at room temperature. The beads naturally settled or centrifuged briefly at low speed, the supernatant was removed, and the beads were blocked with PBS containing 1% BSA for 1 hour. Prepare 15mL 300pM antigen solution and 15mL 240pM Ab2 (38E10E1C11) solution.
  • Example 6 P36-033, 38E10E1C11 mAb competes with LIFR for binding to human LIF protein
  • Example 7 P36-033, 38E10E1C11 mAb competes with GP130 for binding to human LIF protein
  • Human LIF, human IL-6, human OSM, and human CNTF (all purchased from Sino Biological) were coated with an ELISA plate at a concentration of 1 ⁇ g/mL, and different concentrations of LIF antibody 38E10E1C11, P36-033, 5D8 were added and incubated for 1 hour at room temperature After washing four times with PBST, add HRP-labeled goat anti-mouse Fab secondary antibody and incubate at room temperature for 1 hour. After washing four times with PBST, add TMB color developing solution to develop color at room temperature for 10 minutes, and read the absorbance value at 450nm with a microplate reader. Use Origin pro 9 software for analysis and mapping. The results are shown in Figure 7.
  • Example 9 38E10E1C11mAb can be used for western blot detection of human LIF protein
  • HCT116 cells were digested and centrifuged, the cells were resuspended, and a 12-well plate was plated at 1 mL, 5 ⁇ 10 5 cells/well. Incubate overnight at 37°C, 5% CO 2. The next day, discard the original medium, add cell culture medium containing 100ng/mL recombinant human LIF protein and different concentrations of anti-LIF antibody, set up control wells without recombinant human LIF protein and only recombinant human LIF protein without antibody. Incubate at 37°C for 30 min. Then remove the medium, add 100uL 1x lysate to each well of the 12-well plate, and lyse on ice for 30 minutes.
  • FIG. 10 shows that the 38E10E1C11 antibody of the present invention blocks the activation of STAT3 of pancreatic cancer cell KP4 by human LIF secreted by CT26-hLIF cells.
  • the constructed lentivirus containing the human LIF gene was infected with the mouse colon cancer cell line CT26, and the LIF protein expression was detected after 48 hours of infection.
  • the cell line was cloned by the limiting dilution method, and a medium with a final concentration of 1 ⁇ g/mL puromycin was added. Under pressure screening, the CT26 stable cell line CT26-hLIF with high expression of human LIF protein was finally obtained.
  • CT26-hLIF cells were cultured in 10% RPMI-1640 medium containing fetal bovine serum, cells in the logarithmic growth phase were collected, resuspended to PBS vaccinated BABL / c mice were subcutaneously 10 7 cells / mL.
  • the mice were divided into groups and injected with vehicle control and anti-human LIF antibody at a concentration of 15 mg/kg body weight. They were administered twice a week for 4 consecutive weeks, and the tumor volume was measured twice a week. Tumor growth curve, calculate tumor inhibition rate.
  • the results are shown in Figure 13. The results show that the monoclonal antibody 38E10E1C11 can inhibit the proliferation of CT26-hLIF cells in BABL/c mice.
  • the light and heavy chain antibody genes of the 38E10E1C11 and P36-033 monoclonal antibodies were constructed using homologous recombination technology into the eukaryotic expression vector PCDNA3.1+, and the recombinant antibodies were expressed using the thermo ExpiCHO expression system, and the antibodies were purified using Protein G affinity chromatography , Use the Endotoxin Removal Beads of Changzhou Tiandiren and the company to remove endotoxin from the purified antibody.
  • the specific experimental method refers to Example 10.2.
  • Figure 15 shows that the 38E10E1C11 antibody recombinantly expressed by CHO cells (labeled as 38E10E1C11R) can inhibit the phosphorylation of STAT3 in KP4 cells induced by human LIF protein.
  • the cell supernatant was taken for SDS-PAGE electrophoresis and Transfer the membrane, use 293T cell culture supernatant as a negative control, use 38E10E1C11 as the primary antibody, and HRP-labeled goat anti-mouse Fab as the secondary antibody for detection.
  • use the M1 cell proliferation experiment to detect the activity of the hybrid protein and verify the 38E10E1C11 pair of hybrid The neutralizing activity of the protein.
  • the monoclonal antibody 38E10E1C11 obtained from mouse immunization was humanized. Use standard CDR grafting methods for humanization.
  • the heavy chain region and light chain region were cloned from the 38E10E1C11 hybridoma using standard molecular cloning techniques, and sequenced using the Sanger method. Then a BLAST search is performed on the human heavy chain and light chain variable sequences, and 3 or 4 sequences from each sequence are selected as humanized acceptor frameworks.
  • the heavy chain and light chain CDR1, CDR2 and CDR3 of 38E10E1C11 were cloned into 3 different heavy chain acceptor frameworks (H1-H3) and 4 different light chain frameworks (L1-L4), and the HCDR2 (
  • the amino acid sequence before mutation is shown in SEQ ID NO: 45) point mutation (the amino acid sequence after mutation is shown in SEQ ID NO: 5), the heavy chain constant region selects human IgG1 subtype, and the light chain constant region selects human Kappa Chain
  • the expression vector containing the humanized antibody heavy chain and the humanized antibody light chain gene was co-transfected into 293S cells, and the heavy and light chain variable region gene sequence and variable region of the humanized anti-LIF antibody obtained above were
  • the amino acid sequence, full-length gene sequence, and full-length amino acid sequence are shown in Table 1, and then the expression levels, antigen binding ability and thermal stability of 12 different antibody combinations in 293S cells were tested.
  • the 38E10E1C11 chimeric antibody (Chimeric) was used as a positive control.
  • the 38E10E1C11 chimeric antibody was referred to as 38E chimeric antibody or 38E Chimeric (SEQ ID NO: 52 and SEQ ID NO: 50).
  • the culture medium was collected, and the expression level of IgG was quantified on Gator (similar to Octet), and corrected by ELISA.
  • the ELISA method was used to compare the antigen binding ability of different combinations (Table 2, Table 3).
  • the present invention selects the following five candidate antibodies for the next characterization: H1L1, H1L4, H2L4, H3L2, H3L4, and renews these five candidate antibodies.
  • the numbers are 38E HuH1L1 (SEQ ID NOs: 25 and 9), 38E HuH1L4 (SEQ ID NOs: 25 and 21), 38E HuH2L4 (SEQ ID NOs: 29 and 21), 38E HuH3L2 (SEQ ID NOs: 33 and 13), 38E HuH3L4 (SEQ ID NOs: 33 and 21), and then co-transfect the selected VH/VL plasmid into 293S cells, harvest the cell culture supernatant, and purify the antibody by protein A affinity chromatography.
  • the purified antibody was used for binding ELISA analysis to compare the specific binding ability of the humanized antibody and the 38E chimeric antibody.
  • the present invention also conducted some preliminary analyses to compare their thermal stability and non-specific binding. The results showed that the purified candidate antibody has very similar antigen binding properties to the 38E chimeric antibody ( Figure 17A, 17C). After being treated at 70°C for 5 minutes, the 5 humanized antibodies all showed similar binding capacity to the chimeric antibody ( Figure 17B, 17D).
  • LIF-negative HEK293 cell FACS was used as a preliminary test to assess the potential risk of non-specific binding of the antibody.
  • HEK293 cells were trypsinized, they were washed twice with 1% FBS-containing PBS, the cells were resuspended, the cell density was adjusted to 1.5-2 ⁇ 10 6 cells/mL, and the cells were added to a 96-well U-shaped plate. Adjust the concentration of the antibody to be tested to 20 ⁇ g/mL, and then carry out a 3-fold dilution for a total of 8 concentrations, and set a blank control and a negative control (Rituxan). Add the diluted antibody and blank control to the cells in the 96-well plate, and add 100 ⁇ L of antibody to each well.
  • the working concentration of CE-SDS analysis is 1 mg/mL, and the antibody sample is diluted to the specified concentration with loading buffer.
  • Preparation of reduced sample solution Take 95 ⁇ L of diluted sample solution, add 5 ⁇ L of 2-mercaptoethanol solution, 5 ⁇ L of internal control, and vortex to mix. Take 95 ⁇ L of blank control, add 5 ⁇ L of 2-mercaptoethanol solution, 5 ⁇ L of internal control, and vortex to mix well. It is reduced blank control. At 70°C, heat the metal bath for 15 minutes, cool to room temperature, and centrifuge at 6000 rpm for 1 minute.
  • Sample analysis Take 75 ⁇ L of sample into the test tube, put the test tube into the test cup, carefully insert the test cup into the sample tray, and run the test program.
  • the injection time of the reduced sample is 30 seconds
  • the injection time of the non-reduced sample is For 40 seconds
  • the capillary temperature is 20°C
  • the sample temperature is 20°C
  • the focusing voltage is 15KV
  • the focusing time is 40 minutes
  • the data is collected with a PDA detector at 214nm wavelength.
  • the CE results are shown in Table 4 and Table 5.
  • Example 18 Analysis of thermal stability by differential scanning fluorescence (DSF)/static light scattering (SLS)
  • the sample is submitted to the UNcle system (Unchained Labs) for analysis.
  • UNcle system Unchained Labs
  • DSF and SLS are monitored at a temperature of 1°C/min.
  • IgG has multiple domains, and each domain has its own melting temperature (T m ).
  • the CH2 domain usually has a Tm of about 70°C in PBS, and CH3 is more stable with a Tm of about 80°C.
  • Fabs have a large range of Tm due to their large sequence variation, which is about 50-85°C. Therefore, the Tm value measured by various analytical techniques is usually the "apparent" transition temperature, rather than the true Tm value of each domain. Obviously, even this DSF analysis can produce more than one Tm value, and only Tm1 is used to evaluate the thermal stability of therapeutic antibodies.
  • Tagg is the temperature at which SLS starts to detect aggregation.
  • Tagg266 measures SLS at 266nm, it is more sensitive and more suitable for detecting smaller aggregated particles.
  • Tagg473 measures SLS at 473nm, which is more suitable for detecting larger particles.
  • the three humanized candidate antibodies all have a higher melting temperature (Tm1) and a lower risk of aggregation than the 38E chimeric antibody
  • Example 19 uses dynamic light scattering (DLS) to analyze the aggregation tendency of antibodies:
  • DLS Dynamic light scattering
  • Mode diameter refers to the protein particle diameter
  • mass percentage refers to the percentage of each particle size fraction.
  • PDI is the polydispersity index. The higher the index, the stronger the polydispersity of the sample. If the PDI is not greater than 0.25, the sample can be regarded as monodisperse. As shown in Table 7, all 4 antibody samples have a main “peak” (mass fraction exceeds 99%).
  • the PDI of 38E HuH3L4 is better than that of chimeric antibody.
  • 38E HuH3L2 is similar to chimeric antibody.
  • the PDI of 38E HuH1L1 is higher than that of chimeric antibody. Poor combination of antibodies.
  • the Gator was used to determine the affinity of anti-LIF antibody and human LIF protein.
  • the anti-human LIF antibody was diluted with PBS to 5ug/mL, and then added to the second row of AF wells (200 ⁇ L per well) of the 96-well plate; the human LIF protein concentration was diluted with PBS to 100, 50, 25, 12.5, and 12.5, respectively. 6.25 ⁇ g/mL were added to the AE wells in the fourth column of the 96-well plate (100 ⁇ L per well), and PBS was added to the F well as a blank control; the AF wells in the first column and the third column were added PBS (200 ⁇ L per well). Put the 96-well plate into the instrument and use the anti-human Fc biosensor for detection. The experimental results are shown in Table 8. The results show that the affinity of the three humanized antibodies is close to that of the chimeric antibody.
  • variable regions of the light and heavy chains of the humanized antibodies 38E HuH3L2 and 38E HuH3L4 were linked to the mouse antibody constant region (heavy chain constant region is mouse IgG1, light chain constant region is kappa chain), respectively, and cloned into the PCDNA.
  • 38E HuH3L2-m the full-length gene sequences encoding the heavy chain and light chain of the 38E HuH3L2-m antibody are shown in SEQ ID NO: 36 and SEQ ID NO: 38, respectively, and the corresponding 38E HuH3L2-m antibody heavy
  • the full-length amino acid sequences of the chain and the light chain are shown in SEQ ID NO: 35 and SEQ ID NO: 37, respectively) and 38E HuH3L4-m (encoding the full-length gene sequences of the heavy chain and light chain of the 38E HuH3L4-m antibody, respectively, are shown in SEQ ID NO: 35 and SEQ ID NO: 37).
  • the recombinant human LIF protein was coated on the ELISA plate at a concentration of 1 ⁇ g/mL, and 0.6125 ⁇ g/mL recombinant human LIFR (expressed in fusion with human Fc, purchased from ACRO, catalog number: LIR-H4252) protein 50 ⁇ L/well was added at the same time
  • Add different concentrations of LIF antibodies 38E HuH3L2-m (SEQ ID NOs: 35 and 37), 38E HuH3L4-m (SEQ ID NOs: 35 and 39), 38E10E1C11 (SEQ ID NOs: 41 and 43), P36-033 (SEQ ID NOs: 54and 56) 100 ⁇ L/well, with anti-CD3 antibody as a negative control (purchased from BioLegend, catalog number: 317326), incubate at room temperature for 2 hours, wash with PBST four times, add HRP-labeled goat anti-human Fc antibody, and incubate at room temperature for 1 hour After washing four times with PBST,
  • the results are shown in Figure 19.
  • the results show that 38E10E1C11, 38E HuH3L2-m, and 38E HuH3L4-m can inhibit the combination of recombinant human LIF and human LIFR, with IC 50 of 0.074 ⁇ g/ml, 0.145 ⁇ g/ml, and 0.103 ⁇ g/ml, respectively.
  • P36-033 has a weak inhibitory effect.
  • the negative control anti-CD3 antibody cannot inhibit the binding of recombinant human LIF to human LIFR.
  • Example 23 Humanized anti-LIF antibody does not compete with GP130 for binding to human LIF protein
  • Recombinant human LIF protein was coated on the ELISA plate at a concentration of 1 ⁇ g/mL, and 12 ⁇ g/mL recombinant human GP130 (expressed in fusion with human Fc, purchased from Yiqiao Shenzhou, catalog number: 10974-H03H) protein 50 ⁇ L/well, At the same time, add different concentrations of LIF antibody 38E HuH3L2-m ((SEQ ID NOs: 35 and 37), 38E HuH3L4-m (SEQ ID NOs: 35 and 39), P36-033 (SEQ ID NOs: 56 and 54) 100 ⁇ L/well, with Anti-CD28 antibody was used as a negative control (purchased from BioLegend, catalog number: 302914), incubated for 2 hours at room temperature, washed with PBST four times, added HRP-labeled goat anti-human Fc antibody, incubated for 1 hour at room temperature, washed with PBST four times, Add TMB color developing solution to develop color at room
  • Human LIF, human IL-6, human OSM, human CNTF (the four proteins were purchased from Yiqiao Shenzhou, the product numbers are: 14890-HNAH; 10395-HNAE; 10452-HNAH; 11841-H07E) according to 1 ⁇ g/mL
  • Concentrate coated ELISA plate add different concentrations of LIF antibody 38E10E1C11, 38E huH3L2-m, 38E huH3L4-m, incubate for 1 hour at room temperature, wash four times with PBST, add HRP-labeled goat anti-mouse Fab secondary antibody, incubate at room temperature 1 After washing four times with PBST, add TMB color developing solution to develop color at room temperature for 10 minutes.
  • the absorbance value at 450nm is read by the microplate reader, and the data is analyzed and graphed by Origin pro 9 software.
  • the results are shown in Figure 21.
  • the results show that the 38E10E1C11, 38E huH3L2-m, and 38E huH3L4-m antibodies only bind to human LIF protein but not to human IL-6, OSM, CNTF.
  • the present invention found that the 38E10E1C11 antibody recognizes the linear epitope of the LIF protein. Therefore, the present invention first verifies whether the 38E humanized antibody recognizes the linear epitope of the LIF protein. Take the supernatant of 293T cells transfected with human LIF full-length gene sequence, Mut3 and Mut4 protein sequences after 3 days of culture, the negative control is 293T cell culture supernatant, add 5xSDS-PAGE loading buffer, boil for 10 minutes, and then take 10 ⁇ L The sample is subjected to SDS-PAGE electrophoresis, and then the electrophoresis band is transferred to the PVDF membrane for western blot detection.
  • the primary antibody used for detection is 38E huH3L2 or 38E huH3L4 antibody, the antibody concentration is 1 ⁇ g/mL, incubate at room temperature for 2 hours, PBST buffer After washing three times, add HRP-labeled goat anti-human Fc secondary antibody diluted 1:3000, incubate for 2 hours at room temperature, wash three times with PBST buffer, add enhanced chemiluminescence solution (Perice, catalog number: 34079) and incubate with Amersham Imager 600 ultra-sensitive multi-function imager for detection and photographing.
  • the results are shown in Figure 22, Panel A. The results show that both humanized antibodies can recognize the denatured human LIF protein and Mut3 protein but not the Mut4 protein.
  • the M1 cell proliferation experiment also obtained the same result, and the result is shown in sub-panel B in Figure 22. Therefore, it is determined that the epitope sequence recognized by the 38E huH3L2 and 38E huH3L4 antibodies is TYGPDTSGKDVFQKK (SEQ ID NO: 61).
  • Example 27 M1 cell proliferation experiment to detect the activity of humanized anti-LIF antibody
  • RPMI1640 medium was washed twice, 96-well plates were seeded at a density of 2.5x10 5 cells/mL, 80 ⁇ L cells were seeded per well, and the medium containing 4ng/mL recombinant human LIF protein and different concentrations of anti-LIF antibodies was added. Make the final volume of each well 160 ⁇ L, and set a control well without LIF, incubate in a 37°C incubator for 72 hours, add CCK-8 to detect cell proliferation. The results are shown in Figure 24.
  • LIF binds to GP130 and LIFR, and the humanized LIF antibody blocks the binding of LIF to LIFR, but cannot block the binding of LIF to GP130.
  • ADCC buffer RPMI-1640+1% FBS
  • ADCC buffer RPMI-1640+1% FBS
  • ADCC buffer containing LIF protein, three-fold dilution, eight gradients, and use pancreatin (Cat# 25200072, GIBCO) digest DLD-1 cells, after terminating the reaction, blow off the cells and collect them in a centrifuge tube, centrifuge at 1500 rpm for 3 min, discard the supernatant, resuspend the cells in ADCC buffer and count them, adjust the cell concentration, and use them for later use; resuscitate PBMC cells ( Cat#SLB-HP010B, Shanghai Sai Li Biological Technology Co., Ltd.), add 10 mL ADCC buffer, centrifuge at 2000 rpm for 10 min, discard the supernatant, resuspend it in ADCC buffer for counting, adjust the cell concentration
  • the 38E huH3L4 antibody has no ADCC activity.
  • Leukemia inhibitory factor promotes tumor growth and metastasis inhuman osteosarcoma via activating STAT3.APMIS 2015; 123:837–46.
  • Leukemia inhibitory factor protects cholangiocarcinoma cells from drug induced apoptosis via a PI3K/AKT-dependent: Mcl-1 activation.Onco target.2015; 6 26052–64.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Plant Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

本发明提供对LIF具有特异性的结合分子及其用途,具体而言,本发明提供结合LIF且抑制LIF活性的分离的抗体或其抗原结合部分。本发明亦提供本发明分离的抗体或其抗原结合部分在治疗疾病中的用途。

