CN113336851B - Novel fully human anti-human B7H3 antibody, composition containing same and application thereof - Google Patents

Novel fully human anti-human B7H3 antibody, composition containing same and application thereof Download PDF

Info

Publication number
CN113336851B
CN113336851B CN202110736498.8A CN202110736498A CN113336851B CN 113336851 B CN113336851 B CN 113336851B CN 202110736498 A CN202110736498 A CN 202110736498A CN 113336851 B CN113336851 B CN 113336851B
Authority
CN
China
Prior art keywords
antibody
cancer
antigen
binding fragment
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110736498.8A
Other languages
Chinese (zh)
Other versions
CN113336851A (en
Inventor
王刚
郑骏年
赵博
李新宇
康晓彤
王亚楠
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Jicui Juchuang Biotechnology Research Institute Co ltd
Original Assignee
Xuzhou Medical University
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 Xuzhou Medical University filed Critical Xuzhou Medical University
Priority to CN202110736498.8A priority Critical patent/CN113336851B/en
Priority to PCT/CN2021/115806 priority patent/WO2023272924A1/en
Publication of CN113336851A publication Critical patent/CN113336851A/en
Application granted granted Critical
Publication of CN113336851B publication Critical patent/CN113336851B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/6835Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70532B7 molecules, e.g. CD80, CD86

Abstract

The invention discloses a novel fully human anti-human B7H3 antibody, a composition containing the antibody and application thereof, and the composition comprises a novel fully human anti-human B7H3 antibody, a pharmaceutical composition containing the antibody, an antibody-drug conjugate, a polynucleotide molecule for encoding the antibody, an expression vector containing the polynucleotide molecule, a host cell containing the expression vector and a host cell population.

Description

Novel fully human anti-human B7H3 antibody, composition containing same and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, relates to a fully human anti-B7H 3 antibody, and particularly relates to a novel fully human anti-B7H 3 antibody, a composition containing the antibody and application thereof.
Background
B7H3, also known as CD276, is an immune checkpoint from molecules of the B7 family and is also a newly discovered co-stimulatory/inhibitory molecule. B7H3 was originally cloned from a human dendritic cell-derived cDNA library by professor Highetto Huan scientists 2001 (chapeval AI, Ni J, Lau JS, et al. B7-H3: A costimulatory molecule for T cell activation and IFN-gamma production. Nat Immunol.2001; 2: 269. 274.). The B7H3 gene contains 7 exons and 6 introns, and consists of 316 amino acids with a signal peptide at the amino terminus, including the extracellular immunoglobulin-like variable (IgV), constant (IgC), transmembrane, and a cytoplasmic region containing 45 amino acids. Because of its gene structure similarity to the B7 family, the mRNA named B7H3, B7H3, is widely expressed in human tissues and is about 4.1Kb in size. B7H3 has two allosteric splices a and B, a is called 2IgB7H3 and consists of 2 immunoglobulin domains of IgV-IgC extracellularly, B is called 4IgB7H3 and consists of 4 immunoglobulin domains of IgV-IgC-IgV-IgC extracellularly (Zhou YH, Chen YJ, Ma ZY, et al.4IgB7-H3 is the major iso expression on immune cells of immune cells. tissue antibodies 2007; 70:96-104.) recent studies have shown that B7H3 has a co-inhibitory effect on T cells and contributes to tumor Cell immune evasion (Wang C, Li Y, Jia, LF, et al.276. CDexpression of immune cells Cell tissue Cell to Cell antigen of Cell # 1931; Cell # 595; Cell # 2025).
The study shows that the expression of B7H3 can be detected in a plurality of malignant tumor tissues such as respiratory system, digestive system, urogenital system, musculoskeletal system and the like, the clinical statistical analysis shows that the expression of B7H3 in the tumor tissues is closely related to tumor metastasis and prognosis, and the related study reports that B7H3 is related to prostate cancer (Yuan H, Wei X, Zhang G, Li C, Zhang X, Hou J.B7-H3 over expression in promoter tumor tissue and clinical diagnosis, 186:1093-9, breast cancer, pancreatic cancer (Xu L, Ding X, Tan H, Cortion Wen B7-H3 expression and 2expression in cancer tissue, Wa sarcoma, Wa 13, Wa W13, Zhang Q3, shan B, Ding Y, Zhang G, et al, b7-H3 is overexpressed in a substrates summary and associated with a tumor aggregation and a metastasis, plos one 2013; e70689.), non-small cell lung cancer (Sun Y, Wang Y, ZHao J, Gu M, Giscombe R, Lefvert AK, et al. B7-H3 and B7-H4 expression in non-small-cell lung cancer. Lung cancer 2006; 53:143-51.), gastric cancer, renal cancer, colon cancer and other tissues.
