WO2024027835A1 - 靶向EGFRvIII的抗体及其在细胞免疫治疗的应用 - Google Patents

靶向EGFRvIII的抗体及其在细胞免疫治疗的应用 Download PDF

Info

Publication number
WO2024027835A1
WO2024027835A1 PCT/CN2023/111305 CN2023111305W WO2024027835A1 WO 2024027835 A1 WO2024027835 A1 WO 2024027835A1 CN 2023111305 W CN2023111305 W CN 2023111305W WO 2024027835 A1 WO2024027835 A1 WO 2024027835A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
seq
amino acid
acid sequence
chain variable
Prior art date
Application number
PCT/CN2023/111305
Other languages
English (en)
French (fr)
Inventor
宋玉洁
李双
周子珊
张嵘
Original Assignee
北京鼎成肽源生物技术有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京鼎成肽源生物技术有限公司 filed Critical 北京鼎成肽源生物技术有限公司
Publication of WO2024027835A1 publication Critical patent/WO2024027835A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • 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
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens

Definitions

  • This article relates to antibodies targeting EGFRvIII or antigen-binding fragments thereof. This article also relates to CARs including antigen-binding fragments targeting EGFRvIII and their applications.
  • CAR Chimeric antigen receptor
  • Tumor antigens are divided into tumor-associated antigens (TAA) and tumor-specific antigens (TSA). , Tumor-associated antigens are expressed in normal tissues. Cell therapy products targeting tumor-associated antigens are prone to off-target toxicity, while cell therapy products targeting tumor-specific antigens are safer and safer.
  • TAA tumor-associated antigens
  • TSA tumor-specific antigens
  • EGFRvIII antigen is a tumor-specific target antigen and is the most common extracellular region mutation of the EGFR gene (Chistiakov et al., 2017). It removes 801 bases between exons 2-7 of the EGFR coding region, 267 amino acids less in the extracellular region than wild-type EGFR, and creates a fusion region between exons 1 and 8. and a novel glycine residue ( Figure 1, An et al., 2018), forming a tumor-specific deletion mutation. Compared with wild-type EGFR, EGFRvIII is ligand-independent and is constitutively activated (Chistiakov et al., 2017).
  • This mutation is found in a variety of cancers (such as breast cancer, lung cancer, head and neck cancer, etc.), but is most common in glioblastoma (Chistiakov et al., 2017; Del Vecchio et al., 2012).
  • Glioblastoma accounts for approximately 15% of brain tumors and is estimated to be diagnosed in nearly 210,000 people worldwide each year. Glioblastoma is a malignant grade 4 tumor that is highly aggressive, prone to recurrence, and has a high rate of death and disability. Glioblastoma develops rapidly, has one of the worst 5-year overall survival rates among all human cancers, and has no effective treatments. at present, Engineers are planning to solve this world problem through immunotherapy (Feldman et al., 2022; Londhe and Date, 2020).
  • Antibody drugs are an effective anti-cancer drug that have become more mature in recent years. It can target tumor cells with specific antigens and use effects such as ADCC and CDC to stimulate the patient's immune system to fight cancer cells.
  • antibodies targeting EGFRvIII, or antigen-binding fragments thereof comprising a heavy chain variable region (HCVR) including HCDR1, HCDR2, and HCDR3, the HCDR1, HCDR2 and HCDR3 are selected from one of the following combinations:
  • HCDR1 The amino acid sequence of HCDR1 is DFSMH (SEQ ID NO: 1);
  • HCDR2 The amino acid sequence of HCDR2 is WINTETGEPSYADDFKG (SEQ ID NO: 2);
  • amino acid sequence of HCDR3 is YGYDVRGDY (SEQ ID NO: 3);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 5);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 6);
  • HCDR2 The amino acid sequence of HCDR2 is WINTETGEPTYADDFKG (SEQ ID NO: 8);
  • amino acid sequence of HCDR3 is YGYDVRGDY (SEQ ID NO: 9);
  • HCDR1 DYYLH (SEQ ID NO: 10);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 11);
  • amino acid sequence of HCDR3 is GYLTY (SEQ ID NO: 12);
  • HCDR1 The amino acid sequence of HCDR1 is RYWMH (SEQ ID NO: 13);
  • HCDR2 The amino acid sequence of HCDR2 is EINPSNGRANYNEKFMS (SEQ ID NO: 14);
  • HCDR3 The amino acid sequence of HCDR3 is GREITTGFAY (SEQ ID NO: 15);
  • HCDR1 DYYMH (SEQ ID NO: 16);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 17);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 18);
  • HCDR1 DYYMH (SEQ ID NO: 19);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 20);
  • amino acid sequence of HCDR3 is GYLAY (SEQ ID NO: 21);
  • HCDR1 DYYMH (SEQ ID NO: 22);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 23);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 25);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 26);
  • amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 27);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 29);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 30);
  • HCDR1 The amino acid sequence of HCDR1 is DYYMH (SEQ ID NO: 31);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 32);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 33);
  • HCDR1 The amino acid sequence of HCDR1 is DYYMH (SEQ ID NO: 34);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 35);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 36);
  • HCDR1 The amino acid sequence of HCDR1 is NYAMS (SEQ ID NO: 37);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 38);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 39);
  • HCDR1 The amino acid sequence of HCDR1 is GYAMS (SEQ ID NO: 40);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 41);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 42);
  • HCDR1 The amino acid sequence of HCDR1 is GYAMS (SEQ ID NO: 43);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 44);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 45);
  • HCDR1 The amino acid sequence of HCDR1 is SGYSWH (SEQ ID NO: 46);
  • amino acid sequence of HCDR2 is YIHYSGSTNYNPPLKS (SEQ ID NO: 47);
  • the amino acid sequence of HCDR3 is GVVSNYAMGN (SEQ ID NO: 48);
  • HCDR1 The amino acid sequence of HCDR1 is TYWMH (SEQ ID NO: 49);
  • amino acid sequence of HCDR2 is YINPNTAYTEYNQNFKD (SEQ ID NO: 50);
  • HCDR3 The amino acid sequence of HCDR3 is GAYYRTYYAMDY (SEQ ID NO: 51);
  • HCDR1 The amino acid sequence of HCDR1 is NYGMN (SEQ ID NO: 52);
  • amino acid sequence of HCDR2 is WINTYTGEPTYADDFKG (SEQ ID NO: 53);
  • HCDR3 The amino acid sequence of HCDR3 is EEFYSRGAMDY (SEQ ID NO: 54);
  • HCDR1 The amino acid sequence of HCDR1 is DYYIN (SEQ ID NO: 55);
  • HCDR2 The amino acid sequence of HCDR2 is WIYPGSGNTKYNEKFKG (SEQ ID NO: 56);
  • HCDR3 The amino acid sequence of HCDR3 is SSRCDF (SEQ ID NO: 57), or
  • the antibody includes a variant of the HCDR sequence combination in any one of (1)-(19), wherein the variant has at least 90% sequence identity, or a total of at least 1 and no more than 10, or no more than 5, 4, 3, or 2 amino acid changes in the HCDR sequence.
  • the antibody further includes a light chain variable region (LCVR) comprising LCDR1, LCDR2, and LCDR3 selected from one of the following combinations:
  • LCVR light chain variable region
  • LCDR1 The amino acid sequence of LCDR1 is SASSSISSNYLH (SEQ ID NO: 58);
  • LCDR2 The amino acid sequence of LCDR2 is GTSNLAS (SEQ ID NO: 59);
  • the amino acid sequence of LCDR3 is HQGSSIPLT (SEQ ID NO: 60);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNHLA (SEQ ID NO: 61);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRAS (SEQ ID NO: 62);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 63);
  • LCDR1 is SASSGISSNYLH (SEQ ID NO: 64);
  • amino acid sequence of LCDR2 is STSNLAS (SEQ ID NO: 65);
  • LCDR3 The amino acid sequence of LCDR3 is HQGSDIPLT (SEQ ID NO: 66);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQNLLNSSNQKNYLA (SEQ ID NO: 67);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRYS (SEQ ID NO: 68);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 69);
  • LCDR1 The amino acid sequence of LCDR1 is KASQSVSNDVV (SEQ ID NO: 70);
  • LCDR2 The amino acid sequence of LCDR2 is YASNRYT (SEQ ID NO: 71);
  • the amino acid sequence of LCDR3 is QQDYSSPWT (SEQ ID NO: 72);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 73);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRES (SEQ ID NO: 74);
  • the amino acid sequence of LCDR3 is QQHYSIPLT (SEQ ID NO: 75);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 76);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRKS (SEQ ID NO: 77);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 78);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNHLA (SEQ ID NO: 79);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRQS (SEQ ID NO: 80);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 81);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 82);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRQS (SEQ ID NO: 83);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 84);
  • LCDR1 KSSQSLLNSSNQKNHLA (SEQ ID NO: 85);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRGS (SEQ ID NO: 86);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 87);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 88);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRDS (SEQ ID NO: 89);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 90);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 91);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRES (SEQ ID NO: 92);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 93);
  • LCDR1 The amino acid sequence of LCDR1 is RSSQSLVHSDGNTYLH (SEQ ID NO: 94);
  • amino acid sequence of LCDR2 is KVSNRFS (SEQ ID NO: 95);
  • the amino acid sequence of LCDR3 is SQSIHVPWT (SEQ ID NO: 96);
  • LCDR1 The amino acid sequence of LCDR1 is SASSSVSYMH (SEQ ID NO: 97);
  • LCDR2 The amino acid sequence of LCDR2 is STSNLAS (SEQ ID NO: 98);
  • the amino acid sequence of LCDR3 is QQRSSYPLT (SEQ ID NO: 99);
  • the amino acid sequence of LCDR1 is RSSQSLVHSDGNTYLH (SEQ ID NO: 100);
  • the amino acid sequence of LCDR2 is KVSNRFS (SEQ ID NO: 101);
  • the amino acid sequence of LCDR3 is SQTTQVPWT (SEQ ID NO: 102);
  • LCDR1 The amino acid sequence of LCDR1 is ITNTDIDDMN (SEQ ID NO: 103);
  • LCDR2 The amino acid sequence of LCDR2 is EGNTLRP (SEQ ID NO: 104);
  • LCDR3 The amino acid sequence of LCDR3 is LQSDDLLPT (SEQ ID NO: 105);
  • LCDR1 The amino acid sequence of LCDR1 is KASQSVDYDGDSYMN (SEQ ID NO: 106);
  • LCDR2 The amino acid sequence of LCDR2 is AASNLES (SEQ ID NO: 107);
  • LCDR3 LQSNEDPYT (SEQ ID NO: 108);
  • amino acid sequence of LCDR2 is KISNRFS (SEQ ID NO: 110);
  • LCDR3 The amino acid sequence of LCDR3 is FQGSHVPFT (SEQ ID NO: 111).
  • LCDR1 The amino acid sequence of LCDR1 is KASEDIYNRLA (SEQ ID NO: 112);
  • LCDR2 The amino acid sequence of LCDR2 is GATSLET (SEQ ID NO: 113);
  • the amino acid sequence of LCDR3 is QQYWSSPLT (SEQ ID NO: 114), or
  • the antibody includes a variant of the combination of LCDR sequences in any one of (1)-(19), wherein the variant has at least 90% Sequence identity, or a total of at least 1 and no more than 10, or no more than 5, 4, 3, or 2 amino acid changes in the LCDR sequence.
  • amino acid sequence of the heavy chain variable region is selected from any of the following:
  • amino acid sequence of the heavy chain variable region and the amino acid sequence of the light chain variable region of the antibody are selected from any combination of:
  • the antibody has a KD value for binding to the EGFRvIII antigen, as determined by surface plasmon resonance (SPR), of less than 10 ⁇ 6 M, preferably less than 10 ⁇ 7 M.
  • SPR surface plasmon resonance
  • the antibody is selected from a murine antibody, a chimeric antibody, a humanized antibody, or a human antibody.
  • the humanized antibody has a binding KD value for EGFRvIII antigen determined by surface plasmon resonance (SPR) of less than 10 ⁇ 6 M, preferably less than 10 ⁇ 7 M.
  • SPR surface plasmon resonance
  • the antigen-binding fragment includes Fab, Fab', F(ab') 2 , single domain antibody, or single chain antibody.
  • the humanized antibody is a single chain antibody whose amino acid sequence is selected from any of the following:
  • the antibody or antigen-binding fragment thereof further includes an Fc fragment.
  • the antigen-binding fragment is a single-chain antibody; preferably, the linker sequence (linker) used to connect the heavy chain variable region and the light chain variable region in the single-chain antibody includes SEQ ID NO. :The sequence shown in 166.
  • chimeric antigen receptors whose extracellular antigen-binding domains include one or more antibody molecules targeting EGFRvIII, or antigen-binding fragments thereof, whose heavy chains are variable
  • the HCDR1, HCDR2 and HCDR3 of the area are selected from one of the following combinations:
  • HCDR1 The amino acid sequence of HCDR1 is DFSMH (SEQ ID NO: 1);
  • HCDR2 The amino acid sequence of HCDR2 is WINTETGEPSYADDFKG (SEQ ID NO: 2);
  • amino acid sequence of HCDR3 is YGYDVRGDY (SEQ ID NO: 3);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 5);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 6);
  • HCDR2 The amino acid sequence of HCDR2 is WINTETGEPTYADDFKG (SEQ ID NO: 8);
  • amino acid sequence of HCDR3 is YGYDVRGDY (SEQ ID NO: 9);
  • HCDR1 DYYLH (SEQ ID NO: 10);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 11);
  • amino acid sequence of HCDR3 is GYLTY (SEQ ID NO: 12);
  • HCDR1 The amino acid sequence of HCDR1 is RYWMH (SEQ ID NO: 13);
  • HCDR2 The amino acid sequence of HCDR2 is EINPSNGRANYNEKFMS (SEQ ID NO: 14);
  • HCDR3 The amino acid sequence of HCDR3 is GREITTGFAY (SEQ ID NO: 15);
  • HCDR1 DYYMH (SEQ ID NO: 16);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 17);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 18);
  • HCDR1 DYYMH (SEQ ID NO: 19);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 20);
  • amino acid sequence of HCDR3 is GYLAY (SEQ ID NO: 21);
  • HCDR1 DYYMH (SEQ ID NO: 22);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 23);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 25);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 26);
  • amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 27);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 29);
  • the amino acid sequence of HCDR3 is GWFAY (SEQ ID NO: 30);
  • HCDR1 The amino acid sequence of HCDR1 is DYYMH (SEQ ID NO: 31);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 32);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 33);
  • HCDR1 The amino acid sequence of HCDR1 is DYYMH (SEQ ID NO: 34);
  • HCDR2 The amino acid sequence of HCDR2 is WIDPENGNTIYDPKFQG (SEQ ID NO: 35);
  • HCDR3 The amino acid sequence of HCDR3 is GYLVY (SEQ ID NO: 36);
  • HCDR1 The amino acid sequence of HCDR1 is NYAMS (SEQ ID NO: 37);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 38);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 39);
  • HCDR1 The amino acid sequence of HCDR1 is GYAMS (SEQ ID NO: 40);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 41);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 42);
  • HCDR1 The amino acid sequence of HCDR1 is GYAMS (SEQ ID NO: 43);
  • HCDR2 The amino acid sequence of HCDR2 is TITSGGSYTYYPDSVKG (SEQ ID NO: 44);
  • amino acid sequence of HCDR3 is KDYGNYWFAY (SEQ ID NO: 45);
  • HCDR1 The amino acid sequence of HCDR1 is SGYSWH (SEQ ID NO: 46);
  • amino acid sequence of HCDR2 is YIHYSGSTNYNPPLKS (SEQ ID NO: 47);
  • the amino acid sequence of HCDR3 is GVVSNYAMGN (SEQ ID NO: 48);
  • HCDR1 The amino acid sequence of HCDR1 is TYWMH (SEQ ID NO: 49);
  • amino acid sequence of HCDR2 is YINPNTAYTEYNQNFKD (SEQ ID NO: 50);
  • HCDR3 The amino acid sequence of HCDR3 is GAYYRTYYAMDY (SEQ ID NO: 51);
  • HCDR1 The amino acid sequence of HCDR1 is NYGMN (SEQ ID NO: 52);
  • amino acid sequence of HCDR2 is WINTYTGEPTYADDFKG (SEQ ID NO: 53);
  • HCDR3 The amino acid sequence of HCDR3 is EEFYSRGAMDY (SEQ ID NO: 54);
  • HCDR1 The amino acid sequence of HCDR1 is DYYIN (SEQ ID NO: 55);
  • HCDR2 The amino acid sequence of HCDR2 is WIYPGSGNTKYNEKFKG (SEQ ID NO: 56);
  • HCDR3 The amino acid sequence of HCDR3 is SSRCDF (SEQ ID NO: 57), or
  • the antibody includes a variant of the HCDR sequence combination in any one of (1)-(19), wherein the variant has at least 90% sequence identity, or a total of at least 1 and no more than 10, or no more than 5, 4, 3, or 2 amino acid changes in the HCDR sequence.
  • the antibody further includes a light chain variable region (LCVR) comprising LCDR1, LCDR2, and LCDR3 selected from one of the following combinations:
  • LCVR light chain variable region
  • LCDR1 The amino acid sequence of LCDR1 is SASSSISSNYLH (SEQ ID NO: 58);
  • LCDR2 The amino acid sequence of LCDR2 is GTSNLAS (SEQ ID NO: 59);
  • the amino acid sequence of LCDR3 is HQGSSIPLT (SEQ ID NO: 60);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNHLA (SEQ ID NO: 61);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRAS (SEQ ID NO: 62);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 63);
  • LCDR1 is SASSGISSNYLH (SEQ ID NO: 64);
  • amino acid sequence of LCDR2 is STSNLAS (SEQ ID NO: 65);
  • LCDR3 The amino acid sequence of LCDR3 is HQGSDIPLT (SEQ ID NO: 66);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQNLLNSSNQKNYLA (SEQ ID NO: 67);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRYS (SEQ ID NO: 68);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 69);
  • LCDR1 The amino acid sequence of LCDR1 is KASQSVSNDVV (SEQ ID NO: 70);
  • LCDR2 The amino acid sequence of LCDR2 is YASNRYT (SEQ ID NO: 71);
  • the amino acid sequence of LCDR3 is QQDYSSPWT (SEQ ID NO: 72);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 73);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRES (SEQ ID NO: 74);
  • the amino acid sequence of LCDR3 is QQHYSIPLT (SEQ ID NO: 75);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 76);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRKS (SEQ ID NO: 77);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 78);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNHLA (SEQ ID NO: 79);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRQS (SEQ ID NO: 80);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 81);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 82);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRQS (SEQ ID NO: 83);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 84);
  • LCDR1 KSSQSLLNSSNQKNHLA (SEQ ID NO: 85);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRGS (SEQ ID NO: 86);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 87);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 88);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRDS (SEQ ID NO: 89);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 90);
  • LCDR1 The amino acid sequence of LCDR1 is KSSQSLLNSSNQKNYLA (SEQ ID NO: 91);
  • LCDR2 The amino acid sequence of LCDR2 is FASTRES (SEQ ID NO: 92);
  • the amino acid sequence of LCDR3 is QQHYSTPLT (SEQ ID NO: 93);
  • LCDR1 The amino acid sequence of LCDR1 is RSSQSLVHSDGNTYLH (SEQ ID NO: 94);
  • amino acid sequence of LCDR2 is KVSNRFS (SEQ ID NO: 95);
  • the amino acid sequence of LCDR3 is SQSIHVPWT (SEQ ID NO: 96);
  • LCDR1 The amino acid sequence of LCDR1 is SASSSVSYMH (SEQ ID NO: 97);
  • LCDR2 The amino acid sequence of LCDR2 is STSNLAS (SEQ ID NO: 98);
  • the amino acid sequence of LCDR3 is QQRSSYPLT (SEQ ID NO: 99);
  • the amino acid sequence of LCDR1 is RSSQSLVHSDGNTYLH (SEQ ID NO: 100);
  • the amino acid sequence of LCDR2 is KVSNRFS (SEQ ID NO: 101);
  • the amino acid sequence of LCDR3 is SQTTQVPWT (SEQ ID NO: 102);
  • LCDR1 The amino acid sequence of LCDR1 is ITNTDIDDMN (SEQ ID NO: 103);
  • LCDR2 The amino acid sequence of LCDR2 is EGNTLRP (SEQ ID NO: 104);
  • LCDR3 The amino acid sequence of LCDR3 is LQSDDLLPT (SEQ ID NO: 105);
  • LCDR1 The amino acid sequence of LCDR1 is KASQSVDYDGDSYMN (SEQ ID NO: 106);
  • LCDR2 The amino acid sequence of LCDR2 is AASNLES (SEQ ID NO: 107);
  • LCDR3 LQSNEDPYT (SEQ ID NO: 108);
  • amino acid sequence of LCDR2 is KISNRFS (SEQ ID NO: 110);
  • LCDR3 The amino acid sequence of LCDR3 is FQGSHVPFT (SEQ ID NO: 111).
  • LCDR1 The amino acid sequence of LCDR1 is KASEDIYNRLA (SEQ ID NO: 112);
  • LCDR2 The amino acid sequence of LCDR2 is GATSLET (SEQ ID NO: 113);
  • the amino acid sequence of LCDR3 is QQYWSSPLT (SEQ ID NO: 114), or
  • the antibody includes a variant of the combination of LCDR sequences in any one of (1)-(19), wherein the variant has at least 90% Sequence identity, or a total of at least 1 and no more than 10, or no more than 5, 4, 3, or 2 amino acid changes in the LCDR sequence.
  • amino acid sequence of the heavy chain variable region is selected from any of the following:
  • amino acid sequence of the heavy chain variable region and the amino acid sequence of the light chain variable region of the antibody are selected from any combination of:
  • the antibody has a KD value for binding to the EGFRvIII antigen, as determined by surface plasmon resonance (SPR), of less than 10 ⁇ 6 M, preferably less than 10 ⁇ 7 M.
  • SPR surface plasmon resonance
  • the antibody is selected from a murine antibody, a chimeric antibody, a humanized antibody, or a human antibody.
  • the humanized antibody has a binding KD value for EGFRvIII antigen determined by surface plasmon resonance (SPR) of less than 10 ⁇ 6 M, preferably less than 10 ⁇ 7 M.
  • SPR surface plasmon resonance
  • the antigen-binding fragment is a single-chain antibody; preferably, the linker sequence (linker) used to connect the heavy chain variable region and the light chain variable region in the single-chain antibody includes SEQ ID NO. :The sequence shown in 166.
  • the humanized antibody is a single chain antibody whose amino acid sequence is selected from any of the following:
  • the CAR also includes a hinge region, a transmembrane region, a co-stimulatory domain and an activation domain; preferably, the hinge region is a CD8 hinge region, and the transmembrane region is a CD8 ⁇ transmembrane region;
  • the costimulatory domain is a 41BB costimulatory domain or a CD28 costimulatory domain;
  • the activation domain is a CD3 ⁇ activation domain.
  • the hinge region includes the sequence shown in SEQ ID NO: 156; the transmembrane region includes the sequence shown in SEQ ID NO: 158; the costimulatory domain includes SEQ ID NO: 160 and/or The sequence shown in SEQ ID NO: 162; the activation domain includes the sequence shown in SEQ ID NO: 164.
  • the CAR also includes a signal peptide; preferably, the signal peptide is a CD8 ⁇ signal peptide; more preferably, the signal peptide includes the sequence shown in SEQ ID NO: 154.
  • the signal peptide includes the sequence shown in SEQ ID NO: 154.
  • this article provides a nucleic acid molecule encoding: 1) the above-mentioned antibody or antigen-binding fragment thereof, or the heavy chain variable region and/or light chain variable region of the above-mentioned antibody or antigen-binding fragment thereof; or 2) the above-mentioned CAR.
  • the nucleic acid molecule includes SEQ ID NO: 153, 155, 157, 159, 161, 163, 165, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, or 188
  • this article provides an expression vector, which includes the above-mentioned nucleic acid molecule; preferably, the expression vector is a lentiviral vector.
  • cells comprising the above-described expression vectors.
  • this article provides cells expressing the above-mentioned CAR.
  • the cells are immune cells.
  • the cells are T cells or NK cells.
  • the cells are also engineered to express a dominant negative receptor (DNR) or a chimeric switch receptor (CSR); preferably, the DNR is dnTGF ⁇ RII and/or dnPD1; preferably, the DNR is dnTGF ⁇ RII and/or dnPD1; preferably, the DNR is The intracellular domain of the CSR is derived from IL7R ⁇ ; more preferably, the dnTGF ⁇ RII includes the sequence shown in SEQ ID NO: 193, and the intracellular domain of the CSR includes the sequence shown in SEQ ID NO: 197.
  • DNR dominant negative receptor
  • CSR chimeric switch receptor
  • this article provides a pharmaceutical composition, which includes: 1) the above-mentioned antibody or antigen-binding fragment thereof, nucleic acid molecule, expression vector or cell; and 2) a pharmaceutically acceptable carrier.
  • this article provides the use of the above-mentioned antibody or antigen-binding fragment thereof, nucleic acid molecule, expression vector or cell in the preparation of a medicament for treating tumors.
  • the tumor expresses EGFRvIII.
  • the tumor is glioblastoma.
  • a method of treating a tumor in a subject comprising administering to the subject a therapeutically effective amount of the above-mentioned antibody or antigen-binding fragment thereof, nucleic acid molecule, expression vector, cell or pharmaceutical composition. .
  • the tumor expresses EGFRvIII.
  • the tumor is glioblastoma.
  • this article provides a multispecific antibody molecule, which at least includes a first functional part and a second functional part, wherein the first functional part includes the above-mentioned antibody or an antigen-binding fragment thereof; the second functional part has the same effect as the first functional part.
  • Functional moieties differ in antigen binding specificity.
  • the second functional moiety has binding specificity for T cells.
  • immunoconjugates comprising the above-described antibodies or antigen-binding fragments thereof linked to a therapeutic agent.
  • the therapeutic agent is a drug.
  • the therapeutic agent is a cytotoxic agent.
  • the therapeutic agent is a radioactive isotope.
  • this article provides a kit for detecting the presence or content of EGFRvIII in a sample, wherein the kit includes the above-mentioned antibody or antigen-binding fragment thereof.
  • Figure 1 is a schematic structural diagram of EGFRvIII.
  • Figure 2 shows the results of flow cytometric detection of the binding ability of positive subclones supernatants to target cells.
  • Figure 3 shows the results of detecting the affinity of some purified mouse-derived antibodies by the SPR method.
  • FIG. 4 is a schematic structural diagram of the CAR used in this article.
  • Figure 5 shows the CAR positivity rate results of mouse antibody sequence CAR T cells.
  • Figure 6 shows the killing efficiency of murine antibody sequence CAR T cells on U87MG-EGFRvIII target cells.
  • Figure 7 shows the killing efficiency of murine antibody sequence CAR T cells on U87MG-EGFR target cells.
  • Figure 8 shows the in vivo tumor inhibition efficiency of murine antibody sequence CAR T cells.
  • Figure 9 shows the affinity results of the purified #28 clone humanized antibody detected by the SPR method.
  • Figure 10 shows the affinity results of the purified #40-2 clone humanized antibody detected by the SPR method.
  • Figure 11 shows the CAR positivity rate of CAR T cells prepared from the #28 clone humanized antibody.
  • Figure 12 shows the CAR positivity rate of CAR T cells prepared from humanized antibodies from the #40-2 clone.
  • Figure 13 shows the killing efficiency of #28 clone humanized antibody CAR T cells on U87MG EGFRvIII target cells.
  • Figure 14 shows the killing efficiency of #28 clone humanized antibody CAR T cells on U87MG EGFR target cells.
  • Figure 15 shows the killing efficiency of #40-2 clone humanized antibody CAR T cells on U87MG EGFRvIII target cells.
  • Figure 16 shows the killing efficiency of #40-2 clone humanized antibody CAR T cells on U87MG EGFR target cells.
  • Figure 17 shows the in vivo tumor inhibition efficiency of cloned humanized antibody CAR T cells.
  • Figure 18 shows the in vivo tumor inhibition efficiency of cloned humanized antibody CAR T cells.
  • Figure 19 shows the target cell killing efficiency of CAR NK cells prepared from the #28 clone humanized antibody.
  • A U87MG EGFRvIII target cells
  • B U87MG EGFR target cells.
  • Figure 20 shows a schematic diagram of CAR constructs with DNR (A) or CSR (B).
  • Figure 21 shows the CAR positive rate detection results of the fourth generation CAR T cells constructed in this application.
  • A RII cells
  • B T7R cells
  • C CAR T C cells.
  • FIG. 22 shows the results of CAR T cell killing efficiency detection.
  • A Killing rate when TGF ⁇ is not used;
  • B Killing rate when TGF ⁇ is used;
  • C Changes in killing rate.
  • FIG. 23 In vivo efficacy study results of CAR T cells.
  • CAR-T-C refers to CAR T cells in the control group;
  • RII refers to CAR T cells expressing the dominant negative receptor dnTGF- ⁇ RII;
  • T7R refers to CAR T cells expressing the chimeric switch receptor dnTGF- ⁇ RII-IL7R ⁇ .
  • Antibodies refer to immunoglobulins secreted by plasma cells (effector B cells) and used by the body's immune system to neutralize foreign substances (polypeptides, viruses, bacteria, etc.). This foreign substance is accordingly called an antigen.
  • the basic structure of a classic antibody molecule is a tetramer composed of two identical heavy chains and two identical light chains. According to the conservative differences in amino acid sequences, heavy and light chains are divided into variable regions (V) located at the amino terminus and constant regions located at the carboxyl terminus. The variable regions of a heavy chain and a light chain interact to form the antigen-binding site (Fv).
  • variable region the composition and arrangement of amino acid residues in certain regions are more variable than other regions (backbone regions, FR) within the variable region, which are called hypervariable regions (HVR).
  • the hypervariable regions are actually antibodies. Key site for antigen binding. Because these hypervariable region sequences are complementary to antigenic determinants, they are also called complementarity-determining regions (CDRs). Both the heavy chain and the light chain have three complementarity determining regions, called HCDR1, HCDR2, HCDR3 and LCDR1, LCDR2, and LCDR3 respectively.
  • antibody molecules such as PEGylation, glycosylation, formation of antibody molecule conjugates (such as ADC), addition of purification tags, or fusion with other proteins, such as formation of bispecific antibodies.
  • ADC antibody molecule conjugates
  • the sequence of the antibody complementarity determining region is known, those skilled in the art can easily replace part of the sequence of the antibody molecule (such as the constant region or framework region) with sequences from other species, such as through DNA recombinant technology, to form a chimeric antibody.
  • the chimeric antibody can substantially retain the binding specificity of the antibody from which it was derived.
  • the source antibody is a mouse antibody
  • its constant region can be replaced with a human antibody constant region
  • the source antibody is a human antibody
  • its constant region can be replaced with a mouse antibody constant region to weaken the antibody in different species Immunization when used is immunogenic or to exploit a specific function of the constant region, such as ADCC-related activity.
  • all other sequences in the antibody molecule except the CDR sequence can be replaced with the corresponding sequence of another antibody molecule (from the same or different species) (which may include one or several as appropriate). amino acid mutations) while substantially retaining the binding specificity of the antibody from which it was derived.
  • those skilled in the art often use CDR transplantation to humanize murine antibodies.
  • the "antigen-binding fragment" of an antibody molecule refers to the amino acid fragment in the antibody molecule that participates in specific antigen binding, such as Fab, Fab' and (Fab') 2 , etc.
  • the person skilled in the art knows how to obtain these antigen-binding fragments.
  • a classical antibody molecule can be digested with papain to obtain the Fab fragment, and pepsin digested to obtain F(ab') 2 , which is formed by treatment with a reducing agent to break the disulfide bond between the hinge regions of F(ab') 2 Fab' fragment.
  • Single-chain antibodies and single-domain antibodies with antigen-binding ability are single-peptide chain antibody molecules and can be regarded as "antigen-binding fragments" of classic antibody molecules.
  • Fc fragment refers to the handle region of the Y"-shaped antibody molecule, that is, the crystallizable fragment (fragment crystallizable, Fc) includes the second and third constant domains (CH2 and CH3 domains) of the heavy chain. It can be passed through proteolytic enzymes (such as papain) hydrolyzes the antibody molecule to obtain the antibody Fc region.
  • the Fc region can include a hinge, CH2, and CH3. When the Fc region includes a hinge, it can mediate dimerization between two Fc-containing polypeptides.
  • the Fc fragment can be from IgG, IgM, IgD, IgE, or IgA.
  • the Fc region is from IgG1, IgG2, IgG3, or IgG4.
  • Fc fragment also includes fragments from native Fc fragments that have been altered but still retain their effector function.
  • a "variant Fc fragment” includes an amino acid sequence that has at least one amino acid change in the amino acid sequence of a native Fc fragment.
  • a variant Fc fragment has At least one amino acid substitution, for example, about 1 to about 10 amino acids are substituted in the parent Fc fragment, and preferably about 1 to about 5 amino acid substitutions.
  • the variant Fc fragment Fc region has at least about 80% sequence identity, at least about 90% sequence identity, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity.
  • the effector function of the "Fc fragment” can Including binding to Fc receptors, Clq binding, complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), mediating phagocytosis, etc.
  • Single chain fragment variable is composed of an antibody heavy chain variable region and a light chain variable region connected through a short peptide to form a peptide chain. Through correct folding, the variable regions from the heavy chain and light chain form Fv segments through non-covalent interactions, so scFv can better retain its affinity activity for antigens.
  • Single domain antibody also known as “VHH antibody” refers to an antibody molecule with antigen-binding ability, including a heavy chain variable region but no light chain. From a structural point of view, single domain antibodies can also be considered as an antigen-binding fragment of an antibody molecule. It was first discovered in camelids. Subsequently, researchers discovered more single-domain antibodies with antigen-binding ability through screening of antibody libraries (such as phage display libraries). Single domain antibodies have some advantages over ordinary antibody molecules (for example, classic tetrameric antibody molecules) or their antigen-binding fragments, including but not limited to: smaller molecular weight, and when used in the human body, they can easily reach tissues that are difficult for ordinary antibody molecules to reach. or parts, or can access antigenic epitopes in proteins or polypeptides that are difficult for ordinary antibody molecules to access; they are more stable and can withstand changes in temperature and pH, as well as the effects of denaturants and proteases.