Description

对LIF具有特异性的结合分子及其用途 技术领域
本发明涉及与LIF特异性结合的分离的抗体或其抗原结合部分,以及本发明的分离的抗体或其抗原结合部分的用途,以及使用本发明的分离的抗体或其抗原结合部分的治疗方法。
发明背景
白血病抑制因子(LIF,Leukemia Inhibitory Factor)是IL-6类细胞因子的一员,它具有多种生物活性包括刺激或抑制细胞的增殖、分化和存活[1]。人LIF蛋白含有202个氨基酸,它在细胞膜表面的受体有两个,GP130和LIFR,LIF蛋白通过与这两个受体结合,导致这两个受体形成异源二聚体,从而激活下游信号通路,例如MAPK信号通路和JAK/STAT信号通路[2]。高表达和高血清浓度的LIF蛋白已显示与多种肿瘤的不良预后相关[3,4]。LIF是肿瘤干细胞的重要调节因子,在干细胞维持、自我更新和多能性等方面发挥重要作用,并与化疗耐药有关[5,6]。此外,LIF还能促进肿瘤的生长和转移[7]。最近的证据表明在肿瘤中LIF通过自分泌和旁分泌的方式上调JAK–STAT3信号通路,从而发挥促进肿瘤生长和抑制免疫应答的作用[8,9,10]。因此,LIF是极具潜力的治疗靶点,但是目前针对LIF靶点所开发的治疗方法并不乐观。例如已有多篇文献报道,利用RNA干扰降低LIF蛋白的表达,能够抑制肿瘤生长[11,12],但是RNA干扰技术具有靶向性差,半衰期短,过膜性差等弱点成药困难;EC359为针对LIFR的小分子抑制剂,不仅能够抑制LIFR与LIF的结合还能同时抑制OSM,CTF1,CNTF与LIFR的结合[13],这些额外的抑制会不会导致额外的毒性还是未知,而特异性针对LIF蛋白的小分子抑制剂还未见报道;目前只有一款针对LIF蛋白的抗体处于临床开发阶段,相关的安全性和有效性数据还未公开。
因此,仍然需要更多研究来开发针对LIF靶点的药物及组合治疗方法。
发明内容
本发明提供特异性结合LIF的分离的抗体或抗原结合部分及其在治疗疾病中的用途。
在一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,所述分离的抗体或其抗原结合部分结合在人LIF蛋白的氨基酸序列TYGPDTSGKDVFQKK(SEQ ID NO:61)所示的表位或在不同哺乳动物物种的相应氨基酸序列的表位。
在再一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
(a)LCDR1,该LCDR1包含选自以下所组成的组:SEQ ID NO:1或SEQ ID NO:66及其保守修饰形式的氨基酸序列;
(b)LCDR2,该LCDR2包含选自以下所组成的组:SEQ ID NO:2或SEQ ID NO:67及其保守修饰形式的氨基酸序列;
(c)LCDR3,该LCDR3包含选自以下所组成的组:SEQ ID NO:3或SEQ ID NO:68及其保守修饰形式的氨基酸序列;
(d)HCDR1,该HCDR1包含选自以下所组成的组:SEQ ID NO:4或SEQ ID NO:69及其保守修饰形式的氨基酸序列;
(e)HCDR2,该HCDR2包含选自以下所组成的组:SEQ ID NO:5、SEQ ID NO:45或SEQ ID NO:70及其保守修饰形式的氨基酸序列;及
(f)HCDR3,该HCDR3包含选自以下所组成的组:SEQ ID NO:6或SEQ ID NO:71及其保守修饰形式的氨基酸序列。
任选地,所述LCDR1、LCDR2、LCDR3、HCDR1、HCDR2或HCDR3具有17个或更少的氨基酸的添加、取代、删除和/或***。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(a)LCDR1,该LCDR1包含选自以下所组成的组:SEQ ID NO:1及其保守修饰形式的氨基酸序列;
(b)LCDR2,该LCDR2包含选自以下所组成的组:SEQ ID NO:2及其保守修饰形式的氨基酸序列;
(c)LCDR3,该LCDR3包含选自以下所组成的组:SEQ ID NO:3及其保守修饰形式的氨基酸序列;
(d)HCDR1,该HCDR1包含选自以下所组成的组:SEQ ID NO:4及其保守修饰形式的氨基酸序列;
(e)HCDR2,该HCDR2包含选自以下所组成的组:SEQ ID NO:5或SEQ ID NO:45及其保守修饰形式的氨基酸序列;及
(f)HCDR3,该HCDR3包含选自以下所组成的组:SEQ ID NO:6及其保守修饰形式的氨基酸序列。
任选地,所述LCDR1、LCDR2、LCDR3、HCDR1、HCDR2或HCDR3具有17个或更少的氨基酸的添加、取代、删除和/或***。
在某些实施方案中,任选地,所述LCDR1、LCDR2、LCDR3、HCDR1、HCDR2或HCDR3具有9个或更少的氨基酸的添加、取代、删除、和/或***。
在某些实施方案中,任选地,所述所述LCDR1、LCDR2、LCDR3、HCDR1、HCDR2或HCDR3具有5个或更少的氨基酸的添加、取代、删除、和/或***。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
1)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3;
2)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3;或者
3)(a)包含SEQ ID NO:66的LCDR1,(b)包含SEQ ID NO:67的LCDR2,(c)包含SEQ ID NO:68的LCDR3,(d)包含SEQ ID NO:69的HCDR1,(e)包含SEQ ID NO:70的HCDR2,以及(f)包含SEQ ID NO:71的HCDR3。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
1)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3;或者
2)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(a)包含SEQ ID NO:66的LCDR1,(b)包含SEQ ID NO:67的LCDR2,(c)包含SEQ ID NO:68的LCDR3,(d)包含SEQ ID NO:69的HCDR1,(e)包含SEQ ID NO:70的HCDR2,以及(f)包含SEQ ID NO:71的HCDR3。
在某些实施方案中,所述分离的抗体或其抗原结合片段是鼠源抗体或其抗原结合片段、嵌合抗体或其抗原结合片段、全人抗体或其抗原结合片段,或人源化抗体或其抗原结合片段。
在某些实施方案中,所述分离的抗体为人源化抗体,所述人源化抗体包含源自人抗体的框架区或其框架区变体
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(i)轻链可变区(VL),该轻链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:7、11、15、19、46、74或82及其保守修饰形式的氨基酸序列;以及
(ii)重链可变区(VH),该重链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:23、27、31、48、75或83及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(i)轻链可变区(VL),该轻链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:7、11、15、19或46及其保守修饰形式的氨基酸序列;以及
(ii)重链可变区(VH),该重链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:23、27、31或48及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
(i)轻链可变区(VL),该轻链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:7、11、15或19及其保守修饰形式的氨基酸序列;以及
(ii)重链可变区(VH),该重链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:23、27或31及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的轻链可变区包含与选自(i)中的轻链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列;所述重链可变区包含与选自(ii)中的重链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
1)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
2)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
3)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
4)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
5)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
6)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
7)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
8)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
9)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
10)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
11)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
12)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
13)轻链可变区(VL),其包含与SEQ ID NO:46的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:48的氨基酸序列至少85%同源性的氨基酸序列;
14)轻链可变区(VL),其包含与SEQ ID NO:74的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:75的氨基酸序列至少85%同源性的氨基酸序列;或者
15)轻链可变区(VL),其包含与SEQ ID NO:82的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:83的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
1)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
2)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
3)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
4)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
5)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
6)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
7)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
8)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
9)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
10)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
11)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;或者
12)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:46的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:48的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:74的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:75的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链可变区(VL),其包含与SEQ ID NO:82的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:83的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述轻链可变区及重链可变区分别包含与选自1)-15)中轻链可变区及重链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
在一方面,本发明提供一种分离的抗体或其抗原结合片段,其包含选自如下(a)至(c)任一项所述的重链可变区和轻链可变区组合:
(a)轻链可变区(VL),该轻链可变区包含与SEQ ID NOs:7、11、15、19、46或82任一序列中具有相同序列的LCDR1、LCDR2以及LCDR3;以及
(ii)重链可变区(VH),该重链可变区包含与SEQ ID NOs:23、27、31、48或83任一序列中具有相同HCDR1、HCDR2以及HCDR3。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含轻链和重链,其中:
(I)所述轻链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:9、13、17、21、37、39、50或者54及其保守修饰形式的氨基酸序列;以及
(II)所述重链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:25、29、33、35、52或者56及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含轻链和重链,其中:
(I)所述轻链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:9、13、17、21、37、39或者50及其保守修饰形式的氨基酸序列;以及
(II)所述重链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:25、29、33、35或者52及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含轻链和重链,其中:
(I)所述轻链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:9、13、17或21及其保守修饰形式的氨基酸序列;以及
(II)所述重链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:25、29或33及其保守修饰形式的氨基酸序列。
在某些实施方案中,所述的轻链包含与选自(I)中的轻链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列;所述的重链包含与选自(II)的重链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
1)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
2)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
3)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
4)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
5)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
6)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
7)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
8)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
9)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
10)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
11)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
12)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
13)轻链,其包含与SEQ ID NO:37的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列;
14)轻链,其包含与SEQ ID NO:39的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列;
15)轻链,其包含与SEQ ID NO:50的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:52的氨基酸序列至少85%同源性的氨基酸序列;或者
16)轻链,其包含与SEQ ID NO:54的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:56的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述轻链及重链分别包含与选自1)-16)中轻链及重链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:37的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:39的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:50的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:52的氨基酸序列至少85%同源性的氨基酸序列。
在某些实施方案中,所述的分离的抗体或其抗原结合部分包含:
轻链,其包含与SEQ ID NO:54的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:56的氨基酸序列至少85%同源性的氨基酸序列。
在另一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
在又一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
在又一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:7所示的轻链可变区(VL),以及含有SEQ ID NO:23所示的重链可变区(VH)。
在再一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:11所示的轻链可变区(VL),以及含有SEQ ID NO:31所示的重链可变区(VH)。
在另一方面,本发明提供一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:19所示的轻链可变区(VL),以及含有SEQ ID NO:31所示的重链可变区(VH)。
在某些实施方案中,所述分离的抗体是单克隆抗体、嵌合抗体、人源化抗体、人改造抗体、人抗体、双特异性抗体、Fv、单链抗体(scFv)、Fab、Fab’、Fab’-SH或者F(ab’) 2
在某些实施方案中,所述的分离的抗体是IgG。
在某些实施方案中,所述分离的抗体是IgG1、IgG2或IgG4。
在某些实施方案中,所述的分离的抗体或其抗原结合部分为LIF(leukemia inhibitory factor)拮抗剂。
在某些实施方案中,所述的分离的抗体或其抗原结合部分能够抑制LIF表达和/或阻断LIF活性。
在某些实施方案中,所述的分离的抗体或其抗原结合部分能够竞争或交叉竞争用于结合LIF。
在再一方面,本发明提供一种核苷酸组合物,其包含编码本发明所述的分离的抗体或其抗原结合部分的核苷酸分子。在一些实施方式中,所述的核苷酸分子为DNA或RNA。在一些实施方式中,所述的核苷酸分子为DNA。
在某些实施方案中,所述的核苷酸组合物包含:
(i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15、19、46或82所示的轻链可变区(VL)的DNA;以及
(ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27、31、48或83所述的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
(i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15、19或46所示的轻链可变区(VL)的DNA;以及
(ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27、31或48所述的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
(i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15或19所示的轻链可变区(VL)的DNA;以及
(ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27或31所述的重链可变区(VH)的DNA。
在某些实施方案中,第一核酸分子包含与选自(i)中第一核酸分子至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA;第二核酸分子包含与选自(ii)中的第二核酸分子至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
1)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
2)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
3)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
4)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23的重链可变区(VH)的DNA;
5)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
6)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码SEQ ID NO:31所示的重链可变区(VH)的DNA;
7)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
8)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列SEQ ID NO:27所示的重链可变区(VH)的DNA;
9)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
10)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
11)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
12)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
13)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:46所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:48所示的重链可变区(VH)的DNA;
14)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:74所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:75所示的重链可变区(VH)的DNA;或者