The role of B7H3 in the suppression of the immune system and the high expression in various tumor tissues suggest that this molecule may be a target for Cancer therapy, and related studies have reported that antibodies against B7H3 and molecules that modulate B7H3 expression are useful for treating tumors (Loo D, Alderson RF, Chen FZ, Huang L, Zhang W, Gorlatov S, et al. development of an Fc-enhanced antigen-B7-H3 monoclonal antibody with Cancer antigen activity. Clin Cancer Res.2012 Jul 15; 18(14): 3834-45.; Ahmed M, Chemistry M, Zhao Q, et al. humanned antibody-modified monoclonal antibody 8H9 antigen put of mouse strain FG 29, mouse antibody of Biological origin No. 11H 30029, mouse strain FG 27, strain J29, strain IgG 27, mouse strain of animal origin, mouse strain 3729, mouse strain J29, mouse strain G30029, mouse strain G30, mouse strain J29, there remains a need in the art for novel fully human antibodies targeting B7H3 and therapeutic agents that target and kill B7H 3-expressing tumor cells.
Disclosure of Invention
In view of the above, the present invention aims to provide a novel fully human anti-human B7H3 antibody, a composition comprising the same, and uses thereof.
In order to achieve the purpose, the invention adopts the following technical scheme:
in a first aspect, the present invention provides an isolated fully human monoclonal antibody or antigen-binding fragment thereof.
Further, the antibody or antigen-binding fragment thereof specifically binds to B7H 3;
preferably, the antibody or antigen-binding fragment thereof comprises an HCVR, LCVR;
more preferably, the HCVR comprises HCDR1, HCDR2, HCDR 3;
more preferably, the LCVR comprises LCDR1, LCDR2, LCDR 3;
most preferably, the HCDR1 comprises the amino acid sequence of SEQ ID No. 1 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 1;
most preferably, the HCDR2 comprises the amino acid sequence of SEQ ID No. 2 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 2;
most preferably, the HCDR3 comprises the amino acid sequence of SEQ ID No. 3 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 3;
most preferably, the LCDR1 comprises the amino acid sequence of SEQ ID No. 6 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 6;
most preferably, the LCDR2 comprises the amino acid sequence of SEQ ID No. 7 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 7;
most preferably, the LCDR3 comprises the amino acid sequence of SEQ ID No. 8 or an amino acid sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identity to SEQ ID No. 8;
most preferably, the amino acid sequence of the antibody or antigen-binding fragment thereof HCVR is as set forth in SEQ ID NO 4;
most preferably, the amino acid sequence of the LCVR antibody or antigen-binding fragment thereof is as set forth in SEQ ID NO 9;
most preferably, the amino acid sequence of the antibody or antigen-binding fragment thereof is set forth in SEQ ID NO 13.
Further, the nucleotide sequence of the antibody or the antigen-binding fragment HCVR thereof is shown in SEQ ID NO. 5.
Further, the nucleotide sequence of the LCVR of the antibody or the antigen-binding fragment thereof is shown as SEQ ID NO. 10.
Further, the fully human monoclonal antibody or antigen-binding fragment thereof further comprises a linker linking the HCVR and the LCVR;
preferably, the linker is a short peptide of G4S;
more preferably, the amino acid sequence of the G4S short peptide is shown in SEQ ID NO. 11;
more preferably, the amino acid sequence of the G4S short peptide is shown in SEQ ID NO 12.
Further, the nucleotide sequence of the antibody or antigen binding fragment is shown as SEQ ID NO. 14.
In a second aspect of the invention, a pharmaceutical composition is provided.