  • Bispecific antibodies refer to antibody molecules that have two different binding sites and can recognize and bind to two different antigens respectively.
  • one binding site of a bispecific antibody can be used to bind immune cells (such as T cells), and the other binding site is used to bind tumor cells, thereby enhancing the killing effect of immune cells on tumor cells while reducing side effects such as off-target toxicity.
  • This bifunctional antibody usually has higher efficacy than monoclonal antibody drugs as a tumor treatment drug.
  • antibody molecules can be engineered to include multiple different binding sites, creating "trispecific antibodies,”"tetraspecificantibodies,” etc.
  • the "multispecific antibody” used in this article covers these bispecific antibodies, trispecific antibodies, tetraspecific antibodies, etc.
  • the multispecific antibodies used are bispecific antibodies.
  • Humanized antibody refers to an antibody that is less immunogenic in humans while retaining the reactivity of a non-human antibody. This can be accomplished, for example, by retaining the non-human CDR regions and replacing the remainder of the antibody with their human counterparts (ie, the constant regions as well as the framework portions of the variable regions).
  • Immunoconjugate refers to an EGFRvIII-targeting antibody or antigen-binding fragment thereof disclosed herein conjugated to a therapeutic agent.
  • the treatment is, for example, cytotoxic, pharmaceutical (eg immunosuppressive) or radioactive toxin.
  • Fusion protein refers to a protein molecule composed of at least two different peptide segments that is artificially produced (for example, through genetic engineering technology). These peptides do not exist in nature or do not exist in the same protein molecule. Common examples of fusion proteins including antibody fragments include antibody-cytokine fusion proteins, antibody-cytotoxin fusion proteins (also known as immunotoxins), enzyme-labeled antibodies for immunoassays, chimeric antigen receptors (CARs), etc. .
  • Targeting or “specific binding” means that one molecule (e.g., an antibody or antigen-binding fragment thereof) has a higher affinity for another molecule (e.g., a tumor cell surface antigen) relative to other molecules also present in the environment. Binding affinity. "Targeting” or “specific binding” does not exclude that the molecule may have binding affinity for more than one molecule, for example, a bispecific antibody may have high affinity for two different antigens.
  • the modifier "anti” used before an antigen means that the antibody that follows it “targets” or “specifically binds” to the antigen, for example, an anti-EGFRvIII antibody means that the antibody “targets” or “specifically binds” to EGFRvIII.
  • the anti-EGFRvIII antibody does not "target” or “specifically bind” normal EGFR molecules.
  • An antibody “specifically binds an antigen” or the “antigen-binding specificity” of an antibody refers to the antibody's ability to bind the target antigen with a higher or higher binding affinity relative to other antigens.
  • the binding ability of an antibody to the target antigen can be qualitatively or quantitatively identified through various methods, such as measuring the K D value of the antibody binding to the target antigen through surface plasmon resonance (SPR), measuring the ability of the antibody to compete with other antibodies for binding to the target antigen, etc.
  • Kd is the equilibrium dissociation constant, which can be used to measure the binding affinity between the antibody and its antigen. The smaller the K D value, the stronger the affinity.
  • Binding complex refers to a complex formed between a molecule and its binding partner.
  • binding complexes may include antigen-antibody complexes, ligand-receptor complexes, protein dimers, and the like.
  • the forces that form binding complexes mainly include non-covalent bond forces such as hydrogen bonds, van der Waals forces, and ionic bonds.
  • the presence of the target antigen or antibody in the sample can be detected by detecting the complex formed by the antigen and antibody, or the content or concentration of the target antigen or antibody can be determined.
  • Protein segment refers to a polypeptide fragment, which is a short amino acid sequence, for example, about 2-20 amino acids in length, and is usually a part of a polypeptide or protein.
  • EC50 concentration for 50% of maximal effect refers to the concentration that causes 50% of the maximum effect.
  • concentration of the antibody molecule that produces half of the maximum fluorescence intensity. The lower the EC50 value, the greater the binding affinity to the antigen on the cell.
  • Purification tag refers to the amino acid sequence expressed together with the target protein or polypeptide in the form of a fusion protein for purifying the target protein or polypeptide, including but not limited to His6 tag, Flag tag, MBP (maltose binding protein) tag and GST (gluten thione sulfhydryl transferase) tag, SUMO (small ubiquitin related modifier), etc. These tags can be removed by enzymatic digestion after purification, or they can be used with tags (such as His6 tags) without affecting the normal function of the target protein or peptide.
  • Detectable label refers to an amino acid sequence or other chemical group linked to a protein or polypeptide, used to indicate the presence or content of the protein or polypeptide in a sample, or used to track the protein or polypeptide in a subject's body or cells location information.
  • detectable labels include various enzymes that can be used in immunoassays, such as horseradish peroxidase (HRP), alkaline phosphatase (ALP); fluorescent groups (such as FAM, FITC) or fluorescent proteins (such as GFP ); radioactive isotopes (such as 3 H, 14 C, 35 S).
  • HRP horseradish peroxidase
  • ALP alkaline phosphatase
  • fluorescent groups such as FAM, FITC
  • fluorescent proteins such as GFP
  • radioactive isotopes such as 3 H, 14 C, 35 S.
  • polypeptide and “protein” are used interchangeably and refer to a polymer of amino acid residues. Such polymers of amino acid residues may contain natural or unnatural amino acid residues and include, but are not limited to, peptides, oligopeptides, dimers, trimers and multimers of amino acid residues. Full-length proteins and their fragments are included in this definition. The term also includes post-expression modifications of the polypeptide, such as glycosylation, sialylation, acetylation, phosphorylation and similar modifications.
  • polypeptide refers to a protein that includes modifications to the native sequence, such as deletions, additions and substitutions (which are generally conservative in practice) so long as the protein retains the desired activity. These modifications may be purposeful, such as induced via site-directed mutagenesis, or may be accidental, such as via mutation of the host in which the protein is produced or errors due to PCR amplification.
  • variants or functional variants may include one or more sequences compared to the parent sequence (the antibody or antigen-binding fragment thereof). Multiple (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 , 25, 26, 27, 28, 29, 30 or more) insertion, deletion or substitution of amino acid residues while retaining at least 50%, 60%, 70%, 80%, 90%, or even higher than the parent sequence. It is known to those skilled in the art that the VH or VL framework sequence can have considerable room for change, as long as it can maintain the spatial position of the corresponding CDR sequence.
  • the framework region sequence in the VH amino acid sequence can be changed, for example, by replacing the framework regions of antibody molecules of different species with each other, and functional variants that substantially retain the antigen-binding ability can be obtained.
  • nucleic acid molecule As used herein, the terms “nucleic acid molecule,” “nucleic acid,” and “polynucleotide” are used interchangeably to refer to polymers of nucleotides. Such nucleotide polymers may contain natural and/or non-natural nucleotides and include, but are not limited to, DNA, RNA and PNA. "Nucleic acid sequence” refers to a linear sequence of nucleotides contained in a nucleic acid molecule or polynucleotide.
  • vector refers to a nucleic acid molecule that can be engineered to contain a polynucleotide of interest (eg, a coding sequence for a polypeptide of interest) or a nucleic acid molecule that can replicate in a host cell (eg, nucleic acid, plasmid, or virus, etc.).
  • a vector may include one or more of the following components: an origin of replication, one or more regulatory sequences that regulate expression of a polynucleotide of interest (such as a promoter and/or enhancer), and/or one or more Selectable marker genes (such as antibiotic resistance genes and genes useful in colorimetric assays, such as beta-galactose).
  • expression vector refers to a vector used to express a polypeptide of interest in a host cell.
  • “Host cell” refers to a cell that is or has been the recipient of a vector or isolated polynucleotide.
  • the host cell can be a prokaryotic cell or a eukaryotic cell.
  • Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate cells; fungal cells, such as yeast; plant cells; and insect cells.
  • Non-limiting exemplary mammalian cells include, but are not limited to, NSO cells, 293 and CHO cells, and derivatives thereof, such as 293-6E, CHO-DG44, CHO-K1, CHO-S and CHO-DS cells.
  • Host cells include the progeny of a single host cell, and the progeny may not necessarily be identical (in terms of morphology or genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutations.
  • Host cells also include cells transfected in vivo with the nucleic acid molecules or expression vectors provided herein.
  • Treatment refers to the treatment of a subject to obtain a beneficial or desired clinical result.
  • Treatment encompasses a variety of treatments, including administration of any possible drug to the subject, surgery, radiation, etc.
  • beneficial or desired clinical outcomes include, but are not limited to, any one or more of the following: alleviation of one or more symptoms, attenuation of disease severity, prevention or delay of disease spread (e.g. metastasis, e.g. metastasize to the lungs or lymph nodes), prevent or delay disease recurrence, delay or slow down disease progression, improve disease conditions, inhibit disease or disease progression, block its development and remission (whether partial or complete remission).
  • the methods provided herein encompass any one or more of these aspects of treatment. In accordance with the above, “treatment” does not require complete removal of all symptoms of a condition or disease or complete alleviation.
  • terapéuticaally effective amount refers to an amount of active compound sufficient to elicit in a subject the biological or medical response desired by the clinician.
  • the "therapeutically effective dose” of the fusion protein of the present invention can be determined by those skilled in the art based on the route of administration, the subject's weight, age, condition and other factors. For example, a typical daily dosage may range from 0.01 mg to 100 mg or more of active ingredient per kg of body weight.
  • pharmaceutically acceptable carrier refers to solid or liquid diluents, fillers, antioxidants, stabilizers and the like that can be administered safely and are suitable for use by humans and/or Administration to the animal without undue adverse side effects while being suitable for maintaining the viability of the drug or active agent therein.
  • various carriers well known in the art may be administered, including, but not limited to, sugars, starch, cellulose and its derivatives, maltose, gelatin, talc, calcium sulfate, vegetable oils, synthetic oils, polyols , alginic acid, phosphate buffer, emulsifier, isotonic saline, and/or pyrogen-free water, etc.
  • the pharmaceutical compositions provided herein can be made into clinically acceptable dosage forms such as powders and injections.
  • composition of the present invention can be administered to a subject by any appropriate route, for example, by oral administration, intravenous infusion, intramuscular injection, subcutaneous injection, subperitoneal, rectal, sublingual, or by inhalation, transdermal, etc. route of administration.
  • Subject refers to an animal, such as a mammal, including (but not limited to) humans, rodents, simians, felines, canines, equines, bovines, porcines, sheep, goats, mammals Laboratory animals, mammalian farm animals, mammalian sporting animals and mammalian pets.
  • the subject may be male or female and may be of any appropriate age, including infants, juveniles, young adults, adults, and geriatric subjects.
  • a subject refers to an individual in need of treatment of a disease or condition.
  • a subject receiving treatment can be a patient who has a condition associated with the treatment, or is at risk of developing the condition.
  • the subject is a human, such as a human patient. The term is often used interchangeably with "patient,” "subject,” “subject,” etc.
  • Bio sample means a quantity of material derived from a living or previously living organism. Such substances include, but are not limited to, blood (eg, whole blood), plasma, serum, urine, amniotic fluid, synovial fluid, endothelial cells, leukocytes, monocytes, other cells, organs, tissues, bone marrow, lymph nodes, and spleen.
  • sequence identity refers to the identity between two amino acid or nucleotide sequences (such as a query sequence and a reference sequence) A quantity of degree, usually expressed as a percentage. Usually, before calculating the percent identity between two amino acid or nucleotide sequences, the sequences are aligned and gaps (if any) are introduced. If at a certain alignment position, the amino acid residues or bases in the two sequences are the same, the two sequences are considered to be consistent or matching at that position; if the amino acid residues or bases in the two sequences are different, the two sequences are considered to be inconsistent or matching at that position. mismatch.
  • the number of matching positions is divided by the total number of positions in the alignment window to obtain sequence identity.
  • the number of gaps and/or the gap length is also taken into account.
  • the publicly available alignment software BLAST available on the web page ncbi.nlm.nih.gov
  • BLAST can be used to obtain an optimal sequence alignment and calculate two amino acids or nucleotides by using default settings Sequence identity between sequences.
  • Chimeric antigen receptor also known as chimeric T cell receptor, chimeric immune receptor
  • Chimeric antigen receptor is an engineered protein receptor molecule that can confer desired specificity to immune effector cells, such as with The ability to bind to specific tumor antigens.
  • Chimeric antigen receptors usually consist of an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain. In most cases, the antigen-binding domain is a scFv sequence that is responsible for recognizing and binding to a specific antigen.
  • Intracellular signaling domains usually include immunoreceptor tyrosine activation motifs (ITAMs), such as the signaling domain derived from CD3 ⁇ molecules, which are responsible for activating immune effector cells and producing killing effects.
  • ITAMs immunoreceptor tyrosine activation motifs
  • the chimeric antigen receptor may also include a signal peptide at the amino terminus responsible for the intracellular localization of the nascent protein, and a hinge region between the antigen-binding domain and the transmembrane domain.
  • intracellular signaling domains may also include costimulatory domains derived, for example, from the 4-1BB molecule.
  • CAR-T cells refer to T cells expressing CAR, which are usually obtained by transducing T cells with an expression vector encoding CAR.
  • Commonly used expression vectors are viral vectors, such as lentiviral expression vectors.
  • Chimeric antigen receptor-modified T cells (CAR-T) are not restricted by the major histocompatibility complex and have specific targeting killing activity and the ability to sustain expansion.
  • expression vectors encoding CAR can also be used to transform other lymphocytes such as NK cells to obtain targeted killer cells expressing the CAR.
  • DNR Dominant Negative Receptor
  • DNRs usually have a complete ligand-binding domain but lack a domain for intracellular enzymatic activity (such as kinase activity). It can be a mutated form of the full-length receptor or a truncated form of the receptor.
  • some cancers, particularly solid cancers may upregulate inhibitory ligands that bind to inhibitory receptors on CAR T cells. This adaptive resistance compromises the efficacy of CAR T cell therapy.
  • TGF- ⁇ transforming growth factor beta
  • TGF- ⁇ can induce or promote metastasis and effectively suppress the immune system. Therefore, in some embodiments, we use a truncated form of the TGF-beta receptor TGFBRII as a TGF-beta DNR to improve the anti-tumor properties of the CAR T cells disclosed herein.
  • CAR and TGF- ⁇ DNR are co-expressed on the T cell surface using 2A self-cleaving peptide.
  • the CAR and TGF- ⁇ DNR are expressed separately on the T cell surface using two expression vectors.
  • TGF- ⁇ DNR is introduced into the CAR T cells disclosed herein, the cytotoxicity of the CAR T cells against some EGFRvIII-positive cancer cells can be enhanced.
  • CSR Chimeric Switch Receptor
  • CAR T cells e.g., CAR T cells
  • CSR can exploit inhibitory molecules expressed by cancer cells to further stimulate CAR T cells.
  • CAR T cells can be engineered to express a CSR formed by fusion of the PD-1 extracellular ligand-binding domain with the CD28 transmembrane and intracellular costimulatory signaling domains.
  • the expressed CAR can bind to EGFRvIII, while the simultaneously expressed CSR can bind to PD-L1.
  • the properties of the PD-1/CD28 chimeric switching receptor fusion protein prevent normal PD1/PD-L1-mediated T cell suppression and instead promote signaling through the CD28 domain, thereby stimulating CAR T cells. Therefore, PD-L1 is converted into a costimulatory ligand for CAR T cells by switching the transmembrane and intracellular domains of PD-1 with those of CD28. This would induce enhanced toxicity against PD-L1-expressing cancer cells.
  • the transmembrane and intracellular domains of TGFBRII can be switched with those of the IL7R ⁇ receptor, thereby switching the inhibitory signal of TGF- ⁇ in the tumor microenvironment. Stimulate signals for CAR T cells and promote their proliferation or activity.
  • anti-EGFRvIII antibodies that specifically bind EGFRvIII.
  • the EGFRvIII antibody binds the EGFRvIII antigen with relatively high binding affinity.
  • an assay such as surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • the ability of an anti-EGFRvIII antibody to bind to EGFRvIII can be measured by an assay such as surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • it can also be determined by other protein interaction assay methods known in the art, such as biolayer interference (BLI) technology, enzyme-linked immunosorbent assay (ELISA) and flow cytometric fluorescence sorting technology (FACS).
  • BLI biolayer interference
  • ELISA enzyme-linked immunosorbent assay
  • FACS flow cytometric fluorescence sorting technology
  • This article provides the CDR sequences of a variety of anti-EGFRvIII antibodies.
  • their heavy chain CDR sequences are SEQ ID NO: 1-3, 4-6, 7-9, 10-12, 13-15, 16-18, 19. -21, 22-24, 25-27, 28-30, 31-33, 34-36, 37-39, 40-42, 43-45, 46-48, 49-51, 52-54 or 55-57 sequence shown.
  • the corresponding light chain CDR sequences are SEQ ID NO: 58-60, 61-63, 64-66, 67-69, 70-72, 73-75, 76-78, 79-81, 82-84 , 85-87, 88-90, 91-93, 94-96, 97-99, 100-102, 103-105, 106-108, 109-111 or 112-114.
  • the heavy chain variable region of the anti-EGFRvIII antibody provided by the invention includes the same sequence as SEQ ID NO: 115, 117, 119, 121, 123, 125, 127, 129, 131, 133, 135, 137, 139
  • the sequence shown in , 141, 143, 145, 147, 149 or 151 has an amino acid sequence with at least 90% sequence identity (for example, at least 95%, at least 98%, at least 99% or even 100% sequence identity).
  • the corresponding light chain variable region includes SEQ ID NO: 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138,
  • the sequence represented by 140, 142, 144, 146, 148, 150 or 152 is an amino acid sequence that has at least 90% sequence identity (eg, at least 95%, at least 98%, at least 99%, or even 100% sequence identity).
  • CDR sequences of the anti-EGFRvIII antibodies have been provided herein, those skilled in the art can construct various polypeptide constructs with EGFRvIII binding capabilities, which includes using backbone regions (FRs) from different antibody molecules to combine with these CDR sequences.
  • FRs backbone regions
  • These framework regions include native framework region sequences from human antibodies or animal (eg, mouse, rat, sheep, camel, etc.) antibodies. These framework regions may also include framework region sequence variants resulting from alterations of the natural framework region sequence.
  • a polypeptide construct that specifically binds to EGFRvIII can be easily obtained by combining the CDR sequences provided herein with different backbone region sequences to form a heavy chain variable region, and testing its binding ability to EGFRvIII.
  • humanized antibodies such as humanized single chain antibodies (scFV) can be constructed based on these CDR sequences.
  • the humanized single chain antibodies (scFV) provided by the invention include those having the same sequence as SEQ ID NO: 169, 171, 173, 175, 177, 179, 181, 183, 185 or 187. Amino acid sequences that are at least 90% sequence identical (eg, at least 95%, at least 98%, at least 99%, or even 100% sequence identical).
  • anti-EGFRvIII antibodies or antigen-binding fragments thereof described herein may contain conservative amino acid substitutions.
  • Conservative amino acid substitutions can generally be described as the replacement of one amino acid residue by another amino acid residue of similar chemical structure and having little or essentially no effect on the function, activity, or other biological properties of the polypeptide.
  • Conservative amino acid substitutions are well known in the art.
  • Conservative substitutions may, for example, be the substitution of one amino acid in the following groups (a)-(e) by another amino acid in the same group: (a) Small aliphatic non-polar or weakly polar residues: Ala, Ser, Thr, Pro and Gly; (b) polar negatively charged residues and their (uncharged) amides: Asp, Asn, Glu and Gln; (c) polar positively charged residues: His, Arg and Lys; (d) large aliphatic non-polar residues: Met, Leu, Ile, Val and Cys; and (e) aromatic residues: Phe, Tyr and Trp.
  • the antibody molecules or single-chain antibodies provided herein may have at least 1 and no more than 10, such as no more than 5, 4, 3, 2 or 1 amino acids in their full-length or variable region sequences or CDR sequences. changes.
  • the antigen-binding fragment of the anti-EGFRvIII antibody is used to bind to the corresponding antigen (EGFRvIII).
  • the antigen-binding fragment is a single chain antibody (scFv).
  • scFv single chain antibody
  • the single-chain antibody (scFv) (one or more) formed therefrom can be used as the extracellular antigen-binding domain of the CAR.
  • Fusion proteins including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • Anti-EGFRvIII antibodies or antigen-binding fragments thereof can also form fusion proteins with other polypeptides.
  • an antigen-binding fragment with EGFRvIII binding ability and an Fc fragment can be linked to form a fusion protein, that is, a classic antibody molecule form.
  • the Fc fragment can be located at the C-terminal or N-terminal end of the EGFRvIII antibody antigen-binding fragment.
  • the Fc fragment may be located at the C-terminus of the EGFRvIII antibody antigen-binding fragment.
  • the fusion protein formed by the EGFRvIII antibody antigen-binding fragment and the Fc fragment has the ability to specifically bind to EGFRvIII and also has the effector function of the Fc fragment, such as mediating complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity ( ADCC), mediate phagocytosis, etc.
  • fusion with the Fc fragment can increase the half-life of the anti-EGFRvIII antibody antigen-binding fragment in the body, thereby increasing the dosing interval when the anti-EGFRvIII antibody antigen-binding fragment is used as a therapeutic drug.
  • an anti-EGFRvIII antibody or antigen-binding fragment thereof can be linked to a protein tag to form a fusion protein.
  • Protein tags can include purification tags and detectable tags. Purification tags include but are not limited to His6 tag, Flag tag, MBP tag, GST tag, SUMO) tag, etc.
  • the detectable label can be used to indicate the presence or content of the anti-EGFRvIII antibody or its antigen-binding fragment in a sample, or to track the location information of the anti-EGFRvIII antibody or its antigen-binding fragment in the subject's body or cells.
  • detectable labels include various enzymes that can be used in immunoassays, such as horseradish peroxidase (HRP), alkaline phosphatase (ALP), etc.; fluorescent proteins, such as GFP. Due to the specific binding ability of anti-EGFRvIII antibodies or antigen-binding fragments thereof to EGFRvIII, the amount of anti-EGFRvIII antibodies or antigen-binding fragments thereof can be determined by the amount of detectable tags linked to the anti-EGFRvIII antibodies or antigen-binding fragments thereof, and thereby Determine the presence or amount of EGFRvIII in the sample.
  • HRP horseradish peroxidase
  • ALP alkaline phosphatase
  • GFP fluorescent proteins
  • an anti-EGFRvIII antibody or antigen-binding fragment thereof can be linked to a cytokine or therapeutic protein to form a fusion protein.
  • the specific binding ability of anti-EGFRvIII antibodies or antigen-binding fragments thereof to EGFRvIII can be used to deliver cytokines or therapeutic proteins to specific tissues or cells (such as tumor tissues expressing EGFRvIII) in a targeted manner. Therapeutic effects of cytokines or therapeutic proteins.
  • Multispecific antibodies including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • multispecific antibody molecules that include at least one domain (or functional unit) that binds EGFRvIII and one or more additional binding domains.
  • the one or more additional binding domains may bind to a second antigen or protein other than EGFRvIII.
  • multispecific antibody molecules comprising at least two domains that bind EGFRvIII.
  • the two binding domains that bind to EGFRvIII are different in amino acid sequence, and preferably bind to different antigenic epitopes on EGFRvIII respectively.
  • the EGFRvIII-binding domain is in the form of a single-chain antibody; the one or more additional binding domains can be a single-domain antibody, a single-chain antibody, or other antigen-binding fragment.
  • the second antigen is a tumor associated antigen (TAA) or a tumor microenvironment associated antigen (TMEAA).
  • TAA tumor associated antigen
  • TAEAA tumor microenvironment associated antigen
  • the second antigen is an immunomodulatory antigen, wherein the antigen is associated with enhancing or inhibiting signaling pathways in immune cells.
  • the second antigen is a T cell surface molecule, such as a component of a T cell receptor complex, such as CD3 (including gamma, delta, epsilon, zeta, and n chains).
  • the multispecific binding peptide is a bispecific antibody molecule.
  • multispecific binding peptides also include Fc fragments.
  • the presence of the Fc fragment facilitates multimerization of binding domains and may provide associated effector functions.
  • Immunoconjugates including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • conjugates comprising at least one anti-EGFRvIII antibody or antigen-binding fragment thereof that specifically binds EGFRvIII provided herein and one or more other functional moieties.
  • the other moieties may be chemical groups, for example therapeutic agents, such as cytotoxic agents, or may be tracers.
  • the moiety can be a targeting moiety, a small molecule drug (e.g., a non-peptide drug less than 500 Da), a toxin, a cytostatic agent, a cytotoxic agent, an immunosuppressive agent, a radioactive agent suitable for diagnostic purposes, Radioactive metal ions, etc. for therapeutic purposes.
  • the immunoconjugate is an antibody drug conjugate (ADC) containing one or more anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein and a therapeutic agent that is cytotoxic, inhibits cell growth, Or provide some therapeutic benefit.
  • the cytotoxic agent is a chemotherapeutic agent, a drug, a growth inhibitor, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant or animal origin, or a fragment thereof), or a radioactive isotope (i.e., a radioconjugate ).
  • the antibody drug conjugates provided by the present invention allow targeted delivery of drug moieties to tumors. In some cases, this can result in targeted killing of tumor cells.
  • therapeutic agents include, for example, daunomycin, doxorubicin, methotrexate, vindesine, maytansinoid, and the like.
  • the therapeutic agent has intracellular activity.
  • the immunoconjugate coupled to the anti-EGFRvIII antibody or antigen-binding fragment thereof can be internalized by the cell, and the therapeutic agent has the activity of blocking cellular protein synthesis or blocking the activity of nucleic acid synthesis, thereby causing cell growth arrest or die.
  • the immunoconjugate contains one or more anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein and a tracer.
  • the immunoconjugate may be used for research or diagnostic purposes, such as for in vivo detection of cancer. Tracers can produce detectable signals directly or indirectly.
  • the tracer may be a radioisotope, such as 3 H, 14 C, 32 P, 35 S, 123 I; a fluorescent (fluorophore) or chemiluminescent (chromophore) compound, such as a fluorescent isothiocyanate, Rhodamine or fluorescein; developer; or metal ions.
  • the tracer is a radioactive atom, such as 99 Tc or 123 I, for scintillation studies, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, MRI),
  • NMR nuclear magnetic resonance
  • 89 Zr, 123 I, 19 F, 13 C, 15 N, 17 O, 89 Zr can be complexed with a variety of metal chelators and bound to antibodies, for example for PET imaging.
  • connection between the anti-EGFRvIII antibody or its antigen-binding fragment and other functional parts can be covalent or non-covalent.
  • non-covalent linkages may include via the Biotin-Avidin System.
  • covalent linkages may include various chemical linkers, including peptide linkers, cleavable linkers, or non-cleavable linkers.
  • linker components include 6-maleimidocaproyl ("MC”), maleimidopropionyl (“MP”), valine-citrulline (“val” -cit”), alanine-phenylalanine (“ala-phe”), p-aminobenzyloxycarbonyl (“PAB”), N-succinimidyl 4-(2-pyridylthio) methyl)valerate (“SPP”), N-succinimidyl 4-(N-maleimidomethyl)cyclohexane-1carboxylate (“SMCC”) and N -Succinimidyl(4-iodo-acetyl)aminobenzoate (“SIAB”).
  • MC 6-maleimidocaproyl
  • MP maleimidopropionyl
  • val valine-citrulline
  • alanine-phenylalanine ala-phe
  • PAB p-aminobenzyloxycarbonyl
  • SPP N-
  • the linker may comprise amino acid residues.
  • Exemplary amino acid linker components include dipeptides, tripeptides, tetrapeptides, or pentapeptides.
  • Exemplary dipeptides include: valine-citrulline (vc or val-cit), alanine-phenylalanine (afa-phe).
  • Exemplary tripeptides include: glycine-valine-citrulline (gly-val-cit) and glycine-glycine-glycine (gly-gly-gly).
  • Amino acid residues comprising the amino acid linker component include naturally occurring residues as well as non-naturally occurring amino acid analogs, such as citrulline.
  • Amino acid linker components can be designed and optimized in their selectivity for enzymatic cleavage by specific enzymes, such as tumor-associated proteases, cathepsins B, C and D, cytoplasmic proteases.
  • Conjugates of anti-EGFRvIII antibodies or antigen-binding fragments thereof and cytotoxic agents can be prepared using a variety of bifunctional protein coupling agents, such as N-succinimidyl-3-(2-pyridyldithiol)propane Acid esters (SPDP), imidothiolanes (IT), bifunctional derivatives of imide esters (such as dimethyl adipimidate HCl), active esters (such as disuccinimide amine base), aldehydes (such as glutaraldehyde), bisazido compounds (such as bis(p-azidobenzoyl)hexanediamine), biszo derivatives (such as bis-(p-azidobenzoyl)hexanediamine) )-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate) and dual-reactive fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenz
  • CARs Chimeric antigen receptors (CARs) including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • the chimeric antigen receptor (CAR) provided herein is an artificially constructed chimeric protein, including an extracellular antigen-binding domain that specifically binds EGFRvIII, a transmembrane domain, and one or more intracellular signaling structures. area. Characteristics of these CARs include their ability to redirect T cell specificity and responsiveness to EGFRvIII-expressing cells in a non-MHC-restricted manner. Non-MHC-restricted EGFRvIII recognition enables T cells or NK cells expressing the disclosed CARs to recognize antigens independent of antigen processing and kill EGFRvIII-expressing cells (such as tumor cells).
  • Intracellular signaling domains may include T cell receptor signaling domains, T cell costimulatory signaling domains, or both.
  • the T cell receptor signaling domain may include the intracellular domain of the T cell receptor, such as the intracellular portion of the CD3 ⁇ protein.