15)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:82所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:83所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
1)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
2)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
3)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
4)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23的重链可变区(VH)的DNA;
5)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
6)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码SEQ ID NO:31所示的重链可变区(VH)的DNA;
7)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
8)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列SEQ ID NO:27所示的重链可变区(VH)的DNA;
9)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
10)第一核酸序列,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸序列,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
11)第一核酸序列,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸序列,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;或者
12)第一核酸序列,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸序列,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码SEQ ID NO:31所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列SEQ ID NO:27所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:46所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:48所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:74所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:75所示的重链可变区(VH)的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:82所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:83所示的重链可变区(VH)的DNA。
在某些实施方案中,第一核酸序列分别包含与选自1)-15)中第一核酸序列或第二核酸序列至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA序列。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA如SEQ ID NO:8所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA如SEQ ID NO:12所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA如SEQ ID NO:16所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA如SEQ ID NO:20所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:46所示的轻链可变区(VL)的DNA如SEQ ID NO:47所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:74所示的轻链可变区(VL)的DNA如SEQ ID NO:76所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:82所示的轻链可变区(VL)的DNA如SEQ ID NO:72所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA如SEQ ID NO:24所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA如SEQ ID NO:28所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA如SEQ ID NO:32所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:48所示的重链可变区(VH)的DNA如SEQ ID NO:49所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:75所示的重链可变区(VH)的DNA如SEQ ID NO:77所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:83所示的重链可变区(VH)的DNA如SEQ ID NO:73所示。
在某些实施方案中,所述的核苷酸组合物包括:
(I)第一核酸序列,其含有编码氨基酸序列如SEQ ID NOs:9、13、17、21、37、39、50或54所示的轻链的DNA;以及
(II)第二核酸序列,其含有编码氨基酸序列如SEQ ID NOs:25、29、33、35、52或56所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包括:
(I)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:9、13、17、21、37、39或50所示的轻链的DNA;以及
(II)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:25、29、33、35或52所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包括:
(I)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:9、13、17或21所示的轻链的DNA;以及
(II)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:25、29或33所示的重链的DNA。
在某些实施方案中,第一核酸分子包含与选自(I)中的第一核酸分子至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA;第二核酸分子包含与选自(II)中的第二核酸分子至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
1)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
2)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
3)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
4)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
5)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
6)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
7)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
8)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
9)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
10)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
11)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29的重链的DNA;
12)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
13)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:37所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA;
14)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:39所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA;
15)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:50所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:52所示的重链的DNA;或者
16)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:54所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:56所示的重链的DNA。
在某些实施方案中,所述第一核酸分子以及第二核酸分子包含与选自1)-16)中第一核酸分子或第二核酸分子至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:37所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:39所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:50所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:52所示的重链的DNA。
在某些实施方案中,所述的核苷酸组合物包含:
第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:54所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:56所示的重链的DNA。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA如SEQ ID NO:10所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA如SEQ ID NO:14所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA如SEQ ID NO:18所示。
在某些实施方案中,编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA如SEQ ID NO:22所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:37所示的轻链的DNA如SEQ ID NO:38所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:39所示的轻链的DNA如SEQ ID NO:40所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:50所示的轻链的DNA如SEQ ID NO:51所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:54所示的轻链的DNA如SEQ ID NO:55所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:25所示的重链的DNA如SEQ ID NO:26所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:29所示的重链的DNA如SEQ ID NO:30所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:33所示的重链的DNA如SEQ ID NO:34所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:35所示的重链的DNA如SEQ ID NO:36所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:52所示的重链的DNA如SEQ ID NO:53所示。
在某些实施方式中,编码氨基酸序列如SEQ ID NO:56所示的重链的DNA如SEQ ID NO:57所示。
再一方面,本发明提供一种载体,其含有如本发明所述的核苷酸组合物。
在某些实施方式中,所述的载体为真核表达载体、原核表达载体或病毒载体。
再一方面,本发明提供一种宿主细胞,其含有本发明所述的载体。
在一些实施方式中,含所述载体的宿主细胞是经载体转化获得的。
在一些实施方式中,所述宿主细胞为细菌、酵母或者哺乳动物细胞。
在一些实施方式中,所述宿主细胞为大肠杆菌、毕赤酵母、中国仓鼠卵巢细胞或人胚肾293细胞。
再一方面,本发明提供一种用于制备本发明所述的分离的抗体或其抗原结合部分的方法,所述方法包括在本发明所述的宿主细胞中表达所述抗体或其抗原结合片段,并分离所述抗体或其抗原结合片段。
在又一方面,本发明提供一种药物组合物,其包括治疗有效治疗剂量的上述的分离的抗体或其抗原结合部分,以及药学上可接受的赋形剂。
在再一方面,本发明提供一种用于测量生物样品中LIF的试剂,其包含上述的分离的抗体或其抗原结合部分。
在某些实施方案中,生物样本为血液、血清、尿、活检材料、肿瘤或怀疑含有异常LIF水平的任意组织。
在另一方面,本发明提供一种抑制LIF表达和/或阻断LIF活性的方法,包括向有需要的病人给予有效治疗剂量的上述的分离的抗体或其抗原结合部分,和/或上述的药物组合物。
在又一方面,本发明提供上述分离的抗体或其抗原结合部分,和/或上述的药物组合物在治疗药物中的用途,所述药物用于抑制LIF表达和/或阻断LIF活性。
在再一方面,本发明提供用于抑制LIF表达和/或阻断LIF活性的上述的分离的抗体或其抗原结合部分,和/或上述的药物组合物。
在另一方面,本发明提供一种治疗与LIF相关的疾病或病症的方法,所述方法包括向有需要的病人给予有效治疗剂量的上述的分离的抗体或其抗原结合部分,和/或上述的药物组合物。在某些实施方式中,与LIF相关的疾病为肿瘤。在一些实施方式中,所述肿瘤为实体瘤。在一些实施方式,所述的实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
在又一方面,本发明提供上述分离的抗体或其抗原结合部分,和/或上述的药物组合物在治疗与LIF相关的疾病或病症的药物中的用途。在某些实施方式中,与LIF相关的疾病为肿瘤。在一些实施方式中,所述肿瘤为实体瘤。在一些实施方式,所述的实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
在再一方面,本发明提供用于治疗与LIF相关的疾病或病症的上述的分离的抗体或其抗原结合部分,和/或上述的药物组合物。在某些实施方式中,与LIF相关的疾病为肿瘤。在一些实施方式中,所述肿瘤是实体瘤。在一些实施方式中,所述实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
与LIF相关的疾病或病症是指阻断LIF与LIRR和/或GP130能够治疗、减轻、缓解和/或稳定该疾病或病症。
在另一方面,本发明提供一种检测生物样品中LIF的方法,包括:(i)获得受试者的组织或液体样品,(ii)暴露所述组织或液体样品到上述的分离的抗体或其抗原结合部分或上述的试剂;以及(iii)检测从(ii)中获得的所述组织或液体样品结合的LIF与对照样品结合的LIF进行对比,其中与对照进行对比的LIF结合量的增加显示LIF的生成、表达或激活的异常水平。
在某些实施方案中,其中所述组织或液体样品包括血液、血清、尿、活检材料、肿瘤或怀疑含有异常LIF水平的任意组织。
表I 本发明抗体序列说明表
Figure PCTCN2020118247-appb-000001
Figure PCTCN2020118247-appb-000002
Figure PCTCN2020118247-appb-000003
Figure PCTCN2020118247-appb-000004
Figure PCTCN2020118247-appb-000005
Figure PCTCN2020118247-appb-000006
Figure PCTCN2020118247-appb-000007
Figure PCTCN2020118247-appb-000008
Figure PCTCN2020118247-appb-000009
Figure PCTCN2020118247-appb-000010
Figure PCTCN2020118247-appb-000011
Figure PCTCN2020118247-appb-000012
Figure PCTCN2020118247-appb-000013
Figure PCTCN2020118247-appb-000014
Figure PCTCN2020118247-appb-000015
Figure PCTCN2020118247-appb-000016
Figure PCTCN2020118247-appb-000017
Figure PCTCN2020118247-appb-000018
Figure PCTCN2020118247-appb-000019
Figure PCTCN2020118247-appb-000020
Figure PCTCN2020118247-appb-000021
Figure PCTCN2020118247-appb-000022
Figure PCTCN2020118247-appb-000023
Figure PCTCN2020118247-appb-000024
Figure PCTCN2020118247-appb-000025
Figure PCTCN2020118247-appb-000026
Figure PCTCN2020118247-appb-000027
Figure PCTCN2020118247-appb-000028
说明:CDR和框架区所有氨基酸编号均根据Kabat***的EU索引(Kabat,E.A.等人(1991)Sequences of Proteins of Immunological Interest,第5版,U.S.Department of Health and Human Services,NIH公布第91-3242号)注释。所有序列均不包含信号肽。aa:amino acid,nt:nucleotide.
附图说明
图1显示本发明LIF抗体与人LIF蛋白结合能力;
图2显示本发明LIF抗体与小鼠LIF蛋白结合能力;
图3显示本发明LIF抗体与食蟹猴LIF蛋白结合能力;
图4显示本发明38E10E1C11抗体与抗原结合的亲和力;
图5显示本发明38E10E1C11抗体与LIFR竞争结合人LIF蛋白的能力;
图6显示本发明P36-333抗体与受体GP130竞争结合人LIF蛋白的能力;
图7显示本发明38E10E1C11抗体和P36-333抗体与IL-6家族蛋白的交叉反应;
图8显示本发明显示本发明38E10E1C11R抗体识别变性的人LIF蛋白。
图9中的子图A显示本发明38E10E1C11抗体抑制人LIF蛋白诱导的结肠癌细胞(HCT116)的磷酸化;图9中的子图B显示本发明38E10E1C11抗体抑制人LIF蛋白诱导的胰腺癌细胞(KP4)STAT3的磷酸化;
图10显示本发明38E10E1C11抗体阻断CT26-hLIF细胞分泌的人LIF对于胰腺癌细胞KP4的STAT3的激活;
图11显示本发明P36-333抗体抑制人LIF蛋白诱导的结肠癌细胞(HCT116)STAT3的磷酸化;
图12显示本发明38E1E1C11和P36-033抗体逆转LIF对于M1细胞的增殖抑制;
图13显示本发明38E10E1C11抗体抑制CT26-hLIF细胞在BABL/c小鼠体内的生长;
图14显示三种人胰腺癌细胞系对人LIF蛋白刺激的敏感性;
图15显示本发明38E10E1C11R抗体抑制LIF蛋白刺激导致的KP4细胞STAT3的磷酸化;
图16中的子图A显示本发明38E10E1C11抗体识别全长人LIF蛋白以及杂合LIF蛋白的实验结果,图16中的子图B显示本发明38E10E1C11抗体能够逆转全长人LIF蛋白以及杂合LIF蛋白对M1细胞增殖抑制的实验结果;
图17A-图17B显示本发明人源化抗LIF抗体与抗原结合特性的实验结果;
图18显示本发明人源化抗LIF抗体的非特异性结合亲和力的实验结果;
图19显示本发明人源化抗LIF抗体与LIFR竞争结合人LIF蛋白的实验结果;
图20显示本发明人源化抗LIF抗体与GP130竞争结合人LIF蛋白的实验结果;
图21显示本发明人源化抗LIF抗体识别抗原特异性的实验结果;
图22中的子图A显示本发明显示本发明人源化抗LIF抗体识别全长人LIF蛋白以及杂合LIF蛋白的实验结果,图22中的子图B显示本发明人源化抗LIF抗体阻断全长LIF蛋白和杂合蛋白对M1细胞增殖抑制的实验结果;
图23显示本发明人源化抗LIF抗体抑制LIF蛋白诱导的STAT3的磷酸化的实验结果;
图24显示本发明人源化抗LIF抗体能够逆转人LIF蛋白对M1细胞增殖抑制的实验结果。
图25显示HTFR方法检测的人源化抗LIF抗体抑制LIF蛋白诱导的STAT3的磷酸化水平和总STAT3水平。
图26显示人源化抗LIF抗体的ADCC作用。
详细说明
定义
为更容易地理解本说明书,首先定义某些术语。其他定义陈述于说明书通篇中。
本文所用的术语“LIF(Leukemia Inhibitory Factor)”指白血病抑制因子。LIF的氨基酸序列为公开可用的(Ref Seq NM_001257135)。在一些实施方案中,LIF可以是人、小鼠(Ref Seq NM_001039537.2)或食蟹猴LIF(XM_015457518.1)。如本文别处所描述,LIF可以是重组的和/或糖基化或非糖基化的。
如本文所使用的术语“抗体”可包括完整抗体及其任何抗原结合片段(即“抗原结合部分”)或单链。在一个实施方案中,“抗体”指包含藉由二硫键互连的至少两个重(H)链及两个轻(L)链的糖蛋白或其抗原结合部分。每一重链包含重链可变区(在本文中缩写为VH)及重链恒定区。在某些天然存在的IgG、IgD及IgA抗体中,重链恒定区包含三个结构域,CH1、CH2及CH3。在某些天然存在的抗体中,每一轻链包含轻链可变区(在本文中缩写为VL)及轻链恒定区。轻链恒定区包含一个结构域CL。可将VH及VL区进一步细分成超变区(称为互补决定区(CDR))及更保守的区域(称为框架区(FR)),二者间杂排列。在本文中,VH区的CDR缩写为HCDR,即VH区的三个CDR可以缩写为HCDR1、HCDR2、HCDR3;VL区的CDR缩写为LCDR,即VL区的三个CDR可以缩写为LCDR1、LCDR2、LCDR3。每一VH及VL由三个CDR及四个FR构成,其自氨基末端至羧基末端按下列顺序布置:FR1、CDR1、FR2、CDR2、FR3、CDR3、FR4。重链及轻链的可变区含有与抗原相互作用的结合结构域。
抗体的重链可或可不含有末端赖氨酸(K)或末端甘氨酸及赖氨酸(GK)。因此,本文所提供的任何重链序列及重链恒定区序列可以以GK或G结尾,或缺少K或GK,无论该序列的最后一个氨基酸为何。这是因为末端的赖氨酸,有时是甘氨酸与赖氨酸,在抗体表达期间会被切去。
抗体通常以高亲和力特异性结合至其同源抗原,高亲和力由10 -7M至10 -11M或更小的解离常数(K D)所反映。通常认为大于约10 -6M的任何K D指示非特异性结合。如本文所使用的,“特异性结合”至抗原的抗体指以高亲和力结合至抗原且实质上相同的抗原、但不以高亲和力结合至不相关抗原的抗体,高亲和力意指具有10 -7M或更小、优选10 -8M或更小、甚至更优选5×10 -9M或更小、且最佳介于10 -8M与10 -10M之间或更小的K D。若抗原展现与给定抗原的高度序列同一性,例如若其展现与给定抗原的序列至少80%、至少90%、至少95%、至少97%或至少99%或更大的序列同一性,则该抗原与给定抗原“实质上相同”。