Further, the pharmaceutical composition comprises an effective amount of the antibody or antigen-binding fragment thereof according to the first aspect of the invention;
preferably, the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients;
preferably, the pharmaceutical composition is used for treating ovarian cancer, renal cancer, lung cancer, breast cancer, colorectal cancer, esophageal cancer, prostate cancer, oral cancer, gastric cancer, pancreatic cancer, endometrial cancer, liver cancer, bladder cancer, osteosarcoma, acute myeloid leukemia;
more preferably, the pharmaceutical composition can be used in combination with a second therapeutic agent;
most preferably, the pharmaceutical composition is used in combination with a second therapeutic agent to enhance the therapeutic effect;
most preferably, the second therapeutic agent comprises regorafenib, sorafenib tosylate, trastuzumab, apatinib mesylate, imatinib mesylate, vemurafenib, everolimus, perizopanib, cidentamine, thalidomide, lucentinib, bortezomib, cetuximab, bevacizumab, dasatinib, nilotinib, rituximab, ibrutinib, trastuzumab, lapatinib tosylate, nituzumab, sorafenib tosylate.
Further, the effective amount of the antibody or antigen-binding fragment thereof according to the first aspect of the invention refers to an amount of antibody that is sufficient to eliminate B7H3 positive cells but toxicologically tolerable in the treated area of the patient, either as a single dose or according to a multiple dose regimen, alone or in combination with other agents that alleviate the adverse condition.
Further, the patient includes humans, non-human animals, wherein the non-human animals include rabbits, rats, mice, monkeys, or other lower primates.
In a third aspect of the invention, an antibody-drug conjugate is provided.
Further, the antibody-drug conjugate comprises an antibody according to the first aspect of the invention or an antigen-binding fragment thereof;
preferably, the antibody-drug conjugate further comprises a small molecule drug;
more preferably, the antibody-drug conjugate is formed by covalent attachment of an antibody or antigen-binding fragment thereof according to the first aspect of the invention to a small molecule drug;
more preferably, the small molecule drug comprises an alkylating agent, an antimetabolite, an antitumor antibiotic, a mitotic inhibitor, an inhibitor of chromatin function, an antiangiogenic agent, an antiestrogen, an antiandrogen, an immunomodulator;
most preferably, the alkylating agent comprises mechlorethamine, chlorambucil, melphalan, bromopropylpiperazine, prednimustine, estramustine phosphate, cyclophosphamide, hexamethylmelamine, cyclophosphamide chloride, ifosfamide, thiotepa, carmustine, streptozocin, fotemustine, lomustine, busulfan, improsulfan, imizamide, cisplatin, oxaliplatin, carboplatin;
most preferably, the antimetabolite comprises methotrexate, 5-fluorouracil, fluoroglycoside, 5-fluorodeoxyuracil, capecitabine, cytarabine, fludarabine, cytarabine, 6-mercaptopurine (6-MP), 6-mercaptoguanine (6-TG), 2-chlorodeoxyadenosine, 5-azacytidine, 2-difluorodeoxycytidine, cladribine, deoxycoformycin, pentostatin;
most preferably, the antitumor antibiotic comprises doxorubicin, daunorubicin, noroxytetracycline, valrubicin, mitoxantrone hydrochloride, dactinomycin, mithramycin, mitomycin C, bleomycin, procarbazine;
most preferably, the mitotic inhibitor comprises paclitaxel, docetaxel, vinblastine, vincristine, vindesine, vinorelbine;
most preferably, the chromatin function inhibitor comprises topotecan, irinotecan, etoposide phosphate, etoposide;
most preferably, the anti-angiogenic agent comprises propylenimine, marimastat, batimastat, prinomastat, tanostat, ilomastat, CGS-27023A, bromocloperaquine, COL-3, neovastat, BMS-275291, thalidomide;
most preferably, the antiestrogen comprises tamoxifen, toremifene, raloxifene, droloxifene, oxifene, anastrozole, letrozole, exemestane;
most preferably, the antiandrogen comprises flutamide, nilutamide, bicalutamide, spironolactone, cyproterone acetate, finasteride, cimetidine;
most preferably, the immunomodulator comprises interferon, interleukin, tumor necrosis factor, lentinan, tezomib, roquinmerac, pidogrel, methoxypolyethylene glycol succinamide adenosine deaminase, thymosin preparations.
In a fourth aspect, the invention provides an isolated polynucleotide molecule.