  • Costimulatory signaling domains are intracellular domains of costimulatory molecules. Costimulatory molecules are cell surface molecules other than the antigen receptor or its ligand that are required for effective lymphocyte response to antigen.
  • a CAR provided herein includes an extracellular antigen binding domain that specifically binds EGFRvIII.
  • the antigen-binding domain may be a scFv, which may be the heavy and light chain variable regions of any antibody or antigen-binding fragment thereof linked by a peptide linker that specifically binds to EGFRvIII.
  • the extracellular antigen binding domain may include the VHH from the single domain antibody without including any light chain variable region.
  • the extracellular antigen-binding domain of a CAR provided herein includes one or more antigen-binding fragments from an anti-EGFRvIII antibody provided herein, such as unhumanized or humanized single chain Antibodies (scFv).
  • an anti-EGFRvIII antibody provided herein, such as unhumanized or humanized single chain Antibodies (scFv).
  • the extracellular antigen-binding domain When the extracellular antigen-binding domain includes two or more antigen-binding fragments, they may be connected in series directly or through a peptide linker. Using two or more serially connected antigen-binding fragments as the extracellular antigen-binding domain or part thereof helps to promote the recognition and binding of the constructed CAR to target molecules (such as EGFRvIII). Preferably, these antigen-binding fragments each bind to different epitopes on EGFRvIII.
  • the CAR may include a signal peptide sequence, for example, located N-terminal to the antigen-binding domain. Any suitable signal peptide sequence can be used.
  • the signal peptide sequence is a CD8 ⁇ signal peptide.
  • the signal peptide sequence can promote the expression of CAR on the cell surface, the presence of the signal peptide sequence in the expressed CAR is not necessary for the CAR to function.
  • the signal peptide sequence may be cleaved from the CAR.
  • a CAR lacks a signal peptide sequence.
  • a hinge region (or spacer domain), which includes the polypeptide sequence.
  • the hinge region may contain up to 300 amino acids, preferably 10 to 100 amino acids.
  • a CAR provided herein includes a CD8 alpha protein hinge region.
  • the CAR may include a transmembrane domain fused to the extracellular domain of the CAR.
  • transmembrane domains that are naturally associated with other elements in the CAR, such as cross-linking regions or intracellular signaling domains, are used.
  • Transmembrane domains can be derived from natural or synthetic sources. Where the source is natural, the domain may be derived from any membrane-binding or transmembrane protein. Exemplary transmembrane domains for use in the disclosed CARs may include at least the alpha, beta or zeta chain of a T cell receptor, CD28, CD3 ⁇ , CD45, CD4, CD5, CDS, CD9, CD 16, CD22, CD33, Transmembrane domains of CD37, CD64, CD80, CD86, CD 134, CD137, and CD154. Alternatively, the transmembrane domain can be synthetic, in which case it will contain primarily hydrophobic residues such as leucine and valine. In some embodiments, a triplet of phenylalanine, tryptophan, and valine will be present at each end of the synthetic transmembrane domain.
  • a short oligopeptide or polypeptide linker may form the link between the transmembrane domain and the intracellular T cell signaling domain and/or T cell costimulatory domain of the CAR.
  • exemplary linker sequences include one or more glycine-serine doublets.
  • the transmembrane domain includes the transmembrane domain of a T cell receptor, such as the CD8 alpha transmembrane domain.
  • the intracellular region of the CAR includes one or more intracellular T cell signaling domains responsible for activating at least one normal effector function of the CAR-expressing T cells.
  • Exemplary T cell signaling domains are provided herein and are known to those of ordinary skill in the art.
  • the entire intracellular T cell signaling domain can be used in a CAR, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of an intracellular T cell signaling domain is used, the truncated portion can be used in place of the intact chain as long as it transduces the relevant T cell effector function signal.
  • intracellular T cell signaling domains used in CARs include the cytoplasmic sequences of the T cell receptor (TCR) and costimulatory molecules that cooperate to initiate signal transduction upon antigen receptor binding, as well as any of these sequences Derivatives or variants and any synthetic sequence having the same function.
  • TCR T cell receptor
  • T cell receptor signaling domains regulate activation of T cell receptor complexes in a stimulatory or inhibitory manner.
  • the CARs disclosed herein may include cytoplasmic signaling sequences that act in a stimulatory manner, which may contain signaling motifs known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • ITAM-containing primary cytoplasmic signaling sequences that may be included in the disclosed CARs include those from CD3 ⁇ , FcR ⁇ , FcR ⁇ , CD3 ⁇ , Intracellular domains of CD3 ⁇ , CD3 ⁇ , CDS, CD22, CD79a, CD79b and CD66d proteins.
  • the cytoplasmic signaling molecule in the CAR includes the intracellular T cell signaling domain from CD3 ⁇ .
  • the intracellular domain of the CAR provided herein may include a CD3 zeta chain portion and an intracellular costimulatory signaling domain.
  • the costimulatory signaling domain may include an intracellular domain of a costimulatory molecule.
  • Costimulatory molecules are cell surface molecules other than the antigen receptor or its ligand that are required for effective lymphocyte response to antigen. Examples of such molecules include CD27, CD28, 4-1BB (CD137), OX40 (CD134), CD30, CD40, PD-1, ICOS, Lymphocyte Function Associated Antigen 1 (LFA-1), CD2, CD7, LIGHT, NKG2C and B7-H3.
  • a CAR can include a CD3 ⁇ signaling domain, a CD8 signaling domain, a CD28 signaling domain, a 4-1BB signaling domain, or a combination of two or more thereof.
  • the intracellular domain includes the signaling domain of CD3- ⁇ and the signaling domain of CD28.
  • the cytoplasmic domain includes the signaling domain of CD3 ⁇ and the signaling domain of 4-1BB.
  • the cytoplasmic domain includes the signaling domain of CD3- ⁇ and the signaling domains of CD28 and CD137.
  • the cytoplasmic signaling sequences within the cytoplasmic signaling portion of the CAR of the invention can be linked to each other in a random or specified order.
  • short polypeptide linkers preferably between 2 and 10 amino acids in length, can form the linkage. Glycine-serine doublets provide particularly suitable linkers.
  • a spacer domain which includes the polypeptide sequence.
  • the spacer domain may comprise up to 300 amino acids, preferably 10 to 100 amino acids, and most preferably 25 to 50 amino acids.
  • CAR a parent CAR
  • those skilled in the art can modify it, for example, fuse it with other polypeptides, or retain only fragments of the CAR provided herein, and these fusion molecules or fragments have the parent CAR
  • At least part of the biological activity of a CAR is, for example, the activity of recognizing target cells (eg, tumor cells expressing EGFRvIII) or detecting, treating, or preventing disease.
  • the CARs provided herein may also include additional amino acids at the amino or carboxyl terminus, or at both termini, that are not present in the amino acid sequence of the parent CAR.
  • the additional amino acids do not interfere with the biological function of the CAR or functional portion, such as identifying target cells, detecting cancer, treating or preventing cancer, etc. More ideally, the additional amino acid enhances the biological activity compared to the biological activity of the parent CAR.
  • Functional variants of the CARs described herein that have substantial or significant sequence identity or similarity to the parent CAR, which functional variants retain the biological activity of the CAR of which they are a variant.
  • Functional variants encompass, for example, those variants of the CARs described herein (parental CAR) that retain the ability to recognize target cells to a similar degree, to the same degree, or to a greater degree than the parental CAR.
  • the functional variant may be, for example, at least about 30%, about 50%, about 75%, about 80%, about 85%, about 90%, about 91%, about 92% identical in amino acid sequence to the parent CAR. , about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99% or higher consistency.
  • Functional variants may, for example, include the amino acid sequence of a parent CAR with at least one conservative amino acid substitution.
  • functional variants may include the amino acid sequence of the parent CAR with at least one non-conservative amino acid substitution. In this case, it is preferred that the non-conservative amino acid substitutions do not interfere with or inhibit the biological activity of the functional variant. Non-conservative amino acid substitutions can enhance the biological activity of the functional variant, making the biological activity of the functional variant increased compared with the parental CAR.
  • the CARs provided herein may include synthetic amino acids in place of one or more naturally occurring amino acids.
  • Such synthetic amino acids are known in the art and include, for example, aminocyclohexanecarboxylic acid, norleucine, a-amino-n-decanoic acid, homoserine, S-acetamidomethyl-cysteine, trans- 3- and trans-4-hydroxyproline, 4-aminophenylalanine, 4-nitrophenylalanine, 4-chlorophenylalanine, 4-carboxyphenylalanine, ⁇ -phenyl Serine, ⁇ -hydroxyphenylalanine, phenylglycine, ⁇ -naphthylalanine, cyclohexylalanine, cyclohexylglycine, indoline-2-carboxylic acid, 1,2,3,4-tetrakis Hydroisoquinoline-3-carboxylic acid, aminomalonic acid, aminomalonic acid monoamide, N'-benzy
  • CARs provided herein can be glycosylated, amidated, carboxylated, phosphorylated, esterified, N-acylated, cyclized via, for example, disulfide bonds, or converted to acid addition salts and/or optionally dimerized Either polymerization or conjugation.
  • nucleic acid molecules encoding the disclosed chimeric antigen-binding receptors can be included in expression vectors (eg, lentiviral vectors) for expression in host cells, such as T cells, to prepare the disclosed CARs.
  • methods of using chimeric antigen receptors include isolating T cells from a subject, transforming the T cells with an expression vector encoding the chimeric antigen receptor (eg, a lentiviral vector), and expressing the chimeric antigen The recipient's engineered T cells are administered to the subject for treatment, for example, to treat a tumor in the subject.
  • the CARs provided herein may also be used in conjunction with safety mechanisms for controlling the activity of the CAR or CAR-expressing cells.
  • the CAR can be co-expressed with "EGFRt".
  • EGFRt refers to a polypeptide encoding a truncated human epidermal growth factor receptor that lacks the distal membrane EGF binding domain and cytoplasmic signaling tail but retains the extracellular epitope recognized by anti-EGFR antibodies.
  • EGFRt can be used as a non-immunogenic selection tool as well as a tracking marker with the function of genetically modifying cells. On the one hand, it can be used as a marker molecule for CAR-T cells.
  • CAR-T cells can also be used to eliminate CAR-T cells in the body when needed, such as through the ADCC pathway mediated by EGFR antibodies (e.g., cetuximab) ( See US8802374B2), which is used as a safety switch during clinical transformation.
  • EGFR antibodies e.g., cetuximab
  • the CAR-T cells provided in this article can also be considered for use in combination with safety switches of other mechanisms.
  • the CARs provided herein can be engineered so that they can utilize chemical inducers of dimerization (CIDs) to control CAR activity.
  • CIDs chemical inducers of dimerization
  • a complete CAR provided herein can be expressed as a separate fusion protein (e.g., the CD3 ⁇ intracellular domain is expressed separately from other parts) and utilizes a FKBP/FRB-based binding domain and rapamycin (The dimerization mechanism of rapamycin or rapamycin analogs) reconstitutes the intact CAR within the cell, allowing the integrity and activity of the CAR to be controlled by the presence or absence of rapamycin (e.g., see description in U.S. Patent 11,084,880) .
  • CAR-T cell activity can be controlled by degrading CAR molecules or causing CAR-T cell death. Examples of this include the use of PROTAC technology to degrade CAR molecules and the use of proteins with the ability to induce apoptosis, such as caspase-9.
  • compositions and methods of treatment including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof as well as fusion proteins, multispecific antibody molecules, and immunoconjugates including anti-EGFRvIII antibodies or antigen-binding fragments thereof disclosed herein can be used to administer to a subject for tumor prevention or treatment.
  • the effective dosage for this purpose may depend on the severity of the disease and the overall state of the patient's own immune system. Dosage regimens will also vary with the disease state and subject status, and will generally range from a single bolus dose or continuous infusion to multiple doses per day (eg, every 4-6 hours). Clinicians skilled in the art can readily determine whether a subject is a candidate for such treatment, for example, by utilizing clinical trials, physical examinations, and the subject's family history.
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof as well as fusion proteins, multispecific antibody molecules, and immunoconjugates including the anti-EGFRvIII antibodies or antigen-binding fragments thereof disclosed herein can be combined with one or more Co-administration of other drugs (e.g., antineoplastic agents).
  • other drugs e.g., antineoplastic agents
  • the disease or condition treated is a tumor, preferably glioblastoma.
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof (and fusion proteins, multispecific antibody molecules, immunoconjugates comprising the anti-EGFRvIII antibodies or antigen-binding fragments thereof) disclosed herein may be used using any suitable method or approach, and optionally combined with other antineoplastic agents. Routes of administration include, for example, oral, intravenous, intraperitoneal, subcutaneous or intramuscular administration.
  • nucleic acid molecules encoding anti-EGFRvIII antibodies or antigen-binding fragments thereof, expression vectors including the nucleic acid molecules, and host cells transfected with only the nucleic acid molecules or expression vectors provided herein can also be used for the above therapeutic purposes in various ways.
  • the expression vector can be introduced into the subject through gene therapy methods known in the art, and the target protein or polypeptide (anti-EGFRvIII antibody or antigen-binding fragment thereof) can be expressed to achieve therapeutic purposes.
  • Detection, diagnostic kits or treatment kits including anti-EGFRvIII antibodies or antigen-binding fragments thereof
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein, as well as other forms of molecules including the anti-EGFRvIII antibodies or antigen-binding fragments thereof (eg, fusion proteins or bispecific antibody molecules), can specifically bind to EGFRvIII in a sample.
  • detecting the amount of the anti-EGFRvIII antibody or its antigen-binding fragment-EGFRvIII complex formed, or the amount of the anti-EGFRvIII antibody or its antigen-binding fragment in the formed anti-EGFRvIII antibody or its antigen-binding fragment-EGFRvIII complex it is convenient to detect Determine the amount (or presence) of -EGFRvIII in a sample.
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein can be linked to various detection tags to facilitate detection by various means, including but not limited to bioluminescence, fluorescence, radioactive labeling , the product production volume of enzymatic reaction, etc.
  • detection tags including but not limited to bioluminescence, fluorescence, radioactive labeling , the product production volume of enzymatic reaction, etc.
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein, as well as other forms of molecules including the anti-EGFRvIII antibodies or antigen-binding fragments thereof (such as fusion proteins or bispecific antibody molecules), CAR-expressing cells, can be placed in a container to Form test, diagnostic or treatment kits.
  • These containers may be in the form of boxes, ampoules, vials, tubes, bags or other suitable containers known in the art.
  • These containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for preserving medications. If required, instructions for use are also provided with the container.
  • Instructions may generally include information on how to use an anti-EGFRvIII antibody or antigen-binding fragment thereof, a composition including an anti-EGFRvIII antibody or antigen-binding fragment thereof, or CAR-T cells for the treatment or prevention of tumors (e.g., glioblastoma),
  • a composition including an anti-EGFRvIII antibody or antigen-binding fragment thereof, or CAR-T cells for the treatment or prevention of tumors (e.g., glioblastoma)
  • the therapeutic agent such as an anti-EGFRvIII antibody or antigen-binding fragment thereof
  • dosage regimens for the treatment or prevention of neoplasia such as glioblastoma
  • precautions warnings; indications; contraindications; Adverse reactions; animal pharmacology; clinical studies; and/or reference materials.
  • Instructions may be printed directly on the container (if present), or as a label affixed to the container, or as a separate paper, booklet, card or folded print provided in
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein have a binding affinity to EGFRvIII that is close to, or better than, existing control antibodies.
  • the anti-EGFRvIII antibodies or antigen-binding fragments thereof provided herein are humanized and have a binding affinity to EGFRvIII that is close to, or better than, existing control antibodies.
  • this study also genetically modified second-generation CAR T cells in order to improve CAR T cells' resistance to the suppression of the tumor immune microenvironment and improve CAR T cell persistence and infiltration into entities. capabilities within the tumor.
  • This study simultaneously expressed dnTGF ⁇ RII on self-developed EGFRvIII-targeted CAR T cells, or converted the inhibitory signal into an IL7 activation signal through dnTGF ⁇ RII/. The added components significantly improved the anti-tumor efficiency of CAR T cells and resisted the inhibitory effect of TGF ⁇ . .
  • mice Before mouse immunization, 100 ⁇ l of mouse blood was collected, left to stand at 37°C for 5 minutes, and then centrifuged to collect serum as a negative serum control. Three methods were used to immunize mice. Group 1 was immunized using a combination of protein + peptide, group 2 was immunized using overexpressed EGFRvIII target cells, and group 3 was immunized using peptides. The adjuvant and antigen are mixed at a ratio of 1:1, emulsified, and the emulsified immunogen is injected subcutaneously into mice. One week after the second immunization, the blood of the mice was collected, the serum was separated, and the antibody titer was tested. Additional immunization was performed based on the antibody titer until the serum titer met the requirements, and the final vaccination was performed.
  • the positive cells and negative cells were F98npEGFRvIII and F98npEGFR (ATCC) respectively. Collect the digested cells into a centrifuge tube, put it into a centrifuge, and centrifuge at 300g for 5 minutes. Discard the supernatant and add an appropriate amount of 1 x Wash 2 times with PBS, discard the supernatant, resuspend the cells in 1 x PBS, add 50 ⁇ l of cell suspension to the 96-well plate, about 2 x 10 ⁇ 5 cells, add 1/50 and 1/500 dilutions to each well.
  • F98npEGFRvIII and F98npEGFR ATCC
  • Serum, positive control antibody C097VEJ300, isotype control mouse IgG and negative control PBS add 100 ⁇ l diluted fluorescent secondary antibody Alexa Fluor 647 AffiniPure Goat Anti-Mouse IgG, Fc ⁇ fragment specific (min X Hu Bov Hrs SrProt) to each well, shake at 4°C After incubation for 30 minutes, take out the 96-well plate, add 150 ⁇ l 1 x PBS to each well, wash twice, discard the supernatant, add 150 ⁇ l 1 x PBS to each well to resuspend the cells, perform flow cytometry detection, and read with a flow cytometer.
  • mice with the best titer test results were sacrificed and the mouse spleens were removed using autoclaved forceps in a clean bench and then soaked in sterile saline.
  • Use sterile scissors to fully mince the mouse spleen then transfer the minced spleen to a sterile 70 ⁇ m cell mesh, and use the inner tube of a disposable sterile syringe to fully grind the mouse spleen.
  • the mouse spleen cells ground on the cell screen were washed with RPMI-1640 culture medium, washed three times, and the washing fluid was collected to obtain the corresponding cell suspension. Centrifuge at 1000 rpm for 5 min to collect spleen cells.
  • SP 2/0 cells Resuspend and collect SP 2/0 cells in good growth status, centrifuge at 1000 rpm for 5 minutes, and wash and count the collected cells.
  • Mouse splenocytes SP 2/0 cells were mixed at a ratio of 5:1 and centrifuged. Flick to loosen the cell sediment and place in a 37°C water bath for 1 minute. Slowly add PEG-1450 solution (37°C) dropwise; centrifuge the dropped sample and discard the supernatant.
  • HAT screening medium Dilute the cell suspension prepared after spleen fusion of each mouse and inoculate it into a 96-well plate. Add 150 ⁇ l/well of the liquid and place it in a CO 2 cell culture incubator for culture. Replenish the solution every 3-5 days. After the bottom of the plate reaches the appropriate cell density, collect the supernatant for detection.
  • the positive cells and negative cells were F98npEGFRvIII and F98npEGFR respectively.
  • the supernatant sample and control were diluted and added to a 384-well plate. About 1x10 ⁇ 3 cells were added to each well, and an appropriate amount of CFSE was added. The cells are stained, mixed thoroughly, then add the secondary antibody of the corresponding species, incubate at room temperature, and use an instrument to perform detection readings.
  • the positive clones were further screened using FACS method.
  • the positive clones were subcloned using the limiting dilution method, and the cell subclones were cultured in 1xHT medium.
  • the subcloned cells were cultured in a 37°C, 5% carbon dioxide incubator. Clone screening was performed using flow cytometry.
  • the purified monoclonal antibody detection concentration is diluted into a series of gradient concentrations for SPR detection, and the EC50 is calculated (Figure 3).
  • RNA extraction kit to extract total RNA from monoclonal hybridoma cells.
  • a reverse transcription kit to use the extracted total RNA as a template to perform reverse transcription.
  • the antibody sequence is amplified by PCR and the antibody sequence is determined.
  • the maximum antigen concentration added is 16ug, and a total of 8 gradients are diluted.
  • the dilution system is as follows.
  • the CAR structure includes the developed murine antibody sequence ScFv structure against EGFRvIII antigen, and also includes but is not limited to the CD8 hinge region and transmembrane region, 41-BB activation signal and CD3z activation domain ( Figure 4), and can be based on this Other modifications are performed, including secretion of cytokines, secretion of chemokines, modification of immune checkpoint inhibitory genes, blocking of tumor microenvironment inhibitory signals and conversion to activated models and other fourth-generation CAR T.
  • the synthesized CAR gene sequence was connected to the lentiviral expression plasmid through molecular cloning.
  • the synthesized plasmid was transfected into competent cells Stbl3, and the plasmid was maximally extracted after shaking for subsequent lentiviral packaging.
  • Different CAR plasmids and packaging plasmids were co-transfected into 293T cells respectively.
  • the cell supernatants were collected 48 h after transfection and centrifuged at 4000 rpm for 10 min. Collect the supernatant, filter it with a 0.45 ⁇ m filter membrane, and use an ultracentrifuge to collect the lentivirus pellet.
  • the centrifugation conditions were 4°C, 100,000 ⁇ g, no brake, and centrifugation for 90 min. Dispense the concentrated lentivirus into 0.5 mL cryovials at 200 ⁇ l per tube, freeze them at -80°C, and use flow cytometry to detect the lentivirus titer.
  • the number of U87MG-EGFRvIII target cells in the sample well was 1 ⁇ 10 ⁇ 4/well, repeated for 6 wells, and 6 control wells were set at the same time, each well was 100 ⁇ l. After overnight, discard the supernatant and add CAR-T cells.
  • the two clone sequences with the best in vivo drug efficacy were selected for humanization of the antibody sequences.
  • the hybridoma V-type gene was codon optimized to be more suitable for mammalian expression and synthesized through overlap extension PCR.
  • the mouse monoclonal antibody V-type gene will be transplanted to the most suitable framework through CDR transplantation method for humanization. 12-16 humanized variants will be designed with minimal framework differences from the human germline V-gene.
  • Up to 3 variants will be designed for each site to remove key PTM sites in the murine antibody sequence.
  • humanized V-genes and PTM-removed V-genes will be synthesized.
  • the mouse and all variant genes will be cloned into the human IgG expression vector to produce the human IgG1 construct of the required isotype, the antibody gene will be synthesized, and purified after expression. After purification, the antibody affinity will be identified using the SPR method, and the best antibody will be selected. Optimal affinity humanized antibody sequences.
  • the CAR structure includes the developed murine antibody sequence ScFv structure against EGFRvIII antigen, and also includes but is not limited to the CD8 hinge region and transmembrane region, 41-BB activation signal and CD3z activation domain, and other modifications can be made on this basis. Including the secretion of cytokines, the secretion of chemokines, the modification of immune checkpoint inhibitory genes, the blocking and conversion of tumor microenvironment inhibitory signals into activated models and other four generations of CAR T.
  • the synthesized CAR gene sequence was connected to the lentiviral expression plasmid through molecular cloning.
  • the synthesized plasmid was transfected into competent cells Stbl3, and the plasmid was maximally extracted after shaking for subsequent lentiviral packaging.
  • Different CAR plasmids and packaging plasmids were co-transfected into 293T cells respectively.
  • the cell supernatants were collected 48 h after transfection and centrifuged at 4000 rpm for 10 min. Collect the supernatant, filter it with a 0.45 ⁇ m filter membrane, and use an ultracentrifuge to collect the lentivirus pellet.
  • the centrifugation conditions were 4°C, 100,000 ⁇ g, no brake, and centrifugation for 90 min. Dispense the concentrated lentivirus into 0.5 mL cryovials at 200 ⁇ l per tube, freeze them at -80°C, and use flow cytometry to detect the lentivirus titer.
  • the number of U87MG-EGFRvIII target cells in the sample well was 1 ⁇ 10 ⁇ 4/well, repeated for 6 wells, and 6 control wells were set at the same time, each well was 100 ⁇ l. After overnight, discard the supernatant and add CAR-T cells.
  • the number of U87MG-EGFRvIII target cells in the sample well was 1 ⁇ 10 ⁇ 4/well, repeated for 6 wells, and 6 control wells were set at the same time, each well was 100 ⁇ l. After overnight, discard the supernatant and add CAR-NK cells.
  • dnTGF ⁇ RII has the ability to bind TGF ⁇ , but it lacks the ability to convert bound TGF ⁇ into intracellular signals (for example, it lacks an intracellular domain or lacks the kinase activity required for signal transduction).
  • the second is to connect the chimeric switching receptor (dnTGF ⁇ RII-IL7RA TM-IL7RA intracellular domain) through the T2A self-cleaving peptide ( Figure 20B).
  • the existence of this chimeric switching receptor can resist TGF ⁇ inhibition, and can also convert bound TGF ⁇ into an intracellular IL7RA receptor stimulating signal to promote the proliferation and/or activity of CAR T cells.
  • the specific method is the same as the preparation process of humanized antibody CAR T cells.
  • the number of U87MG-EGFRvIII target cells in the sample well was 1 ⁇ 10 ⁇ 4/well, repeated for 6 wells, and 6 control wells were set at the same time, each well was 100 ⁇ l. After overnight, discard the supernatant and add CAR-T cells.
  • TGF ⁇ inhibition group was set up for comparison during killing. Before the CAR T cell killing experiment, some cells were taken and 30ng/ml TGF ⁇ was used to inhibit CAR T cells. , and then monitor the killing efficiency of CAR T cells after inhibition and CAR T cells without inhibition.
  • the CAR T cells prepared by detecting the mouse antibody sequence were tested for killing efficiency against target cells U87MG-EGFRvIII overexpressing EGFRvIII and target cells U87MG-EGFR expressing EGFR.
  • the test results showed that the prepared CAR T cells specifically killed overexpressed EGFRvIII. target cells, but almost no killing efficiency against target cells expressing EGFR, indicating that the CAR T cells prepared by the developed antibodies have specific killing efficiency ( Figures 6 and 7).
  • 20-2-1, 20-2-4, 20-2-25, 20-2-28, 20-2-34 and 20-2-40-2 have higher killing efficiency.
  • the mouse antibody was humanized through CDR transplantation and post-translational modification site removal.
  • the results showed that after humanization of clone #28, a humanized antibody sequence with the same affinity as the mouse antibody was obtained, #40-2
  • the affinity of cloned murine antibodies dropped approximately 2-fold after humanization (Tables 2 and 3, Figures 9 and 10).
  • the CAR T cells prepared by detecting human antibody sequences were tested for killing efficiency against target cells U87MG-EGFRvIII overexpressing EGFRvIII and target cells U87MG-EGFR expressing EGFR.
  • the test results showed that the prepared CAR T cells specifically killed overexpressed EGFRvIII. target cells, but almost no killing efficiency against target cells expressing EGFR, indicating that the CAR T cells prepared by the developed humanized antibodies have specific killing efficiency ( Figures 13, 14, 15 and 16).
  • CAR NK cells prepared from human antibody sequences specifically kill target cells that overexpress EGFRvIII, but have almost no killing efficiency against target cells that express EGFR ( Figures 19A and 19B).
  • U87MG-EGFRvIII was used to form subcutaneous tumors in mice.
  • the tumor volume grew to about 100 mm3 and CAR T cells were reinfused. After the CAR T cells were reinfused, the tumor volume of the mice was measured every other week.
  • the test results showed that 28-H9, 40- CAR T cells prepared from cloned antibody sequences 2-H10, 40-2-H12, 40-2-H13, and 40-2-H15 have significant tumor-inhibitory effects in vivo ( Figures 17 and 18).
  • CAR positivity rate detection results are shown in Table 4 and Figure 21A-C.
  • PE-EGFRvIII antigen protein is used to detect RII (CAR T cells expressing dnTGF ⁇ RII), T7R (CAR T cells expressing chimeric switching receptor dnTGF ⁇ RII-IL7R ⁇ ) or CAR T C cells (traditional second-generation CAR T cells, not expressing DNR or CSR) CAR positivity rate.
  • EGFRvIII is a mutated form of EGFR. It is a deletion of exons 2-7 of EGFR. A glycine is added at the junction of the deleted sequence to form a new antigenic epitope. Therefore, there is a high degree of similarity between the sequences of EGFRvIII and EGFR. Homology makes it very difficult to screen antibody sequences because EGFR is expressed in all epithelial cells, while EGFRvIII is a tumor-specific antigen and is only expressed on tumors. For the development of drugs targeting the EGFRvIII target, screening only specifically recognizes the EGFRvIII target antigen, but does not recognize the wild-type EGFR antigen. Therefore, the specificity of the antibody is required to be high.
  • this study optimized a variety of mouse immunization strategies and antibody screening strategies to screen out mouse-derived antibody sequences with high specificity and high affinity.
  • the use of mouse-derived sequences can prepare immune cells with significant anti-tumor effects. cell.
  • this study humanized the mouse antibody sequence based on the mouse antibody sequence. Humanized antibodies with high affinity and specificity were screened, and immune cells prepared from humanized antibodies with significant anti-tumor effects were developed, laying the foundation for the subsequent development of anti-tumor drugs.
  • Antibody drugs and cellular immunotherapy have made significant progress in the field of tumor treatment in recent years.
  • Glioma is a highly malignant tumor with no effective treatment. Therefore, there is an urgent need to develop drugs that can effectively treat brain glioma.
  • Research shows EGFRvIII antigen is expressed in 30-60% of glioma patients. Therefore, the developed EGFRvIII-targeting antibodies and immune cells prepared using this antibody sequence will be applicable to EGFRvIII-expressing gliomas and other patients with EGFRvIII-expressing gliomas. on EGFRvIII-expressing tumors.
  • antibodies make drugs targeted, antibody drugs currently come in many forms, including monoclonal antibodies, double antibodies, polyclonal antibodies, ADCs, and immune cells. Therefore, the antibodies developed in this study can be applied in different fields. In addition, since current targeted therapies require corresponding targeted diagnosis, the antibodies developed in this study can also be used in the development of diagnostic kits and companion diagnostic applications.
  • CAR T cells have made significant progress in hematological tumors, and CAR T cells are already on the market.
  • the CAR structure consists of an antigen recognition domain, a hinge region, a transmembrane region, a costimulatory domain and an activation domain.
  • the CAR T cells developed in this study Antibodies can be assembled with different structures to prepare various types of immune cells, including but not limited to T cells, NK cells, macrophages, monocytes, B cells, red blood cells, etc.