免疫球蛋白可来自普遍已知同型中的任一者,包括(但不限于)IgA、分泌性IgA、IgG及IgM。IgG同型在某些物种中分成多个子类:在人中分成IgG1、IgG2、IgG3及IgG4,在小鼠中分成IgG1、IgG2a、IgG2b及IgG3。在某些实施方案中,本文所述抗LIF抗体具有人IgG1或小鼠IgG1子型。免疫球蛋白(例如人IgG1)以若干异型存在,其彼此相差至多几个氨基酸。举例而言,“抗体”可包括天然存在的及非天然存在的抗体;单克隆及多株抗体;嵌合及人源化抗体;人及非人抗体;全合成抗体;及单链抗体。
如本文所使用,术语抗体的“抗原结合部分”指抗体的保留特异性结合至抗原的能力的一个或多个片段。已显示,抗体的抗原结合功能可由全长抗体的片段来实施。抗体(例如本文所述的抗LIF抗体)的“抗原结合部分”这一术语所涵盖的结合片段的实例包括(i)Fab片段,其是由VL、VH、CL及CH1结构域所组成的单价片段;(ii)F(ab') 2片段,其是包含藉由二硫桥在铰链区处连接的两个Fab片段的二价片段;(iii)Fd片段,其由VH及CH1结构域组成;(iv)Fv片段,其由抗体的一条单臂的VL及VH结构域组成,(v)dAb片段(Ward等人(1989)Nature 341:544-546),其由VH结构域组成;及(vi)分离的互补决定区(CDR),或(vii)两个或更多个分离的CDR的组合,这些CDR可视情况藉由合成接头连结。另外,尽管Fv片段的两个结构域VL及VH由不同的基因编码,但其可利用重组方法藉由合成接头来连结这些结构域,使它们能够被制备成为单一蛋白链,其中VL及VH区配对形成单价分子(称为单链Fv(scFv));参见例如Bird等人,(1988)Science 242:423-426;及Huston等人(1988)Proc.Natl.Acad.Sci.USA 85:5879-5883)。这些单链抗体亦欲涵盖于术语抗体的“抗原结合部分”内。这些及其他潜在构建体阐述于Chan及Carter(2010)Nat.Rev.Immunol.10:301。这些抗体片段系使用本领域技术人员已知的习用技术来获得,并且从这些片段中筛选可以按照与完整抗体相同的方式使用者。抗原结合部分可通过重组DNA技术或通过酶或化学裂解完整免疫球蛋白来产生。
术语“保守修饰形式的氨基酸序列”指这样的氨基酸修饰,它不显著影响或改变包含该氨基酸序列的抗体的结合特性,该修饰包括氨基酸置换、增加和缺失。修饰可通过例如定点诱变和PCR介导的诱变等标准技术引入本发明的抗体中。保守氨基酸置换系其中的氨基酸残基被具有相似侧链的氨基酸残基替换的置换。在本领域中已经确定具有相似侧链的氨基酸残基家族。这些家族包括具有碱性侧链的氨基酸(如赖氨酸、精氨酸、组氨酸),具有酸性侧链的氨基酸(如天冬氨酸、谷氨酸),具有不带电的极性侧链的氨基酸(如甘氨酸、天冬酰胺、谷氨酰胺、丝氨酸、苏氨酸、酪氨酸、半胱氨酸、色氨酸),具有非极性侧链的氨基酸(如丙氨酸、缬氨酸、亮氨酸、异亮氨酸、脯氨酸、苯丙氨酸、甲硫氨酸),具有β-支化侧链的氨基酸(如苏氨酸、缬氨酸、异亮氨酸)、和具有芳香侧链的氨基酸(如酪氨酸、苯丙氨酸、色氨酸、组氨酸)。因此,本发明的抗体的CDR区中的一个或多个氨基酸残基可以被来自相同侧链家族的其他氨基酸残基替代,并且使可用此处说明的功能测定方法对经改变的抗体的保留功能进行测试。优选的是,保守修饰在数目上不超过1个或2个。
修饰本文所描述的氨基酸序列是本发明希望的,特别是那些人重链恒定区以改编序列到所希望的异型、如在亚洲人群中发现的异型。
例如,抗体的一个或多个CDR或CDR组可以被嫁接到框架内(如人框架)以提供抗体分子。框架区可以是人生殖系的基因序列,或非生殖系的。这样框架可以是生殖的,其中框架内的一个或多个残基可以被交换以匹配最相似人生殖系框架内的相当位置的残基。这样,本发明的结合成员可以是分离的具有在人生殖系框架内包含HCDR组的VH结构域,例如,人生殖系IgG VH框架。结合成员还可以带有包含LCDR组的VL结构域,如在人生殖系IgG VL框架内。
VH和/或VL支架残基可以如所讨论的被修饰,本文举例,如使用位点导向诱变。本发明的VH或VL结构域,或结合成员包括这样的VL结构域。
可以在一个或多个框架区和/或一个或多个CDR内进行变化,所述变化通常不导致功能的丧失,所以包含这种变化氨基酸序列的结合成员应保持结合和/或中和LIF的能力。它可以保持与没有进行变化的结合成员具有相同数量结合和/或中和能力,如按本文所述的分析法进行测量。包含这样变化的氨基酸序列的结合成员可以具有改善的结合和/或中和LIF的能力。
变化可以包括有非天然存在的或非标准氨基酸替换一个或多个氨基酸残基,将一个或多个氨基酸残基修饰到非天然存在的或非标准的形式内,或将一个或多个非天然存在的或非标准的氨基酸***该序列。本发明序列中变化的位置和数量的例子在本文别处进行描述。天然存在的氨基酸包括通过其标准单字母代码被确定为G、A、V、L、I、M、P、F、W、S、T、N、Q、Y、C、K、R、H、D、E的20“标准”L-氨基酸。非标准氨基酸包括可以被合并入多肽骨架或来自现有氨基酸残基修饰而成的任意的其他残基。非标准氨基酸可以是天然存在或非天然存在。几个天然存在非标准氨基 酸已为本领域所知,4-羟基脯氨酸、5-羟基赖氨酸、3-甲基组氨酸、N-乙基丝氨酸,等(Voet&Voet,1995,Biochemistry,2nd Edition,(Wiley))。在其N-α位进行衍生的那些氨基酸残基将仅被定位在氨基酸序列的N-末端。通常本发明中的氨基酸为L-氨基酸,但可以是D-氨基酸。因此变化可以包括用L-氨基酸进行修饰或D-氨基酸替换L-氨基酸。氨基酸的甲酰化、乙酰化和/或磷酸化形式是已知的,本发明氨基酸可以进行这样的修饰。
本发明结合成员和抗体结构域中的氨基酸序列可以包含以上所描述的非天然或非标准的氨基酸。非标准氨基酸(如D-氨基酸)可以在合成过程中被并入氨基酸序列,或通过在氨基酸合成之后进行“原始”标准氨基酸的修饰或替换。
使用非标准和/或非天然存在的氨基酸提高结构和功能的多样性,并能提高潜能以实现本发明结合成员中所希望的LIF结合和中和性能。另外,与标准L-氨基酸相比,D-氨基酸及类似物已显示具有更好的药代动力学特性,原因是在向动物如人施用之后,具有L-氨基酸的多肽的体内降解。
本发明带有CDR-衍生序列的新的VH或VL区域的生成可以采用一种或多种选自VH和/或VL基因的随机诱变以生成全部变异区内的突变体。这样的技术描述在Gram等人(Gram等人,1992,Proc.Natl.Acad.Sci.,USA,89:3576-3580),其使用了易错PCR。在一些实施方案中,一种或多种氨基酸替代在全部变异区或CDR组内进行。
可以使用的另一种方法为VH或VL基因的CDR区的导向诱变。这样的方法公布在Barbas等人(Barbas等人,1994,Proc.Natl Acad.Sci.,USA,91:3809-3813)和Schier等人(Schier等人,1996,J.Mol.Biol.263:551-567)。
以上所述描述的所有方法为本领域已知,本领域技术人员将能够使用这样的方法并采用本领域的常规方法以提供本发明的结合成员。
根据本发明可以使用本文所公开的特定序列的任意VH和VL结构域的变异结构域氨基酸序列变异体,如所讨论的。特定的变异体可以包括一种或多种氨基酸序列的变化(氨基酸残基的增加、删除、取代和/或***)。在某些实施方式中,变异体具有少于约17、少于9或少于5的这样的变化。
如以上所示,实质上如本文所述的CDR氨基酸序列可以被携带作为人抗体变异结构区或其大部分中的CDR。实质上如本文所述的HCDR3序列代表本发明的实施方式,这些中的每一个可以被携带作为人抗体变异区或其大部分中的CDR,任选地联合本发明的HCDR1、HCDR2、LCDR1、LCDR2和LCDR3。
如本文所使用,术语“单克隆抗体”指展示针对特定表位的单一结合特异性及亲和力的某一抗体,或者是抗体组合物,其中抗体组合物中的所有的抗体皆展示针对特定表位的单一结合特异性及亲和力。通常,这些单克隆抗体将源自单一抗体编码细胞或核酸,且将在不有意引入任何序列变化 的条件下增殖。因此,术语“人单克隆抗体”指具有源自人种系免疫球蛋白序列的可变区、及任选的恒定区的单克隆抗体。在一个实施方案中,人单克隆抗体是通过例如通过融合来自转基因或转染色体非人动物(例如具有包含人重链转基因及轻链转基因的基因组的转基因小鼠)获得的B细胞与永生细胞获得的杂交瘤产生的。术语“mAb”指单克隆抗体。
如本文所使用的,术语“重组人抗体”包括通过重组方式制备、表达、产生或分离的所有人抗体,例如(a)自对人免疫球蛋白基因转基因或转染色体的动物(例如小鼠)或自其制备的杂交瘤分离的抗体,(b)自经转化以表达该抗体的宿主细胞,例如自转染瘤分离的抗体,(c)自重组、组合人抗体文库分离的抗体,及(d)通过涉及将人免疫球蛋白基因序列剪接至其他DNA序列的任何其他方式制备、表达、产生或分离的抗体。这些重组人抗体包含利用具体人种系免疫球蛋白序列且由种系基因编码、但包括在例如抗体成熟期间发生的后续重排及突变的可变区及恒定区。如本领域已知(例如,参见Lonberg(2005)Nature Biotech.23(9):1117-1125),可变区含有由重排以形成特异性针对外源抗原的抗体的不同基因编码的抗原结合结构域。除重排之外,可变区可进一步经多个单一氨基酸变化(称为体细胞突变或超突变)修饰以增加抗体对外源抗原的亲和力。恒定区将进一步因应抗原而改变(即同型切换)。因此,因应抗原的编码轻链及重链免疫球蛋白多肽的经重排及体细胞突变的核酸序列可与原始种系序列不相同,但将实质上相同或相似(即具有至少80%同一性)。
“人”抗体(HuMAb)指具有这样的可变区的抗体:可变区中的框架区及CDR区皆源自人种系免疫球蛋白序列。另外,若抗体含有恒定区,则恒定区亦源自人种系免疫球蛋白序列。本文所述抗体可包括不由人种系免疫球蛋白序列编码的氨基酸残基(例如,通过体外随机诱变或位点特异性诱变或通过体内体细胞突变引入的突变)。然而,如本文所使用的,术语“人抗体”并不欲包括源自另一哺乳动物物种(例如小鼠)种系的CDR序列已移植至人框架序列上的抗体。“人”抗体与“全人”抗体两个术语以同义使用。
“人源化”抗体指这样的抗体,其中,非人抗体的CDR结构域以外的一些、大部分或所有氨基酸被源自人免疫球蛋白的相应氨基酸替代。在抗体的人源化形式的一个实施方案中,CDR结构域以外的一些、大部分或所有氨基酸已被来自人免疫球蛋白的氨基酸替代,而一个或多个CDR区内的一些、大部分或所有氨基酸未发生变化。可容许存在氨基酸的小添加、缺失、***、取代或修饰,只要其不消除抗体结合具体抗原的能力即可。“人源化”抗体保留类似于原始抗体的抗原特异性。
“嵌合抗体”指可变区源自一个物种且恒定区源自另一物种的抗体,例如可变区源自小鼠抗体且恒定区源自人抗体的抗体。
本发明功能性抗体片段包括通过诸如通过PEG化,或并入到脂质体中的化学修饰提高半衰期的任意的功能性片段。
本发明抗体包括双特异性抗体。双特异性或双功能性抗体形成第二代单克隆抗体,其中两个不同变异区被结合到相同的分子中(Holliger and Bohlen,1999 Cancer and metastasis rev.18:411-419)。从它们的招募新效应子功能或靶向一些分子到肿瘤细胞表面上的能力它们在诊断领域和治疗领域的应用已经被阐述。其中使用双特异性抗体时,这些可以是常规的双特异性抗体,其可以是以各种方式进行制造(HolligerP.& Winter G.Current Opinion Biotechnol.4,446-449:1993),如化学制备、或来自杂交的杂交瘤,或可以是以上所提到的任意的双特异性抗体片段。这些抗体的获得可以通过化学方法或体细胞的方法,但同样地并优选基因工程方法,所述方法允许实施异源二聚化,这样有利于所得抗体的纯化工艺。双特异性抗体的例子包括那些BiTETM方法,该方法中带有不同的特异性两个抗体的结合结构域可以被使用并直接地通过短的柔性肽连接。这在短的单个多肽链上结合两个抗体。双链抗体和scFc在没有Fc区域下被构建,仅使用变异区,潜在地降低了抗独特型反应的效果。
双特异性抗体可以被构建为全IgG,双特异性(Fab’)2、(Fab’)PEG、双链抗体或其他的双特异性scFv。进一步可以使用本领域已知常规方法两个双特异性抗体进行连接以形成四价抗体。
相对于双特异性全抗体,双特异性双链抗体也是特别有用的,因为它们能够轻易地在大肠杆菌中被构建和表达。使用噬菌体展示库(WO1994/13804,适当结合特异性的双链抗体(和许多其他的多肽,如抗体片段)能够轻易地被选择。如果双链抗体的一个臂被保持恒定,然后制备库,其中其他的臂被变异,选择适当特异性的抗体。双特异性全抗体可以通过不同的工程方法进行制备,所述方法描述在Ridgeway等人(Ridgeway,J.B.B.等人,Protein Eng.9,616-621,1996)或WO1996/27011,WO1998/50431和WO2006/028936。
“经修饰的重链恒定区”指包含恒定结构域CH1、铰链、CH2及CH3的重链恒定区,其中一个或多个恒定结构域来自不同同型(例如IgG1、IgG2、IgG3、IgG4)。在某些实施方案中,经修饰的恒定区包括人IgG2CH1结构域及融合至人IgG1CH2结构域的人IgG2铰链及人IgG1CH3结构域。在某些实施方案中,这些经修饰恒定区亦包括相对于野生型氨基酸序列,一个或多个结构域内的氨基酸修饰。
如本文所使用的,“同型”指由重链恒定区基因编码的抗体类别(例如IgG1、IgG2、IgG3、IgG4、IgM、IgA1、IgA2、IgD及IgE抗体)。
“异型”指特定同型群内的天然存在的变体,这些变体相差几个氨基酸(例如,参见Jefferis等人(2009)mAbs 1:1)。本文所述抗体可具有任何异型。
除非本文在别处另有说明,否则所有氨基酸编号均根据Kabat***的EU索引(Kabat,E.A.等人(1991)Sequences of Proteins of Immunological Interest,第5版,U.S.Department of Health and Human Services,NIH公布第91-3242号)。
“识别抗原的抗体”及“特异性针对抗原的抗体”在本文中可与术语“特异性结合至抗原的抗体”互换使用。
如本文所使用的,术语“分离的抗体”欲指这样的抗体,其实质游离于其他具有不同抗原特异性的抗体(例如,特异性结合至LIF的分离的抗体实质游离于特异性结合除LIF外的抗原的抗体)。然而,特异性结合至LIF的表位的分离的抗体可具有针对不同物种的其他LIF蛋白的交叉反应性。
“效应物功能”指抗体Fc区与Fc受体或配体的相互作用,或源自其的生物化学事件。示例性“效应物功能”包括C1q结合、补体依赖性细胞毒性(CDC)、Fc受体结合、FcγR介导的效应物功能(例如ADCC及抗体依赖性细胞介导的吞噬作用(ADCP))及下调细胞表面受体(例如B细胞受体;BCR)。这些效应物功能通常需要组合Fc区与结合结构域(例如抗体可变结构域)。
“Fc受体”或“FcR”是结合至免疫球蛋白的Fc区的受体。结合至IgG抗体的FcR包括FcγR家族的受体,包括这些受体的等位基因变体及可变剪接形式。FcγR家族由三种活化性受体(在小鼠中为FcγRⅠ、FcγRⅢ及FcγRⅣ;在人类中为FcγRⅠA、FcγRⅡA及FcγRⅢA)及一种抑制性受体(FcγRⅡB)组成。人FcγR的多种特性概述于表A中。大部分固有效应细胞类型共表达一种或多种活化FcγR及抑制性FcγRⅡB,而天然杀伤(NK)细胞选择性表达一种活化Fc受体(小鼠中的FcγRⅢ及人中的FcγRⅢA),但不表达小鼠及人中的抑制性FcγRⅡB。人IgG1结合至大部分人Fc受体且认为其所结合的活化Fc受体类型等效于鼠类IgG2a。
表A.人FcγR的特性
Figure PCTCN2020118247-appb-000029
“铰链”、“铰链结构域”或“铰链区”或“抗体铰链区”指重链恒定区的连结CH1结构域与CH2结构域且包括铰链之上部分、中间部分及下部分的结构域(Roux等人,J.Immunol.1998  161:4083)。铰链提供抗体的结合区域与效应物区域之间的不同柔性且亦提供两个重链恒定区之间的分子间二硫键连接的位点。
术语“铰链”包括野生型铰链以及其变体(例如非天然存在的铰链或经修饰铰链)。例如,术语“IgG2铰链”包括野生型IgG2铰链及具有1个、2个、3个、4个、5个、1-3个、1-5个、3-5个和/或至多5个、4个、3个、2个或1个突变(例如取代、缺失或添加)的变体。
术语“CH1结构域”指将可变结构域与重链恒定结构域中的铰链连接的重链恒定区。术语“CH1结构域”包括野生型CH1结构域以及其变体(例如非天然存在的CH1结构域或经修饰的CH1结构域)。例如,术语“CH1结构域”包括野生型CH1结构域及其具有1个、2个、3个、4个、5个、1-3个、1-5个、3-5个和/或至多5个、4个、3个、2个或1突变(例如取代、缺失或添加)的变体。示例性CH1结构域包括具有改变抗体的生物活性(例如ADCC、CDC或半衰期)的突变的CH1结构域。
术语“CH2结构域”指连接重链恒定结构域中的铰链与CH3结构域的重链恒定区。术语“CH2结构域”包括野生型CH2结构域以及其变体(例如非天然存在的CH2结构域或经修饰的CH2结构域)。例如,术语“CH2结构域”包括野生型CH2结构域及其具有1个、2个、3个、4个、5个、1-3个、1-5个、3-5个和/或至多5个、4个、3个、2个或1个突变(例如取代、缺失或添加)的变体。示例性CH2结构域包括具有改变抗体的生物活性(例如ADCC、CDC或半衰期)的突变的CH2结构域。
术语“CH3结构域”指在重链恒定结构域中为CH2结构域的C末端的重链恒定区。术语“CH3结构域”包括野生型CH3结构域以及其变体(例如非天然存在的CH3结构域或经修饰的CH3结构域)。例如,术语“CH3结构域”包括野生型CH3结构域及其具有1个、2个、3个、4个、5个、1-3个、1-5个、3-5个和/或至多5个、4个、3个、2个或1个突变(例如取代、缺失或添加)的变体。示例性CH3结构域包括具有改变抗体的生物活性(例如ADCC、CDC或半衰期)的突变的CH3结构域。
“CL结构域”指轻链的恒定结构域。术语“CL结构域”包括野生型CL结构域及其变体。
“天然序列Fc区”或“天然序列Fc”包含与自然界中发现的Fc区的氨基酸序列相同的氨基酸序列。天然序列人Fc区包括天然序列人IgG1Fc区;天然序列人IgG2Fc区;天然序列人IgG3Fc区;及天然序列人IgG4Fc区以及其天然变体。天然序列Fc包括Fc的多个异型(例如,参见Jefferis等人(2009)mAbs 1:1)。
术语“表位”或“抗原决定簇”指抗原(例如LIF)上免疫球蛋白或抗体特异性结合的位点。蛋白质抗原内的表位可自连续氨基酸(通常为线性表位)或通过蛋白质的三级折叠并置的非连续氨基 酸二者形成(通常为构象表位)。在暴露于变性溶剂时,通常但不始终保留自连续氨基酸形成的表位,而在用变性溶剂处理时通常丢失由三级折叠形成的表位。在独特空间构象中,表位通常包括至少3个、4个、5个、6个、7个、8个、9个、10个、11个、12个、13个、14个或15个氨基酸。确定由给定抗体结合表位的方法(即表位定位)为本领域所熟知,且包括例如免疫印迹及免疫沉淀分析,其中测试重叠或连续肽(例如来自LIF)与给定抗体(例如抗LIF抗体)的反应性。确定表位的空间构象的方法包括本领域中的技术及那些本文所述者,例如x射线结晶学、2维核磁共振及HDX-MS(例如,参见Epitope Mapping Protocolsin Methods in Molecular Biology,第66卷,G.E.Morris编辑(1996))。
“与另一抗体竞争与靶结合”的抗体指抑制(部分抑制或完全抑制)另一抗体与靶的结合的抗体。可使用已知竞争实验(例如实施例中所述的那些)来确定两种抗体是否彼此竞争结合至靶,即一种抗体是否抑制另一抗体与靶的结合以及抑制程度。在某些实施方案中,抗体与另一抗体竞争与靶的结合,且抑制至少10%、20%、30%、40%、50%、60%、70%、80%、90%或100%的该结合。抑制或竞争的程度可依赖于哪种抗体为“封闭抗体”(即先与靶一起温育的冷抗体)而不同。竞争测定可如例如以下文献中所述来实施:Ed Harlow及David Lane,Cold Spring Harb Protoc;2006;doi:10.1101/pdb.prot4277或“UsingAntibodies”,Ed Harlow及David Lane,Cold Spring Harbor Laboratory Press,ColdSpring Harbor,NY,USA 1999的第11章。竞争性抗体结合至相同表位、重叠表位或毗邻表位(例如,由位阻作用所证实的)。
其他竞争性结合测定包括:固相直接或间接放射性免疫测定(RIA)、固相直接或间接酶免疫测定(EIA)、夹心竞争测定(参见Stahli等人,Methods in Enzymology 9:242(1983));固相直接生物素-抗生物素蛋白EIA(参见Kirkland等人,J.Immunol.137:3614(1986));固相直接标记测定、固相直接标记夹心分析(参见Harlow及Lane,Antibodies:ALaboratory Manual,Cold Spring Harbor Press(1988));使用I-125标记的固相直接标记RIA(参见Morel等人,Mol.Immunol.25(1):7(1988));固相直接生物素-抗生物素蛋白EIA(Cheung等人,Virology 176:546(1990));及直接标记RIA。(Moldenhauer等人,Scand.J.Immunol.32:77(1990))。
如本文所使用的,术语“Kassoc”或“Ka”欲指具体抗体-抗原相互作用的缔合速率常数(association rate constant),而如本文所使用,术语“Kdis”或“Kd”欲指具体抗体-抗原相互作用的解离速率常数(dissociation rate constant)。如本文所使用,术语“K D”欲指平衡解离常数,其系自Kd对Ka的比率(即Kd/Ka)获得且以摩尔浓度(M)表示。可使用本领域充分确立的方法测定抗体的K D值。测定抗体的K D的优选方法系通过使用表面等离子共振,优选使用生物传感器***,例如
Figure PCTCN2020118247-appb-000030
表面等离子共振***或流式细胞术或Scatchard分析。
术语“EC50”在使用抗体或其抗原结合片段的体外或体内分析背景下指诱导50%最大反应,即介于最大反应与基线之间之中途的反应的抗体或其抗原结合部分的浓度。
术语“IC50”,在功能性分析中,IC50为能将生物应答降低到其最大值50%的结合成员的浓度,以nM为单位。在配体-结合研究中,IC50为降低受体结合到最大特异性结合水平的50%的浓度。IC50可以通过绘制最大生物活性应答的百分数作为结合成员浓度的log的函数,并使用软件程序如Origin(OriginLab软件公司,Northampton,Massachusetts,USA),以将S函数拟合数据以生成IC50值。使用一种或多种本领域技术人员已知的和/或本文所描述或参考的分析法进行测定或测量效价。结合成员的中和效价可用表述为几何平均数(Geomean)。
如本文所使用的术语“天然存在”在应用于目标时指可在自然界中发现目标的事实。例如,存在于可自自然界来源分离且尚未在实验室中经人有意修饰的生物体(包括病毒)中的多肽或多核苷酸序列是天然存在的。
“多肽”指包含至少两个连续连接的氨基酸残基的链,且链的长度无上限。蛋白质中之一或多个氨基酸残基可含有修饰,例如(但不限于)糖基化、磷酸化或二硫键。“蛋白质”可包含一个或多个多肽。
如本文所使用,术语“核酸分子”意欲包括DNA分子及RNA分子。核酸分子可为单链或双链,且可为cDNA。亦提供本文所述SEQ ID NOs中所述序列的“保守序列修饰”,即不消除由核苷酸序列编码或含有氨基酸序列的抗体与抗原的结合的核苷酸及氨基酸序列修饰。这些保守序列修饰包括保守核苷酸及氨基酸取代以及核苷酸及氨基酸添加及缺失。例如,可通过本领域已知的标准技术(例如定点诱变及PCR介导的诱变)将修饰引入本文所述的SEQ ID NOs中。保守序列修饰包括保守氨基酸取代,其中氨基酸残基被替换为具有类似侧链的氨基酸残基。具有类似侧链的氨基酸残基的家族是本领域中已有定义的。这些家族包括具有碱性侧链的氨基酸(例如赖氨酸、精氨酸、组氨酸)、具有酸性侧链的氨基酸(例如天冬氨酸、谷氨酸)、具有不带电极性侧链的氨基酸(例如甘氨酸、天冬酰胺、谷氨酰胺、丝氨酸、苏氨酸、酪氨酸、半胱氨酸、色氨酸)、具有非极性侧链的氨基酸(例如丙氨酸、缬氨酸、亮氨酸、异亮氨酸、脯氨酸、苯丙氨酸、甲硫氨酸)、具有β分枝侧链的氨基酸(例如苏氨酸、缬氨酸、异亮氨酸)及具有芳香族侧链的氨基酸(例如酪氨酸、苯丙氨酸、色氨酸、组氨酸)。
对于核酸而言,术语“实质同源性”表示,两个核酸或其指定序列在最佳比对及比较(其中适当***或缺失核苷酸)时有至少约80%的核苷酸、通常至少约90%至95%、且更优选至少约98%至99.5%的核苷酸相同。作为另一选择,当区段在选择性杂交条件下与链的互补物杂交,则存在实质同源性。
对于多肽而言,术语“实质同源性”表示,两个多肽或其指定序列在最佳比对及比较(其中适当***或缺失核苷酸)时有至少约80%的氨基酸、通常至少约90%至95%、且更优选至少约98%至99.5%的氨基酸相同。
两条序列之间的同一性%在最佳比对序列时随这些序列所共享的相同位置数而变化(即同源性%=相同位置数/总位置数×100),其中最佳比对系考虑到为达成两条序列的最佳比对而需要引入的空位数及每一空位的长度来确定。两条序列之间的序列比较及同一性百分数测定可使用数学算法来完成,如下文非限制性实施例中所述。