Further, the polynucleotide molecule comprises a nucleotide sequence encoding a HCVR of the antibody or antigen-binding fragment thereof of the first aspect of the invention, a nucleotide sequence encoding a LCVR of the antibody or antigen-binding fragment thereof of the first aspect of the invention;
preferably, the nucleotide sequence of the polynucleotide molecule is shown as SEQ ID NO. 5;
preferably, the nucleotide sequence of the polynucleotide molecule is shown as SEQ ID NO. 10;
preferably, the nucleotide sequence of the polynucleotide molecule is shown in SEQ ID NO. 14.
In a fifth aspect, the present invention provides an expression vector.
Further, the expression vector comprises a polynucleotide molecule according to the fourth aspect of the invention;
preferably, the vector comprises a plasmid, a vector of viral origin;
more preferably, the virus-derived vector includes a lentiviral vector, a retroviral vector, an adenoviral vector, an adeno-associated viral vector.
In a sixth aspect, the invention provides a host cell.
Further, the host cell comprises an expression vector according to the fifth aspect of the present invention;
preferably, the host cell includes prokaryotic cells, eukaryotic cells;
more preferably, the prokaryotic cell comprises a bacterium, actinomycete, cyanobacterium, mycoplasma, chlamydia, rickettsia;
most preferably, the bacteria include escherichia coli, bacillus subtilis, salmonella typhimurium, pseudomonas, streptomyces, staphylococcus;
more preferably, the eukaryotic cell comprises a mammalian cell, an insect cell, a plant cell, a yeast cell.
In a seventh aspect of the invention, a population of host cells is provided.
Further, the population of host cells comprises host cells according to the sixth aspect of the invention;
preferably, the population of host cells further comprises host cells that do not comprise an expression vector according to the fifth aspect of the invention;
more preferably, the host cell includes prokaryotic cells, eukaryotic cells;
most preferably, the prokaryotic cell comprises a bacterium, actinomycete, cyanobacterium, mycoplasma, chlamydia, rickettsia;
most preferably, the bacteria include escherichia coli, bacillus subtilis, salmonella typhimurium, pseudomonas, streptomyces, staphylococcus;
most preferably, the eukaryotic cell comprises a mammalian cell, an insect cell, a plant cell, a yeast cell.
An eighth aspect of the invention provides a kit.
Further, the kit comprises the antibody or antigen-binding fragment thereof according to the first aspect of the invention, the polynucleotide molecule according to the fourth aspect of the invention, the expression vector according to the fifth aspect of the invention, the host cell according to the sixth aspect of the invention, and the population of host cells according to the seventh aspect of the invention.
A ninth aspect of the present invention provides any one of the following methods:
(1) a method of producing an antibody or antigen-binding fragment thereof according to the first aspect of the invention, comprising the steps of: culturing a host cell according to the sixth aspect of the invention or a population of host cells according to the seventh aspect of the invention, and isolating the antibody or antigen-binding fragment thereof according to the first aspect of the invention from the culture;
(2) a method for detecting B7H3 in a test sample in a non-diagnostic and non-therapeutic destination, said method comprising the steps of: contacting a sample to be tested with an antibody or antigen-binding fragment thereof according to the first aspect of the invention, and detecting the formation of a complex between the antibody or antigen-binding fragment thereof and B7H 3;
preferably, the antibody or antigen-binding fragment thereof is an antibody or antigen-binding fragment thereof labeled with a label that can be used for detection;
more preferably, the label for detection comprises a fluorescent dye, avidin, paramagnetic atom, radioisotope;
most preferably, the fluorescent pigment is fluorescein, rhodamine, Texas Red, phycoerythrin, phycocyanin, allophycocyanin, and polyanemxanthin-chlorophyll protein;
most preferably, the avidin is biotin, avidin, streptavidin, vitellin, avidin;
most preferably, the radioisotope is radioactive iodine, radioactive cesium, radioactive iridium, radioactive cobalt;
(3) a method for producing a host cell according to the sixth aspect of the present invention, comprising the steps of: introducing an expression vector according to the fifth aspect of the invention into a host cell;
preferably, the means of introduction includes calcium phosphate transfection, DEAE, dextrose-mediated transfection, electroporation, phage infection;
(4) a method of specifically inhibiting B7H3 activity, comprising the steps of: the polynucleotide molecule of the fourth aspect of the invention is introduced into a cell of an organism and the activity of B7H3 is inhibited by expression of the antibody or antigen-binding fragment thereof of the first aspect of the invention.