Abstract

提供了靶向EGFRvIII的抗体或其抗原结合片段。还提供了包括靶向EGFRvIII的抗原结合片段的CAR及其治疗和检测应用。

Description

靶向EGFRvIII的抗体及其在细胞免疫治疗的应用
相关申请的交叉援引
本申请要求申请号为CN202210936403.1、申请日为2022年8月5日的中国专利申请的优先权,在此通过引用将其全文并入本文。
技术领域
本文涉及靶向EGFRvIII的抗体或其抗原结合片段。本文还涉及包括靶向EGFRvIII的抗原结合片段的CAR及其应用。
背景技术
细胞免疫治疗是肿瘤治疗新兴技术,在肿瘤治疗领域展现出良好的应用前景,工程T细胞在治疗血液肿瘤方面取得了成功,基于其在临床试验中前所未有的疗效,截止目前,全球已有8款嵌合抗原受体(CAR)-T细胞疗法被批准用于治疗B细胞恶性肿瘤和骨髓瘤。
细胞免疫治疗在血液肿瘤上取得显著的疗效,但是在实体肿瘤的治疗上受到一些限制因素,比如实体肿瘤免疫抑制微环境的存在以及CAR-T细胞在肿瘤内持续性和浸润能力差等,这些限制因素需要在细胞免疫治疗产品上进行创新型修饰和突破(Akhavan et al.,2019)。
细胞免疫治疗另一个关键的决定因素是靶点的选择,靶点的选择决定了细胞治疗产品的安全性和有效性,肿瘤抗原的分为肿瘤相关抗原(TAA)和肿瘤特异性抗原(TSA),肿瘤相关抗原在正常组织中有表达,细胞治疗产品靶向肿瘤相关抗原易产生脱靶毒性,而靶向肿瘤特异性抗原的细胞治疗产品的安全高兴更高。
EGFRvIII抗原是肿瘤特异性靶抗原,是EGFR基因最常见胞外区域突变形式(Chistiakov et al.,2017)。它在EGFR编码区域的外显子2-7之间移去了801个碱基,较野生型EGFR在胞外区域少了267个氨基酸,近而在外显子1和8之间产生了融合区域以及一个新的甘氨酸残基(图1,An et al.,2018),形成肿瘤特异的缺失突变。与野生型EGFR相比,EGFRvIII不依赖于配体,是组成型激活(Chistiakov et al.,2017)。该突变在多种癌症中(如乳腺癌,肺癌,头颈癌等)都有发现,但在胶质母细胞瘤中最为常见(Chistiakov et al.,2017;Del Vecchio et al.,2012)。
胶质母细胞瘤约占脑肿瘤的15%,全球预计每年将近210000人被诊断出来。胶质母细胞瘤是恶性4级肿瘤,具有高度侵袭性、易复发、致死致残率高等特点。胶质母细胞瘤发展迅速,5年总生存率是所有人类癌症中最差的肿瘤之一,没有有效的治疗手段。目前, 科学工作者正在计划通过免疫疗法来解决这一世界难题(Feldman et al.,2022;Londhe and Date,2020)。
研究结果显示EGFRvIII在胶质母细胞瘤中表达,而在正常组织中不表达,恶性程度越高的患者EGFRvIII表达率越高,因此,靶向EGFRvIII抗原的CAR-T细胞用于治疗脑胶质瘤,具有特异性强,脱靶毒性低,副作用小等优势(Zhu et al.,2021)。
已有临床研究表明靶向EGFRvIII的抗肿瘤药物具有显著的安全性和初步的疗效(O'Rourke et al.,2017),抗体药物是近年来发展比较成熟的一种有效的抗癌药物,它可以靶向特定抗原的肿瘤细胞并利用ADCC和CDC等效应,激发患者的免疫系统对抗癌细胞。
目前,市场仍亟需有更佳安全性和疗效的靶向EGFRvIII的抗肿瘤药物。
发明内容
在一方面,本文提供了靶向EGFRvIII的抗体或其抗原结合片段,所述抗体包括重链可变区(HCVR),所述重链可变区包括HCDR1、HCDR2和HCDR3,所述HCDR1、HCDR2和HCDR3选自如下组合之一:
(1)HCDR1的氨基酸序列为DFSMH(SEQ ID NO:1);
HCDR2的氨基酸序列为WINTETGEPSYADDFKG(SEQ ID NO:2);
HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:3);
(2)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:4);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:5);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:6);
(3)HCDR1的氨基酸序列为DYSIH(SEQ ID NO:7);
HCDR2的氨基酸序列为WINTETGEPTYADDFKG(SEQ ID NO:8);
HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:9);
(4)HCDR1的氨基酸序列为DYYLH(SEQ ID NO:10);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:11);
HCDR3的氨基酸序列为GYLTY(SEQ ID NO:12);
(5)HCDR1的氨基酸序列为RYWMH(SEQ ID NO:13);
HCDR2的氨基酸序列为EINPSNGRANYNEKFMS(SEQ ID NO:14);
HCDR3的氨基酸序列为GREITTGFAY(SEQ ID NO:15);
(6)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:16);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:17);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:18);
(7)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:19);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:20);
HCDR3的氨基酸序列为GYLAY(SEQ ID NO:21);
(8)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:22);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:23);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:25);
(9)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:25);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:26);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:27);
(10)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:28);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:29);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:30);
(11)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:31);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:32);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:33);
(12)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:34);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:35);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:36);
(13)HCDR1的氨基酸序列为NYAMS(SEQ ID NO:37);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:38);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:39);
(14)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:40);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:41);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:42);
(15)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:43);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:44);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:45);
(16)HCDR1的氨基酸序列为SGYSWH(SEQ ID NO:46);
HCDR2的氨基酸序列为YIHYSGSTNYNPPLKS(SEQ ID NO:47);
HCDR3的氨基酸序列为GVVSNYAMGN(SEQ ID NO:48);
(17)HCDR1的氨基酸序列为TYWMH(SEQ ID NO:49);
HCDR2的氨基酸序列为YINPNTAYTEYNQNFKD(SEQ ID NO:50);
HCDR3的氨基酸序列为GAYYRTYYAMDY(SEQ ID NO:51);
(18)HCDR1的氨基酸序列为NYGMN(SEQ ID NO:52);
HCDR2的氨基酸序列为WINTYTGEPTYADDFKG(SEQ ID NO:53);
HCDR3的氨基酸序列为EEFYSRGAMDY(SEQ ID NO:54);以及
(19)HCDR1的氨基酸序列为DYYIN(SEQ ID NO:55);
HCDR2的氨基酸序列为WIYPGSGNTKYNEKFKG(SEQ ID NO:56);
HCDR3的氨基酸序列为SSRCDF(SEQ ID NO:57),或者
所述抗体包括(1)-(19)任一项中的HCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的HCDR序列相比,具有至少90%的序列一致性,或在HCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
在一些实施方案中,所述抗体还包括轻链可变区(LCVR),所述轻链可变区包括LCDR1、LCDR2和LCDR3,所述LCDR1、LCDR2和LCDR3选自如下组合之一:
(1)LCDR1的氨基酸序列为SASSSISSNYLH(SEQ ID NO:58);
LCDR2的氨基酸序列为GTSNLAS(SEQ ID NO:59);
LCDR3的氨基酸序列为HQGSSIPLT(SEQ ID NO:60);
(2)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:61);
LCDR2的氨基酸序列为FASTRAS(SEQ ID NO:62);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:63);
(3)LCDR1的氨基酸序列为SASSGISSNYLH(SEQ ID NO:64);
LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:65);
LCDR3的氨基酸序列为HQGSDIPLT(SEQ ID NO:66);
(4)LCDR1的氨基酸序列为KSSQNLLNSSNQKNYLA(SEQ ID NO:67);
LCDR2的氨基酸序列为FASTRYS(SEQ ID NO:68);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:69);
(5)LCDR1的氨基酸序列为KASQSVSNDVV(SEQ ID NO:70);
LCDR2的氨基酸序列为YASNRYT(SEQ ID NO:71);
LCDR3的氨基酸序列为QQDYSSPWT(SEQ ID NO:72);
(6)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:73);
LCDR2的氨基酸序列为FASTRES(SEQ ID NO:74);
LCDR3的氨基酸序列为QQHYSIPLT(SEQ ID NO:75);
(7)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:76);
LCDR2的氨基酸序列为FASTRKS(SEQ ID NO:77);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:78);
(8)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:79);
LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:80);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:81);
(9)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:82);
LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:83);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:84);
(10)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:85);
LCDR2的氨基酸序列为FASTRGS(SEQ ID NO:86);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:87);
(11)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:88);
LCDR2的氨基酸序列为FASTRDS(SEQ ID NO:89);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:90);
(12)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:91);
LCDR2的氨基酸序列为FASTRES(SEQ ID NO:92);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:93);
(13)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:94);
LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:95);
LCDR3的氨基酸序列为SQSIHVPWT(SEQ ID NO:96);
(14)LCDR1的氨基酸序列为SASSSVSYMH(SEQ ID NO:97);
LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:98);
LCDR3的氨基酸序列为QQRSSYPLT(SEQ ID NO:99);
(15)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:100);
LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:101);
LCDR3的氨基酸序列为SQTTQVPWT(SEQ ID NO:102);
(16)LCDR1的氨基酸序列为ITNTDIDDDMN(SEQ ID NO:103);
LCDR2的氨基酸序列为EGNTLRP(SEQ ID NO:104);
LCDR3的氨基酸序列为LQSDDLPLT(SEQ ID NO:105);
(17)LCDR1的氨基酸序列为KASQSVDYDGDSYMN(SEQ ID NO:106);
LCDR2的氨基酸序列为AASNLES(SEQ ID NO:107);
LCDR3的氨基酸序列为LQSNEDPYT(SEQ ID NO:108);
(18)LCDR1的氨基酸序列为RSSQFIVHSNGNTYLE(SEQ ID NO:109);
LCDR2的氨基酸序列为KISNRFS(SEQ ID NO:110);
LCDR3的氨基酸序列为FQGSHVPFT(SEQ ID NO:111);以及
(19)LCDR1的氨基酸序列为KASEDIYNRLA(SEQ ID NO:112);
LCDR2的氨基酸序列为GATSLET(SEQ ID NO:113);
LCDR3的氨基酸序列为QQYWSSPLT(SEQ ID NO:114),或者
所述抗体包括(1)-(19)任一项中的LCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的LCDR序列相比,具有至少90%的序列一致性,或在LCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
在一些实施方案中,所述重链可变区的氨基酸序列选自如下任一项:
(1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
(19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
在一些实施方案中,所述抗体的重链可变区的氨基酸序列和轻链可变区的氨基酸序列选自如下任一组合:
(1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:116所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:118所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:120所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:122所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:124所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:126所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:128所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:130所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:132所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:134所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:136所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:138所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:140所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:142所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:144所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:146所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:148所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:150所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;以及
(19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:152所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列。
在一些实施方案中,通过表面等离子共振(SPR)测定的所述抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
在一些实施方案中,所述抗体选自鼠源抗体、嵌合抗体、人源化抗体或人抗体。
在一些实施方案中,通过表面等离子共振(SPR)测定的所述人源化抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
在一些实施方案中,所述抗原结合片段包括Fab、Fab’、F(ab’)2、单域抗体或单链抗体。
在一些实施方案中,所述人源化抗体为单链抗体,其氨基酸序列选自如下任一项:
(1)SEQ ID NO:167所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(2)SEQ ID NO:169所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(3)SEQ ID NO:171所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(4)SEQ ID NO:173所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(5)SEQ ID NO:175所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(6)SEQ ID NO:177所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(7)SEQ ID NO:179所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(8)SEQ ID NO:181所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(9)SEQ ID NO:183所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(10)SEQ ID NO:185所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
(11)SEQ ID NO:187所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
在一些实施方案中,所述抗体或其抗原结合片段还包括Fc片段。
在一些实施方案中,所述抗原结合片段为单链抗体;优选地,用于连接所述单链抗体中的重链可变区和轻链可变区的接头序列(linker)包括SEQ ID NO:166所示序列。
另一方面,本文提供了嵌合抗原受体(CAR),其胞外抗原结合结构域包括一个或更多个靶向EGFRvIII的抗体分子或其抗原结合片段,所述抗体分子的重链可变区的HCDR1、HCDR2和HCDR3选自如下组合之一:
(1)HCDR1的氨基酸序列为DFSMH(SEQ ID NO:1);
HCDR2的氨基酸序列为WINTETGEPSYADDFKG(SEQ ID NO:2);
HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:3);
(2)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:4);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:5);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:6);
(3)HCDR1的氨基酸序列为DYSIH(SEQ ID NO:7);
HCDR2的氨基酸序列为WINTETGEPTYADDFKG(SEQ ID NO:8);
HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:9);
(4)HCDR1的氨基酸序列为DYYLH(SEQ ID NO:10);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:11);
HCDR3的氨基酸序列为GYLTY(SEQ ID NO:12);
(5)HCDR1的氨基酸序列为RYWMH(SEQ ID NO:13);
HCDR2的氨基酸序列为EINPSNGRANYNEKFMS(SEQ ID NO:14);
HCDR3的氨基酸序列为GREITTGFAY(SEQ ID NO:15);
(6)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:16);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:17);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:18);
(7)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:19);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:20);
HCDR3的氨基酸序列为GYLAY(SEQ ID NO:21);
(8)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:22);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:23);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:25);
(9)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:25);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:26);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:27);
(10)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:28);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:29);
HCDR3的氨基酸序列为GWFAY(SEQ ID NO:30);
(11)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:31);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:32);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:33);
(12)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:34);
HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:35);
HCDR3的氨基酸序列为GYLVY(SEQ ID NO:36);
(13)HCDR1的氨基酸序列为NYAMS(SEQ ID NO:37);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:38);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:39);
(14)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:40);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:41);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:42);
(15)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:43);
HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:44);
HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:45);
(16)HCDR1的氨基酸序列为SGYSWH(SEQ ID NO:46);
HCDR2的氨基酸序列为YIHYSGSTNYNPPLKS(SEQ ID NO:47);
HCDR3的氨基酸序列为GVVSNYAMGN(SEQ ID NO:48);
(17)HCDR1的氨基酸序列为TYWMH(SEQ ID NO:49);
HCDR2的氨基酸序列为YINPNTAYTEYNQNFKD(SEQ ID NO:50);
HCDR3的氨基酸序列为GAYYRTYYAMDY(SEQ ID NO:51);
(18)HCDR1的氨基酸序列为NYGMN(SEQ ID NO:52);
HCDR2的氨基酸序列为WINTYTGEPTYADDFKG(SEQ ID NO:53);
HCDR3的氨基酸序列为EEFYSRGAMDY(SEQ ID NO:54);以及
(19)HCDR1的氨基酸序列为DYYIN(SEQ ID NO:55);
HCDR2的氨基酸序列为WIYPGSGNTKYNEKFKG(SEQ ID NO:56);
HCDR3的氨基酸序列为SSRCDF(SEQ ID NO:57),或者
所述抗体包括(1)-(19)任一项中的HCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的HCDR序列相比,具有至少90%的序列一致性,或在HCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
在一些实施方案中,所述抗体还包括轻链可变区(LCVR),所述轻链可变区包括LCDR1、LCDR2和LCDR3,所述LCDR1、LCDR2和LCDR3选自如下组合之一:
(1)LCDR1的氨基酸序列为SASSSISSNYLH(SEQ ID NO:58);
LCDR2的氨基酸序列为GTSNLAS(SEQ ID NO:59);
LCDR3的氨基酸序列为HQGSSIPLT(SEQ ID NO:60);
(2)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:61);
LCDR2的氨基酸序列为FASTRAS(SEQ ID NO:62);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:63);
(3)LCDR1的氨基酸序列为SASSGISSNYLH(SEQ ID NO:64);
LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:65);
LCDR3的氨基酸序列为HQGSDIPLT(SEQ ID NO:66);
(4)LCDR1的氨基酸序列为KSSQNLLNSSNQKNYLA(SEQ ID NO:67);
LCDR2的氨基酸序列为FASTRYS(SEQ ID NO:68);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:69);
(5)LCDR1的氨基酸序列为KASQSVSNDVV(SEQ ID NO:70);
LCDR2的氨基酸序列为YASNRYT(SEQ ID NO:71);
LCDR3的氨基酸序列为QQDYSSPWT(SEQ ID NO:72);
(6)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:73);
LCDR2的氨基酸序列为FASTRES(SEQ ID NO:74);
LCDR3的氨基酸序列为QQHYSIPLT(SEQ ID NO:75);
(7)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:76);
LCDR2的氨基酸序列为FASTRKS(SEQ ID NO:77);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:78);
(8)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:79);
LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:80);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:81);
(9)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:82);
LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:83);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:84);
(10)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:85);
LCDR2的氨基酸序列为FASTRGS(SEQ ID NO:86);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:87);
(11)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:88);
LCDR2的氨基酸序列为FASTRDS(SEQ ID NO:89);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:90);
(12)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:91);
LCDR2的氨基酸序列为FASTRES(SEQ ID NO:92);
LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:93);
(13)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:94);
LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:95);
LCDR3的氨基酸序列为SQSIHVPWT(SEQ ID NO:96);
(14)LCDR1的氨基酸序列为SASSSVSYMH(SEQ ID NO:97);
LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:98);
LCDR3的氨基酸序列为QQRSSYPLT(SEQ ID NO:99);
(15)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:100);
LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:101);
LCDR3的氨基酸序列为SQTTQVPWT(SEQ ID NO:102);
(16)LCDR1的氨基酸序列为ITNTDIDDDMN(SEQ ID NO:103);
LCDR2的氨基酸序列为EGNTLRP(SEQ ID NO:104);
LCDR3的氨基酸序列为LQSDDLPLT(SEQ ID NO:105);
(17)LCDR1的氨基酸序列为KASQSVDYDGDSYMN(SEQ ID NO:106);
LCDR2的氨基酸序列为AASNLES(SEQ ID NO:107);
LCDR3的氨基酸序列为LQSNEDPYT(SEQ ID NO:108);
(18)LCDR1的氨基酸序列为RSSQFIVHSNGNTYLE(SEQ ID NO:109);
LCDR2的氨基酸序列为KISNRFS(SEQ ID NO:110);
LCDR3的氨基酸序列为FQGSHVPFT(SEQ ID NO:111);以及
(19)LCDR1的氨基酸序列为KASEDIYNRLA(SEQ ID NO:112);
LCDR2的氨基酸序列为GATSLET(SEQ ID NO:113);
LCDR3的氨基酸序列为QQYWSSPLT(SEQ ID NO:114),或者
所述抗体包括(1)-(19)任一项中的LCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的LCDR序列相比,具有至少90%的序列一致性,或在LCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