两条核苷酸序列之间的同一性百分数可使用GCG软体包(可在http://www.gcg.com上获得)中的GAP程序、使用NWSgapdna.CMP矩阵及空位权重40、50、60、70或80以及长度权重1、2、3、4、5或6来测定。两条核苷酸或氨基酸序列之间之同一性百分数亦可使用已纳入比对程序(ALIGN program,2.0版)中的E.Meyers及W.Miller(CABIOS,4:11-17(1989))算法、使用PAM120权重残基表、空位长度罚分12及空位罚分4来测定。另外,两条氨基酸序列之间的同一性百分数可使用已纳入GCG软体包(可在http://www.gcg.com上获得)中的空位程序中的Needleman及Wunsch(J.Mol.Biol.(48):444-453(1970))算法、使用Blossum 62矩阵或PAM250矩阵及空位权重16、14、12、10、8、6或4以及长度权重1、2、3、4、5或6来测定。
本文所述的核酸及蛋白质序列可进一步用作“询问序列”来实施针对公共数据库的检索,以例如鉴定相关序列。这些检索可使用Altschul等人(1990)J.Mol.Biol.215:403-10的NBLAST及XBLAST程序(2.0版)来实施。BLAST核苷酸检索可使用NBLAST程序、评分=100、字长=12来实施,以获得与本文所述核酸分子同源的核苷酸序列。BLAST蛋白质检索可使用XBLAST程序、评分=50、字长=3来实施,以获得与本文所述蛋白质分子同源的氨基酸序列。为获得用于比较目的的空位比对,可如Altschul等人(1997)Nucleic Acids Res.25(17):3389-3402中所述使用空位BLAST。在使用BLAST及空位BLAST程序时,可使用各程序(例如XBLAST及NBLAST)的预设参数。参见www.ncbi.nlm.nih.gov。
这些核酸可存在于完整细胞中,存在于细胞溶解物中,或以部分纯化或实质上纯净的形式存在。当通过标准技术实施纯化以清除其他细胞组分或其他污染物例如其他细胞核酸(例如染色体的其他部分)或蛋白质)时,核酸是“分离的”或“实质上纯净”的,这些标准技术包括碱/SDS处理、CsCl显带、柱层析、琼脂糖凝胶电泳及本领域熟知的其他方法。参见F.Ausubel等人编辑.Current Protocols in Molecular Biology,Greene Publishingand Wiley Interscience,New York(1987)。
核酸(例如cDNA)可根据标准技术进行突变以提供基因序列。对于编码序列,这些突变可视需要影响氨基酸序列。具体而言,涵盖与天然V序列、D序列、J序列、恒定序列、切换序列及本文所述的其他这些序列实质上同源或源自其的DNA序列。
如本文所使用,术语“载体”欲指能够转运与其连接的另一核酸的核酸分子。一类载体为“质粒”,其指其他DNA区段可连接至其中的环形双链DNA环。另一类载体为病毒载体,其中其他DNA区段可连接至病毒基因组中。某些载体能够在已引入其的宿主细胞中进行自主复制(例如,具有细菌复制起点的细菌载体及游离型哺乳动物载体)。其他载体(例如非游离型哺乳动物载体)可在引入宿主细胞中时整合至该宿主细胞的基因组中,并藉此随宿主基因组一同复制。另外,某些载体能够引导与其可操作连接的基因的表达。这些载体在本文中称为“重组表达载体”(或简称为“表达载体”)。一般而言,用于重组DNA技术中的表达载体通常呈质粒形式。在本说明书中,“质粒”与“载体”可互换使用,这是因为质粒是载体的最常用形式。然而,本发明亦包括提供等效功能的其他形式的表达载体,例如病毒载体(例如复制缺陷型反转录病毒、腺病毒及腺相关病毒)。
如本文所使用,术语“重组宿主细胞”(或简称为“宿主细胞”)欲指包含并非天然存在于细胞中的核酸的细胞,且可为已引入重组表达载体中的细胞。应理解,这些术语不仅欲指具体个体细胞,亦欲指此细胞的子代。由于突变或环境影响可使后续各代发生某些改变,故该子代实际上可能与亲代细胞不同,但仍包括于如本文所使用的术语“宿主细胞”的范畴内。
如本文所使用的,术语“抗原”指任何天然或合成免疫原性物质,例如蛋白质、肽或半抗原。抗原可为LIF或其片段。
如本文所使用,术语“抑制”或“阻断”(例如,提及LIF结合或活性的抑制/阻断)可互换使用,且涵盖部分及完全抑制/阻断二者。
如本文所使用,“癌症”指以身体中的不受控异常细胞生长为特征的一大类疾病。由于失调的细胞***可形成恶性的肿瘤或细胞,它们侵袭邻近组织且可经由淋巴***或血流转移至身体远端。
如本文所使用,术语“治疗”或“处理”指对受试者实施任何类型的干预或过程,或向受试者施用活性剂,以逆转、缓和、改善、抑制或减缓或预防与疾病相关的症状、并发症、病况或生物化学标记的进展、发展、严重程度或复发。预防指对未患疾病的受试者施用来预防疾病发生,或者若疾病发生,最小化其影响。
术语“有效剂量(effective dose或effective dosage)”定义为足以达成或至少部分达成期望效应的量。药物或治疗剂的“治疗有效量”或“治疗有效剂量”是该药物在单独使用或与另一治疗剂组合使用时可促进疾病消退的任何量,该疾病消退由疾病症状的严重程度的减轻、无疾病症状期的频率及持续时间的增加、或因患病所致的损害或失能的防止所证实。药物的“预防有效量”或“预防 有效剂量”是该药物在单独施用或与另一治疗剂组合施用于具有罹患疾病或遭受疾病复发的风险的受试者时,抑制该疾病的发展或复发的量。治疗剂或预防剂促进疾病消退或抑制疾病的发展或复发的能力可使用本领域技术人员已知的多种方法来评估,例如在临床试验期间在人受试者中评估,在可预测在人中的功效的动物模型***中评估,或通过在体外测定中测定药剂的活性来评估。
术语“患者”及“受试者”指接受预防或治疗性治疗的任何人或非人动物。例如,可使用本文所述的方法及组合物来治疗患有癌症的受试者。术语“非人动物”包括所有脊椎动物,例如哺乳动物及非哺乳动物,例如非人灵长类动物、绵羊、狗、牛、鸡、两栖动物、爬行动物等。
实施例
实施例1 抗人LIF单克隆抗体的筛选与鉴定
1.1杂交瘤法制备抗人LIF单克隆抗体38E10E1C11
按单克隆抗体制备方法(Kohler和Milstein(1975)Nature256:495),用重组人LIF蛋白(购自于Sino Biological)免疫BABL/c小鼠,初次免疫使用25μg重组人LIF蛋白与等体积的弗氏完全佐剂,背部皮下多点注射,四周后第二次免疫,使用弗氏不完全佐剂加25μg重组人LIF蛋白,20天后通过间接ELISA方法检测小鼠血清抗体效价。间隔2-3周后,经腹腔注射50μg重组人LIF蛋白加强免疫,3天后处死动物取脾细胞进行融合。
取对数生长期的小鼠骨髓瘤细胞SP2/0计数,同时制备免疫鼠的脾细胞悬液,按常规方法用50%PEG使脾细胞与SP2/0细胞进行融合。融合后的细胞加入滋养层细胞(六周龄BABL/c鼠腹腔巨噬细胞)的96孔板内,已含1%HAT、20%胎牛血清的DMEM筛选培养。当克隆长至1/3板底时,收集培养上清。以重组人LIF蛋白包被ELISA板,间接ELISA法检测培养上清中抗LIF抗体,筛选分泌抗人LIF抗体的克隆,进一步利用有限稀释法进行单克隆化获得一株可稳定分泌高亲和力的抗人LIF单克隆抗体的细胞株,将其分泌的抗体标记为38E10E1C11,经测定,编码38E10E1C11抗体轻链及重链的全长基因序列分别为SEQ ID NO:42和SEQ ID NO:44所示,对应的38E10E1C11抗体的轻链及重链的全长氨基酸序列分别如SEQ ID NO:41和SEQ ID NO:43所示;编码38E10E1C11抗体轻链及重链的可变区基因序列分别如SEQ ID NO:76和SEQ ID NO:77所示,对应的38E10E1C11抗体的轻链及重链的可变区的氨基酸序列分别如SEQ ID NO:74和SEQ ID NO:75所示;按Kabat编码规则获知38E10E1C11抗体的LCDR1的氨基酸序列如SEQ ID NO:1所示,LCDR2的氨基酸序列如SEQ ID NO:2所示,LCDR3的氨基酸序列如SEQ ID NO:3所示,HCDR1的氨基酸序列如SEQ ID NO:4所示,HCDR2的氨基酸序列如SEQ ID NO:45所示,HCDR3的氨基酸序列如SEQ ID NO:6所示,对该抗体进行免疫球蛋白类型及亚型鉴定(结果为IgG1亚类,к型轻链)。
在获得能够稳定分泌抗体的杂交瘤细胞株之后,使用thermo fisher的CD hybridoma无血清培养基驯化细胞,让其适应无血清悬浮震荡培养,之后利用无血清培养基表达纯化抗体。
1.2噬菌体展示技术制备抗人LIF单克隆抗体P36-033
用重组人LIF蛋白免疫BABL/c小鼠,初次免疫使用25μg纯重组人LIF蛋白与等体积的弗氏完全佐剂,背部皮下多点注射,四周后第二次免疫,使用弗氏不完全佐剂加25μg重组人LIF蛋白,20天后通过间接ELISA方法检测小鼠血清抗体效价。间隔2-3周后,经腹腔注射50μg重组人LIF蛋白加强免疫,3天后处死动物取脾细胞。采用invitrogen公司的TRIZOL Reagent提取脾细胞总RNA,invitrogen cDNA反转录试剂盒进行反转录,合成cDNA,利用小鼠轻重链可变区简并引物扩增抗体基因,构建到噬菌体展示载体中,构建噬菌体抗体库,利用thermo的全自动磁珠分选***进行噬菌体抗体库淘筛,利用噬菌体ELISA挑选能够结合重组人LIF蛋白的大肠杆菌克隆,测定抗体基因序列。进一步通过ELISA和细胞活性鉴定获得了一株抗体标记为P36-033,P36-033轻链及重链的全长基因序列分别如SEQ ID NO:55和SEQ ID NO:57所示,对应的P36-033抗体的轻链及重链的全长氨基酸序列分别如SEQ ID NO:54和SEQ ID NO:56所示;编码P36-033抗体轻链及重链的可变区基因序列分别如SEQ ID NO:72和SEQ ID NO:73所示,对应的P36-033抗体的轻链及重链的可变区的氨基酸序列分别如SEQ ID NO:82和SEQ ID NO:83所示;按Kabat编码规则获知P36-033抗体的LCDR1的氨基酸序列如SEQ ID NO:66所示,LCDR2的氨基酸序列如SEQ ID NO:67所示,LCDR3的氨基酸序列如SEQ ID NO:68所示,HCDR1的氨基酸序列如SEQ ID NO:69所示,HCDR2的氨基酸序列如SEQ ID NO:70所示,HCDR3的氨基酸序列如SEQ ID NO:71所示。
1.3阳性对照抗体5D8的表达纯化
根据专利文献WO 2018/115960A1的报道,5D8抗体为一株阻断LIF蛋白与其受体GP130结合的抗体,参考专利文献中的序列本发明合成了多条基因序列,进行不同的轻重链组合配对,构建为人IgG1形式的全长抗体,最终发现其中一组配对具有最好的LIF蛋白结合活性,同时能够阻断重组人LIF蛋白与人GP130蛋白的结合,经过细胞活性验证能够阻断重组人LIF蛋白对HCT116细胞的STAT3的磷酸化,因此本发明将其命名为5D8,用于后续实验中作为阳性对照抗体,经测定,编码5D8抗体重链及轻链的全长基因序列分别如SEQ ID NO:63和SEQ ID NO:65所示,对应的5D8抗体的重链及轻链的全长氨基酸序列分别如SEQ ID NO:62和SEQ ID NO:64所示;编码5D8抗体重链及轻链的可变区基因序列分别如SEQ ID NO:80和SEQ ID NO:81所示,对应的5D8抗体的重链及轻链的可变区的氨基酸序列分别如SEQ ID NO:78和SEQ ID NO:79所示。
实施例2 抗人LIF抗体与人LIF蛋白的结合试验
将重组人LIF蛋白稀释为1μg/ml,包被酶标板,每孔加入100μLl蛋白,4℃冰箱孵育过夜,第二天取出酶标板,弃去液体,PBST洗涤三次,加入含2%BSA的PBS室温封闭一个小时,PBST洗涤三次,加入不同浓度的抗人LIF抗体38E10E1C11、5D8、P36-033,室温孵育一个小时,弃去液体,PBST洗涤四次,加入HRP标记的山羊抗小鼠Fab抗体或山羊抗人FC抗体,室温孵育1个小时,弃去液体,PBST洗涤四次,加入TMB显色液,室温孵育10分钟,加入2mol/L浓硫酸终止显色,酶标仪读取450nm吸光值,结果详见图1。结果显示38E10E1C11和P36-033对人LIF蛋白的结合活性要强于5D8。
实施例3 抗人LIF抗体与鼠LIF蛋白的结合试验
将重组小鼠LIF(购自于ACRO Biosystems)蛋白稀释为1μg/mL,包被酶标板,每孔加入100μL蛋白,4℃冰箱孵育过夜,第二天取出酶标板,弃去液体,PBST洗涤三次,加入含2%BSA的PBS室温封闭一个小时,PBST洗涤三次,加入不同浓度的抗人LIF抗体38E10E1C11、5D8、P36-033,室温孵育1个小时,弃去液体,PBST洗涤四次,加入HRP标记的山羊抗小鼠Fab抗体或山羊抗人FC抗体,室温孵育1个小时,弃去液体,PBST洗涤四次,加入TMB显色液,室温孵育10分钟,加入2mol/L浓硫酸终止显色,酶标仪读取450nm吸光值,结果详见图2。结果显示38E10E1C11抗体对小鼠LIF蛋白没有结合活性。P36-033对小鼠LIF蛋白有结合活性,但结合活性要弱于5D8抗体。
实施例4 抗人LIF抗体与食蟹猴LIF蛋白的结合试验
将重组猴LIF蛋白(购自于Sino Biological)稀释为0.5μg/mL,包被酶标板,每孔加入100μL蛋白,4℃冰箱孵育过夜,第二天取出酶标板,弃去液体,PBST洗涤三次,加入含2%BSA的PBS室温封闭一个小时,PBST洗涤三次,加入不同浓度的抗人LIF抗体38E10E1C11、5D8、P36-033,室温孵育1个小时,弃去液体,PBST洗涤四次,加入HRP标记的山羊抗小鼠Fab抗体或山羊抗人Fc抗体,室温孵育1个小时,弃去液体,PBST洗涤四次,加入TMB显色液,室温孵育10分钟,加入2mol/L浓硫酸终止显色,酶标仪读取450nm吸光值,结果详见图3。结果显示38E10E1C11和P36-033的结合活性好于5D8。
实施例5 抗人LIF抗体亲和力测定
用KinExA 4000测定纯化单克隆抗体与重组人LIF蛋白亲和力,向200mg PMMA hard beads中加入30ug 38E10E1C11抗体,补加包被溶液至1.0mL。缓冲液组成为1×PBS,pH7.4,0.02%NaN3。确保珠子完全悬浮在溶液中。室温旋转珠子2小时。珠子自然沉降或低速短暂离心,去上清,用含1%BSA的PBS封闭1小时。准备15mL 300pM抗原溶液和15mL 240pM Ab2(38E10E1C11)溶液。取0.6mL 300pM抗原和0.6mL 240pM抗体Ab2(38E10E1C11)各自放入不同的样品管中。 将两个管中的样品充分混匀,合到一管,此时抗原的浓度为150pM,抗体Ab2(38E10E1C11)的浓度为120pM,放在管架中相应的位置。准备16组,每组的样品孵育时间不同。加入1μg/ml Streptavidin Protein,DyLight 650溶液,上机检测,设置孵育时间为24小时。在KinExatm Pro软件中,用未知配体模型计算平衡离解常数(Kd)进行n-曲线分析,结果详见图4。结果显示38E10E1C11单抗与人LIF蛋白结合的亲和力为4.52x10 -12M。
实施例6 P36-033、38E10E1C11mAb与LIFR竞争结合人LIF蛋白
将重组人LIF蛋白按照1μg/mL的浓度包被酶标板,加入0.6125μg/mL的LIFR(与人F C融合表达)蛋白50uL/孔,同时加入不同浓度的LIF抗体38E10E1C11、P36-033和经过CHO细胞重组表达的38E10E1C11R(SEQ ID NOs:41and 43),50μL/孔,室温孵育2小时,PBST洗涤四次后,加入HRP标记的羊抗人F C二抗,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图5,结果显示38E10E1C11mAb和38E10E1C11R能够抑制人LIF与人LIFR的结合,而P36-033不能抑制人LIF与人LIFR的结合。
实施例7 P36-033、38E10E1C11mAb与GP130竞争结合人LIF蛋白
将重组人LIF蛋白按照1μg/mL的浓度包被酶标板,加入20μg/mL的GP130(与人Fc融合表达)蛋白50μL/孔,同时加入不同浓度的P36-033抗体或不同浓度的38E10E1C11抗体,50μL/孔,同时设置只加抗体不加GP130的对照孔,室温孵育2小时,PBST洗涤四次后,加入HRP标记的羊抗人Fc二抗,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图6,结果显示P36-033和38E10E1C11能够抑制人LIF与人GP130的结合。
实施例8 38E10E1C11mAb特异性检测
将人LIF、人IL-6、人OSM、人CNTF(均购自于Sino Biological)按照1μg/mL浓度包被酶标板,加入不同浓度的LIF抗体38E10E1C11、P36-033、5D8室温孵育1小时,PBST洗涤四次后,加入HRP标记的羊抗鼠Fab二抗,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图7,结果显示38E10E1C11单抗、P36-033单抗仅与人LIF蛋白结合而与人IL-6、OSM、CNTF没有结合,5D8抗体除结合人LIF蛋白外还与人OSM及人CNTF蛋白结合。
实施例9 38E10E1C11mAb能够用于人LIF蛋白的western blot检测
将重组人LIF蛋白稀释为图8中所示浓度,或将培养三天的CT26和CT26-hLIF细胞上清,加入5XSDS-PAGE上样缓冲液,煮沸10分钟,然后取10μL样品进行SDS-PAGE电泳,然后将电泳 条带转移到PVDF膜上进行western blot检测,检测用的一抗为38E10E1C11抗体,抗体浓度为1μg/mL,室温孵育2小时,TBST缓冲液洗涤三次后加入1:3000稀释的HRP标记的羊抗小鼠二抗,室温孵育2小时,室温孵育1小时,TBST缓冲液洗涤三次后,加入增强型化学发光液(Perice)孵育,利用Amersham Imager 600超灵敏多功能成像仪进行检测和拍照。结果详见图8,结果显示38E10E1C11mAb能够用于人LIF蛋白的western blot检测。
实施例10 抗人LIF抗体细胞活性检测
10.1 HCT116细胞STAT3激活抑制试验检测
HCT116细胞消化并离心后,重悬细胞,以1mL,5x10 5个细胞/孔,铺12孔板。37℃,5%CO 2过夜培养。第二天弃去原培养基,加入含有100ng/mL重组人LIF蛋白和不同浓度抗LIF抗体的细胞培养基,设置不加重组人LIF蛋白和只加重组人LIF蛋白不加抗体的对照孔,37℃培养箱孵育30min。然后将培养基取出,向12孔板中每孔加100uL 1x裂解液,冰上裂解30min。将裂解液转移到1.5mL离心管中,13,000rpm,10min离心,收集上清液。取上清液进行western blot检测STAT3的磷酸化情况。结果详见图9中的子图A和图11,由结果可知38E10E1C11抗体和P36-033抗体能够抑制人LIF蛋白诱导的HCT116细胞中STAT3的磷酸化。
10.2 KP4细胞STAT3激活抑制试验检测LIF抗体活性
KP4细胞消化并离心后,重悬细胞,以1mL,5x10 5个细胞/孔,铺12孔板。37℃,5%CO 2过夜培养。第二天弃去原培养基,加入含有50ng/mL重组人LIF蛋白和不同浓度抗LIF抗体的细胞培养基,设置不加重组人LIF蛋白和只加重组人LIF蛋白不加抗体的对照孔,37℃培养箱孵育30min。然后将培养基取出,向细胞中加入1x细胞裂解液,每孔100uL,冰上裂解30min。将裂解液转移到1.5mL离心管中,13,000rpm,10min离心,收集上清液。取上清液进行western blot检测STAT3的磷酸化情况。图9中的子图B结果显示38E10E1C11抗体能够抑制人LIF蛋白诱导的KP4细胞中STAT3的磷酸化。
10.3 KP4细胞STAT3激活抑制试验检测LIF抗体活性
KP4细胞消化并离心后,重悬细胞,以1mL,5x10 5个细胞/孔,铺12孔板。37℃,5%CO 2过夜培养。第二天弃去原培养基,将不同浓度抗LIF抗体和CT26-hLIF细胞培养基按体积比1:1配置后加入细胞中,设置CT26培养上清的对照孔,37℃培养箱孵育30min。然后将培养基取出,向细胞中加入1x细胞裂解液,每孔100uL,冰上裂解30min。将裂解液转移到1.5mL离心管中,13,000rpm,10min离心,收集上清液。取上清液进行western blot检测STAT3的磷酸化情况。图10显示本发明38E10E1C11抗体阻断CT26-hLIF细胞分泌的人LIF对于胰腺癌细胞KP4的STAT3的激活。
10.4 M1细胞增殖实验检测LIF抗体活性
M1离心后,RPMI1640培养基洗涤两次,按照2x10 5/mL的密度接种96孔板,每孔接种100ul细胞,加入含有10ng/mL重组人LIF蛋白和不同浓度抗LIF抗体的培养基,使每孔的最终体积为200uL,同时设置不加LIF的对照孔,37℃培养箱培养72小时,加入CCK-8检测细胞增殖情况。结果详见图12,结果显示单抗38E10E1C11和单抗P36-033都能够逆转人LIF蛋白对M1细胞的增殖抑制。
实施例11 抗人LIF抗体体内抗肿瘤活性检测
经过ELISA检测38E10E1C11抗体与小鼠LIF蛋白没有交叉反应,为进行体内活性评价,需构建过表达人LIF蛋白的CT26细胞株,根据文献报道人LIF蛋白能够结合小鼠细胞表面的LIFR和GP130从而激活下有信号,因此推测高表达人LIF的CT26分泌的人LIF蛋白能够抑制小鼠的免疫***,加入抗LIF蛋白的抗体能够解除这种抑制效果,从而发挥抗肿瘤作用。
11.1过表达人LIF基因的CT26细胞株的建立
将构建好的含有人LIF基因的慢病毒感染小鼠结肠癌细胞株CT26,感染48小时候检测LIF蛋白表达情况,有限稀释法对细胞株进行克隆化,加入终浓度为1μg/mL puromycin的培养基进行加压筛选,最终得到高表达人LIF蛋白的CT26稳定细胞株CT26-hLIF。
11.2 CT26-hLIF BABL/C皮下种植模型检测抗人LIF抗体的抗肿瘤活性
CT26-hLIF细胞培养在含10%胎牛血清的RPMI-1640培养液中,收集对数生长期的细胞,PBS重悬至10 7个细胞/mL后接种BABL/c小鼠皮下。接种一天后对小鼠进行分组,分别注射溶媒对照,抗人LIF抗体,给药浓度为15mg/kg体重,每周给药两次,连续给药4周,每周测量肿瘤体积两次,绘制肿瘤生长曲线,计算肿瘤抑制率。结果详见图13,结果显示单抗38E10E1C11能够抑制CT26-hLIF细胞在BABL/c小鼠体内增殖。
11.3测试不同胰腺癌细胞株对于LIF蛋白刺激的敏感性
将人胰腺癌细胞株Panc02.03、KP4、MIA paca2接种6孔板,每孔接种10 6个细胞,过夜培养后更换新鲜培养基,加入50ng/mL重组人LIF蛋白和抗人LIF抗体38E10E1C11,同时设置不加LIF的对照孔,37℃培养箱孵育30min,然后移除培养基,向细胞中加入1x细胞裂解液,每孔200uL,冰上裂解30min。将裂解液转移到1.5mL离心管中,13,000rpm,10min离心,收集上清液。取上清液进行western blot检测STAT3的磷酸化情况。结果详见图14,由结果可知,KP4细胞株对于人LIF蛋白刺激最为敏感。
实施例12 38E10E1C11单克隆抗体和P36-033单克隆抗体的重组表达及验证
将38E10E1C11和P36-033单克隆抗体的轻重链抗体基因利用同源重组技术构建到真核表达载体PCDNA3.1+中,利用thermo的ExpiCHO表达***表达重组抗体,利用Protein G亲和层析纯化 抗体,利用常州天地人和公司的Endotoxin Removal Beads对纯化的抗体进行内毒素去除。具体实验方法参照实施例10.2,图15显示经CHO细胞重组表达的38E10E1C11抗体(标注为38E10E1C11R)能够抑制人LIF蛋白诱导的KP4细胞中STAT3的磷酸化。
实施例13 38E10E1C11抗体识别表位的鉴定
前期实验证实38E10E1C11(SEQ ID NOs:41和43)抗体识别的是人LIF蛋白(SEQ ID NO:58)表面的线性表位,此抗体不能识别小鼠LIF蛋白,能够阻断人LIF蛋白与人LIFR蛋白的结合,根据此三点提示,同时结合多种在线蛋白线性表位预测软件分析,推测此抗体的识别表位位于人LIF蛋白160-202这段氨基酸序列中,于是合成了如下的杂合LIF蛋白,Mut3(SEQ ID NO:59)为将人LIF蛋白的182-202位点的氨基酸序列替换为小鼠LIF蛋白氨基酸序列,Mut4(SEQ ID NO:60)为将人LIF蛋白的166-202位点的氨基酸序列替换为小鼠LIF蛋白氨基酸序列,将含有Mut3,Mut4杂合蛋白以及全长人LIF蛋白的质粒转染293T细胞,培养三天后取细胞上清进行SDS-PAGE电泳和转膜,以293T细胞培养上清作为阴性对照,使用38E10E1C11作为一抗,HRP标记的羊抗小鼠Fab作为二抗进行检测,同时利用M1细胞增殖实验检测杂合蛋白的活性和验证38E10E1C11对杂合蛋白的中和活性。同时设置多组对照孔,加入人重组LIF蛋白(rhLIF,购自于义翘神州,货号分别为:14890-HNAH)的对照孔,加入rhLIF和38E10E1C11的对照孔,不加rhLIF和不加LIF抗体的对照孔。结果显示38E10E1C11抗体能够识别变性全长LIF蛋白,Mut3蛋白,但不能识别Mut4蛋白(图16中的子图A),M1细胞增殖实验显示38E10E1C11抗体能够逆转全长LIF蛋白和Mut3蛋白对M1细胞增值的抑制而不能逆转Mut4蛋白的抑制作用(图16中的子图B),综上所述38E10E1C11抗体的识别表位位于LIF蛋白的的167-181位氨基酸之中,即TYGPDTSGKDVFQKK这段氨基酸之中(SEQ ID NO:61)
实施例14人源化抗LIF抗体设计及表达纯化