The invention also provides a method of diagnosing whether a subject has a B7H 3-expressing cancer.
Further, the method comprises the steps of: detecting the amount of B7H3 in a sample from the subject using an antibody or antigen-binding fragment thereof according to the first aspect of the invention;
preferably, the method further comprises: comparing the amount of the B7H3 in a sample from the subject to its amount in a known standard or reference sample and determining whether the level of B7H3 of the sample from the subject falls within the level of B7H3 associated with cancer;
more preferably, the sample may be selected from: urine, blood, serum, plasma, saliva, ascites fluid, circulating tumor cells, non-tissue associated cells, tissue, histological preparations.
The invention also provides a method of treating a subject having a B7H 3-expressing cancer.
Further, the method comprises administering to a subject in need thereof a pharmaceutical composition according to the second aspect of the invention, an antibody-drug conjugate according to the third aspect of the invention.
A tenth aspect of the invention provides the use of any one of the following aspects:
(1) use of an antibody or antigen-binding fragment thereof according to the first aspect of the invention, a polynucleotide molecule according to the fourth aspect of the invention, an expression vector according to the fifth aspect of the invention, a host cell according to the sixth aspect of the invention, or a population of host cells according to the seventh aspect of the invention, in the manufacture of a medicament for the prevention and/or treatment of a tumour;
(2) use of an antibody or antigen-binding fragment thereof according to the first aspect of the invention, a polynucleotide molecule according to the fourth aspect of the invention, an expression vector according to the fifth aspect of the invention, a host cell according to the sixth aspect of the invention, a population of host cells according to the seventh aspect of the invention for the preparation of an antibody-drug conjugate for the prevention and/or treatment of a tumour;
(3) use of an antibody or antigen-binding fragment thereof according to the first aspect of the invention, a polynucleotide molecule according to the fourth aspect of the invention, an expression vector according to the fifth aspect of the invention, a host cell according to the sixth aspect of the invention, a population of host cells according to the seventh aspect of the invention in the manufacture of a kit for detecting B7H3 in a test sample and/or diagnosing whether a subject has a tumor;
(4) the use of a pharmaceutical composition according to the second aspect of the present invention in a medicament for the prevention and/or treatment of a tumor;
(5) the use of the antibody-drug conjugate according to the third aspect of the present invention in a medicament for the prevention and/or treatment of tumors;
(6) the kit of the eighth aspect of the invention is used for detecting B7H3 in a sample to be detected and/or diagnosing whether a subject has a tumor;
(7) use of an antibody or antigen-binding fragment thereof according to the first aspect of the invention in the preparation of a polynucleotide molecule, an expression vector, a host cell, a population of host cells;
(8) the use of a polynucleotide molecule according to the fourth aspect of the invention for the preparation of an expression vector, a host cell, a population of host cells;
(9) use of an expression vector according to the fifth aspect of the invention in the preparation of a host cell, a population of host cells;
(10) use of a host cell according to the sixth aspect of the invention in the preparation of a population of host cells;
preferably, the tumor is a B7H 3-expressing tumor;
more preferably, the tumor comprises ovarian cancer, renal cancer, lung cancer, breast cancer, colorectal cancer, esophageal cancer, prostate cancer, oral cancer, gastric cancer, pancreatic cancer, endometrial cancer, liver cancer, bladder cancer, osteosarcoma, acute myeloid leukemia;
preferably, the polynucleotide molecule is a polynucleotide molecule according to the fourth aspect of the invention;
preferably, the expression vector is an expression vector according to the fifth aspect of the invention;
preferably, the host cell is a host cell according to the sixth aspect of the invention;
preferably, the population of host cells is a population of host cells according to the seventh aspect of the invention.
The invention has the following advantages and beneficial effects:
the invention provides a novel fully human anti-human B7H3 antibody, a pharmaceutical composition containing the antibody, an antibody-drug conjugate, a polynucleotide molecule for encoding the antibody, an expression vector containing the polynucleotide molecule, a host cell containing the expression vector and a host cell population.