在一些实施方案中,所述重链可变区的氨基酸序列选自如下任一项:
(1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
(19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
在一些实施方案中,所述抗体的重链可变区的氨基酸序列和轻链可变区的氨基酸序列选自如下任一组合:
(1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:116所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:118所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:120所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:122所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:124所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:126所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:128所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:130所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:132所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:134所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:136所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:138所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:140所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:142所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:144所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:146所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:148所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
(18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:150所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;以及
(19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:152所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列。
在一些实施方案中,通过表面等离子共振(SPR)测定的所述抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
在一些实施方案中,所述抗体选自鼠源抗体、嵌合抗体、人源化抗体或人抗体。
在一些实施方案中,通过表面等离子共振(SPR)测定的所述人源化抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
在一些实施方案中,所述抗原结合片段为单链抗体;优选地,用于连接所述单链抗体中的重链可变区和轻链可变区的接头序列(linker)包括SEQ ID NO:166所示序列。
在一些实施方案中,所述人源化抗体为单链抗体,其氨基酸序列选自如下任一项:
(1)SEQ ID NO:167所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(2)SEQ ID NO:169所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(3)SEQ ID NO:171所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(4)SEQ ID NO:173所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(5)SEQ ID NO:175所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(6)SEQ ID NO:177所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(7)SEQ ID NO:179所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(8)SEQ ID NO:181所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
(9)SEQ ID NO:183所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
(10)SEQ ID NO:185所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
(11)SEQ ID NO:187所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
在一些实施方案中,所述CAR还包括铰链区、跨膜区、共刺激结构域和激活结构域;优选地,所述铰链区为CD8铰链区,所述跨膜区为CD8α跨膜区;所述共刺激结构域为41BB共刺激结构域或CD28共刺激结构域;所述激活结构域为CD3ζ激活结构域。
在一些实施方案中,所述铰链区包括SEQ ID NO:156所示序列;所述跨膜区包括SEQ ID NO:158所示序列;所述共刺激结构域包括SEQ ID NO:160和/或162所示序列;所述激活结构域包括SEQ ID NO:164所示序列。
在一些实施方案中,所述CAR还包括信号肽;优选地,所述信号肽为CD8α信号肽;更优选地,所述信号肽包括SEQ ID NO:154所示序列。
在一些实施方案中,所述信号肽包括SEQ ID NO:154所示序列。
另一方面,本文提供了核酸分子,其编码:1)述抗体或其抗原结合片段,或上述抗体或其抗原结合片段的重链可变区和/或轻链可变区;或2)上述CAR。
在一些实施方案中,所述核酸分子包括SEQ ID NO:153、155、157、159、161、163、165、168、170、172、174、176、178、180、182、184、186或188任一项所示的核苷酸序列。
另一方面,本文提供了表达载体,其包括上述核酸分子;优选地,所述表达载体为慢病毒载体。
另一方面,本文提供了包括上述表达载体的细胞。
另一方面,本文提供了表达上述CAR的细胞。
在一些实施方案中,所述细胞为免疫细胞。
在一些实施方案中,所述细胞为T细胞或NK细胞。
在一些实施方案中,所述细胞还经改造以表达显性负性受体(DNR)或嵌合转换受体(CSR);优选地,所述DNR为dnTGFβRII和/或dnPD1;优选地,所述CSR的胞内结构域来自IL7Rα.;更优选地,所述dnTGFβRII包括SEQ ID NO:193所示序列,所述CSR的胞内结构域包括SEQ ID NO:197所示序列。
另一方面,本文提供了药物组合物,其包括:1)上述抗体或其抗原结合片段、核酸分子、表达载体或细胞;以及2)药学上可接受的载体。
另一方面,本文提供了上述抗体或其抗原结合片段、核酸分子、表达载体或细胞在制备用于治疗肿瘤的药物中的用途。
在一些实施方案中,所述肿瘤表达EGFRvIII。
在一些实施方案中,所述肿瘤为胶质母细胞瘤。
另一方面,本文提供了在受试者中治疗肿瘤的方法,包括以治疗有效量的上述抗体或其抗原结合片段、核酸分子、表达载体、细胞或药物组合物向所述受试者给药。
在一些实施方案中,所述肿瘤表达EGFRvIII。
在一些实施方案中,所述肿瘤为胶质母细胞瘤。
另一方面,本文提供了多特异性抗体分子,其至少包括第一功能部分和第二功能部分,其中第一功能部分包括上述抗体或其抗原结合片段;第二功能部分具有与所述第一功能部分不同的抗原结合特异性。
在一些实施方案中,所述第二功能部分对T细胞具有结合特异性。
另一方面,本文提供了免疫偶联物,其包括与治疗剂连接的上述抗体或其抗原结合片段。
在一些实施方案中,所述治疗剂是药物。
在一些实施方案中,所述治疗剂是细胞毒素。
在一些实施方案中,所述治疗剂是放射性同位素。
另一方面,本文提供了试剂盒,其用于检测样品中是否存在EGFRvIII或其含量,其中所述试剂盒包括上述抗体或其抗原结合片段。
附图说明
图1为EGFRvIII的结构示意图。
图2显示了流式检测阳性亚克隆上清对靶细胞的结合能力的结果。
图3显示了通过SPR方法检测纯化的部分鼠源抗体亲和力的结果。
图4为本文所采用的CAR的结构示意图。
图5显示了鼠源抗体序列CAR T细胞的CAR阳性率结果。
图6显示了鼠源抗体序列CAR T细胞对U87MG-EGFRvIII靶细胞的杀伤效率。
图7显示了鼠源抗体序列CAR T细胞对U87MG-EGFR靶细胞的杀伤效率。
图8显示了鼠源抗体序列CAR T细胞的体内抑瘤效率。
图9显示了SPR方法检测纯化的#28克隆人源化抗体亲和力结果。
图10显示了SPR方法检测纯化的#40-2克隆人源化抗体亲和力结果。
图11显示了#28克隆人源化抗体制备CAR T细胞的CAR阳性率。
图12显示了#40-2克隆人源化抗体制备CAR T细胞的CAR阳性率。
图13显示了#28克隆人源化抗体CAR T细胞对U87MG EGFRvIII靶细胞杀伤效率。
图14显示了#28克隆人源化抗体CAR T细胞对U87MG EGFR靶细胞杀伤效率。
图15显示了#40-2克隆人源化抗体CAR T细胞对U87MG EGFRvIII靶细胞杀伤效率。
图16显示了#40-2克隆人源化抗体CAR T细胞对U87MG EGFR靶细胞杀伤效率。
图17显示了克隆人源化抗体CAR T细胞体内抑瘤效率。
图18显示了克隆人源化抗体CAR T细胞体内抑瘤效率。
图19显示了#28克隆人源化抗体制备CAR NK细胞的靶细胞杀伤效率。(A)U87MG EGFRvIII靶细胞;(B)U87MG EGFR靶细胞。
图20显示了带有DNR(A)或CSR(B)的CAR构建体示意图。
图21显示了本申请构建的第四代CAR T细胞的CAR阳性率检测结果。(A)RII细胞(B);T7R细胞;(C)CAR T C细胞。
图22显示了CAR T细胞杀伤效率检测结果。(A)不使用TGFβ处理时的杀伤率;(B)使用TGFβ处理时的杀伤率;(C)杀伤率变化。
图23CAR T细胞的体内药效研究结果。图中CAR-T-C指对照组CAR T细胞;RII指表达显性负性受体dnTGF-βRII的CAR T细胞;T7R指表达嵌合转换受体dnTGF-βRII-IL7Rα的CAR T细胞。
具体实施方式
除非另有说明,本文使用的所有技术和科学术语具有本领域普通技术人员所通常理解的含义。
抗体指由浆细胞(效应B细胞)分泌、被机体免疫系统用来中和外来物质(多肽、病毒、细菌等)的免疫球蛋白。该外来物质相应地称作抗原。经典抗体分子的基本结构是由2个相同重链和2个相同轻链组成的4聚体。根据氨基酸序列的保守性差异,将重链和轻链分为位于氨基端的可变区(V)和位于羧基端的恒定区一条重链和一条轻链的可变区相互作用形成了抗原结合部位(Fv)。在可变区中,某些区域氨基酸残基的组成和排列次序比可变区内的其它区域(骨架区,FR)更易变化,称为高变区(HVR),高变区实际上是抗体与抗原结合的关键部位。由于这些高变区序列与抗原决定簇互补,故又称为互补决定区(complementarity-determining region,CDR)。重链和轻链均具有三个互补决定区,分别称为HCDR1、HCDR2、HCDR3和LCDR1、LCDR2、LCDR3。本领域已知还可以对抗体分子进行多种修饰,例如PEG化、糖基化、形成抗体分子偶联物(例如ADC)、加纯化标签、或者与其他蛋白融合,例如形成双特异性抗体。在本公开提供的抗体分子基础上进行修饰获得的这些抗体衍生物也应涵盖在本发明的范围内。
在已知抗体互补决定区序列的情况下,本领域技术人员可容易地例如通过DNA重组技术将抗体分子的部分序列(例如恒定区或骨架区)替换为来自其他物种的序列,形成嵌合抗体,而该嵌合抗体可基本上保留其来源抗体的结合特异性。例如,在来源抗体为鼠抗体时,可将其恒定区替换为人抗体恒定区,或者相反,在来源抗体为人抗体时,可将其恒定区替换为鼠抗体恒定区,以减弱抗体在不同物种中使用时的免疫免疫原性或者为了利用恒定区的特定功能,例如ADCC相关活性。出于进一步降低抗体免疫原性或其他目的,可将抗体分子中除CDR序列之外的其他序列都用另一抗体分子(来自相同或不同物种)的对应序列(视情况可包括一个或数个氨基酸突变)替换,同时基本上保留其来源抗体的结合特异性。在一个具体实例中,本领域技术人员经常采用CDR移植来将鼠源抗体人源化。
抗体分子的“抗原结合片段”指抗体分子中参与抗原特异性结合的氨基酸片段,例如,Fab、Fab’和(Fab’)2等。本领域技术人员已知如何获得这些抗原结合片段。例如,经典抗体分子可经木瓜蛋白酶消化而得到Fab片段,经胃蛋白酶消化得到F(ab’)2,通过以还原剂处理断开F(ab’)2铰链区之间的二硫键而形成Fab’片段。具有抗原结合能力的单链抗体和单域抗体为单肽链抗体分子,可视为经典抗体分子的“抗原结合片段”。
“Fc片段”指Y”形抗体分子的柄部区域,即可结晶片段(fragment crystallizable,Fc)包括重链的第二和第三恒定结构域(CH2和CH3结构域)。可通过蛋白水解酶(如木瓜蛋白酶)水解抗体分子得到抗体Fc区。在一些实例中,Fc区可包含铰链、CH2和CH3。当Fc区包含铰链时可介导两个含Fc的多肽之间的二聚化。Fc片段可来自IgG、IgM、IgD、IgE或IgA。在一些实例中,Fc区来自IgG1、IgG2、IgG3或IgG4。“Fc片段”还包括来自天然Fc片段,经改动但仍保持其效应功能的变体Fc片段。“变体Fc片段”包含在天然Fc片段的氨基酸序列上具有至少一个氨基酸变动的氨基酸序列。在一些实例中,变体Fc片段相比于亲本Fc片段(天然Fc片段)具有至少一个氨基酸取代,例如在亲本Fc片段中约1至约10个氨基酸被取代,且优选约1至约5个氨基酸取代。在一些实例中,变体Fc片段Fc区与亲本Fc片段具有至少约80%序列一致性、至少约90%序列一致性、至少约95%、至少约96%、至少约97%、至少约98%或至少约99%序列一致性。“Fc片段”的效应功能可包括与Fc受体的结合、Clq结合和补体依赖性细胞毒性(CDC)、抗体依赖性细胞介导的细胞毒性(ADCC)、介导吞噬作用等。
单链抗体(single chain fragment variable,scFv),是由抗体重链可变区和轻链可变区通过短肽连接成一条肽链而构成。通过正确折叠,来自重链和轻链的可变区通过非共价键相互作用形成Fv段,因而scFv能较好地保留其对抗原的亲和活性。
“单域抗体(single domain antibody,sdAb)”,或者也称为“VHH抗体”,指具有抗原结合能力,包括重链可变区而无轻链的抗体分子。从结构上看,单域抗体也可以认为是抗体分子的一种抗原结合片段。其首先在骆驼科动物中被发现,随后,研究人员通过抗体库(例如噬菌体展示文库)筛选发现了更多的具有抗原结合能力的单域抗体。单域抗体相对于普通抗体分子(例如,经典四聚体抗体分子)或其抗原结合片段具有一些优势,例如包括但不限于:分子量更小,使用于人体时易于到达普通抗体分子难以到达的组织或部位,或者,能够接触到蛋白或多肽中普通抗体分子难以接触到的抗原表位;更加稳定,能够耐受例如温度和pH的变化以及变性剂和蛋白酶的作用。
“双特异性抗体”指具有两个不同的结合位点、能分别识别和结合两种不同的抗原的抗体分子。例如,双特异性抗体的一个结合位点可用于结合免疫细胞(例如T细胞),另一个结合位点用于结合肿瘤细胞,进而增强免疫细胞对肿瘤细胞的杀伤作用,同时减少脱靶毒性等副作用。这种具有双功能的抗体作为治疗肿瘤的药物通常比单抗药物有更高的疗效。类似地,可对抗体分子进行改造以使其包括多个不同的结合位点,生成“三特异性抗体”、“四特异性抗体”等。本文使用的“多特异性抗体”涵盖这些双特异性抗体、三特异性抗体、四特异性抗体等。优选地,所采用的多特异性抗体为双特异性抗体。
“人源化抗体”在本文中指在保留非人抗体的反应性的同时在人中具有较低免疫原性的抗体。例如,可以通过保留非人CDR区并用其人对应物(即,恒定区以及可变区的框架区部分)替换抗体的其余部分来实现。
“免疫偶联物”指与治疗剂偶联的本文公开的靶向EGFRvIII的抗体或其抗原结合片段。该治疗例如为细胞毒素、药物(例如,免疫抑制剂)或放射性毒素。
“融合蛋白”指人为生成(例如通过基因工程技术)的由至少两个不同肽段构成的蛋白分子。这些肽段在自然界中不存在,或者不存在于同一个蛋白分子中。常见的包括抗体片段的融合蛋白的实例包括抗体-细胞因子融合蛋白、抗体-细胞毒素融合蛋白(也称为免疫毒素)、用于免疫检测的酶标抗体、嵌合抗原受体(CAR)等。
“靶向”或“特异性结合”指,相对于环境中同时存在的其他分子,一种分子(例如抗体或其抗原结合片段)对另一种分子(如肿瘤细胞表面抗原)具有更高的结合亲和力。“靶向”或“特异性结合”并不排除该分子可以对一种以上的分子具有结合亲和力,例如双特异性抗体可以对两种不同抗原具有高亲和力。用在抗原前的修饰语“抗”表面其后的抗体“靶向”或“特异性结合”该抗原,例如抗EGFRvIII抗体指该抗体“靶向”或“特异性结合”EGFRvIII。优选地,该抗EGFRvIII抗体不“靶向”或“特异性结合”正常EGFR分子。抗体“特异结合抗原”或抗体的“抗原结合特异性”指该抗体相对于其他抗原,能够以更高或较高结合亲和力结合靶抗原。抗体与靶抗原的结合能力可通过各种方法进行定性或定量鉴定,例如通过表面等离子共振(SPR)测定抗体与靶抗原结合的KD值,测定抗体与其他抗体竞争结合靶抗原的能力等。这里Kd为平衡解离常数,可用于衡量抗体和其抗原之间的结合亲和力强弱。KD值越小,表明亲和力越强。抗体特异结合靶抗原并不意味着其不可以结合其他抗原,例如免疫交叉反应在本领域内也是已知的。
“结合复合物”指某分子与其结合对象之间形成的包括这二者的复合体。结合复合物的实例可包括抗原抗体复合物、配体受体复合物、蛋白二聚体等。形成结合复合物的作用力主要包括氢键、范德华力、离子键等非共价键作用力。可通过检测抗原抗体形成的复合物来检测样品中是否存在目的抗原或抗体,或者确定目的抗原或抗体的含量或浓度。
“肽段”,指多肽片段,为短氨基酸序列,例如长度约2-20个氨基酸,通常为多肽或蛋白的一部分。
EC50(concentration for 50%of maximal effect)指引起50%最大效应的浓度。例如在FACS测定中用于表示抗体分子与细胞上对应抗原的结合能力时,可指产生最大荧光强度一半时的抗体分子浓度。EC50值越低,则与细胞上抗原的结合亲和力越大。
“纯化标签”指与目的蛋白或多肽以融合蛋白形式一起表达的用于纯化该目的蛋白或多肽的氨基酸序列,包括但不限于His6标签、Flag标签、MBP(麦芽糖结合蛋白)标签和GST(谷胱甘肽巯基转移酶)标签、SUMO(小泛素相关修饰物(small ubiquitin related modifier))等。这些标签可在纯化后通过酶切去除,或者在不影响目的蛋白或多肽正常功能情况下可带标签使用(例如His6标签)。
“可检测标签”指与蛋白或多肽连接的氨基酸序列或其他化学基团,用于指示样品中该蛋白或多肽的存在或含量,或者用于跟踪该蛋白或多肽在受试者体内或细胞内的位置信息。可检测标签的实例包括免疫检测中可使用的各种酶,例如辣根过氧化物酶(HRP)、碱性磷酸酶(ALP);荧光基团(如FAM、FITC)或荧光蛋白(如GFP);放射性同位素(例如3H、14C、35S)。当可检测标签为酶时,可通过酶的酶学活性来确定与酶连接的蛋白或多肽的存在或含量。
术语“多肽”和“蛋白质”可互换使用且指氨基酸残基的聚合物。氨基酸残基的此类聚合物可含有天然或非天然氨基酸残基且包括但不限于氨基酸残基构成的肽、寡肽、二聚体、三聚体和多聚体。全长蛋白与其片段皆涵盖于该定义中。该术语亦包括多肽的表达后修饰,例如糖基化、唾液酸化、乙酰化、磷酸化和类似修饰。此外,出于本发明的目的,“多肽”指一种蛋白质,其包括对天然序列的修饰,诸如缺失、添加和取代(实际上通常为保守的),只要该蛋白质保持所需活性即可。这些修饰可为有目的的,如经由定点突变诱发;或可为偶然的,诸如经由产生蛋白质的宿主的突变或因PCR扩增所致的误差。
提及抗体或其抗原结合片段时,所用的“变体”或“功能性变体”这些变体或功能性变体与其亲本序列(抗体或其抗原结合片段)相比,可包括一个或更多个(1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20、21、22、23、24、25、26、27、28、29、30或更多个)氨基酸残基的插入、缺失或替换,同时保留了亲本序列与对应抗原(如EGFRvIII)的结合亲和力的至少50%、60%、70%、80%、90%,或甚至高于亲本序列。本领域技术人员已知,VH或VL框架区序列可以有相当大的变动余地,只要其可以维持相应CDR序列的空间位置。例如,可改变VH氨基酸序列中的框架区序列,例如以不同种属抗体分子的框架区互相替换,可获得基本上保留了抗原结合能力的功能性变体。此外,还可以通过改变少数几个CDR序列中的残基,例如1个、2个或3个氨基酸残基,并检测改动后的抗体分子与相应抗原的结合能力,来获得依然具有抗原结合能力的功能性变体。
本文中,术语“核酸分子”、“核酸”和“多核苷酸”可互换使用,指核苷酸聚合物。此类核苷酸聚合物可含有天然和/或非天然核苷酸且包括(但不限于)DNA、RNA和PNA。“核酸序列”指包含于核酸分子或多核苷酸中的核苷酸线性序列。
术语“载体”指可经工程改造以含有目的多核苷酸(例如目的多肽的编码序列)的核酸分子或可在宿主细胞中复制的核酸分子(例如,核酸、质粒、或病毒等)。载体可包括以下组件中的一个或更多个:复制起点、一或更多个调控目的多核苷酸的表达的调控序列(诸如启动子和/或增强子子)和/或一个或更多个可选择标记物基因(诸如抗生素抗性基因和可用于比色分析中的基因,例如β-半乳糖)。术语“表达载体”指用于在宿主细胞中表达目的多肽的载体。
“宿主细胞”指可为或已为载体或经分离多核苷酸的接受体的细胞。宿主细胞可为原核细胞或真核细胞。示例性真核细胞包括哺乳动物细胞,诸如灵长类动物或非灵长类动物细胞;真菌细胞,诸如酵母;植物细胞;以及昆虫细胞。非限制性示例性哺乳动物细胞包括(但不限于)NSO细胞、293以及CHO细胞,以及其衍生细胞,诸如293-6E、CHO-DG44、CHO-K1、CHO-S和CHO-DS细胞。宿主细胞包括单个宿主细胞的后代,且后代可能由于自然、偶然或故意突变而不一定与原始母细胞完全一致(在形态或基因组DNA互补方面)。宿主细胞也包括在活体内经本文提供的核酸分子或表达载体转染的细胞。
“治疗”指对受试者进行处理以获得有益的或所期望的临床结果。本文所用的“治疗”涵盖各种处理手段,包括以任何可能的药物向受试者给药、手术、辐射等。出于本发明的目的,有益或所期望的临床结果包括但不限于以下的任一种或多种:减轻一种或更多种症状、减弱疾病程度、预防或延迟疾病扩散(例如转移,例如转移至肺或淋巴结)、预防或延迟疾病复发、延迟或减缓疾病进展、改善疾病病况、抑制疾病或疾病进展、阻滞其发展和缓解(无论部分抑或完全缓解)。本文所提供的方法涵盖这些治疗方面中的任一种或多种。按照以上内容,“治疗”不需要完全去除病症或疾病的所有症状或完全缓解。
术语“治疗有效量”指足以在受试者体内引起临床医师所期望的生物学或医学反应的活性化合物的量。本发明融合蛋白的“治疗有效量”可由本领域技术人员根据给药途径、受试者的体重、年龄、病情等因素而确定。例如,典型的日剂量范围可以为每kg体重0.01mg至100mg或更多活性成分。
提及药物组合物,所使用的术语“药学上可接受的载体”指可以安全地进行施用的固体或液体稀释剂、填充剂、抗氧化剂、稳定剂等物质,这些物质适合于人和/或动物给药而无过度的不良副反应,同时适合于维持位于其中的药物或活性剂的活力。依照给药途径,可以施用本领域众所周知的各种不同的载体,包括,但不限于糖类、淀粉、纤维素及其衍生物、麦芽糖、明胶、滑石、硫酸钙、植物油、合成油、多元醇、藻酸、磷酸缓冲液、乳化剂、等渗盐水、和/或无热原水等。本文所提供的药物组合物可以制成粉末、注射剂等临床可接受的剂型。可以使用任何适当的途径向受试者施用本发明的药物组合物,例如可通过口服、静脉内输注、肌肉内注射、皮下注射、腹膜下、直肠、舌下,或经吸入、透皮等途径给药。
“受试者”指动物,例如哺乳动物,包括(但不限于)人类、啮齿动物、猿猴、猫科动物、犬科动物、马科动物、牛科动物、猪科动物、绵羊、山羊、哺乳类实验动物、哺乳类农畜、哺乳类运动动物和哺乳类宠物。受试者可为雄性或雌性且可为任何适龄受试者,包括婴儿、幼年、青年、成年和老年受试者。在一些实例中,受试者指需要治疗疾病或病症的个体。在一些实例中,接受治疗的受试者可为患者,其患有与该治疗有关联的病症,或有风险患上该病症。在特定实例中,受试者为人类,诸如人类患者。该术语通常可与“患者”、“检测对象”、“治疗对象”等互换使用。
“生物样品”意谓一定量的来自活物或先前活物的物质。所述物质包括(但不限于)血液(例如全血)、血浆、血清、尿液、羊水、滑液、内皮细胞、白血球、单核细胞、其他细胞、器官、组织、骨髓、淋巴结和脾脏。
提及氨基酸或核苷酸序列时,术语“序列一致性(sequence identity)”(也称为“序列同一性”)指两氨基酸或核苷酸序列(例如查询序列和参照序列)之间一致性程度的量,一般以百分比表示。通常,在计算两氨基酸或核苷酸序列之间的一致性百分比之前,先进行序列比对(alignment)并引入缺口(gap)(如果有的话)。如果在某个比对位置,两序列中的氨基酸残基或碱基相同,则认为两序列在该位置一致或匹配;两序列中的氨基酸残基或碱基不同,则认为在该位置不一致或错配。在一些算法中,用匹配位置数除以比对窗口中的位置总数以获得序列一致性。在另一些算法中,还将缺口数量和/或缺口长度考虑在内。出于本发明的目的,可以采用公开的比对软件BLAST(可在网页ncbi.nlm.nih.gov找到),通过使用缺省设置来获得最佳序列比对并计算出两氨基酸或核苷酸序列之间的序列一致性。
嵌合抗原受体(CAR),也称为嵌合T细胞受体、嵌合免疫受体,为一种工程化的蛋白受体分子,其可将期望的特异性赋予免疫效应细胞,例如与特定肿瘤抗原结合的能力。嵌合抗原受体通常由胞外抗原结合结构域、跨膜结构域和胞内信号结构域构成。多数情况下,抗原结合结构域为一段scFv序列,负责识别和结合特定的抗原。胞内信号结构域通常包括免疫受体酪氨酸活化基序(ITAM),例如来源于CD3ζ分子的信号传导结构域,负责激活免疫效应细胞,产生杀伤作用。另外,嵌合抗原受体还可在氨基端包括负责新生蛋白在细胞内定位的信号肽,以及在抗原结合结构域和跨膜结构域之间包括铰链区。除了信号传导结构域,胞内信号结构域还可包括例如来源于4-1BB分子的共刺激结构域。
“CAR-T细胞”指表达CAR的T细胞,通常采用编码CAR的表达载体转导T细胞获得。常用的表达载体为病毒载体,例如慢病毒表达载体。经嵌合抗原受体修饰的T细胞(CAR-T)不受主要组织相容性复合体的限制,具有特异性靶向杀伤活性和持久扩增的能力。除T细胞外,也可以用编码CAR的表达载体转化诸如NK细胞的其他淋巴细胞,获得表达该CAR的靶向杀伤性细胞。
“显性阴性受体(Dominant Negative Receptor,DNR)”指能够结合其配体但不会在细胞内诱导信号级联的受体。DNR通常具有完整的配体结合区域,但缺少细胞内酶活性(如激酶活性)区域。它可以是全长受体的突变形式或受体的截短形式。在CAR T细胞免疫治疗之后,一些癌症,尤其是实体癌,可能会上调与CAR T细胞上的抑制性受体结合的抑制性配体。这种适应性抗性损害了CAR T细胞疗法的功效。一些癌症,特别是实体癌,已知会分泌转化生长因子β(TGF-β),从而产生免疫抑制环境。TGF-β可诱导或促进转移并有效抑制免疫系统。因此,在一些实施方案中,我们使用截短形式的TGF-β受体TGFBRII作为TGF-βDNR来改善本文公开的CAR T细胞的抗肿瘤性能。在一些实施方案中,CAR和TGF-βDNR通过使用2A自剪切肽在T细胞表面上共表达。在一些实施方案中,CAR和TGF-βDNR通过使用两个表达载体分别在T细胞表面上表达。我们发现,当将TGF-βDNR引入本文公开的CAR T细胞时,能够增强CAR T细胞对一些EGFRvIII阳性癌细胞的细胞毒性。类似地,在一些实施方案中,我们使用PD-1受体的截短形式作为PD-1 DNR来改善本文公开的CAR T细胞的抗肿瘤性能。
“嵌合转换受体(Chimeric Switch Receptor,CSR)”指这样的受体,其被创建以逆转其原始信号通路的结果,从而赋予免疫细胞(例如,CAR T细胞)所需的活性,例如,克服免疫抑制性肿瘤微环境并使其具有更大的体内持久性的能力。在一些实施方案中,CSR可以利用癌细胞表达的抑制分子来进一步刺激CAR T细胞。在一个非限制性实例中,CAR T细胞可以被工程化以表达PD-1细胞外配体结合域与CD28跨膜和胞内共刺激信号结构域融合的形成的CSR。当将该CAR T细胞给药至患有表达EGFRvIII和PD-L1的癌症的受试者时,表达的CAR可以与EGFRvIII结合,而同时表达的CSR可以与PD-L1结合。PD-1/CD28嵌合转换受体融合蛋白的性质阻止了正常的PD1/PD-L1介导的T细胞抑制,而是通过CD28结构域促进信号传导,从而刺激CAR T细胞。因此,通过将PD-1的跨膜和细胞内结构域与CD28的跨膜和细胞内结构域进行转换,将PD-L1转化为CAR T细胞的共刺激配体。这将诱导对表达PD-L1的癌细胞的毒性增强。在另一个非限制性实例中,可以将TGFBRII的跨膜和细胞内结构域与IL7Rα受体的跨膜和细胞内结构域进行转换,从而可以在肿瘤微环境中将TGF-β的抑制信号转换为CAR T细胞刺激信号,促进其增殖或活性。
抗EGFRvIII抗体及其抗原结合片段
本文提供了特异性结合EGFRvIII的抗EGFRvIII抗体。该EGFRvIII抗体以相对较高的结合亲和力结合EGFRvIII抗原。如下文实施例中所阐述的,抗EGFRvIII抗体与EGFRvIII的结合能力可通过诸如表面等离子体共振技术(SPR)的测定方法来进行测量。另外,也可通过本领域已知的其他蛋白相互作用测定方法来测定,例如,生物层干涉(BLI)技术、酶联免疫吸附测定(ELISA)和流式细胞荧光分选技术(FACS)。
本文提供了多种抗EGFRvIII抗体的CDR序列,例如它们的重链CDR序列分别为SEQ ID NO:1-3、4-6、7-9、10-12、13-15、16-18、19-21、22-24、25-27、28-30、31-33、34-36、37-39、40-42、43-45、46-48、49-51、52-54或55-57所示序列。优选地,对应的轻链CDR序列分别为SEQ ID NO:58-60、61-63、64-66、67-69、70-72、73-75、76-78、79-81、82-84、85-87、88-90、91-93、94-96、97-99、100-102、103-105、106-108、109-111或112-114所示序列。
在一些实施方案中,本发明提供的抗EGFRvIII抗体的重链可变区包括与SEQ ID NO:115、117、119、121、123、125、127、129、131、133、135、137、139、141、143、145、147、149或151所示序列有至少90%序列一致性(例如,至少95%、至少98%、至少99%或甚至100%序列一致性)的氨基酸序列。优选地,对应的轻链可变区包括与SEQ ID NO:116、118、120、122、124、126、128、130、132、134、136、138、 140、142、144、146、148、150或152所示序列有至少90%序列一致性(例如,至少95%、至少98%、至少99%或甚至100%序列一致性)的氨基酸序列。
在本文已提供该抗EGFRvIII抗体的CDR序列基础上,本领域技术人员可构建各种具有EGFRvIII结合能力的多肽构建体,这包括使用来自不同抗体分子的骨架区(FR)与这些CDR序列进行组合。这些骨架区包括来自人抗体或动物(如小鼠、大鼠、羊、骆驼等)抗体的天然骨架区序列。这些骨架区还可以包括对天然骨架区序列改动所产生的骨架区序列变体。让本文提供的CDR序列与不同的骨架区序列组合形成重链可变区,并检测其与EGFRvIII的结合能力可容易地获得特异性结合EGFRvIII的多肽构建体。优选地,可以在这些CDR序列基础上构建人源化抗体,例如人源化的单链抗体(scFV)。
在一些实施方案中,本发明提供的经人源化的单链抗体(scFV)包括与SEQ ID NO:169、171、173、175、177、179、181、183、185或187所示序列有至少90%序列一致性(例如,至少95%、至少98%、至少99%或甚至100%序列一致性)的氨基酸序列。
预期本文描述的抗EGFRvIII抗体或其抗原结合片段可包含保守氨基酸取代。保守氨基酸取代通常可被描述为一种氨基酸残基被类似化学结构的另一种氨基酸残基取代,并且对多肽的功能、活性或其他生物学性质几乎没有或基本上没有影响。保守氨基酸取代是本领域众所周知的。保守性取代可例如是下列(a)-(e)组中的一个氨基酸被同组内的另一个氨基酸取代:(a)小的脂肪族非极性或弱极性残基:Ala、Ser、Thr、Pro和Gly;(b)极性带负电荷的残基及其(不带电荷的)酰胺:Asp、Asn、Glu和Gln;(c)极性带正电荷的残基:His、Arg和Lys;(d)大的脂肪族非极性残基:Met、Leu、Ile、Val和Cys;和(e)芳族残基:Phe、Tyr和Trp。因此,本领域技术人员可以理解的是,在本文提供的具体序列基础上,可以通过对少数氨基酸进行替换、删除、添加并验证或筛选所得产物与相应抗原EGFRvIII的结合能力或生物学活性,从而获得本文提供的靶向EGFRvIII的抗体分子的相应变体,这些变体也应包括在本发明的范围内。例如,本文提供的抗体分子或单链抗体在其全长或可变区序列或CDR序列上,可以有至少1个且不超过10个,例如不超过5、4、3、2或1个氨基酸的改变。
在一些与该抗EGFRvIII抗体的应用相关的实施方案中,以该抗EGFRvIII抗体的抗原结合片段来与对应抗原(EGFRvIII)结合。优选地,抗原结合片段为单链抗体(scFv)。例如,在将该抗体用于CAR时,可将其构成的单链抗体(scFv)(一个或多个)作为CAR的胞外抗原结合结构域。
包括抗EGFRvIII抗体或其抗原结合片段的融合蛋白
抗EGFRvIII抗体或其抗原结合片段还可与其他多肽形成融合蛋白。
在一些实施方案中,可以将具有EGFRvIII结合能力的抗原结合片段与Fc片段(经修饰的或不同种属来源的)连接形成融合蛋白,即经典抗体分子形式。Fc片段可位于EGFRvIII抗体抗原结合片段的C末端或N末端。优选地,Fc片段可位于EGFRvIII抗体抗原结合片段的C末端。EGFRvIII抗体抗原结合片段与Fc片段形成的融合蛋白具有特异性结合EGFRvIII的能力,同时具有Fc片段的效应功能,例如介导补体依赖性细胞毒性(CDC)、抗体依赖性细胞介导的细胞毒性(ADCC)、介导吞噬作用等。另外,与Fc片段的融合可增加抗EGFRvIII抗体抗原结合片段在体内的半衰期,以便在将抗EGFRvIII抗体抗原结合片段用作治疗药物时增加其给药间隔时间。
在一些实施方案中,抗EGFRvIII抗体或其抗原结合片段可以与蛋白标签连接形成融合蛋白。蛋白标签可包括纯化标签和可检测标签。纯化标签包括但不限于His6标签、Flag标签、MBP标签、GST标签、SUMO)标签等。可检测标签可用于指示样品中抗EGFRvIII抗体或其抗原结合片段的存在或含量,或者用于跟踪抗EGFRvIII抗体或其抗原结合片段在受试者体内或细胞内的位置信息。可检测标签的实例包括免疫检测中可使用的各种酶,例如辣根过氧化物酶(HRP)、碱性磷酸酶(ALP)等;荧光蛋白,例如GFP。由于抗EGFRvIII抗体或其抗原结合片段与EGFRvIII的特异性结合能力,可通过与抗EGFRvIII抗体或其抗原结合片段连接的可检测标签的量来确定抗EGFRvIII抗体或其抗原结合片段的量,并进而确定样品中EGFRvIII是否存在或其含量。
在另一些实施方案中,抗EGFRvIII抗体或其抗原结合片段可以与细胞因子或治疗性蛋白连接形成融合蛋白。这种情况下,可以借助抗EGFRvIII抗体或其抗原结合片段与EGFRvIII的特异性结合能力,将细胞因子或治疗性蛋白有目的地输送至特定的组织或细胞(例如表达EGFRvIII的肿瘤组织),实现细胞因子或治疗性蛋白的治疗作用。
包括抗EGFRvIII抗体或其抗原结合片段的多特异性抗体
在一些实施方案中,本文提供了多特异性抗体分子,其至少包括一个结合EGFRvIII的结构域(或功能单元)和一个或更多个额外的结合结构域。该一个或更多个额外的结合结构域可结合至除EGFRvIII外的第二抗原或蛋白质。