将从小鼠免疫得到的单克隆抗体38E10E1C11进行人源化。采用标准CDR嫁接方法进行人源化。用标准分子克隆技术从38E10E1C11杂交瘤中克隆重链区和轻链区,并用Sanger法进行测序。然后对人类重链和轻链可变序列进行BLAST搜索,并从每个序列中选择3或4个序列作为人源化的受体框架。将38E10E1C11的重链和轻链CDR1、CDR2和CDR3克隆到3个不同的重链受体框架(H1-H3)和4个不同的轻链框架(L1-L4)中,同时对38E10E1C11的HCDR2(突变前的氨基酸序列如SEQ ID NO:45所示)进行点突变(突变后的氨基酸序列如SEQ ID NO:5所示),重链恒定区选择人IgG1亚型,轻链恒定区选择人Kappa链,将含有人源化抗体重链和人源化抗体轻链基因的表达载体共转染293S细胞,上述所获得的人源化抗LIF抗体的重轻链可变区基因序列、可变区氨基酸序列、全长基因序列以及全长氨基酸序列详见表1,然后检测12种不同的抗体组合在293S细胞 中的表达水平,抗原结合能力和热稳定性。以38E10E1C11嵌合抗体(Chimeric)作为阳性对照,后续试验中38E10E1C11嵌合抗体均简称为38E嵌合抗体或38E Chimeric(SEQ ID NO:52and SEQ ID NO:50)。收集培养基,在Gator(类似于Octet)上定量其中IgG的表达水平,并用ELISA进行修正。用ELISA的方法比较不同组合的抗原结合能力(表2,表3)。
酶联免疫吸附试验(ELISA):
在每个孔包被100μL 0.5μg/ml抗原,4℃孵育过夜,用300μL洗涤缓冲液洗涤板三次。用200μL封闭缓冲液(2%牛血清白蛋白)在室温下封闭60分钟。用300μL洗涤缓冲液洗涤板三次。加入100μL不同浓度的稀释抗LIF抗体每孔100μL,室温孵育1小时。用300μL洗涤缓冲液洗涤板4次。加入100μL 1:5000稀释的HRP标记的羊抗人Fc二抗,室温孵育1小时。用300μL清洗缓冲液清洗板6次。在黑暗条件下,在室温下加入100μL H 2O 2-Amplx显色液显色10分钟。酶标仪读取OD 450值。热处理:表达培养基在70℃下在PCR机上加热5分钟,然后快速冷却到室温。进行后续ELISA检测,方法同上述。
表1.不同人源化轻重链组合在293S细胞中表达水平
Figure PCTCN2020118247-appb-000031
Figure PCTCN2020118247-appb-000032
Figure PCTCN2020118247-appb-000033
表2.不同人源化轻重链组合经293S细胞表达上清的ELISA结果(非加热)
Figure PCTCN2020118247-appb-000034
表3.不同人源化轻重链组合经293S细胞表达上清的ELISA结果(加热)
Figure PCTCN2020118247-appb-000035
实施例15用纯化的IgG样品表征所选的人源化候选抗体
基于结合亲和力、人源化百分比、抗体表达水平和热稳定性数据,本发明选择了以下5个候选抗体进行下一步表征:H1L1、H1L4、H2L4、H3L2、H3L4,并将这5个候选抗体重新编号为38E HuH1L1(SEQ ID NOs:25和9)、38E HuH1L4(SEQ ID NOs:25和21)、38E HuH2L4(SEQ ID  NOs:29和21)、38E HuH3L2(SEQ ID NOs:33和13)、38E HuH3L4(SEQ ID NOs:33和21),然后将所选的VH/VL质粒共转染293S细胞,收获细胞培养上清,protein A亲和层析纯化抗体。用纯化的抗体进行结合ELISA分析,比较人源化抗体与38E嵌合抗体的特异结合能力。本发明还进行了一些初步分析,以比较它们的热稳定性和非特异性结合。结果表明,用纯化的候选抗体与38E嵌合抗体具有非常相似的抗原结合特性(图17A,17C)。在70℃处理5分钟后,5个人源化抗体均表现出与嵌合抗体相似的结合能力(图17B,17D)。
实施例16评价人源化候选抗体的非特异性结合
采用LIF阴性的HEK293细胞FACS作为初步检测,以评估抗体潜在的非特异性结合风险。
将HEK293细胞用胰酶消化后,用含1%FBS的PBS洗涤两次,重悬细胞,调整细胞密度为1.5-2x10 6个/mL,将细胞加入到96孔U形板中。将待检测的抗体浓度调整为20μg/mL,然后进行3倍倍比稀释,共稀释8个浓度,设置空白对照和阴性对照(Rituxan)。将稀释好的抗体和空白对照加入到96孔板中的细胞中,每孔加入100μL抗体。4℃孵育1小时,1000rpm离心5分钟,小心弃去上清,用含1%FBS的PBS洗涤两次,最终用200μL含1%FBS的PBS重悬细胞,进行流式细胞分析。在HEK293细胞非特异性结合FACS检测中,38E HuH1L1、38E HuH3L2、38E HuH3L4、38E HuH1L4、38E嵌合抗体和阴性对照(Rituxan)对HEK293细胞具有相似的非特异性结合亲和力,而38E HuH2L4对HEK293细胞具有更高的非特异性结合(图18中的子图A和图18中的子图B)。
实施例17 CE-SDS分析抗体纯度
CE-SDS分析的工作浓度为1mg/mL,将抗体样品用上样缓冲液稀释到指定浓度。
非还原CE-SDS电泳样品的制备:取稀释好的样品溶液95μL,加入0.8M碘乙酸铵水溶液5μL,5μL内参,涡旋混匀。取空白对照95μL,加入0.8M碘乙酸铵水溶液5μL,5μL内参,涡旋混匀,为非还原空白对照。70℃,金属浴加热5分钟,冷却至室温,6000rpm离心1分钟。
还原样品溶液制备:取稀释好的样品溶液95μL,加入2-巯基乙醇溶液5μL,内参5μL,涡旋混匀。取空白对照95μL,加入2-巯基乙醇溶液5μL,内参5μL,涡旋混匀,为还原空白对照。70℃,金属浴加热15分钟,冷却至室温,6000rpm离心1分钟。
样品分析:取75μL样品加入到测试管中,将测试管放入测试杯中,测试杯小心***进样盘中,运行测试程序,还原样品进样时长为30秒,非还原样品进样时长为40秒,毛细管温度为20℃,样品温度为20℃,聚焦电压为15KV,聚焦时间为40分钟,用PDA检测器214nm波长采集数据。CE结果表明见表4,表5。
表4.还原CE-SDS结果
Figure PCTCN2020118247-appb-000036
表5.非还原CE-SDS结果
  Size(KDa) Peak Area(%) Peak ID
38E chimeric 166.39 >99 intact AB
38E HuH1L1 165.93 92.46 intact AB
38E HuH3L2 167.63 93.01 intact AB
38E HuH3L4 166.37 98.82 intact AB
实施例18用差示扫描荧光法(DSF)/静态光散射技术(SLS)分析热稳定性
样品被提交给UNcle***(Unchained Labs)进行分析。在25℃至95℃的温度范围内,对DSF和SLS进行1℃/min的升温监测。UNcle在266nm和473nm处测量SLS。利用UNcle分析软件对Tm和Tagg进行了计算和分析。
IgG是具有多个结构域,每个结构域都有自己的熔化温度(T m)。CH2结构域在PBS中通常有约70℃的Tm,CH3更稳定,其Tm约为80℃。Fabs由于其序列变化较大,Tm的范围较大,约为50-85℃。因此,通过各种分析技术测量的Tm值通常是“表观”转变温度,而不是每个结构域的真实Tm值。很明显,即使这种DSF分析也能产生一个以上的Tm值,只有Tm1才用于评价治疗用抗体的热稳定性。Tagg是SLS开始检测聚集的温度。Tagg266在266nm处测量SLS,它更敏感,更适合于检测较小的聚集粒子。Tagg473在473nm处测量SLS,更适合于检测较大的粒子。
如表6所示,三种人源化候选抗体均比38E嵌合抗体具有更高的融解温度(Tm1)和更小的聚集风险
表6.差示扫描荧光法(DSF)/静态光散射技术(SLS)分析结果
Figure PCTCN2020118247-appb-000037
实施例19用动态光散射技术(DLS)分析抗体的聚集倾向:
DLS是在UNcle***(Unchained Labs)上执行的。在25℃下测量DLS。使用UNcle分析软件计算和分析数据。动态光散射(DLS)用于检测抗体样品中的聚集。“模式直径”是指蛋白质颗粒直径,“质量百分比”是指每种粒径分数的百分比。“PDI”是多分散指数,该指数越高,样品的多分散性越强。如果PDI不大于0.25,则样品可视为单分散。如表7所示,所有4个抗体样本都有一个主“峰”(质量分数超过99%),38E HuH3L4的PDI优于嵌合抗体,38E HuH3L2与嵌合抗体相似,38E HuH1L1的PDI比嵌合抗体差。
表7.动态光散射技术(DLS)分析结果
Figure PCTCN2020118247-appb-000038
实施例20抗体亲和力测定
利用Gator测定抗LIF抗体与人LIF蛋白的亲和力。首先将抗人LIF抗体用PBS稀释至5ug/mL,然后分别加入96孔板的第二列A-F孔(每孔200μL);以PBS梯度稀释人LIF蛋白浓度分别为100、50、25、12.5、6.25μg/mL,分别加入到96孔板第四列的A-E孔(每孔100μL),F孔加PBS作为空白对照;第一列与第三列的A-F孔均加入PBS(每孔200μL)。将96孔板放入到仪器中,用抗人Fc生物传感器进行检测。实验结果如表8所示,结果显示3株人源化抗体与嵌合抗体亲和力接近。
表8.人源化抗LIF抗体亲和力测定
Sample Koff(1/s) Kon(1/Ms) KD(M)
38E HuH1L1 4.65E-05 5.69E+05 8.17E-11
38E HuH3L2 3.39E-05 6.31E+05 5.38E-11
38E HuH3L4 4.44E-05 5.79E+05 7.66E-11
38E chimeric 1.57E-05 6.77E+05 2.33E-11
实施例21人源化抗体和38E10E1C11抗体的表达纯化
将人源化抗体38E HuH3L2和38E HuH3L4的轻重链的可变区与小鼠抗体恒定区(重链恒定区为小鼠IgG1,轻链恒定区为kappa链)连接分别克隆到PCDNA.3.4载体中,分别命名为38E HuH3L2-m(编码38E HuH3L2-m抗体重链及轻链的全长基因序列分别如SEQ ID NO:36和SEQ ID NO:38所示,对应的38E HuH3L2-m抗体的重链及轻链的全长氨基酸序列分别如SEQ ID NO:35和 SEQ ID NO:37所示)和38E HuH3L4-m(编码38E HuH3L4-m抗体重链及轻链的全长基因序列分别如SEQ ID NO:36和SEQ ID NO:40所示,对应的38E HuH3L4-m抗体的重链及轻链的全长氨基酸序列分别如SEQ ID NO:35和SEQ ID NO:39所示)利用thermo fisher公司的Expi CHO表达试剂盒进行基因的转染和抗体表达,收集细胞培养上清,利用protein G亲和层析柱对抗体进行纯化,纯化后的抗体利用
Figure PCTCN2020118247-appb-000039
Ultra超滤管进行超滤浓缩和换液,最终将抗体溶解在pH7.4的PBS中。38E10E1C11抗体也利用相同的方式表达纯化。
实施例22人源化抗LIF抗体与LIFR竞争结合人LIF蛋白
将重组人LIF蛋白按照1μg/mL的浓度包被酶标板,加入0.6125μg/mL的重组人LIFR(与人Fc融合表达,购自于ACRO,货号:LIR-H4252)蛋白50μL/孔,同时加入不同浓度的LIF抗体38E HuH3L2-m(SEQ ID NOs:35and 37)、38E HuH3L4-m(SEQ ID NOs:35and 39)、38E10E1C11(SEQ ID NOs:41and 43)、P36-033(SEQ ID NOs:54and 56)100μL/孔,以anti-CD3抗体作为阴性对照(购自于BioLegend,货号:317326)室温孵育2小时,PBST洗涤四次后,加入HRP标记的羊抗人Fc抗体,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图19,结果显示38E10E1C11、38E HuH3L2-m、38E HuH3L4-m能够抑制重组人LIF与人LIFR的结合,IC 50分别为0.074μg/ml、0.145μg/ml、0.103μg/ml。P36-033具有微弱的抑制作用,阴性对照anti-CD3抗体不能抑制重组人LIF与人LIFR的结合。
实施例23人源化抗LIF抗体不与GP130竞争结合人LIF蛋白
将重组人LIF蛋白按照1μg/mL的浓度包被酶标板,加入12μg/mL的重组人GP130(与人Fc融合表达,购自于义翘神州,货号:10974-H03H)蛋白50μL/孔,同时加入不同浓度的LIF抗体38E HuH3L2-m((SEQ ID NOs:35and 37)、38E HuH3L4-m(SEQ ID NOs:35and 39)、P36-033(SEQ ID NOs:56and 54)100μL/孔,以anti-CD28抗体作为阴性对照(购自于BioLegend,货号:302914),室温孵育2小时,PBST洗涤四次后,加入HRP标记的羊抗人Fc抗体,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图20,结果显示人源化抗体38E HuH3L2-m、38E HuH3L4-m和阴性对照抗体anti-CD28抗体不能抑制重组人LIF与人GP130蛋白的结合,P36-033能够抑制重组人LIF与人GP130蛋白的结合。
实施例24人源化抗LIF抗体抗原识别特异性检测
将人LIF、人IL-6、人OSM、人CNTF(四种蛋白均购自于义翘神州,货号分别为:14890-HNAH;10395-HNAE;10452-HNAH;11841-H07E)按照1μg/mL浓度包被酶标板,加入不同浓度的LIF 抗体38E10E1C11、38E huH3L2-m、38E huH3L4-m室温孵育1小时,PBST洗涤四次后,加入HRP标记的羊抗小鼠Fab二抗,室温孵育1小时,PBST洗涤四次后,加入TMB显色液室温显色10分钟,酶标仪读取450nm吸光值,数据采用Origin pro 9软件进行分析和作图。结果详见图21,结果显示38E10E1C11、38E huH3L2-m、38E huH3L4-m抗体仅与人LIF蛋白结合而与人IL-6、OSM、CNTF没有结合。
实施例25人源化抗LIF抗体识别表位的鉴定
在前期实验中本发明发现38E10E1C11抗体识别的是LIF蛋白的线性表位,因此本发明首先验证38E人源化抗体是否识别的仍然是LIF蛋白的线性表位。取转染人LIF全长基因序列,Mut3和Mut4蛋白序列的293T细胞培养3天后的上清,阴性对照为293T细胞培养上清,加入5xSDS-PAGE上样缓冲液,煮沸10分钟,然后取10μL样品进行SDS-PAGE电泳,然后将电泳条带转移到PVDF膜上进行western blot检测,检测用的一抗为38E huH3L2或38E huH3L4抗体,抗体浓度为1μg/mL,室温孵育2小时,PBST缓冲液洗涤三次后加入1:3000稀释的HRP标记的羊抗人Fc二抗,室温孵育2小时,PBST缓冲液洗涤三次后,加入增强型化学发光液(Perice,货号为:34079)孵育,利用Amersham Imager 600超灵敏多功能成像仪进行检测和拍照。结果见图22中的子图A,结果显示两株人源化抗体均能识别变性的人LIF蛋白和Mut3蛋白而不能识别Mut4蛋白。M1细胞增值实验也得到相同的结果,结果详见图22中的子图B。因此确定38E huH3L2和38E huH3L4抗体识别的表位序列为TYGPDTSGKDVFQKK(SEQ ID NO:61)。
实施例26 KP4细胞STAT3激活抑制试验检测人源化抗LIF抗体活性
KP4细胞消化并离心后,重悬细胞,以1mL,5x10 5个细胞/孔,铺12孔板。37℃,5%CO 2过夜培养。第二天弃去原培养基,加入含有50ng/mL重组人LIF蛋白和不同浓度抗LIF人源化抗体的细胞培养基,设置不加重组人LIF蛋白和只加重组人LIF蛋白不加抗体的对照孔,37℃培养箱孵育30min。然后将培养基取出,向细胞中加入1x细胞裂解液,每孔100μL,冰上裂解30min。将裂解液转移到1.5mL离心管中,13,000rpm,10min离心,收集上清液。取上清液进行western blot检测STAT3的磷酸化情况。结果显示人源化抗体38E huH3L4和38E huH3L2能够抑制LIF蛋白诱导的STAT3的磷酸化(图23)。
实施例27 M1细胞增殖实验检测人源化抗LIF抗体的活性
M1离心后,RPMI1640培养基洗涤两次,按照2.5x10 5个/mL的密度接种96孔板,每孔接种80μL细胞,加入含有4ng/mL重组人LIF蛋白和不同浓度抗LIF抗体的培养基,使每孔的最终体积为160μL,同时设置不加LIF的对照孔,37℃培养箱培养72小时,加入CCK-8检测细胞增殖情况。结果详见图24,结果显示人源化抗体38E huH3L4-m和38E huH3L2-m都能够逆转人LIF蛋白对M1细胞的增殖抑制,EC50分别为6.52μg/mL和8.93μg/mL。
实施例28:LIF抗体对LIF诱导的STAT3磷酸化的抑制作用
接种100,000个KP4细胞于96孔板中;倍比稀释的LIF抗体与20-100ng/ml LIF蛋白室温孵育0.5-1h,形成LIF-Ab混合溶液;加入LIF-Ab混合溶液于细胞孔中,37℃刺激5-30min;按P-STAT3(TYR705)KITS(Cisbio)和Total-STAT3KITS(Cisbio)说明书检测P-STAT3和Total-STAT3蛋白表达水平,并计算每个孔的供体和受体的发射信号比:Ratio=Signal 665nm/Signal 620nm*10 4。用GraphPad Prism软件生成数据图并统计LIF抗体的抑制率。结果显示LIF抗体38E HuH3L4对LIF诱导STAT3磷酸化有抑制作用,如图25所示,IC 50为3.415nM。
实施例29:检测LIF抗体的ADCC活性
LIF与GP130和LIFR结合,人源化LIF抗体阻断LIF与LIFR结合,但不能阻断LIF与GP130结合。检测人源化LIF抗体是否通过LIF介导,结合至细胞表面,从而发生ADCC作用。用ADCC缓冲液(RPMI-1640+1%FBS)依次稀释抗体Erbitux(
Figure PCTCN2020118247-appb-000040
Merck Serono,阳性对照)和Human IgG2(Cat#HG2K,Sino,阴性对照);再用含LIF蛋白的ADCC缓冲液稀释38E huH3L4抗体,三倍稀释,八个梯度,备用;用胰酶(Cat#25200072,GIBCO)消化DLD-1细胞,终止反应后吹散细胞收集到离心管中,1500rpm离心3min,弃上清,用ADCC缓冲液重悬细胞并计数,调整细胞浓度,备用;复苏PBMC细胞(Cat#SLB-HP010B,上海赛笠生物科技有限公司),加入10mL ADCC缓冲液,2000rpm离心10min,弃上清,重悬至ADCC缓冲液中计数,调整细胞浓度,备用;取出96孔U底细胞培养板,依次加入50μl靶细胞DLD-1、50μl抗体、50μl效应细胞PBMC,效靶比=30:1,混匀置于37℃5%CO 2孵箱反应6h,用Cyto Tox96Non-Radioactive Cytotoxicity Assay试剂盒(Cat#G1780,Promega)检测LDH,用酶标仪在490nm测量吸光值。
计算各平行孔吸光值均值,使所有实验孔、靶细胞LDH自发释放孔(TCR)、效应细胞LDH自发释放孔(ECR)的吸光值均值减去空白培养基(CMB)的吸光值均值。靶细胞LDH最大释放孔(TCM)的吸光值均值减去体积校正对照孔(VCC)的吸光值均值。用上述校正后的值按照以下公式,计算各浓度抗体产生的Cytotoxicity(%):
Cytotoxicity(%)=(A–B–C)/(D–C)×100%
A:实验孔校正后的平均吸光值
B:效应细胞LDH自发释放孔校正后的平均吸光值
C:靶细胞LDH自发释放孔校正后的平均吸光值
D:靶细胞LDH最大释放孔校正后的平均吸光值
如图26所示,38E huH3L4抗体无ADCC活性。
参考文献
1.Nicola NA,Babon JJ.Leukemia inhibitory factor(LIF).Cytokine Growth Factor Rev.2015;26(5):533-44.
2.Pastuschek J,Poetzsch J,Morales-Prieto DM,Schleuβner E,Markert UR,Georgiev G.Stimulation of the JAK/STAT pathway by LIF and OSM in the human granulosa cell line COV434.J Reprod Immunol.2015;108:48-55.
3.Liu SC,Tsang NM,Chiang WC,Chang KP,Hsueh C,Liang Y,Juang JL,Chow KP,Chang YS.Leukemia inhibitory factor promotes nasopharyngeal carcinoma progression and radioresistance.J Clin Invest.2013;123(12):5269-83.
4.Shi Y,Gao W,Lytle NK,Huang P,Yuan X,Dann AM,et al.Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring.Nature.2019;569(7754):131-135.
5.Cartwright P,McLean C,Sheppard A,RivettD,Jones K,Dalton S.LIF/STAT3controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism.Development 2005;132:885–96.
6.Kuphal S,Wallner S,Bosserhoff AK.Impact of LIF(leukemia inhibitory factor)expression in malignant melanoma.Exp Mol Pathol 2013;95:156–65.
7.Liu B,Lu Y,Li J,Liu Y,Liu J,WangW.Leukemia inhibitory factor promotes tumor growth and metastasis inhuman osteosarcoma via activating STAT3.APMIS 2015;123:837–46.
8.Morton SD,Cadamuro M,Brivio S,Vismara M,Stecca T,Massani M,et al.Leukemia inhibitory factor protects cholangiocarcinoma cells from drug induced apoptosis via a PI3K/AKT-dependent Mcl-1activation.Oncotarget.2015;6:26052–64.
9.Kamohara H,Ogawa M,Ishiko T,Sakamoto K,Baba H.Leukemia inhibitory factor functions as a growth factor in pancreas carcinoma cells:involvement of regulation of LIF and its receptor expression.Int J Oncol.2007;30:977–83.
10.Shin JE,Park SH,Jang YK.Epigenetic up-regulation of leukemia inhibitory factor(LIF)gene during the progression to breast cancer.Mol Cells 2011;31:181–9.
11.Li X,Yang Q,Yu H,Wu L,Zhao Y,Zhang C,Yue X,Liu Z,Wu H,Haffty BG,Feng Z,Hu W.LIF promotes tumorigenesis and metastasis of breast cancer through the AKT-mTOR pathway.Oncotarget.2014;5(3):788-801.
12.Liu SC,Hsu T,Chang YS,Chung AK,Jiang SS,OuYang CN,Yuh CH,Hsueh C,Liu YP,Tsang NM.Cytoplasmic LIF reprograms invasive mode to enhance NPC dissemination through modulating YAP1-FAK/PXN signaling.Nat Commun.2018;9(1):5105.
13.Viswanadhapalli S,Luo Y,Sareddy GR,Santhamma B,Zhou M,et al.EC359:A First-in-Class Small-Molecule Inhibitor for Targeting Oncogenic LIFR Signaling in Triple-Negative Breast Cancer.Mol Cancer Ther.2019;18(8):1341-1354.

Claims (121)

  1. 一种分离的抗体或其抗原结合部分,其中,所述分离的抗体或其抗原结合部分结合在人LIF蛋白的氨基酸序列TYGPDTSGKDVFQKK(SEQ ID NO:61)所示的表位或在不同哺乳动物物种的相应氨基酸序列的表位。
  2. 一种分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (a)LCDR1,该LCDR1包含选自以下所组成的组:SEQ ID NO:1或SEQ ID NO:66及其保守修饰形式的氨基酸序列;
    (b)LCDR2,该LCDR2包含选自以下所组成的组:SEQ ID NO:2或SEQ ID NO:67及其保守修饰形式的氨基酸序列;
    (c)LCDR3,该LCDR3包含选自以下所组成的组:SEQ ID NO:3或SEQ ID NO:68及其保守修饰形式的氨基酸序列;
    (d)HCDR1,该HCDR1包含选自以下所组成的组:SEQ ID NO:4或SEQ ID NO:69及其保守修饰形式的氨基酸序列;
    (e)HCDR2,该HCDR2包含选自以下所组成的组:SEQ ID NO:5、SEQ ID NO:45或SEQ ID NO:70及其保守修饰形式的氨基酸序列;及
    (f)HCDR3,该HCDR3包含选自以下所组成的组:SEQ ID NO:6或SEQ ID NO:71及其保守修饰形式的氨基酸序列。
  3. 根据权利要求2所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (a)LCDR1,该LCDR1包含选自以下所组成的组:SEQ ID NO:1及其保守修饰形式的氨基酸序列;
    (b)LCDR2,该LCDR2包含选自以下所组成的组:SEQ ID NO:2及其保守修饰形式的氨基酸序列;
    (c)LCDR3,该LCDR3包含选自以下所组成的组:SEQ ID NO:3及其保守修饰形式的氨基酸序列;
    (d)HCDR1,该HCDR1包含选自以下所组成的组:SEQ ID NO:4及其保守修饰形式的氨基酸序列;
    (e)HCDR2,该HCDR2包含选自以下所组成的组:SEQ ID NO:5或SEQ ID NO:45及其保守修饰形式的氨基酸序列;
    (f)HCDR3,该HCDR3包含选自以下所组成的组:SEQ ID NO:6及其保守修饰形式的氨基酸序列。
  4. 根据权利要求2或3所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    1)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3;
    2)(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3;或者
    3)(a)包含SEQ ID NO:66的LCDR1,(b)包含SEQ ID NO:67的LCDR2,(c)包含SEQ ID NO:68的LCDR3,(d)包含SEQ ID NO:69的HCDR1,(e)包含SEQ ID NO:70的HCDR2,以及(f)包含SEQ ID NO:71的HCDR3。
  5. 根据权利要求4所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
  6. 根据权利要求4所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
  7. 根据权利要求4所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (a)包含SEQ ID NO:66的LCDR1,(b)包含SEQ ID NO:67的LCDR2,(c)包含SEQ ID NO:68的LCDR3,(d)包含SEQ ID NO:69的HCDR1,(e)包含SEQ ID NO:70的HCDR2,以及(f)包含SEQ ID NO:71的HCDR3。
  8. 根据权利要求2-7中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (i)轻链可变区(VL),该轻链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:7、11、15、19、46、74或82及其保守修饰形式的氨基酸序列;以及
    (ii)重链可变区(VH),该重链可变区包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:23、27、31、48、75或83及其保守修饰形式的氨基酸序列。
  9. 根据权利要求2-8中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    (i)轻链可变区,该轻链可变区(VL)包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:7、11、15、19或46及其保守修饰形式的氨基酸序列;以及
    (ii)重链可变区,该重链可变区(VH)包含选自以下所组成的组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:23、27、31或48及其保守修饰形式的氨基酸序列。
  10. 根据权利要求2-9中任一项所述的分离的抗体或其抗原结合部分,其中,所述分离的抗体包括轻链可变区和重链可变区,所述的轻链可变区包含与选自权利要求8或9中(i)中的轻链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列;所述重链可变区包含与选自权利要求8或9中(ii)中的重链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
  11. 根据权利要求2-10中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    1)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
    2)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
    3)轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
    4)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
    5)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
    6)轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
    7)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
    8)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
    9)轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
    10)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列;
    11)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列;
    12)轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列;
    13)轻链可变区(VL),其包含与SEQ ID NO:46的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:48的氨基酸序列至少85%同源性的氨基酸序列;
    14)轻链可变区(VL),其包含与SEQ ID NO:74的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:75的氨基酸序列至少85%同源性的氨基酸序列;或者
    15)轻链可变区(VL),其包含与SEQ ID NO:82的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:83的氨基酸序列至少85%同源性的氨基酸序列。
  12. 根据权利要求2-11中任一项所述的分离的抗体或其抗原结合部分,其中,所述分离的抗体或其抗原结合片段包括轻链可变区以及重链可变区,所述轻链可变区及重链可变区分别包含与选自权利要求11中的1)-15)中轻链可变区及重链可变区至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
  13. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
  14. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
  15. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:7的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
  16. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
  17. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
  18. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:11的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
  19. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
  20. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
  21. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:15的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
  22. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:23的氨基酸序列至少85%同源性的氨基酸序列。
  23. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:27的氨基酸序列至少85%同源性的氨基酸序列。
  24. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:19的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:31的氨基酸序列至少85%同源性的氨基酸序列。
  25. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:46的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:48的氨基酸序列至少85%同源性的氨基酸序列。
  26. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:74的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:75的氨基酸序列至少85%同源性的氨基酸序列。
  27. 根据权利要求11所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链可变区(VL),其包含与SEQ ID NO:82的氨基酸序列至少85%同源性的氨基酸序列;以及重链可变区(VH),其包含与SEQ ID NO:83的氨基酸序列至少85%同源性的氨基酸序列。
  28. 根据权利要求2-27中任一项所述的分离的抗体或其抗原结合部分,所述的分离的抗体或其抗原结合部分包含轻链和重链,其中:
    (I)所述轻链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:9、13、17、21、37、39、50或者54及其保守修饰形式的氨基酸序列;以及
    (II)所述重链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:25、29、33、35、52或者56及其保守修饰形式的氨基酸序列。
  29. 根据权利要求2-28任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含轻链和重链,其中:
    (I)所述轻链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:9、13、17、21、37、39或者50及其保守修饰形式的氨基酸序列;以及
    (II)所述重链包含选自以下所组成的下组的氨基酸序列至少85%同源性的氨基酸序列:SEQ ID NOs:25、29、33、35、或者52及其保守修饰形式的氨基酸序列。
  30. 根据权利要求2-29任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含轻链和重链,所述的轻链包含与选自权利要求28或29中(I)中的轻链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列;所述的重链包含与选自权利要求28或29中(II)中的重链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
  31. 根据权利要求2-30中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    1)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
    2)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
    3)轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
    4)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
    5)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
    6)轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
    7)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
    8)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
    9)轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
    10)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列;
    11)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列;
    12)轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列;
    13)轻链,其包含与SEQ ID NO:37的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列;
    14)轻链,其包含与SEQ ID NO:39的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列;
    15)轻链,其包含与SEQ ID NO:50的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:52的氨基酸序列至少85%同源性的氨基酸序列;或者
    16)轻链,其包含与SEQ ID NO:54的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:56的氨基酸序列至少85%同源性的氨基酸序列。
  32. 根据权利要求2-31中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含轻链和重链,所述轻链及重链分别包含与选自权利要求31中1)-16)中轻链及重链至少90%、至少95%、至少96%、至少97%、至少98%、至少99%或者100%同源性的氨基酸序列。
  33. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
  34. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
  35. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:9的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
  36. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
  37. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
  38. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:13的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
  39. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
  40. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
  41. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:17的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
  42. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:25的氨基酸序列至少85%同源性的氨基酸序列。
  43. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:29的氨基酸序列至少85%同源性的氨基酸序列。
  44. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:21的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:33的氨基酸序列至少85%同源性的氨基酸序列。
  45. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:37的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列。
  46. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:39的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:35的氨基酸序列至少85%同源性的氨基酸序列。
  47. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:50的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:52的氨基酸序列至少85%同源性的氨基酸序列。
  48. 根据权利要求31所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:
    轻链,其包含与SEQ ID NO:54的氨基酸序列至少85%同源性的氨基酸序列;以及重链,其包含与SEQ ID NO:56的氨基酸序列至少85%同源性的氨基酸序列。
  49. 一种分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:5的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
  50. 一种分离的抗体或抗原结合部分,其中,所述的分离的抗体或其抗原结合部分包含:(a)包含SEQ ID NO:1的LCDR1,(b)包含SEQ ID NO:2的LCDR2,(c)包含SEQ ID NO:3的LCDR3,(d)包含SEQ ID NO:4的HCDR1,(e)包含SEQ ID NO:45的HCDR2,以及(f)包含SEQ ID NO:6的HCDR3。
  51. 一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:7所示的轻链可变区(VL),以及含有SEQ ID NO:23所示的重链可变区(VH)。
  52. 一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:11所示的轻链可变区(VL),以及含有SEQ ID NO:31所示的重链可变区(VH)。
  53. 一种分离的抗体或其抗原结合部分,其中,含有SEQ ID NO:19所示的轻链可变区(VL),以及含有SEQ ID NO:31所示的重链可变区(VH)。
  54. 根据权利要求1-53中任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体是IgG。
  55. 根据权利要求1-54中任一项所述的分离的抗体或其抗原结合部分,其中,所述分离的抗体是IgG1、IgG2或IgG4。
  56. 根据权利要求1-55任一项所述的分离的抗体或其抗原结合部分,其中,所述分离的抗体是单克隆抗体、嵌合抗体、人源化抗体、人改造抗体、人抗体、双特异性抗体、Fv、单链抗体(scFv)、Fab、Fab’、Fab’-SH或者F(ab’) 2
  57. 根据权利要求1-56任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分为LIF(leukemia inhibitory factor)拮抗剂。
  58. 根据权利要求1-56任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分能够抑制LIF表达和/或阻断LIF活性。
  59. 根据权利要求1-56任一项所述的分离的抗体或其抗原结合部分,其中,所述的分离的抗体或其抗原结合部分能够竞争或交叉竞争用于结合LIF。
  60. 一种核苷酸组合物,其包含编码权利要求1-59中任一项所述的分离的抗体或其抗原结合部分的核苷酸分子。
  61. 根据权利要求60所述的核苷酸组合物,其包含:
    (i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15、19、46或82所示的轻链可变区(VL)的DNA;以及
    (ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27、31、48或83所述的重链可变区(VH)的DNA。
  62. 根据权利要求60或61所述的核苷酸组合物,其包含:
    (i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15、19或46所示的轻链可变区(VL)的DNA;以及
    (ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27、31或48所述的重链可变区(VH)的DNA。
  63. 根据权利要求60-62中任一项所述的核苷酸组合物,其包含:
    (i)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:7、11、15或19所示的轻链可变区(VL)的DNA;以及
    (ii)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:23、27或31所述的重链可变区(VH)的DNA。
  64. 根据权利要求60-63中任一项所述的核苷酸组合物,其包含:
    1)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
    2)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
    3)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
    4)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23的重链可变区(VH)的DNA;
    5)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
    6)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码SEQ ID NO:31所示的重链可变区(VH)的DNA;
    7)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
    8)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列SEQ ID NO:27所示的重链可变区(VH)的DNA;
    9)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
    10)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA;
    11)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA;
    12)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA;
    13)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:46所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:48所示的重链可变区(VH)的DNA;
    14)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:74所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:75所示的重链可变区(VH)的DNA;或者
    15)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:82所示的轻链可变区(VL)的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:83所示的重链可变区(VH)的DNA。
  65. 根据权利要求60-64中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:7所示的轻链可变区(VL)的DNA如SEQ ID NO:8所示。
  66. 根据权利要求60-65中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:11所示的轻链可变区(VL)的DNA如SEQ ID NO:12所示。
  67. 根据权利要求60-66中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:15所示的轻链可变区(VL)的DNA如SEQ ID NO:16所示。
  68. 根据权利要求60-67中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:19所示的轻链可变区(VL)的DNA如SEQ ID NO:20所示。
  69. 根据权利要求60-68中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:46所示的轻链可变区(VL)的DNA如SEQ ID NO:47所示。
  70. 根据权利要求60-69中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:74所示的轻链可变区(VL)的DNA如SEQ ID NO:76所示。
  71. 根据权利要求60-70中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:82所示的轻链可变区(VL)的DNA如SEQ ID NO:72所示。
  72. 根据权利要求60-71中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:23所示的重链可变区(VH)的DNA如SEQ ID NO:24所示。
  73. 根据权利要求60-72中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:27所示的重链可变区(VH)的DNA如SEQ ID NO:28所示。
  74. 根据权利要求60-73中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:31所示的重链可变区(VH)的DNA如SEQ ID NO:32所示。
  75. 根据权利要求60-74中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:48所示的重链可变区(VH)的DNA如SEQ ID NO:49所示。
  76. 根据权利要求60-75中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:75所示的重链可变区(VH)的DNA如SEQ ID NO:77所示。
  77. 根据权利要求60-76中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:83所示的重链可变区(VH)的DNA如SEQ ID NO:73所示。
  78. 根据权利要求60-77中任一项所述的核苷酸组合物,其中,所述的核苷酸组合物包括:
    (I)第一核酸序列,其含有编码氨基酸序列如SEQ ID NOs:9、13、17、21、37、39、50或54所示的轻链的DNA;以及
    (II)第二核酸序列,其含有编码氨基酸序列如SEQ ID NOs:25、29、33、35、52或56所示的重链的DNA。
  79. 根据权利要求60-78中任一项所述的核苷酸组合物,其中,所述的核苷酸组合物包括:
    (I)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:9、13、17、21、37、39或50所示的轻链的DNA;以及
    (II)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:25、29、33、35或52所示的重链的DNA。
  80. 根据权利要求60-79中任一项所述的核苷酸组合物,其中,所述的核苷酸组合物包括:
    (I)第一核酸分子,其含有编码氨基酸序列如SEQ ID NOs:9、13、17或21所示的轻链的DNA;以及
    (II)第二核酸分子,其含有编码氨基酸序列如SEQ ID NOs:25、29或33所示的重链的DNA。
  81. 根据权利要求60-80中任一项所述的核苷酸组合物,其中,所述的核苷酸组合物包含:
    1)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
    2)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
    3)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:9所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
    4)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
    5)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
    6)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
    7)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
    8)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29所示的重链的DNA;
    9)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
    10)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:25所示的重链的DNA;
    11)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:29的重链的DNA;
    12)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:33所示的重链的DNA;
    13)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:37所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA;
    14)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:39所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:35所示的重链的DNA;
    15)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:50所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:52所示的重链的DNA;或者
    16)第一核酸分子,其含有编码氨基酸序列如SEQ ID NO:54所示的轻链的DNA;以及第二核酸分子,其含有编码氨基酸序列如SEQ ID NO:56所示的重链的DNA。
  82. 根据权利要求78-81中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:13所示的轻链的DNA如SEQ ID NO:14所示。
  83. 根据权利要求78-82中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:17所示的轻链的DNA如SEQ ID NO:18所示。
  84. 根据权利要求78-83中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:21所示的轻链的DNA如SEQ ID NO:22所示。
  85. 根据权利要求78-84中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:37所示的轻链的DNA如SEQ ID NO:38所示。
  86. 根据权利要求78-85中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:39所示的轻链的DNA如SEQ ID NO:40所示。
  87. 根据权利要求78-86中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:50所示的轻链的DNA如SEQ ID NO:51所示。
  88. 根据权利要求78-87中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:54所示的轻链的DNA如SEQ ID NO:55所示。
  89. 根据权利要求78-88中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:25所示的重链的DNA如SEQ ID NO:26所示。
  90. 根据权利要求78-89中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:29所示的重链的DNA如SEQ ID NO:30所示。
  91. 根据权利要求78-90中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:33所示的重链的DNA如SEQ ID NO:34所示。
  92. 根据权利要求78-91中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:35所示的重链的DNA如SEQ ID NO:36所示。
  93. 根据权利要求78-92中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:52所示的重链的DNA如SEQ ID NO:53所示。
  94. 根据权利要求78-93中任一项所述的核苷酸组合物,其中,编码氨基酸序列如SEQ ID NO:56所示的重链的DNA如SEQ ID NO:57所示。
  95. 一种载体,其含有权利要求60-94中任一项所述的核苷酸组合物。
  96. 根据权利要求95所述的载体,其中,所述的载体为真核表达载体、原核表达载体或病毒载体。
  97. 一种宿主细胞,其含有权利要求95或96中所述的载体。
  98. 根据权利要求97所述的宿主细胞,其中,含所述载体的宿主细胞是经载体转化获得的。
  99. 根据权利要求97或98所述的宿主细胞,其中,所述宿主细胞为细菌、酵母或者哺乳动物细胞。
  100. 根据权利要求97-99中任一项所述的宿主细胞,其中,所述宿主细胞为大肠杆菌、毕赤酵母、中国仓鼠卵巢细胞或人胚肾293细胞。
  101. 一种用于制备权利要求1-59中任一项所述的分离的抗体或其抗原结合部分的方法,所述方法包括在权利要求97-100中任一项所述的宿主细胞中表达所述抗体或其抗原结合片段,并分离所述抗体或其抗原结合片段。
  102. 一种药物组合物,包括治疗有效治疗剂量的权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,以及药学上可接受的赋形剂。
  103. 一种用于测量生物样品中LIF的试剂,其包含权利要求1-59中任一项所述的分离的抗体或其抗原结合部分。
  104. 根据权利要求103所述的试剂,其中,所述的生物样本为血液、血清、尿、活检材料、肿瘤或怀疑含有异常LIF水平的任意组织。
  105. 一种抑制LIF表达和/或阻断LIF活性的方法,包括向有需要的病人给予有效治疗剂量的权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物。
  106. 权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物在治疗药物中的用途,所述药物用于抑制LIF表达和/或阻断LIF活性。
  107. 用于抑制LIF表达和/或阻断LIF活性的权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物。
  108. 一种检测生物样品中LIF的方法,包括:(i)获得受试者的组织或液体样品,(ii)暴露所述组织或液体样品到权利要求1-59任一项所述的分离的抗体或其抗原结合部分或权利要求103或104 所述的试剂;以及(iii)检测从(ii)中获得的所述组织或液体样品结合的LIF与对照样品结合的LIF进行对比,其中与对照进行对比的LIF结合量的增加显示LIF的生成、表达或激活的异常水平。
  109. 根据权利要求108所述的方法,其中所述组织或液体样品包括血液、血清、尿、活检材料、肿瘤或怀疑含有异常LIF水平的任意组织。
  110. 一种治疗与LIF相关的疾病或病症的方法,所述方法包括向有需要的病人给予有效治疗剂量的权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物。
  111. 根据权利要求110所述的方法,其中,与LIF相关的疾病或病症为肿瘤。
  112. 根据权利要求111所述的方法,其中,所述肿瘤为实体瘤。
  113. 根据权利要求112所述的方法,其中,所述的实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
  114. 权利要求1-59中任一项所述的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物在治疗与LIF相关的疾病或病症的药物中的用途。
  115. 根据权利要求114所述的用途,其中,与LIF相关的疾病或病症为肿瘤。
  116. 根据权利要求115所述的用途,其中,所述肿瘤为实体瘤。
  117. 根据权利要求116所述的用途,其中,所述的实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
  118. 用于治疗与LIF相关的疾病或病症的权利要求1-59中任一项的分离的抗体或其抗原结合部分,和/或权利要求102所述的药物组合物。
  119. 根据权利要求118所述的用途,其中,与LIF相关的疾病或病症为肿瘤。
  120. 根据权利要求119所述的用途,其中,所述肿瘤是实体瘤。
  121. 根据权利要求120所述的用途,其中,所述实体瘤包括胶质母细胞瘤、肺癌、卵巢癌、结直肠癌、胰腺癌或***癌。
PCT/CN2020/118247 2019-09-29 2020-09-28 对lif具有特异性的结合分子及其用途 WO2021057991A1 (zh)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BR112022005909A BR112022005909A2 (pt) 2019-09-29 2020-09-28 “anticorpos isolados ou fragmento de ligação ao antígeno dos mesmos, método de preparação e usos dos mesmos, composição nucleotídica, vetor, célula hospedeira, composição farmacêutica, reagente para detecção de lif em amostras biológicas, e método exvivo para detectar o lif em amostras biológicas
CN202080046803.4A CN114040924A (zh) 2019-09-29 2020-09-28 对lif具有特异性的结合分子及其用途
AU2020354255A AU2020354255A1 (en) 2019-09-29 2020-09-28 Binding molecule specific for LIF and use thereof
KR1020227014353A KR20220087457A (ko) 2019-09-29 2020-09-28 Lif에 특이적인 결합 분자 및 이의 용도
CA3156080A CA3156080A1 (en) 2019-09-29 2020-09-28 LIF SPECIFIC BINDING MOLECULE AND ITS USE
EP20870311.6A EP4036114A4 (en) 2019-09-29 2020-09-28 LIFE-SPECIFIC BINDING MOLECULE AND USE THEREOF
MX2022003762A MX2022003762A (es) 2019-09-29 2020-09-28 Molécula de unión específica para el factor inhibidor de la leucemia (lif) y uso de la misma.
JP2022519435A JP2022550121A (ja) 2019-09-29 2020-09-28 Lifに特異的な結合分子及びその使用
US17/407,264 US20210403582A1 (en) 2019-09-29 2021-08-20 Binding molecule specific for lif and use thereof
IL291710A IL291710A (en) 2019-09-29 2022-03-27 LIF-SPECIFIC BINDING COMPOUND AND USE THEREOF
ZA2022/03636A ZA202203636B (en) 2019-09-29 2022-03-29 Binding molecule specific for lif and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2019108904 2019-09-29
CNPCT/CN2019/108904 2019-09-29
CNPCT/CN2020/077049 2020-02-27
CN2020077049 2020-02-27