在一些实施方案中,本文提供了多特异性抗体分子,其至少包括两个结合EGFRvIII的结构域。该两个结合EGFRvIII的结合结构域在氨基酸序列上有差异,优选分别结合EGFRvIII上的不同抗原表位。
在一些实施方案中,所述结合EGFRvIII的结构域为单链抗体形式;所述一个或更多个额外的结合结构域可为单域抗体、单链抗体或其他抗原结合片段。
在一些实施方案中,所述第二抗原为肿瘤相关抗原(TAA)或肿瘤微环境相关抗原(TMEAA)。在一些实施方案中,所述第二抗原为免疫调节抗原,其中所述抗原与增强或抑制免疫细胞中的信号传导路径相关。在一些实施方案中,所述第二抗原为T细胞表面分子,诸如T细胞受体复合物的组分,例如CD3(包括γ、δ、ε、ζ和η链)。
优选地,所述多特异性的结合肽为双特异性抗体分子。
在一些实施方案中,多特异性的结合肽还包括Fc片段。Fc片段的存在可方便地形成结合结构域的多聚化,并且可提供相关的效应功能。
包括抗EGFRvIII抗体或其抗原结合片段的免疫偶联物(或抗体偶联物)
本文提供了包括至少一个本文提供的特异性结合EGFRvIII的抗EGFRvIII抗体或其抗原结合片段和一个或更多个其他功能部分的偶联物。所述其他部分可为化学基团,例如治疗剂,诸如细胞毒性剂,或可为示踪剂。在一些实例中,所述部分可为靶向部分、小分子药物(例如小于500Da的非多肽药物)、毒素、细胞生长抑制剂、细胞毒性剂、免疫抑制剂、适合于诊断目的的放射性试剂、用于治疗目的的放射性金属离子等。
在一些实施方案中,免疫偶联物为含有一个或更多个本文提供的抗EGFRvIII抗体或其抗原结合片段和治疗剂的抗体药物偶联物(ADC),其具有细胞毒性,抑制细胞生长,或者提供一些治疗益处。在一些实施例中,细胞毒性剂为化学治疗剂、药物、生长抑制剂、毒素(例如细菌、真菌、植物或动物来源的酶活性毒素或其片段)或放射性同位素(亦即,放射性偶联物)。在一些实例中,本发明提供的抗体药物偶联物允许药物部分靶向递送至肿瘤。在一些情况下,此可引起肿瘤细胞的靶向杀死。
在一些实例中,治疗剂包括例如道诺霉素(daunomycin)、阿霉素(doxorubicin)、甲氨喋呤(methotrexate)、长春地辛(vindesine)、类美登素(maytansinoid)等。在一些实例中,治疗剂具有细胞内活性。在一些实例中,与抗EGFRvIII抗体或其抗原结合片段偶联的免疫偶联物可经细胞内化,且治疗剂具有阻断细胞蛋白质合成活性或阻断核酸合成活性,从而导致细胞生长停滞或死亡。
在一些实施方案中,免疫偶联物含有一个或更多个本文提供的抗EGFRvIII抗体或其抗原结合片段和示踪剂。该免疫偶联物可用于研究或诊断目的,例如用于活体内检测癌症。示踪剂可直接或间接产生可侦测信号。例如,示踪剂可为射性同位素,如3H、14C、32P、35S、123I;荧光(荧光团)或化学发光(发色团)化合物,诸如荧光异硫氰酸盐、若丹明或荧光素;显影剂;或金属离子。在一些实施例中,示踪剂为用于闪烁摄影研究的放射性原子,例如99Tc或123I,或用于核磁共振(NMR)成像(亦称为磁共振成像,MRI)的自旋标记,例如89Zr、123I、19F、13C、15N、17O、89Zr可与多种金属螯合剂错合且结合至抗体,例如用于PET成像。
抗EGFRvIII抗体或其抗原结合片段与其他功能部分的连接可为共价或非共价连接。非共价连接的实例可包括通过生物素-亲和素系统(Biotin-Avidin System)。共价连接的实例可包括各种化学接头,包括肽接头、可裂解接头、或不可裂解接头。示例性接头组分包括6-顺丁烯二酰亚胺基己酰基(“MC”)、顺丁烯二酰亚胺基丙酰基(“MP”)、缬氨酸-瓜氨酸(“val-cit”)、丙氨酸-苯丙氨酸(“ala-phe”)、对氨基苯甲氧基羰基(“PAB”)、N-丁二酰亚胺基4-(2-吡啶基硫基)戊酸酯(“SPP”)、N-丁二酰亚胺基4-(N-顺丁烯二酰亚胺基甲基)环己烷-1甲酸酯(“SMCC”)和N-丁二酰亚胺基(4-碘-乙酰基)胺基苯甲酸酯(“SIAB”)。
在一些实施方案中,接头可包含氨基酸残基。例示性氨基酸接头组分包括二肽、三肽、四肽或五肽。示例性二肽包括:缬氨酸-瓜氨酸(vc或val-cit)、丙氨酸-苯丙氨酸(afa-phe)。示例性三肽包括:甘氨酸-缬氨酸-瓜氨酸(gly-val-cit)和甘氨酸-甘氨酸-甘氨酸(gly-gly-gly)。包含氨基酸接头组分的氨基酸残基包括天然存在的残基以及非天然存在的氨基酸类似物,诸如瓜氨酸。氨基酸接头组分可在其选择性方面经设计和优化,以通过特定酶,例如肿瘤相关蛋白酶、组织蛋白酶B、C和D、胞浆素蛋白酶进行酶促裂解。
抗EGFRvIII抗体或其抗原结合片段与细胞毒性剂的偶联物可使用多种双官能蛋白质偶合剂制得,诸如N-丁二酰亚胺基-3-(2-吡啶基二硫醇)丙酸酯(SPDP)、亚胺基硫杂环戊烷(IT)、酰亚胺酯的双官能衍生物(诸如己二酰亚胺酸二甲酯HCl)、活性酯(诸如二丁二酰亚胺基基质)、醛(诸如戊二醛)、双叠氮基化合物(诸如双(对叠氮基苯甲酰基)己二胺)、双重氮衍生物(诸如双-(对重氮苯甲酰基)-乙二胺)、二异氰酸酯(诸如甲苯2,6-二异氰酸酯)和双活性氟化合物(诸如1,5-二氟-2,4-二硝基苯)。
包括抗EGFRvIII抗体或其抗原结合片段的嵌合抗原受体(CAR)
本文提供的嵌合抗原受体(CAR),其是人工构建的嵌合蛋白,包括特异性结合EGFRvIII的胞外抗原结合结构域、跨膜结构域、以及一个或更多个细胞内信号传导结构域。这些CAR的特征包括它们以非MHC限制的方式将T细胞特异性和反应性重定向至表达EGFRvIII的细胞的能力。非MHC限制的EGFRvIII识别使表达所公开的CAR的T细胞或NK细胞能够独立于抗原加工而识别抗原,并杀灭表达EGFRvIII的细胞(如肿瘤细胞)。
胞内信号传导结构域可以包括T细胞受体信号传导结构域、T细胞共刺激信号传导结构域或两者。T细胞受体信号传导结构域可包括T细胞受体的细胞内结构域,例如CD3ζ蛋白的细胞内部分。共刺激信号传导结构域为共刺激分子的细胞内结构域。共刺激分子是淋巴细胞对抗原的有效反应所需的抗原受体或其配体以外的细胞表面分子。
一些实施方案中,在本文提供的CAR包括与EGFRvIII特异性结合的胞外抗原结合结构域。例如,抗原结合结构域可以是scFv,其可为与EGFRvIII特异性结合的通过肽接头连接的任何抗体或其抗原结合片段的重链可变区和轻链可变区。又例如,对于利用特异性结合EGFRvIII的单域抗体构建CAR,胞外抗原结合结构域可包括来自该单域抗体的VHH,而不包括任何轻链可变区。
在一些实施方案中,本文提供的CAR的胞外抗原结合结构域包括一个或更多个来自本文提供的抗EGFRvIII抗体的抗原结合片段,如未经人源化的或经人源化的单链抗体(scFv)。
在胞外抗原结合结构域包括两个或更多个抗原结合片段时,它们可直接串联连接或通过肽接头串联连接。采用两个或多个串联连接的抗原结合片段作为胞外抗原结合结构域或其部分,有助于促进构建的CAR对靶分子(例如EGFRvIII)的识别和结合。优选地,这些抗原结合片段分别结合EGFRvIII上的不同表位。
CAR可以包括信号肽序列,例如,位于抗原结合结构域的N端。可以使用任何合适的信号肽序列。在一个实施方案中,信号肽序列是CD8α信号肽。虽然信号肽序列可以促进CAR在细胞表面表达,但在表达的CAR中信号肽序列的存在对于CAR发挥功能并不是必需的。当CAR在细胞表面表达后,信号肽序列可能会从CAR上切下。因此,在一些实施方案中,CAR缺乏信号肽序列。
在CAR的抗原结合结构域和跨膜结构域之间,可能存在铰链区(或间隔结构域),其包括多肽序列。铰链区可包含多达300个氨基酸,优选10至100个氨基酸。在一个实施方案中,本文提供的CAR包括CD8α蛋白铰链区。
CAR可以包括与CAR的胞外结构域融合的跨膜结构域。在一个实施方案中,使用与CAR中其他元件(如交联区或胞内信号传导结构域)天然相关的跨膜结构域。
跨膜结构域可以源自天然或合成来源。在来源是天然的情况下,该结构域可以源自任何膜结合或跨膜蛋白。用在所公开的CAR中的示例性跨膜结构域可以至少包括T细胞受体的α、β或ζ链、CD28、CD3ε、CD45、CD4、CD5、CDS、CD9、CD 16、CD22、CD33、CD37、CD64、CD80、CD86、CD 134、CD137、CD154的跨膜结构域。或者,跨膜结构域可以是合成的,在这种情况下,它将主要包含疏水残基,例如亮氨酸和缬氨酸。在一些实施方案中,苯丙氨酸、色氨酸和缬氨酸的三联体将出现在合成跨膜结构域的每一端。
任选地,优选长度为2至10个氨基酸的短寡肽或多肽接头可以形成跨膜结构域与CAR的细胞内T细胞信号传导结构域和/或T细胞共刺激结构域之间的连接。示例性接头序列包括一个或更多个甘氨酸-丝氨酸双联体。
在一些实施方案中,跨膜结构域包括T细胞受体的跨膜结构域,例如CD8α跨膜结构域。
CAR的细胞内区域包括一个或更多个胞内T细胞信号传导结构域,负责激活表达CAR的T细胞的至少一种正常效应功能。示例性T细胞信号传导结构域在本文中有提供,并且是本领域普通技术人员已知的。
虽然在CAR中可以使用整个细胞内T细胞信号传导结构域,但在许多情况下,没有必要使用整个链。就使用细胞内T细胞信号传导结构域的截短部分而言,该截短部分可代替完整链使用,只要它转导相关的T细胞效应器功能信号即可。
用在CAR中的细胞内T细胞信号传导结构域的例子包括T细胞受体(TCR)的细胞质序列和协同作用以在抗原受体结合后启动信号转导的共刺激分子,以及这些序列的任何衍生物或变体和任何具有相同功能的合成序列。
T细胞受体信号传导结构域以刺激方式或抑制方式调节T细胞受体复合物的激活。本文所公开的CAR可以包括以刺激方式起作用的细胞质信号传导序列,其可以包含被称为基于免疫受体酪氨酸的激活基序或ITAM的信号传导基序。可包含在所公开的CAR中的含有ITAM的初级细胞质信号传导序列的实例包括来自CD3ζ、FcRγ、FcRβ、CD3γ、 CD3δ、CD3ε、CDS、CD22、CD79a、CD79b和CD66d蛋白的胞内结构域。在一些实施方案中,CAR中的细胞质信号传导分子包括来自CD3ζ的细胞内T细胞信号传导结构域。
本文提供的CAR的胞内结构域可以包括CD3ζ链部分和细胞内共刺激信号传导结构域。该共刺激信号传导结构域可包括共刺激分子的细胞内结构域。共刺激分子是淋巴细胞对抗原的有效反应所需的抗原受体或其配体以外的细胞表面分子。此类分子的实例包括CD27、CD28、4-1BB(CD137)、OX40(CD134)、CD30、CD40、PD-1、ICOS、淋巴细胞功能相关抗原1(LFA-1)、CD2、CD7、LIGHT、NKG2C和B7-H3。
在一些实施方案中,CAR可以包括CD3ζ信号传导结构域、CD8信号传导结构域、CD28信号传导结构域、4-1BB信号传导结构域或其两种或更多种的组合。在一个实施方案中,胞内结构域包括CD3-ζ的信号传导结构域和CD28的信号传导结构域。在另一个实施方案中,细胞质结构域包括CD3ζ的信号传导结构域和4-1BB的信号传导结构域。在又一个实施方案中,细胞质结构域包括CD3-ζ的信号传导结构域以及CD28和CD137的信号传导结构域。本领域普通技术人员可以根据需要改变CAR上一个或更多个T细胞信号传导结构域的顺序。本发明的CAR的细胞质信号传导部分内的细胞质信号传导序列可以以随机或指定的顺序相互连接。任选地,短的,优选长度在2和10个氨基酸之间的多肽接头,可以形成该连接。甘氨酸-丝氨酸双联体提供了特别适合的接头。此外,在CAR的信号传导结构域和跨膜结构域之间,可以存在间隔结构域,其包括多肽序列。间隔结构域可包含多达300个氨基酸,优选10至100个氨基酸,最优选25至50个氨基酸。
基于本文提供的CAR(作为亲本CAR),本领域技术人员可对其进行改动,例如,将其与其他多肽融合,或者仅保留本文提供的CAR的片段,而这些融合分子或片段具有所述亲本CAR的至少部分生物活性,例如识别靶细胞(例如表达EGFRvIII的肿瘤细胞)或检测、治疗或预防疾病的活性。
本文提供的CAR还可以在氨基或羧基末端,或在两个末端包括附加氨基酸,这些附加氨基酸不存在于亲本CAR的氨基酸序列中。理想地,附加氨基酸不干扰CAR或功能部分的生物学功能,例如识别靶细胞、检测癌症、治疗或预防癌症等。更理想地,与亲本CAR的生物学活性相比,附加氨基酸增强生物学活性。
还提供了本文描述的CAR的功能性变体,其与亲本CAR具有实质或显著的序列一致性或相似性,该功能变体保留了其作为变体的CAR的生物学活性。功能性变体涵盖例如本文描述的CAR(亲本CAR)的那些变体,其保留与亲本CAR相似程度、相同程度或更高程度识别靶细胞的能力。参考亲本CAR,功能性变体在氨基酸序列上与亲本CAR例如可以有至少约30%、约50%、约75%、约80%、约85%、约90%、约91%、约92%、约93%、约94%、约95%、约96%、约97%、约98%、约99%或更高一致性。
功能性变体例如可以包括具有至少一个保守氨基酸替换的亲本CAR的氨基酸序列。或者或另外,功能变体可包括具有至少一个非保守氨基酸替换的亲本CAR的氨基酸序列。 在这种情况下,优选非保守氨基酸替换不干扰或抑制功能变体的生物学活性。非保守氨基酸替换可以增强功能变体的生物活性,使得功能变体的生物活性与亲本CAR相比增加。
本文提供的CAR可以包括合成氨基酸来代替一种或更多种天然存在的氨基酸。此类合成氨基酸是本领域已知的,包括例如氨基环己烷羧酸、正亮氨酸、a-氨基正癸酸、高丝氨酸、S-乙酰氨基甲基-半胱氨酸、反式-3-和反式-4-羟脯氨酸、4-氨基苯丙氨酸、4-硝基苯丙氨酸、4-氯苯丙氨酸、4-羧基苯丙氨酸、β-苯基丝氨酸、β-羟基苯丙氨酸、苯基甘氨酸、α-萘丙氨酸、环己基丙氨酸、环己基甘氨酸、二氢吲哚-2-羧酸、1,2,3,4-四氢异喹啉-3-羧酸、氨基丙二酸、氨基丙二酸单酰胺、N’-苄基-N’-甲基-赖氨酸、N’,N’-二苄基-赖氨酸、6-羟基赖氨酸、鸟氨酸、α-氨基环戊烷羧酸、α-氨基环己烷羧酸、oc-氨基环庚烷羧酸、-(2-氨基-2-降冰片烷)-羧酸、γ-二氨基丁酸、α,β-二氨基丙酸、高苯丙氨酸和α-叔丁基甘氨酸等。
本文提供的CAR可以被糖基化、酰胺化、羧化、磷酸化、酯化、N-酰化、经由例如二硫键环化,或转化成酸加成盐和/或任选地二聚或多聚,或偶联。
产生嵌合抗原受体的方法、包括此类受体的T细胞及其用途(例如,用于治疗癌症)是本领域已知的。例如,编码所公开的嵌合抗原结合受体的核酸分子可以包括在表达载体中(例如慢病毒载体)用于在宿主细胞例如T细胞中表达,以制备所公开的CAR。在一些实施方案中,使用嵌合抗原受体的方法包括从受试者中分离T细胞,用编码嵌合抗原受体的表达载体(例如慢病毒载体)转化T细胞,并以表达嵌合抗原受体的工程化T细胞向受试者给药用于治疗,例如治疗受试者中的肿瘤。
本文提供的CAR还可以与用于控制CAR或表达CAR的细胞的活性的安全机制联合使用。在一个实施方案中,可以让CAR与“EGFRt”共表达。“EGFRt”指编码截短的人表皮生长因子受体多肽,其缺乏远端膜EGF结合域和细胞质信号传导尾,但保留了由抗EGFR抗体识别的细胞外表位。EGFRt可用作具有遗传修饰细胞功能的非免疫原性选择工具以及追踪标记。其一方面可作为CAR-T细胞的标记分子,另一方面还可以在需要时用于清除体内的CAR-T细胞,例如通过EGFR抗体(例如,西妥昔单抗)介导的ADCC途径(参见US8802374B2),即在临床转化时作为安全开关使用。除了使用tEGFR外,本文提供的CAR-T细胞还可以考虑与其他机制的安全开关联合使用。例如,可以对本文提供的CAR进行加工以使其可以利用二聚化化学诱导剂(CID)来控制CAR活性。在一个实例中,可以将本文提供的完整CAR以分开的融合蛋白进行表达(例如将CD3ζ胞内结构域与其他部分分开表达),并利用基于FKBP/FRB的结合结构域和雷帕霉素(或雷帕霉素类似物)的二聚化机制在细胞内重建完整CAR,从而可通过雷帕霉素的存在与否来控制CAR的完整性和活性(例如,参见美国专利11,084,880中的描述)。另外,当采用自剪切肽使得能够同时表达具有自杀能力的融合蛋白的时,可以通过降解CAR分子或者导致CAR-T细胞死亡来控制CAR-T细胞活性。这方面的实例包括使用PROTAC技术降解CAR分子以及使用具有诱导细胞凋亡能力的蛋白,如caspase-9。
包括抗EGFRvIII抗体或其抗原结合片段的药物组合物和治疗方法
本文公开的抗EGFRvIII抗体或其抗原结合片段以及包括抗EGFRvIII抗体或其抗原结合片段的融合蛋白、多特异性抗体分子、免疫偶联物(可以与药学上可接收的载体一起配制为药物组合物)可用于对受试者给药以进行肿瘤预防或治疗。该用途的有效用量可取决于疾病的严重性和患者自身免疫系统的总体状态。给药方案将也随疾病状态和受试者状态而变动,并且一般范围将从单次大量(bolus)给药或连续输注至每天多次给药(例如每隔4-6小时)。本领域熟练的临床医生可以例如通过利用临床试验、身体检查和受试者家族史,容易地确定某受试者是否为这种治疗的候选者。
在一些实施方式中,本文公开的抗EGFRvIII抗体或其抗原结合片段以及包括该抗EGFRvIII抗体或其抗原结合片段的融合蛋白、多特异性抗体分子、免疫偶联物可以与一种或更多种其他药物(例如抗肿瘤剂)联合给药。
在一些实施方案中,所治疗的疾病或病症为肿瘤,优选胶质母细胞瘤。可以使用任何合适的方法或途径本文公开的抗EGFRvIII抗体或其抗原结合片段(以及包括该抗EGFRvIII抗体或其抗原结合片段的融合蛋白、多特异性抗体分子、免疫偶联物),并任选地联用其他抗肿瘤剂。给药途径包括,例如,口服、静脉内、腹膜内、皮下或者肌内给药。
本文提供的编码抗EGFRvIII抗体或其抗原结合片段的核酸分子、包括该核酸分子的表达载体以及仅所述核酸分子或表达载体转染的宿主细胞也可通过各种方式用于上述治疗目的。例如,对于表达载体,可通过本领域已知的基因治疗手段引入受试者体内,表达目的蛋白或多肽(抗EGFRvIII抗体或其抗原结合片段),从而达到治疗目的。
包括抗EGFRvIII抗体或其抗原结合片段的检测、诊断试剂盒或治疗试剂盒
本文提供的抗EGFRvIII抗体或其抗原结合片段以及包括该抗EGFRvIII抗体或其抗原结合片段的其他形式分子(例如融合蛋白或双特异性抗体分子)可与样品中的EGFRvIII特异性结合。通过检测所形成的抗EGFRvIII抗体或其抗原结合片段-EGFRvIII复合物的量,或者所形成的抗EGFRvIII抗体或其抗原结合片段-EGFRvIII复合物中抗EGFRvIII抗体或其抗原结合片段的量,可方便的确定样品中的-EGFRvIII含量(或是否存在)。
如上文所描述的,用于该目的,本文提供的抗EGFRvIII抗体或其抗原结合片段可与各种检测标签连接,方便通过各种手段进行检测,这包括但不限于生物发光、荧光、放射性标记、酶促反应的产物生产量等。检测样品中EGFRvIII含量后,与正常人群中正常EGFRvIII含量进行比较,可用于确定提供该样品的受试者的疾病状况或严重程度。通过在受试者治疗过程随时间反复检测EGFRvIII含量变化,还可用于确定治疗手段是否有效,从而为治疗方案的改动提供依据。
本文提供的抗EGFRvIII抗体或其抗原结合片段以及包括该抗EGFRvIII抗体或其抗原结合片段的其他形式分子(例如融合蛋白或双特异性抗体分子)、表达CAR的细胞可被置于容器中,以形成检测、诊断或治疗试剂盒。这些容器可以是盒、安瓿瓶、小瓶、管子、袋子或本领域已知的合适容器形式。这些容器可以由塑料、玻璃、层压纸、金属箔或适用于保存药物的其他材料制成。如果需要,与该容器一起提供的还包括使用说明书。说明书通常可包括关于如何使用抗EGFRvIII抗体或其抗原结合片段、包括抗EGFRvIII抗体或其抗原结合片段的组合物或CAR-T细胞用于治疗或预防肿瘤(例如胶质母细胞瘤)的信息,例如可包括对治疗剂(如抗EGFRvIII抗体或其抗原结合片段)的描述;用于治疗或预防瘤形成(例如胶质母细胞瘤)的剂量方案;注意事项;警告;适应症;禁忌症;不良反应;动物药理学;临床研究;和/或参考资料。说明书可直接打印在容器(如果存在的话)上,或作为贴于容器的标签,或者作为提供于容器中或与容器在一起的单独的纸、册、卡或折叠印刷品。
在一些实施方案中,本文提供的抗EGFRvIII抗体或其抗原结合片段与EGFRvIII的结合亲和力与现有对照抗体接近,或优于对照抗体。本文提供的抗EGFRvIII抗体或其抗原结合片段经人源化,并且与EGFRvIII的结合亲和力与现有对照抗体接近,或优于对照抗体。
本研究通过不同的免疫方式免疫小鼠,比较不同免疫方式免疫出抗体的特异性和亲和力,筛选出最佳的免疫条件,通过政府筛选的方法筛选出一系列针对EGFRvIII目的抗原的新型鼠源单克隆抗体,经过ELISA、FACS和SPR等方法检测抗体亲和力,筛选出高亲和力抗体后进行抗体序列测定,应用抗体序列设计制备靶向EGFRvIII CAR T细胞,通过体外和体内药效研究筛选出最佳的抗体序列,选择最佳的鼠源抗体序列再进行人源化改造,通过SPR技术检测人源化抗体的亲和力,筛选出最佳亲和力的人源化抗体,进行抗体序列的测定,选择亲和力较高的人源化抗体进行一系列靶向EGFRvIII抗原的CAR T细胞,通过体外和体内药效研究筛选出最佳的人源化抗体序列。筛选到的最佳的抗体序列制备CAR NK细胞,通过体外药效确定CAR NK在治疗实体肿瘤的药效。
为了克服实体肿瘤抑制细胞免疫治疗的效果,本研究同时在二代CAR T细胞的基础上进行了基因修饰,以期提高CAR T细胞抵抗肿瘤免疫微环境的抑制,提高CAR T细胞持续性和浸润实体肿瘤内部的能力。本研究在自主开发的靶向EGFRvIII CAR T细胞上同时表达dnTGFβRII,或者通过dnTGFβRII/将抑制信号转换成IL7激活信号,增加的元件显著提高了CAR T细胞的抑瘤效率,抵抗了TGFβ的抑制作用。
实施例
1.实验流程
1.1鼠源抗体开发
1.1.1小鼠免疫
在小鼠免疫前,采集小鼠血液100μl,37℃静置5min后离心,收集血清,作为阴性血清对照。采用3种方式对小鼠进行免疫,1组应用蛋白+多肽结合方式进行免疫,2组应用过表达EGFRvIII靶细胞进行免疫,3组应用多肽进行免疫。将佐剂和抗原按照1:1的比例进行配比,乳化,乳化后的免疫原进行小鼠皮下注射。2次免疫后1周,采集小鼠血液,分离血清,进行抗体效价检测,根据抗体效价的高低进行追加免疫,直至血清效价满足要求,进行终免。
1.1.2效价检测
流式细胞术检测血清效价,阳性细胞和阴性细胞分别为F98npEGFRvIII和F98npEGFR(ATCC),将细胞消化收集至离心管中,放入离心机,300g离心5min,弃掉上清,加入适量的1 ⅹ PBS清洗2次,弃调上清,应用1ⅹPBS重悬细胞,向96孔板中加入50μl细胞悬液,约2 ⅹ 10^5细胞,每个孔分别加入1/50和1/500稀释的血清,阳性对照抗体C097VEJ300,同型对照mouse IgG和阴性对照PBS,每孔加入100μl稀释好的荧光二抗Alexa Fluor 647 AffiniPure Goat Anti-Mouse IgG,Fcγfragment specific(min X Hu Bov Hrs SrProt),4℃震荡孵育30min后,取出96孔板,每孔加入150μl 1 ⅹ PBS,清洗2次,弃去上清,每孔加入150μl 1 ⅹ PBS重悬细胞,流式检测,流式细胞仪进行读数。
1.1.3杂交瘤细胞制备
将效价检测结果最好的两只小鼠处死并在超净台中使用高压灭菌过的镊子取出小鼠脾脏,然后将其浸泡在无菌的生理盐水中。用无菌的剪刀将小鼠脾脏充分剪碎,再将剪碎后的脾脏转移至无菌的70μm细胞筛网,使用一次性无菌注射器内管充分研磨小鼠脾脏。用RPMI-1640培养基洗涤细胞筛网上研磨的小鼠脾脏细胞,反复冲洗3次,将冲洗液收集起来得到相应细胞悬液。1000rpm离心5min收集脾脏细胞。将生长状态良好的SP 2/0细胞重悬收集,1000rpm离心5min,收集的细胞清洗计数。小鼠脾细胞:SP 2/0细胞为5:1混合后离心。轻弹使细胞沉淀松散,37℃水浴1min。缓慢滴加PEG-1450溶液(37℃);滴加后的样品离心弃上清。HAT筛选培养基将每只小鼠脾脏融合后制备的细胞悬液稀释成接种至96孔板,加入液体为150μl/孔,放入CO2细胞培养箱内培养。每3-5天进行补液,板底达到合适细胞密度后,收取上清检测。
1.1.4杂交瘤上清抗体效价检测
通过mirrorball实验方法对上清进行阳性筛选,阳性细胞和阴性细胞分别为F98npEGFRvIII和F98npEGFR,将上清样品和对照进行滴度稀释加入384孔板,每孔加入约1ⅹ10^3细胞,加入适量CFSE对细胞进行染色,充分混匀后加入相应种属的二抗,室温孵育,应用仪器进行检测读数。筛选阳性克隆进一步应用FACS方法进行筛选。
1.1.5亚克隆制备及检测
采用有限稀释法将阳性克隆进行亚克隆,用1 ⅹ HT培养基培养细胞亚克隆,亚克隆细胞放置37℃,5%二氧化碳培养箱中进行培养。应用流式细胞术进行克隆筛选。
1.1.6亚克隆抗体表达纯化及检测
取亚克隆上清进行蛋白亲和纯化,将纯化以后的单克隆抗体检测浓度,稀释成一系列梯度浓度进行SPR检测,计算EC50(图3)。
1.1.7候选克隆抗体序列测定
使用总RNA提取试剂盒进行单克隆杂交瘤细胞总RNA提取,应用反转录试剂盒以提取的总RNA为模板,进行逆转录,通过PCR方法扩增抗体序列,进行抗体序列测定。
1.1.8流式检测抗体亲和力
(1)分别取F98npEGFRvIII细胞各1*10^5个/Well,用2mL 2%BAS BSA洗两遍,用100μL PBS重悬;
(2)最大抗原浓度加入量为16ug,共稀释8个梯度,稀释体系如下表。
(3)加入上述稀释抗体,4℃作用30min;
(4)用2mL 2%BSA洗细胞三遍,然后加入BV421标记的anti-Human IgG Fc Secondary Antibody二抗,4℃作用30min;Mouse Antibody二抗用BV421 Anti-Mouse IgG;
(5)PBS洗细胞两遍,500μL PBS重悬细胞,准备上机检测分析;
(6)计算平均荧光强度MFI,根据MFI和抗体浓度作图,计算出EC50值,从而比较不同抗体的亲和力。
1.2鼠源抗体CAR T细胞制备和功能研究
1.2.1 CAR结构设计
CAR结构包括开发的抗EGFRvIII抗原的鼠源抗体序列ScFv结构,还包括但不限于CD8铰链区和跨膜区,41-BB激活信号以及CD3z激活结构域(图4),同时可以在此基础上进行其它修饰,包括细胞因子的分泌,趋化因子的分泌,免疫检查点抑制基因的修饰,肿瘤微环境抑制信号封闭和转化为激活型号等四代CAR T。
1.2.2表达质粒构建
将合成的CAR基因序列通过分子克隆连接至慢病毒表达质粒上,合成的质粒转染至感受态细胞Stbl3,摇菌后大提质粒,用于后续慢病毒包装。
1.2.3慢病毒包装和滴度测定
分别将不同CAR质粒与包装质粒共转染到293T细胞中,转染后48h收集细胞上清,4000rpm离心10min。收集上清,用0.45μm的滤膜过滤,用超速离心机收集慢病毒沉淀。离心条件为4℃,100000×g,无刹车,离心90min。将浓缩后的慢病毒按每管200μl分装到0.5mL冻存管中,-80℃分装冻存,应用流式细胞术检测慢病毒滴度。
1.2.4 CAR T细胞制备
从一份健康人外周血中取出20ml加入到50ml离心管中,向血液中加入1mL RosetteSepTM Cocktail混匀,使抗体浓度为50μL/mL。将20mL GE Ficoll液加入新的50mL离心管中,用吸管小心吸取20mL稀释好的血样按1:1缓慢加入Ficoll液的上层。将加好血样的离心管放入离心机内,转速400g,离心30min。用无菌吸管吸掉上层血浆和血小板,不要碰到单个核细胞层,小心吸取第二层环状乳白色淋巴细胞层到新的离心管内。每管淋巴细胞悬液加入三倍体积PBS清洗细胞,400g离心10min。弃上清,用5mL PBS重悬所得细胞,1200rpm离心10min。最后用X-VIVO+100U IL2完全培养液悬浮细胞,细胞计数。将1mL分离的密度为1×10^6T淋巴细胞接种至24孔板的1个孔中,并加入25μl CD3/CD28磁珠。将细胞培养于5%CO2 37℃培养箱中,48h后用于慢病毒感染。按照MOI=0.5加入相应的慢病毒,第二天细胞补液,每孔补加至30ml并转移至T 75瓶中,水平放置培养。
1.2.5 CAR T细胞CAR阳性率表达检测
3天后,将CAR-T细胞转移至无菌流式管内,每管细胞放置于biolegend磁铁上2min去除磁珠。之后去部分细胞染流式抗体(AF647-protein L)检测CAR表达率。
1.2.6 CAR T细胞体外杀伤效率检测
在检测白板中,样品孔接种U87MG-EGFRvIII靶细胞个数为1×10^4个/孔,重复6个孔,同时设置6个对照孔,每孔100μl。过夜后,吸弃上清后,加入CAR-T细胞,效靶比为E/T ratio=2.5:1;每孔100μl。6h后,300g离心5min,所有孔尽量去掉上清。每孔加入100μl裂解液,室温裂解10min,开始上机检测,每孔加入萤火虫荧光素酶检测试剂100μL。杀伤效率计算公式:杀伤效率(%)=(对照孔荧光强度-杀伤荧光强度)/(对照孔荧光强度-空白孔荧光强度)×100%。
1.2.7 CAR T细胞体内药效研究
选用6-8周雄性NOG小鼠作为实验动物。U87MG-EGFRvIII细胞消化后用无菌1×PBS重悬清洗细胞2次进行活细胞计数,调整细胞密度为5×10^5/50μl/只,细胞注射时按照1:1体积每只混合50μl基质胶,即最终每只小鼠注射100μl总体积的细胞混悬液。移植部位为小鼠背部右下侧皮下。肿瘤移植后,肿瘤体积用卡尺每周三次进行测量,肿瘤体积计算公式为TV=0.5a×b2。当肿瘤大小达到100mm3时按5ⅹ10^6/只回输CAR-T细胞或生理盐水,并持续观察肿瘤大小与小鼠存活状况。
1.3鼠源抗体序列人源化
选择体内药效最佳2个克隆序列进行抗体序列人源化,(1)杂交瘤V型基因进行密码子优化优化为更适合哺乳动物表达,并通过重叠延伸PCR合成。(2)小鼠单克隆抗体V型基因将通过CDR移植方法移植至最适合的框架上进行人源化。将设计12-16个人源化的变体,其框架与人类种系V-基因的差异最小。(3)每个位点最多将设计3个变体,以去除鼠类抗体序列中的关键PTM位点。(4)在针对哺乳动物表达的密码子优化之后,将合成人源化的V-基因和去除PTM的V-基因。小鼠和所有的变体基因将被克隆到人IgG表达载体中,以产生所需同型的人IgG1构建体,合成抗体基因,进行表达后纯化,纯化后同SPR方法鉴定抗体亲和力,筛选出最佳亲和力人源化抗体序列。
1.4人源化抗体CAR T细胞制备和功能研究
1.4.1 CAR结构设计
CAR结构包括开发的抗EGFRvIII抗原的鼠源抗体序列ScFv结构,还包括但不限于CD8铰链区和跨膜区,41-BB激活信号以及CD3z激活结构域,同时可以在此基础上进行其它修饰,包括细胞因子的分泌,趋化因子的分泌,免疫检查点抑制基因的修饰,肿瘤微环境抑制信号封闭和转化为激活型号等四代CAR T。
1.4.2 CAR基因合成
将合成的CAR基因序列通过分子克隆连接至慢病毒表达质粒上,合成的质粒转染至感受态细胞Stbl3,摇菌后大提质粒,用于后续慢病毒包装。
1.4.3慢病毒包装和滴度测定
分别将不同CAR质粒与包装质粒共转染到293T细胞中,转染后48h收集细胞上清,4000rpm离心10min。收集上清,用0.45μm的滤膜过滤,用超速离心机收集慢病毒沉淀。离心条件为4℃,100000×g,无刹车,离心90min。将浓缩后的慢病毒按每管200μl分装到0.5mL冻存管中,-80℃分装冻存,应用流式细胞术检测慢病毒滴度。
1.4.4 CAR T细胞制备
从一份健康人外周血中取出20ml加入到50ml离心管中,向血液中加入1mL RosetteSepTM Cocktail混匀,使抗体浓度为50μL/mL。将20mL GE Ficoll液加入新的50mL离心管中,用吸管小心吸取20mL稀释好的血样按1:1缓慢加入Ficoll液的上层。将加好血样的离心管放入离心机内,转速400g,离心30min。用无菌吸管吸掉上层血浆和血小板,不要碰到单个核细胞层,小心吸取第二层环状乳白色淋巴细胞层到新的离心管内。每管淋巴细胞悬液加入三倍体积PBS清洗细胞,400g离心10min。弃上清,用5mL PBS重悬所得细胞,1200rpm离心10min。最后用X-VIVO+100U IL2完全培养液悬浮细胞,细胞计数。将1mL分离的密度为1×10^6T淋巴细胞接种至24孔板的1个孔中,并加入25μl CD3/CD28磁珠。将细胞培养于5%CO2 37℃培养箱中,48h后用于慢病毒感染。 按照MOI=0.5加入相应的慢病毒,第二天细胞补液,每孔补加至30ml并转移至T 75瓶中,水平放置培养。
1.4.5 CAR T细胞CAR阳性率表达检测
3天后,将CAR-T细胞转移至无菌流式管内,每管细胞放置于biolegend磁铁上2min去除磁珠。之后去部分细胞染流式抗体(AF647-protein L)检测CAR表达率。
1.4.6 CAR T细胞体外杀伤效率检测
在检测白板中,样品孔接种U87MG-EGFRvIII靶细胞个数为1×10^4个/孔,重复6个孔,同时设置6个对照孔,每孔100μl。过夜后,吸弃上清后,加入CAR-T细胞,效靶比为E/T ratio=2.5:1;每孔100μl。6h后,300g离心5min,所有孔尽量去掉上清。每孔加入100μl裂解液,室温裂解10min,开始上机检测,每孔加入萤火虫荧光素酶检测试剂100μL。杀伤效率计算公式:杀伤效率(%)=(对照孔荧光强度-杀伤荧光强度)/(对照孔荧光强度-空白孔荧光强度)×100%。
1.4.7 CAR T细胞体内药效研究
选用6-8周雄性NOG小鼠作为实验动物。U87MG-EGFRvIII细胞消化后用无菌1×PBS重悬清洗细胞2次进行活细胞计数,调整细胞密度为5×10^5/50μl/只,细胞注射时按照1:1体积每只混合50μl基质胶,即最终每只小鼠注射100μl总体积的细胞混悬液。移植部位为小鼠背部右下侧皮下。肿瘤移植后,肿瘤体积用卡尺每周三次进行测量,肿瘤体积计算公式为TV=0.5a×b2。当肿瘤大小达到100mm3时按5ⅹ10^6/只回输CAR-T细胞或生理盐水,并持续观察肿瘤大小与小鼠存活状况。
1.5人源化抗体CAR NK细胞制备和功能研究
1.5.1 CAR NK细胞制备
(1)培养NK细胞,NK细胞加入Retronectin包被的24孔板中,再按照MOI=20在培养的NK细胞中加入慢病毒,32℃离心30min,将细胞转移至37℃,5%CO2培养箱中进行培养;
(2)感染48h后,应用完全培养基清洗细胞,应用含有300IU/ml的IL2的完全培养基中进行培养。
1.5.2 CAR NK细胞CAR阳性率表达检测
将CAR-NK细胞转移至无菌流式管内,细胞染流式抗体(AF647-protein L)检测CAR表达率。
1.5.3 CAR NK细胞体外杀伤效率检测
在检测白板中,样品孔接种U87MG-EGFRvIII靶细胞个数为1×10^4个/孔,重复6个孔,同时设置6个对照孔,每孔100μl。过夜后,吸弃上清后,加入CAR-NK细胞,效靶比为E/T ratio=1:1;每孔100μl。6h后,300g离心5min,所有孔尽量去掉上清。每孔加入100μl裂解液,室温裂解10min,开始上机检测,每孔加入萤火虫荧光素酶检测试剂100μL。杀伤效率计算公式:杀伤效率(%)=(对照孔荧光强度-杀伤荧光强度)/(对照孔荧光强度-空白孔荧光强度)×100%。
1.6抵抗TGFβ抑制的CAR T细胞制备和功能研究
1.6.1 CAR结构设计
设计了两种抵抗TGFβ抑制的CAR结构。第一种为通过T2A自剪切肽连接显性负性受体dnTGFβRII(图20A)。dnTGFβRII作为受体具有结合TGFβ的能力,但其缺乏将结合TGFβ转换为胞内信号的能力(例如缺少胞内结构域或缺乏信号传导所需的激酶活性)。第二种为通过T2A自剪切肽连接嵌合转换受体(dnTGFβRII-IL7RA TM-IL7RA胞内区)(图20B)。该嵌合转换受体的存在可以抗TGFβ抑制,还可将结合TGFβ转换为胞内IL7RA受体刺激信号,促进CAR T细胞的增殖和/或活性。
1.6.2 CAR T细胞制备
具体方法同人源化抗体CAR T细胞制备流程。
1.6.3 CAR T细胞功能研究
在检测白板中,样品孔接种U87MG-EGFRvIII靶细胞个数为1×10^4个/孔,重复6个孔,同时设置6个对照孔,每孔100μl。过夜后,吸弃上清后,加入CAR-T细胞,效靶比为E/T ratio=2.5:1;每孔100μl。6h后,300g离心5min,所有孔尽量去掉上清。每孔加入100μl裂解液,室温裂解10min,开始上机检测,每孔加入萤火虫荧光素酶检测试剂100μL。杀伤效率计算公式:杀伤效率(%)=(对照孔荧光强度-杀伤荧光强度)/(对照孔荧光强度-空白孔荧光强度)×100%。
为了比较修饰型四代CAR T细胞的抵抗TGFβ抑制作用,在做杀伤同时设置TGFβ抑制组进行比较,在CAR T细胞进行杀伤实验前,取部分细胞应用30ng/ml的TGFβ进行CAR T细胞的抑制,然后进行抑制后CAR T细胞和未抑制CAR T细胞的杀伤效率监测。
1.6.4 CAR T细胞体内功能研究
选用6-8周雄性NOG小鼠作为实验动物。U87MG-EGFRvIII细胞消化后用无菌1×PBS重悬清洗细胞2次进行活细胞计数,调整细胞密度为5×10^5/50μl/只,细胞注射时按照1:1体积每只混合50μl基质胶,即最终每只小鼠注射100μl总体积的细胞混悬液。移植部位为小鼠背部右下侧皮下。肿瘤移植后,肿瘤体积用卡尺每周三次进行测量,肿瘤体积计算公式为TV=0.5a×b2。当肿瘤大小达到100mm3时按3ⅹ10^6/只回输CAR-T细胞或生理盐水,并持续观察肿瘤大小与小鼠存活状况。
本实施例涉及的部分序列信息如下:


◆实验结果
1.鼠源抗体亲和力测定结果
1.1鼠源抗体亚克隆上清抗体亲和力检测结果
通过流式细胞术检测亚克隆上清分别对F98npEGFR和F98npEGFRvIII的结合能力,结果显示筛选的阳性亚克隆可特异性结合过表达EGFRvIII的靶细胞F98npEGFRvIII,而与过表达EGFR的靶细胞F98npEGFR不结合,说明筛选到可特异性结合突变形式EGFRvIII靶抗原的特异性抗体(图2)。
1.2鼠源抗体纯化抗体亲和力检测结果
SPR结果显示体外药效佳的鼠源抗体对EGFRvIII靶抗原均有较高的亲和力(表1和图3)。
表1 SPR方法检测纯化鼠源抗体亲和力
2.鼠源抗体序列(采用IMGT编码方案)


注:对20-2-40单个克隆进行测序时获得了不同的抗体序列,分别记为20-2-40-1和20-2-40-2。
3.CAR结构序列
4.鼠源抗体制备CAR T细胞体外药效研究结果
4.1鼠源抗体制备CAR T细胞的CAR阳性率
应用流式细胞术检测CAR T细胞CAR阳性率,结果显示鼠源抗体序列制备的CAR T细胞的CAR阳性率为50-90%之间(图5)。
4.2鼠源抗体制备CAR T细胞的杀伤效率检测结果
分别检测鼠源抗体序列制备的CAR T细胞对过表达EGFRvIII的靶细胞U87MG-EGFRvIII和表达EGFR的靶细胞U87MG-EGFR进行杀伤效率检测,检测结果显示制备的CAR T细胞均特异性杀伤过表达EGFRvIII的靶细胞,而对表达EGFR的靶细胞几乎没有杀伤效率,说明开发的抗体制备的CAR T细胞具有特异性的杀伤效率(图6和图7)。其中20-2-1,20-2-4,20-2-25,20-2-28,20-2-34和20-2-40-2杀伤效率较高。
5.鼠源抗体制备的CAR T细胞体内抑瘤效率
应用U87MG-EGFRvIII在小鼠体内进行皮下成瘤,肿瘤体积生长至100mm3左右进行CAR T细胞回输,回输CAR T细胞后每隔一周检测小鼠肿瘤体积,检测结果显示4、28、40-2号克隆抗体序列制备的CAR T细胞在体内具有显著的抑制肿瘤作用(图8)。
6.人源化抗体亲和力测定结果
通过CDR移植和翻译后修饰位点移除方法进行鼠源抗体的人源化,结果显示#28克隆人源化后获得可和鼠源抗体亲和力保持一致的人源化抗体序列,#40-2克隆鼠源抗体人源化后有2倍左右的亲和力下降(表2和3,图9和图10)。
表2 SPR方法检测纯化的#28克隆人源化抗体亲和力
表3 SPR方法检测纯化的#40-2克隆人源化抗体亲和力

7.人源化抗体序列






8.人源化抗体制备CAR T细胞体外药效研究结果
8.1人源化抗体CAR T细胞CAR阳性率检测
应用流式细胞术检测CAR T细胞CAR阳性率,结果显示鼠源抗体序列制备的CAR T细胞的CAR阳性率为50-90%之间(图11和图12)。
8.2人源化抗体CAR T和CAR NK细胞体外杀伤效率检测结果
分别检测人源抗体序列制备的CAR T细胞对过表达EGFRvIII的靶细胞U87MG-EGFRvIII和表达EGFR的靶细胞U87MG-EGFR进行杀伤效率检测,检测结果显示制备的CAR T细胞均特异性杀伤过表达EGFRvIII的靶细胞,而对表达EGFR的靶细胞几乎没有杀伤效率,说明开发的人源化抗体制备的CAR T细胞具有特异性的杀伤效率(图13、14、15和16)。类似地,人源抗体序列制备的CAR NK细胞特异性杀伤过表达EGFRvIII的靶细胞,而对表达EGFR的靶细胞几乎没有杀伤效率(图19A和19B)。
9.人源化抗体制备的CAR T细胞体内抑瘤效率研究
应用U87MG-EGFRvIII在小鼠体内进行皮下成瘤,肿瘤体积生长至100mm3左右进行CAR T细胞回输,回输CAR T细胞后每隔一周检测小鼠肿瘤体积,检测结果显示28-H9、40-2-H10、40-2-H12、40-2-H13、40-2-H15号克隆抗体序列制备的CAR T细胞在体内具有显著的抑制肿瘤作用(图17和18)。
10.抵抗TGFβ的CAR T细胞体外药效研究结果
10.1 CAR阳性率检测结果
CAR阳性率检测结果显示在表4和图21A-C中。分别应用PE-EGFRvIII抗原蛋白检测RII(表达dnTGFβRII的CAR T细胞)、T7R(表达嵌合转换受体dnTGFβRII-IL7Rα的CAR T细胞)或CAR T C细胞(传统二代CAR T细胞,不表达DNR或CSR)的CAR阳性率。
表4四代CAR T细胞CAR阳性率检测结果
结果显示:制备的CAR T细胞CAR阳性率在50%以上。
10.2 CAR T细胞体外杀伤效率检测结果
结果显示:过表达TGFβRII和TGFβRII-IL7R的CAR T细胞在位加TGEβ抑制时未见杀伤效率显著差异(图22A),加入TGFβ抑制后CAR T细胞整体杀伤效率显著下降(图22B),但过表达TGFβRII和TGFβRII-IL21R的CAR T细胞杀伤效率下降率差显著降低(图22C)。
10.3 CAR T细胞体内抑瘤作用检测结果
结果显示:过表达TGFβRII和TGFβRII-IL21R的CAR T细胞在体内的抑制肿瘤效率显著高于对照组CAR T细胞(图23)。
讨论
EGFRvIII是EGFR的一种突变形式,是EGFR的2-7外显子删除,在删除后的序列连接处添加一个甘氨酸,形成新的抗原表位,因此EGFRvIII和EGFR的序列之间具有很高的同源性,对抗体序列的筛选造成很大的难度,由于EGFR在所有的上皮细胞中均有表达,而EGFRvIII是肿瘤特异性抗原,仅在肿瘤上表达。对于靶向EGFRvIII靶点的药物开发需要筛选仅特异性识别EGFRvIII靶抗原,而对野生型EGFR抗原是不识别的,因此对抗体的特异性要求较高,如果抗体同时识别EGFR抗原会造成脱靶的风险,因此本研究进行多种小鼠免疫策略的优化以及抗体筛选策略的优化,筛选到具有高特异性高亲和力的鼠源抗体序列,同时应用鼠源序列可以制备出具有显著抑瘤作用的免疫细胞。为了降低药物的免疫源性,本研究在鼠源抗体序列的基础上进行了鼠源抗体序列的人源化, 筛选到具有高亲和力高特异性的人源化抗体,同时研发出具有显著抑瘤作用的人源化抗体制备的免疫细胞,为后续抗肿瘤药物的开发奠定了基础。
抗体药物和细胞免疫治疗近年来在肿瘤治疗领域取得了显著的进展,胶质瘤是目前恶性程度很高,没有有效治疗手段的肿瘤,因此急需研发可以有效治疗脑胶质瘤的药物,研究表明在30-60%的脑胶质瘤患者中均有EGFRvIII抗原的表达,因此研发的靶向EGFRvIII抗体和应用此抗体序列制备的免疫细胞将将可以应用于EGFRvIII表达的脑胶质瘤及其他具有EGFRvIII表达的肿瘤上。
由于抗体使得药物具有靶向性,因此目前抗体药物具有多种形式,包括单抗、双抗、多抗、ADC和免疫细胞等,因此本研究开发的抗体可以应用于不同的领域。另外由于目前靶向治疗都需要相应靶向诊断,因此本研究开发的抗体同样可以用于诊断试剂盒的开发盒伴随诊断的应用。
目前CAR T细胞在血液肿瘤上取得了显著的进展,已经有CAR T细胞上市,CAR结构由抗原识别结构域、铰链区、跨膜区、共刺激结构域和激活结构域组成,本研究开发的抗体可以和不同的结构进行组装,然后制备不容类型的免疫细胞,包括但不限于T细胞、NK细胞、巨噬细胞、单核细胞、B细胞、红细胞等。由于实体肿瘤的治疗受到了免疫抑制微环境、浸润性和持久性的限制,需要在研发的免疫细胞上进行不同的修饰,包括但不限于基因编辑、基因敲除、RNAi等,赋予CAR T细胞在体内持久性、浸润性以及解除免疫微环境抑制的能力,因此本研究开发的抗体制备的免疫细胞会进行不同的修饰来提高免疫细胞的抑制肿瘤的能力。
参考文献:
1.Akhavan,D.,Alizadeh,D.,Wang,D.,Weist,M.R.,Shepphird,J.K.,and Brown,C.E.(2019).CAR T cells for brain tumors:Lessons learned and road ahead.Immunological reviews 290,60-84.
An,Z.,Aksoy,O.,Zheng,T.,Fan,Q.,and Weiss,W.J.O.(2018).Epidermal growth factor receptor and EGFRvIII in glioblastoma:signaling pathways and targeted therapies.37,1561-1575.
Chistiakov,D.,Chekhonin,I.,and Chekhonin,V.J.E.j.o.p.(2017).The EGFR variant III mutant as a target for immunotherapy of glioblastoma multiforme.810,70-82.
Del Vecchio,C.,Li,G.,and Wong,A.J.E.r.o.v.(2012).Targeting EGF receptor variant III:tumor-specific peptide vaccination for malignant gliomas.11,133-144.
Feldman,L.,Brown,C.,and Badie,B.(2022).Chimeric Antigen Receptor(CAR)T Cell Therapy for Glioblastoma.Neuromolecular Med 24,35-40.
Londhe,V.,and Date,V.J.E.r.o.v.(2020).Personalized neoantigen vaccines:a glimmer of hope for glioblastoma.19,407-417.
O'Rourke,D.M.,Nasrallah,M.P.,Desai,A.,Melenhorst,J.J.,Mansfield,K.,Morrissette,J.J.D.,Martinez-Lage,M.,Brem,S.,Maloney,E.,Shen,A.,et al.(2017).A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma.Science translational medicine 9.
Zhu,G.,Zhang,Q.,Zhang,J.,and Liu,F.(2021).Targeting Tumor-Associated Antigen:A Promising CAR-T Therapeutic Strategy for Glioblastoma Treatment.Frontiers in pharmacology 12,661606.

Claims (46)

  1. 靶向EGFRvIII的抗体或其抗原结合片段,其中所述抗体包括重链可变区(HCVR),所述重链可变区包括HCDR1、HCDR2和HCDR3,所述HCDR1、HCDR2和HCDR3选自如下组合之一:
    (1)HCDR1的氨基酸序列为DFSMH(SEQ ID NO:1);
    HCDR2的氨基酸序列为WINTETGEPSYADDFKG(SEQ ID NO:2);
    HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:3);
    (2)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:4);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:5);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:6);
    (3)HCDR1的氨基酸序列为DYSIH(SEQ ID NO:7);
    HCDR2的氨基酸序列为WINTETGEPTYADDFKG(SEQ ID NO:8);
    HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:9);
    (4)HCDR1的氨基酸序列为DYYLH(SEQ ID NO:10);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:11);
    HCDR3的氨基酸序列为GYLTY(SEQ ID NO:12);
    (5)HCDR1的氨基酸序列为RYWMH(SEQ ID NO:13);
    HCDR2的氨基酸序列为EINPSNGRANYNEKFMS(SEQ ID NO:14);
    HCDR3的氨基酸序列为GREITTGFAY(SEQ ID NO:15);
    (6)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:16);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:17);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:18);
    (7)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:19);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:20);
    HCDR3的氨基酸序列为GYLAY(SEQ ID NO:21);
    (8)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:22);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:23);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:25);
    (9)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:25);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:26);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:27);
    (10)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:28);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:29);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:30);
    (11)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:31);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:32);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:33);
    (12)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:34);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:35);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:36);
    (13)HCDR1的氨基酸序列为NYAMS(SEQ ID NO:37);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:38);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:39);
    (14)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:40);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:41);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:42);
    (15)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:43);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:44);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:45);
    (16)HCDR1的氨基酸序列为SGYSWH(SEQ ID NO:46);
    HCDR2的氨基酸序列为YIHYSGSTNYNPPLKS(SEQ ID NO:47);
    HCDR3的氨基酸序列为GVVSNYAMGN(SEQ ID NO:48);
    (17)HCDR1的氨基酸序列为TYWMH(SEQ ID NO:49);
    HCDR2的氨基酸序列为YINPNTAYTEYNQNFKD(SEQ ID NO:50);
    HCDR3的氨基酸序列为GAYYRTYYAMDY(SEQ ID NO:51);
    (18)HCDR1的氨基酸序列为NYGMN(SEQ ID NO:52);
    HCDR2的氨基酸序列为WINTYTGEPTYADDFKG(SEQ ID NO:53);
    HCDR3的氨基酸序列为EEFYSRGAMDY(SEQ ID NO:54);以及
    (19)HCDR1的氨基酸序列为DYYIN(SEQ ID NO:55);
    HCDR2的氨基酸序列为WIYPGSGNTKYNEKFKG(SEQ ID NO:56);
    HCDR3的氨基酸序列为SSRCDF(SEQ ID NO:57),或者
    所述抗体包括(1)-(19)任一项中的HCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的HCDR序列相比,具有至少90%的序列一致性,或在HCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
  2. 如权利要求1所述的抗体或其抗原结合片段,其中所述抗体还包括轻链可变区(LCVR),所述轻链可变区包括LCDR1、LCDR2和LCDR3,所述LCDR1、LCDR2和LCDR3选自如下组合之一:
    (1)LCDR1的氨基酸序列为SASSSISSNYLH(SEQ ID NO:58);
    LCDR2的氨基酸序列为GTSNLAS(SEQ ID NO:59);
    LCDR3的氨基酸序列为HQGSSIPLT(SEQ ID NO:60);
    (2)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:61);
    LCDR2的氨基酸序列为FASTRAS(SEQ ID NO:62);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:63);
    (3)LCDR1的氨基酸序列为SASSGISSNYLH(SEQ ID NO:64);
    LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:65);
    LCDR3的氨基酸序列为HQGSDIPLT(SEQ ID NO:66);
    (4)LCDR1的氨基酸序列为KSSQNLLNSSNQKNYLA(SEQ ID NO:67);
    LCDR2的氨基酸序列为FASTRYS(SEQ ID NO:68);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:69);
    (5)LCDR1的氨基酸序列为KASQSVSNDVV(SEQ ID NO:70);
    LCDR2的氨基酸序列为YASNRYT(SEQ ID NO:71);
    LCDR3的氨基酸序列为QQDYSSPWT(SEQ ID NO:72);
    (6)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:73);
    LCDR2的氨基酸序列为FASTRES(SEQ ID NO:74);
    LCDR3的氨基酸序列为QQHYSIPLT(SEQ ID NO:75);
    (7)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:76);
    LCDR2的氨基酸序列为FASTRKS(SEQ ID NO:77);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:78);
    (8)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:79);
    LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:80);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:81);
    (9)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:82);
    LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:83);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:84);
    (10)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:85);
    LCDR2的氨基酸序列为FASTRGS(SEQ ID NO:86);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:87);
    (11)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:88);
    LCDR2的氨基酸序列为FASTRDS(SEQ ID NO:89);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:90);
    (12)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:91);
    LCDR2的氨基酸序列为FASTRES(SEQ ID NO:92);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:93);
    (13)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:94);
    LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:95);
    LCDR3的氨基酸序列为SQSIHVPWT(SEQ ID NO:96);
    (14)LCDR1的氨基酸序列为SASSSVSYMH(SEQ ID NO:97);
    LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:98);
    LCDR3的氨基酸序列为QQRSSYPLT(SEQ ID NO:99);
    (15)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:100);
    LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:101);
    LCDR3的氨基酸序列为SQTTQVPWT(SEQ ID NO:102);
    (16)LCDR1的氨基酸序列为ITNTDIDDDMN(SEQ ID NO:103);
    LCDR2的氨基酸序列为EGNTLRP(SEQ ID NO:104);
    LCDR3的氨基酸序列为LQSDDLPLT(SEQ ID NO:105);
    (17)LCDR1的氨基酸序列为KASQSVDYDGDSYMN(SEQ ID NO:106);
    LCDR2的氨基酸序列为AASNLES(SEQ ID NO:107);
    LCDR3的氨基酸序列为LQSNEDPYT(SEQ ID NO:108);
    (18)LCDR1的氨基酸序列为RSSQFIVHSNGNTYLE(SEQ ID NO:109);
    LCDR2的氨基酸序列为KISNRFS(SEQ ID NO:110);
    LCDR3的氨基酸序列为FQGSHVPFT(SEQ ID NO:111);以及
    (19)LCDR1的氨基酸序列为KASEDIYNRLA(SEQ ID NO:112);
    LCDR2的氨基酸序列为GATSLET(SEQ ID NO:113);
    LCDR3的氨基酸序列为QQYWSSPLT(SEQ ID NO:114),或者
    所述抗体包括(1)-(19)任一项中的LCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的LCDR序列相比,具有至少90%的序列一致性,或在LCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
  3. 如权利要求1或2所述的抗体或其抗原结合片段,其中所述重链可变区的氨基酸序列选自如下任一项:
    (1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
    (19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
  4. 如权利要求1-3任一项所述的抗体或其抗原结合片段,其中所述抗体的重链可变区的氨基酸序列和轻链可变区的氨基酸序列选自如下任一组合:
    (1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:116所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:118所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:120所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:122所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:124所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:126所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:128所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:130所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:132所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:134所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:136所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:138所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:140所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:142所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:144所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:146所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:148所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:150所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;以及
    (19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:152所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列。
  5. 如权利要求1-4任一项所述的抗体或其抗原结合片段,其中通过表面等离子共振(SPR)测定的所述抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
  6. 如权利要求1-5任一项所述的抗体或其抗原结合片段,其中所述抗体选自鼠源抗体、嵌合抗体、人源化抗体或人抗体。
  7. 如权利要求1-6任一项所述的抗体或其抗原结合片段,其中通过表面等离子共振(SPR)测定的所述人源化抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
  8. 如权利要求1-7任一项所述的抗体或其抗原结合片段,其中所述抗原结合片段包括Fab、Fab'、F(ab')2、单域抗体或单链抗体。
  9. 如权利要求1-8任一项所述的抗体或其抗原结合片段,其中所述人源化抗体为单链抗体,其氨基酸序列选自如下任一项:
    (1)SEQ ID NO:167所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (2)SEQ ID NO:169所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (3)SEQ ID NO:171所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (4)SEQ ID NO:173所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (5)SEQ ID NO:175所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (6)SEQ ID NO:177所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (7)SEQ ID NO:179所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (8)SEQ ID NO:181所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (9)SEQ ID NO:183所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (10)SEQ ID NO:185所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
    (11)SEQ ID NO:187所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
  10. 如权利要求1-9任一项所述的抗体或其抗原结合片段,还包括Fc片段。
  11. 如权利要求1-10任一项所述的抗体或其抗原结合片段,其中所述抗原结合片段为单链抗体;优选地,用于连接所述单链抗体中的重链可变区和轻链可变区的接头序列(linker)包括SEQ ID NO:166所示序列。
  12. 嵌合抗原受体(CAR),其胞外抗原结合结构域包括一个或更多个靶向EGFRvIII的抗体分子或其抗原结合片段,所述抗体分子的重链可变区的HCDR1、HCDR2和HCDR3选自如下组合之一:
    (1)HCDR1的氨基酸序列为DFSMH(SEQ ID NO:1);
    HCDR2的氨基酸序列为WINTETGEPSYADDFKG(SEQ ID NO:2);
    HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:3);
    (2)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:4);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:5);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:6);
    (3)HCDR1的氨基酸序列为DYSIH(SEQ ID NO:7);
    HCDR2的氨基酸序列为WINTETGEPTYADDFKG(SEQ ID NO:8);
    HCDR3的氨基酸序列为YGYDVRGDY(SEQ ID NO:9);
    (4)HCDR1的氨基酸序列为DYYLH(SEQ ID NO:10);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:11);
    HCDR3的氨基酸序列为GYLTY(SEQ ID NO:12);
    (5)HCDR1的氨基酸序列为RYWMH(SEQ ID NO:13);
    HCDR2的氨基酸序列为EINPSNGRANYNEKFMS(SEQ ID NO:14);
    HCDR3的氨基酸序列为GREITTGFAY(SEQ ID NO:15);
    (6)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:16);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:17);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:18);
    (7)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:19);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:20);
    HCDR3的氨基酸序列为GYLAY(SEQ ID NO:21);
    (8)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:22);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:23);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:25);
    (9)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:25);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:26);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:27);
    (10)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:28);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:29);
    HCDR3的氨基酸序列为GWFAY(SEQ ID NO:30);
    (11)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:31);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:32);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:33);
    (12)HCDR1的氨基酸序列为DYYMH(SEQ ID NO:34);
    HCDR2的氨基酸序列为WIDPENGNTIYDPKFQG(SEQ ID NO:35);
    HCDR3的氨基酸序列为GYLVY(SEQ ID NO:36);
    (13)HCDR1的氨基酸序列为NYAMS(SEQ ID NO:37);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:38);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:39);
    (14)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:40);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:41);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:42);
    (15)HCDR1的氨基酸序列为GYAMS(SEQ ID NO:43);
    HCDR2的氨基酸序列为TITSGGSYTYYPDSVKG(SEQ ID NO:44);
    HCDR3的氨基酸序列为KDYGNYWFAY(SEQ ID NO:45);
    (16)HCDR1的氨基酸序列为SGYSWH(SEQ ID NO:46);
    HCDR2的氨基酸序列为YIHYSGSTNYNPPLKS(SEQ ID NO:47);
    HCDR3的氨基酸序列为GVVSNYAMGN(SEQ ID NO:48);
    (17)HCDR1的氨基酸序列为TYWMH(SEQ ID NO:49);
    HCDR2的氨基酸序列为YINPNTAYTEYNQNFKD(SEQ ID NO:50);
    HCDR3的氨基酸序列为GAYYRTYYAMDY(SEQ ID NO:51);
    (18)HCDR1的氨基酸序列为NYGMN(SEQ ID NO:52);
    HCDR2的氨基酸序列为WINTYTGEPTYADDFKG(SEQ ID NO:53);
    HCDR3的氨基酸序列为EEFYSRGAMDY(SEQ ID NO:54);以及
    (19)HCDR1的氨基酸序列为DYYIN(SEQ ID NO:55);
    HCDR2的氨基酸序列为WIYPGSGNTKYNEKFKG(SEQ ID NO:56);
    HCDR3的氨基酸序列为SSRCDF(SEQ ID NO:57),或者
    所述抗体包括(1)-(19)任一项中的HCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的HCDR序列相比,具有至少90%的序列一致性,或在HCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
  13. 如权利要求12所述的CAR,其中所述抗体还包括轻链可变区(LCVR),所述轻链可变区包括LCDR1、LCDR2和LCDR3,所述LCDR1、LCDR2和LCDR3选自如下组合之一:
    (1)LCDR1的氨基酸序列为SASSSISSNYLH(SEQ ID NO:58);
    LCDR2的氨基酸序列为GTSNLAS(SEQ ID NO:59);
    LCDR3的氨基酸序列为HQGSSIPLT(SEQ ID NO:60);
    (2)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:61);
    LCDR2的氨基酸序列为FASTRAS(SEQ ID NO:62);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:63);
    (3)LCDR1的氨基酸序列为SASSGISSNYLH(SEQ ID NO:64);
    LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:65);
    LCDR3的氨基酸序列为HQGSDIPLT(SEQ ID NO:66);
    (4)LCDR1的氨基酸序列为KSSQNLLNSSNQKNYLA(SEQ ID NO:67);
    LCDR2的氨基酸序列为FASTRYS(SEQ ID NO:68);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:69);
    (5)LCDR1的氨基酸序列为KASQSVSNDVV(SEQ ID NO:70);
    LCDR2的氨基酸序列为YASNRYT(SEQ ID NO:71);
    LCDR3的氨基酸序列为QQDYSSPWT(SEQ ID NO:72);
    (6)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:73);
    LCDR2的氨基酸序列为FASTRES(SEQ ID NO:74);
    LCDR3的氨基酸序列为QQHYSIPLT(SEQ ID NO:75);
    (7)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:76);
    LCDR2的氨基酸序列为FASTRKS(SEQ ID NO:77);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:78);
    (8)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:79);
    LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:80);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:81);
    (9)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:82);
    LCDR2的氨基酸序列为FASTRQS(SEQ ID NO:83);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:84);
    (10)LCDR1的氨基酸序列为KSSQSLLNSSNQKNHLA(SEQ ID NO:85);
    LCDR2的氨基酸序列为FASTRGS(SEQ ID NO:86);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:87);
    (11)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:88);
    LCDR2的氨基酸序列为FASTRDS(SEQ ID NO:89);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:90);
    (12)LCDR1的氨基酸序列为KSSQSLLNSSNQKNYLA(SEQ ID NO:91);
    LCDR2的氨基酸序列为FASTRES(SEQ ID NO:92);
    LCDR3的氨基酸序列为QQHYSTPLT(SEQ ID NO:93);
    (13)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:94);
    LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:95);
    LCDR3的氨基酸序列为SQSIHVPWT(SEQ ID NO:96);
    (14)LCDR1的氨基酸序列为SASSSVSYMH(SEQ ID NO:97);
    LCDR2的氨基酸序列为STSNLAS(SEQ ID NO:98);
    LCDR3的氨基酸序列为QQRSSYPLT(SEQ ID NO:99);
    (15)LCDR1的氨基酸序列为RSSQSLVHSDGNTYLH(SEQ ID NO:100);
    LCDR2的氨基酸序列为KVSNRFS(SEQ ID NO:101);
    LCDR3的氨基酸序列为SQTTQVPWT(SEQ ID NO:102);
    (16)LCDR1的氨基酸序列为ITNTDIDDDMN(SEQ ID NO:103);
    LCDR2的氨基酸序列为EGNTLRP(SEQ ID NO:104);
    LCDR3的氨基酸序列为LQSDDLPLT(SEQ ID NO:105);
    (17)LCDR1的氨基酸序列为KASQSVDYDGDSYMN(SEQ ID NO:106);
    LCDR2的氨基酸序列为AASNLES(SEQ ID NO:107);
    LCDR3的氨基酸序列为LQSNEDPYT(SEQ ID NO:108);
    (18)LCDR1的氨基酸序列为RSSQFIVHSNGNTYLE(SEQ ID NO:109);
    LCDR2的氨基酸序列为KISNRFS(SEQ ID NO:110);
    LCDR3的氨基酸序列为FQGSHVPFT(SEQ ID NO:111);以及
    (19)LCDR1的氨基酸序列为KASEDIYNRLA(SEQ ID NO:112);
    LCDR2的氨基酸序列为GATSLET(SEQ ID NO:113);
    LCDR3的氨基酸序列为QQYWSSPLT(SEQ ID NO:114),或者
    所述抗体包括(1)-(19)任一项中的LCDR序列组合的变体,其中所述变体与(1)-(19)任一项中的LCDR序列相比,具有至少90%的序列一致性,或在LCDR序列上共包含至少1个且不超过10,或不超过5、4、3、或2个氨基酸改变。
  14. 如权利要求12或13所述的CAR,其中所述重链可变区的氨基酸序列选自如下任一项:
    (1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
    (19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
  15. 如权利要求12-14任一项所述的CAR,其中所述抗体的重链可变区的氨基酸序列和轻链可变区的氨基酸序列选自如下任一组合:
    (1)SEQ ID NO:115所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:116所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (2)SEQ ID NO:117所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:118所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (3)SEQ ID NO:119所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:120所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (4)SEQ ID NO:121所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:122所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (5)SEQ ID NO:123所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:124所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (6)SEQ ID NO:125所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:126所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (7)SEQ ID NO:127所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:128所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (8)SEQ ID NO:129所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:130所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (9)SEQ ID NO:131所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:132所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (10)SEQ ID NO:133所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:134所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (11)SEQ ID NO:135所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:136所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (12)SEQ ID NO:137所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:138所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (13)SEQ ID NO:139所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:140所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (14)SEQ ID NO:141所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:142所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (15)SEQ ID NO:143所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:144所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (16)SEQ ID NO:145所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:146所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (17)SEQ ID NO:147所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:148所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;
    (18)SEQ ID NO:149所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:150所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列;以及
    (19)SEQ ID NO:151所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列和SEQ ID NO:152所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的轻链可变区序列。
  16. 如权利要求12-15任一项所述的CAR,其中通过表面等离子共振(SPR)测定的所述抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
  17. 如权利要求12-16任一项所述的CAR,其中所述抗体选自鼠源抗体、嵌合抗体、人源化抗体或人抗体。
  18. 如权利要求12-17任一项所述的CAR,其中通过表面等离子共振(SPR)测定的所述人源化抗体对EGFRvIII抗原的结合KD值低于10-6M,优选低于10-7M。
  19. 如权利要求12-18任一项所述的CAR,其中所述抗原结合片段为单链抗体;优选地,用于连接所述单链抗体中的重链可变区和轻链可变区的接头序列(linker)包括SEQ ID NO:166所示序列。
  20. 如权利要求12-19任一项所述的CAR,其中所述人源化抗体为单链抗体,其氨基酸序列选自如下任一项:
    (1)SEQ ID NO:167所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (2)SEQ ID NO:169所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (3)SEQ ID NO:171所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (4)SEQ ID NO:173所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (5)SEQ ID NO:175所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (6)SEQ ID NO:177所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (7)SEQ ID NO:179所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (8)SEQ ID NO:181所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
    (9)SEQ ID NO:183所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;
    (10)SEQ ID NO:185所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列;以及
    (11)SEQ ID NO:187所示序列或与其有至少90%、95%、96%、97%、98%或99%序列一致性的重链可变区序列。
  21. 如权利要求12-20任一项所述的CAR,其还包括铰链区、跨膜区、共刺激结构域和激活结构域;优选地,所述铰链区为CD8铰链区,所述跨膜区为CD8α跨膜区;所述共刺激结构域为41BB共刺激结构域或CD28共刺激结构域;所述激活结构域为CD3ζ激活结构域。
  22. 如权利要求12-21任一项所述的CAR,其中所述铰链区包括SEQ ID NO:156所示序列;所述跨膜区包括SEQ ID NO:158所示序列;所述共刺激结构域包括SEQ ID NO:160和/或162所示序列;所述激活结构域包括SEQ ID NO:164所示序列。
  23. 如权利要求12-22任一项所述的CAR,其中所述CAR还包括信号肽;优选地,所述信号肽为CD8α信号肽;更优选地,所述信号肽包括SEQ ID NO:154所示序列。
  24. 如权利要求12-23任一项所述的CAR,其中所述信号肽包括SEQ ID NO:154所示序列。
  25. 核酸分子,其编码:
    1)权利要求1-11任一项的抗体或其抗原结合片段,或所述抗体或其抗原结合片段的重链可变区和/或轻链可变区;或
    2)权利要求12-24任一项所述的CAR。
  26. 如权利要求25所述的核酸分子,其包括SEQ ID NO:153、155、157、159、161、163、165、168、170、172、174、176、178、180、182、184、186或188任一项所示的核苷酸序列。
  27. 表达载体,其包括权利要求25或26所述的核酸分子;优选地,所述表达载体为慢病毒载体。
  28. 包括权利要求27所述的表达载体的细胞。
  29. 表达权利要求12-24任一项所述的CAR的细胞。
  30. 如权利要求28或29所述的细胞,其为免疫细胞。
  31. 如权利要求28-30任一项所述的细胞,其为T细胞或NK细胞。
  32. 如权利要求29-31任一项所述的细胞,其还经改造以表达显性负性受体(DNR)或嵌合转换受体(CSR);优选地,所述DNR为dnTGFβRII和/或dnPD1;优选地,所述CSR的胞内结构域来自IL7Rα.;更优选地,所述dnTGFβRII包括SEQ ID NO:193所示序列,所述CSR的胞内结构域包括SEQ ID NO:197所示序列。
  33. 药物组合物,其包括:
    1)权利要求1-11任一项的抗体或其抗原结合片段、权利要求25或26的核酸分子、权利要求27的表达载体、或者权利要求29-32任一项的细胞;以及
    2)药学上可接受的载体。
  34. 权利要求1-11任一项的抗体或其抗原结合片段、权利要求25或26的核酸分子、权利要求27的表达载体、或者权利要求29-32任一项的细胞在制备用于治疗肿瘤的药物中的用途。
  35. 如权利要求34所述的用途,其中所述肿瘤表达EGFRvIII。
  36. 如权利要求34或35所述的用途,其中所述肿瘤为胶质母细胞瘤。
  37. 在受试者中治疗肿瘤的方法,包括以治疗有效量的权利要求1-11任一项的抗体或其抗原结合片段、权利要求25或26的核酸分子、权利要求27的表达载体、权利要求29-32任一项的细胞、或权利要求33所述的药物组合物向所述受试者给药。
  38. 如权利要求37所述的方法,其中所述肿瘤表达EGFRvIII。
  39. 如权利要求37或38所述的方法,其中所述肿瘤为胶质母细胞瘤。
  40. 多特异性抗体分子,至少包括第一功能部分和第二功能部分,其中第一功能部分包括权利要求1-11任一项所述的抗体或其抗原结合片段;第二功能部分具有与所述第一功能部分不同的抗原结合特异性。
  41. 如权利要求40所述的多特异性抗体分子,其中第二功能部分对T细胞具有结合特异性。
  42. 免疫偶联物,包括与治疗剂连接的权利要求1-11中任一项所述的抗体或其抗原结合片段。
  43. 如权利要求42所述的免疫偶联物,其中所述治疗剂是药物。
  44. 如权利要求42或43所述的免疫偶联物,其中所述治疗剂是细胞毒素。
  45. 如权利要求42-44任一项所述的免疫偶联物,其中所述治疗剂是放射性同位素。
  46. 试剂盒,用于检测样品中是否存在EGFRvIII或其含量,其中所述试剂盒包括权利要求1-11任一项所述的抗体或其抗原结合片段。
PCT/CN2023/111305 2022-08-05 2023-08-04 靶向EGFRvIII的抗体及其在细胞免疫治疗的应用 WO2024027835A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210936403 2022-08-05
CN202210936403.1 2022-08-05

Publications (1)

Publication Number Publication Date
WO2024027835A1 true WO2024027835A1 (zh) 2024-02-08

Family

ID=89848556

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/111305 WO2024027835A1 (zh) 2022-08-05 2023-08-04 靶向EGFRvIII的抗体及其在细胞免疫治疗的应用

Country Status (1)

Country Link
WO (1) WO2024027835A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105358576A (zh) * 2013-02-20 2016-02-24 诺华股份有限公司 使用人源化抗EGFRvIII嵌合抗原受体治疗癌症
CN108699124A (zh) * 2016-01-21 2018-10-23 辉瑞大药厂 靶向表皮生长因子受体变体iii的嵌合抗原受体
CN111548417A (zh) * 2020-04-03 2020-08-18 首都医科大学附属北京胸科医院 EGFRvIII和EGFR的双特异性人源抗体及其应用
WO2020191485A1 (en) * 2019-03-27 2020-10-01 National Research Council Of Canada Anti-egfrviii antibodies and antigen-binding fragments thereof
US20220380474A1 (en) * 2019-07-02 2022-12-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Monoclonal antibodies that bind egfrviii and their use

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105358576A (zh) * 2013-02-20 2016-02-24 诺华股份有限公司 使用人源化抗EGFRvIII嵌合抗原受体治疗癌症
CN108699124A (zh) * 2016-01-21 2018-10-23 辉瑞大药厂 靶向表皮生长因子受体变体iii的嵌合抗原受体
WO2020191485A1 (en) * 2019-03-27 2020-10-01 National Research Council Of Canada Anti-egfrviii antibodies and antigen-binding fragments thereof
US20220380474A1 (en) * 2019-07-02 2022-12-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Monoclonal antibodies that bind egfrviii and their use
CN111548417A (zh) * 2020-04-03 2020-08-18 首都医科大学附属北京胸科医院 EGFRvIII和EGFR的双特异性人源抗体及其应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MAGGS LUKE, CATTANEO GIULIA, DAL ALI EMRE, MOGHADDAM ALI SANJARI, FERRONE SOLDANO: "CAR T Cell-Based Immunotherapy for the Treatment of Glioblastoma", FRONTIERS IN NEUROSCIENCE, FRONTIERS RESEARCH FOUNDATION, CH, vol. 15, CH , XP093135238, ISSN: 1662-453X, DOI: 10.3389/fnins.2021.662064 *
ZHAN, DUQING: "Construction and Application of Anti-EGFRvIII Modified Chimeric Antigen Receptor Jurkat T Cells", CHINA MASTER’S THESES FULL-TEXT DATABASE (BASIC SCIENCES), 15 April 2019 (2019-04-15) *

Similar Documents

Publication Publication Date Title
US20240010728A1 (en) Anti-ctla4 and anti-pd-1 bifunctional antibody, pharmaceutical composition thereof and use thereof
TWI793062B (zh) Dll3及cd3抗體構築體
TWI796283B (zh) Msln及cd3抗體構築體
IL264956A (en) Anti-pd1 monoclonal antibody, pharmaceutical preparation thereof and use thereof
JP2021536242A (ja) 抗pd−1/抗vegfa二官能性抗体、その医薬組成物およびその使用
KR20230113578A (ko) Cldn18.2 항체 및 그의 용도
CN111454358A (zh) 一种嵌合抗原受体及其应用
US20230348627A1 (en) Anti-4-1bb-anti-pd-l1 bispecific antibody, and pharmaceutical composition and use thereof
WO2023116782A1 (zh) 靶向gprc5d的全人源抗体和嵌合抗原受体(car)及其应用
JP2023539501A (ja) 抗vegf-抗pd-l1二重特異性抗体、その医薬組成物およびその使用
WO2023151693A1 (zh) 包含抗tigit抗体和抗pd-1-抗vegfa双特异性抗体的药物组合物及用途
JP2022535005A (ja) 抗bcma免疫療法によりがんを処置するための組成物および方法
TW202110895A (zh) 一種抗ceacam5的單殖株抗體及其製備方法和用途
KR20230166096A (ko) Cldn18.2 항원 결합 단백질 및 이의 적용
IL304095A (en) Mesothelin binding molecule and its application
WO2023045370A1 (zh) 靶向tigit的单克隆抗体
CN113321735B (zh) 一种多功能融合蛋白
WO2024027835A1 (zh) 靶向EGFRvIII的抗体及其在细胞免疫治疗的应用
US20230192903A1 (en) Bispecific antibody against cldn18.2 and cd3
CA3181591A1 (en) Antibodies specific to abcb5 and uses thereof
WO2021170146A1 (zh) 新型抗cd19抗体和cd19-car-t细胞的制备及其应用
KR102548256B1 (ko) Cd22에 특이적인 항체 및 이의 용도
WO2024094159A1 (zh) 靶向人源ror1的单域抗体
US20230212286A1 (en) Antibodies specific for cd22 and uses thereof
KR102393776B1 (ko) Cd22에 특이적인 인간화 항체 및 이를 이용한 키메라 항원 수용체

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: 23849539

Country of ref document: EP

Kind code of ref document: A1