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/407,264 Continuation US20210403582A1 (en) 2019-09-29 2021-08-20 Binding molecule specific for lif and use thereof

Publications (1)

Publication Number Publication Date
WO2021057991A1 true WO2021057991A1 (zh) 2021-04-01

Family

ID=75165589

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/118247 WO2021057991A1 (zh) 2019-09-29 2020-09-28 对lif具有特异性的结合分子及其用途

Country Status (13)

Country Link
US (1) US20210403582A1 (zh)
EP (1) EP4036114A4 (zh)
JP (1) JP2022550121A (zh)
KR (1) KR20220087457A (zh)
CN (1) CN114040924A (zh)
AU (1) AU2020354255A1 (zh)
BR (1) BR112022005909A2 (zh)
CA (1) CA3156080A1 (zh)
CL (1) CL2022000752A1 (zh)
IL (1) IL291710A (zh)
MX (1) MX2022003762A (zh)
TW (1) TWI770619B (zh)
WO (1) WO2021057991A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022228514A1 (zh) * 2021-04-29 2022-11-03 北京浩古元方生物医药科技有限公司 抗人白血病抑制因子抗体及其用途

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994013804A1 (en) 1992-12-04 1994-06-23 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
WO1998050431A2 (en) 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
WO2006028936A2 (en) 2004-09-02 2006-03-16 Genentech, Inc. Heteromultimeric molecules
CN103797031A (zh) * 2010-04-05 2014-05-14 瓦尔德希伯伦肿瘤研究所 识别人类白血病抑制因子(lif)的抗体和抗lif抗体在与有害细胞增殖相关疾病的治疗中的应用
WO2017089614A1 (en) * 2015-11-27 2017-06-01 Fundació Privada Institut D'investigació Oncològica De Vall Hebron Agents for the treatment of diseases associated with undesired cell proliferation
WO2018115960A1 (en) 2016-12-19 2018-06-28 Mosaic Biomedicals, S.L. Antibodies against lif and uses thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8039218B2 (en) * 2002-11-14 2011-10-18 John Wayne Cancer Institute Detection of cancer cells in body fluids
WO2006113747A2 (en) * 2005-04-19 2006-10-26 Prediction Sciences Llc Diagnostic markers of breast cancer treatment and progression and methods of use thereof
WO2007011856A2 (en) * 2005-07-15 2007-01-25 The Trustees Of Columbia University In The City Of New York Compositions and methods for differential diagnosis of chronic lymphocytic leukemia
WO2011035433A1 (en) * 2009-09-23 2011-03-31 University Health Network Selected strains on serum-free growth media for proteomics analysis of lung cancer biomarkers
TW201623329A (zh) * 2014-06-30 2016-07-01 亞佛瑞司股份有限公司 針對骨調素截斷變異體的疫苗及單株抗體暨其用途
WO2019243893A1 (en) * 2018-06-18 2019-12-26 Mosaic Biomedicals Slu Methods of quantifying lif and uses thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994013804A1 (en) 1992-12-04 1994-06-23 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
WO1998050431A2 (en) 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
WO2006028936A2 (en) 2004-09-02 2006-03-16 Genentech, Inc. Heteromultimeric molecules
CN103797031A (zh) * 2010-04-05 2014-05-14 瓦尔德希伯伦肿瘤研究所 识别人类白血病抑制因子(lif)的抗体和抗lif抗体在与有害细胞增殖相关疾病的治疗中的应用
WO2017089614A1 (en) * 2015-11-27 2017-06-01 Fundació Privada Institut D'investigació Oncològica De Vall Hebron Agents for the treatment of diseases associated with undesired cell proliferation
WO2018115960A1 (en) 2016-12-19 2018-06-28 Mosaic Biomedicals, S.L. Antibodies against lif and uses thereof

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Molecular Biology", 1987, WILEY INTERSCIENCE
"Epitope Mapping Protocols in Methods in Molecular Biology", vol. 66, 1996
"Using Antibodies", 1999, COLD SPRING HARBOR LABORATORY PRESS
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, no. 17, 1997, pages 3389 - 3402
BARBAS ET AL., PROC. NATL ACAD. SCI., USA, vol. 91, 1994, pages 3809 - 3813
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
CARTWRIGHT PMCLEAN CSHEPPARD ARIVETTDJONES KDALTON S: "LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism", DEVELOPMENT, vol. 132, 2005, pages 885 - 96
CHANCARTER, NAT. REV. IMMUNOL., vol. 10, 2010, pages 301
CHEUNG ET AL., VIROLOGY, vol. 176, 1990, pages 546
E. MEYERSW. MILLER, CABIOS, vol. 4, 1989, pages 11 - 17
GRAM ET AL., PROC. NATL. ACAD. SCI., USA, vol. 89, 1992, pages 3576 - 3580
HOLLIGERBOHLEN, CANCER AND METASTASIS REV, vol. 18, 1999, pages 411 - 419
HOLLIGERP.WINTER G., CURRENT OPINION BIOTECHNOL, vol. 4, 1993, pages 446 - 449
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
JEFFERIS ET AL., MABS, vol. 1, 2009, pages 1
KABAT, E. A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
KAMOHARA HOGAWA MISHIKO TSAKAMOTO KBABA H: "Leukemia inhibitory factor functions as a growth factor in pancreas carcinoma cells: involvement of regulation of LIF and its receptor expression", INT J ONCOL, vol. 30, 2007, pages 977 - 83, XP009136730
KIRKLAND ET AL., J. IMMUNOL., vol. 137, 1986, pages 3614
KOHLERMILSTEIN, NATURE, vol. 256, 1975, pages 495
KUPHAL SWALLNER SBOSSERHOFF AK: "Impact of LIF (leukemia inhibitory factor) expression in malignant melanoma", EXP MOL PATHOL, vol. 95, 2013, pages 156 - 65, XP055265012, DOI: 10.1016/j.yexmp.2013.06.012
LI XYANG QYU HWU LZHAO YZHANG CYUE XLIU ZWU HHAFFTY BG: "LIF promotes tumorigenesis and metastasis of breast cancer through the AKT-mTOR pathway", ONCOTARGET, vol. 5, no. 3, 2014, pages 788 - 801
LIU BLU YLI JLIU YLIU JWANGW: "Leukemia inhibitory factor promotes tumor growth and metastasis inhuman osteosarcoma via activating STAT3", APMIS, vol. 123, 2015, pages 837 - 46
LIU SCHSU TCHANG YSCHUNG AKJIANG SSOUYANG CNYUH CHHSUEH CLIU YPTSANG NM: "Cytoplasmic LIF reprograms invasive mode to enhance NPC dissemination through modulating YAP1 -FAK/PXN signaling", NAT COMMUN, vol. 9, no. 1, 2018, pages 5105
LIU SCTSANG NMCHIANG WCCHANG KPHSUEH CLIANG YJUANG JLCHOW KPCHANG YS: "Leukemia inhibitory factor promotes nasopharyngeal carcinoma progression and radioresistance", J CLIN INVEST, vol. 123, no. 12, 2013, pages 5269 - 83
LONBERG, NATURE BIOTECH, vol. 23, no. 9, 2005, pages 1117 - 1125
MOLDENHAUER ET AL., SCAND. J. IMMUNOL., vol. 32, 1990, pages 77
MOREL ET AL., MOL. IMMUNOL., vol. 25, no. 1, 1988, pages 7
MORTON SDCADAMURO MBRIVIO SVISMARA MSTECCA TMASSANI M ET AL.: "Leukemia inhibitory factor protects cholangiocarcinoma cells from drug induced apoptosis via a PI3K/AKT-dependent Mcl-1 activation", ONCOTARGET, vol. 6, 2015, pages 26052 - 64
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 444 - 453
NICOLA NABABON JJ: "Leukemia inhibitory factor (LIF", CYTOKINE GROWTH FACTOR REV, vol. 26, no. 5, 2015, pages 533 - 44, XP029276456, DOI: 10.1016/j.cytogfr.2015.07.001
PASTUSCHEK JPOETZSCH JMORALES-PRIETO DMSCHLEUBNER EMARKERT URGEORGIEV G: "Stimulation of the JAK/STAT pathway by LIF and OSM in the human granulosa cell line COV434", J REPROD IMMUNOL, vol. 108, 2015, pages 48 - 55
RIDGEWAY, J.B.B. ET AL., PROTEIN ENG, vol. 9, 1996, pages 616 - 621
ROUX ET AL., J. IMMUNOL., vol. 161, 1998, pages 4083
SCHIER ET AL., J. MOL. BIOL., vol. 263, 1996, pages 551 - 567
SHI YGAO WLYTLE NKHUANG PYUAN XDANN AM ET AL.: "Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring", NATURE, vol. 569, no. 7754, 2019, pages 131 - 135, XP036771230, DOI: 10.1038/s41586-019-1130-6
SHIN JEPARK SHJANG YK: "Epigenetic up-regulation of leukemia inhibitory factor (LIF) gene during the progression to breast cancer", MOL CELLS, vol. 31, 2011, pages 181 - 9, XP055244105, DOI: 10.1007/s10059-011-0020-z
STAHLI ET AL., METHODS IN ENZYMOLOGY, vol. 9, 1983, pages 242
VISWANADHAPALLI SLUO YSAREDDY GRSANTHAMMA BZHOU M ET AL.: "EC359: A First-in-Class Small-Molecule Inhibitor for Targeting Oncogenic LIFR Signaling in Triple-Negative Breast Cancer", MOL CANCER THER, vol. 18, no. 8, 2019, pages 1341 - 1354
VOETVOET: "Biochemistry", 1995, WILEY
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022228514A1 (zh) * 2021-04-29 2022-11-03 北京浩古元方生物医药科技有限公司 抗人白血病抑制因子抗体及其用途

Also Published As

Publication number Publication date
TW202112813A (zh) 2021-04-01
AU2020354255A1 (en) 2022-04-21
CL2022000752A1 (es) 2022-11-18
JP2022550121A (ja) 2022-11-30
KR20220087457A (ko) 2022-06-24
IL291710A (en) 2022-05-01
US20210403582A1 (en) 2021-12-30
MX2022003762A (es) 2022-06-24
CN114040924A (zh) 2022-02-11
BR112022005909A2 (pt) 2022-10-11
EP4036114A4 (en) 2023-11-08
CA3156080A1 (en) 2021-04-01
TWI770619B (zh) 2022-07-11
EP4036114A1 (en) 2022-08-03

Similar Documents

Publication Publication Date Title
WO2020043188A1 (zh) 抗cd47抗体及其应用
AU2014368449B2 (en) Canine antibodies with modified CH2-CH3 sequences
WO2021170082A1 (zh) 抗cd47/抗pd-l1抗体及其应用
WO2021063352A1 (zh) 一种抗pd-l1抗原结合蛋白及其应用
EP4292611A1 (en) Anti-cd112r antibody and use thereof
WO2021057991A1 (zh) 对lif具有特异性的结合分子及其用途
WO2022095970A1 (zh) 双特异抗体及其应用
US20230287126A1 (en) Anti-pd-l1 antibody and use thereof
WO2022100694A1 (zh) 抗体及其制备方法
TW202200615A (zh) 用於治療和預防患者的crs之方法
JP6529602B2 (ja) 抗cd20/抗baff二重特異性抗体
NZ788350A (en) Binding molecule specific for LIF and use thereof
WO2022199527A1 (zh) 靶向肺炎链球菌溶血素的抗原结合蛋白及其用途
WO2024094159A1 (zh) 靶向人源ror1的单域抗体
TWI843182B (zh) 一種抗b7-h4抗體及其製備方法和應用
WO2023273913A1 (zh) 抗b7-h3单克隆抗体及其用途
WO2022262828A1 (zh) 抗il-36r抗体及其用途
WO2022095698A1 (zh) 抗人cd38抗体及其制备方法和用途
WO2023036215A1 (zh) 双特异性抗原结合分子及其应用
WO2022174808A1 (zh) 针对IL-13Rα2的抗体及其应用
WO2024094017A1 (zh) 一种针对磷脂酰肌醇蛋白聚糖3的双特异性抗体及其应用
WO2022148480A1 (zh) 靶向金黄色葡萄球菌α-毒素的抗原结合蛋白及其应用
WO2024140031A1 (zh) 一种靶向cd112r的抗体或其抗原结合片段及其应用
IL310427A (en) Pharmaceutical preparation of anti-R4IL antibody and use thereof
TW202313690A (zh) 一種抗b7-h4抗體及其製備方法和應用

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: 20870311

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022519435

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3156080

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112022005909

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2020354255

Country of ref document: AU

Date of ref document: 20200928

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20227014353

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2020870311

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112022005909

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20220328