Drawings
Embodiments of the invention are described in detail below with reference to the attached drawing figures, wherein:
FIG. 1 shows a statistical chart of the enrichment results of phage clones screened for specific binding antibodies;
FIG. 2 is a graph showing the results of a color reaction using ELISA to detect B7H3-02 monoclonal phage recognition and binding to B7H3 target antigen;
FIG. 3 shows a statistical plot of chromogenic response data for detection of B7H3-02 monoclonal phage recognition and binding to B7H3 target antigen by ELISA;
FIG. 4 shows the result of detecting PCR amplified B7H3-02 scFv and its prokaryotic expression vector identification by DNA electrophoresis, wherein, A is: B7H3-02 scFv, Panel B: pET22B-B7H3-02 scFv;
FIG. 5 is a graph showing the results of purification of a prokaryotic expression of B7H3-02 scFv protein;
FIG. 6 is a graph showing the results of a color reaction for detecting the ability of the purified B7H3-02 scFv protein to recognize the target antigen of B7H3 by ELISA;
FIG. 7 shows a statistical plot of chromogenic responses to ELISA to detect the ability of purified B7H3-02 scFv protein to recognize the target antigen of B7H 3;
figure 8 shows a graph of binding constant versus dissociation constant results for purified B7H3-02 antibody and B7H3 target antigen using Biacore assay, wherein, a is: results plot, panel B: a result statistical chart;
FIG. 9 shows graphs of the results of flow cytometry to detect the ability of B7H3-02 scFv expressed on the surface of eukaryotic cells to bind to B7H3 target antigen and a statistical plot of mean fluorescence intensity, wherein, A is: flow cytometry detection result graph, B graph: mean fluorescence intensity histogram.
Detailed Description
The present invention is further illustrated below with reference to specific examples, which are intended to be illustrative only and are not to be construed as limiting the invention. As will be understood by those of ordinary skill in the art: various changes, modifications, substitutions and alterations can be made to the embodiments without departing from the principles and spirit of the invention, the scope of which is defined by the claims and their equivalents. The following examples are examples of experimental methods not indicating specific conditions, and the detection is usually carried out according to conventional conditions or according to the conditions recommended by the manufacturers.
Example 1 screening of fully human B7H3 Single chain antibody (B7H3 scFv)
1. Experimental conditions set
Respectively setting an experimental group, a control group 1 and a control group 2, wherein the experimental conditions of the groups are respectively as follows:
experimental groups: B7H3 antigen + B7H3-Phage
Control group 1: other biotin-free antigens (PRPS1) + B7H3-Phage
Control group 2: antigen-free + B7H3-Phage
2. Experimental methods
Enriching specific binding antibody sequences through four rounds of screening, wherein the conditions of the total amount of added phage, the amount of added antigen, reaction time and the like are changed in each round;
the final result was obtained by counting the number of phages having infectious ability contained in 0.1M HCl (PH 2.0) eluate per 100 μ L of the control group of the experimental group, and determining the enrichment.
3. Results of the experiment
(1) And (4) analysis of screening results: the experimental results are respectively shown in table 1 and fig. 1, and the results show that enrichment occurs in the third round of screening, and the ratio of the number of phases (antigen-antibody specific binding) eluted by the experimental group to the number of phases (non-specific binding between antigen-antibody or no affinity) eluted by the control group is close to 10 times; after the experimental conditions are changed in the fourth round, the experimental group and the control group still keep 10-fold difference, which indicates that the screened phase has scFv with affinity with the target protein of B7H 3;
(2) scFv sequence analysis: 24 monoclonal clones were selected for sequencing, 13 of which expressed the scFv sequences in their entirety, the enrichment sequences:
clone 02 (clone B7H 3-02): VH IGHV3-23 × 01/IGHV3-23D × 01, IGHJ 4/IGHJ 4 × 0303; VK, IGKV1-39 × 01/IGKV1D-39 × 01, IKJ1 × 01;
the scFv amino acid sequence of clone 02 is shown as SEQ ID NO. 13 by sequencing;
wherein the amino acid sequence of HCDR1 of the Heavy Chain Variable Region (HCVR) of clone 02 is as set forth in SEQ ID NO:1, the amino acid sequence of HCDR2 is shown in SEQ ID NO:2, the amino acid sequence of HCDR3 is shown in SEQ ID NO:3, the amino acid sequence of the HCVR is shown as SEQ ID NO:4, the nucleotide sequence of the HCVR is shown as SEQ ID NO:5, the amino acid sequence of LCDR1 of the Light Chain Variable Region (LCVR) of clone 02 is set forth in SEQ ID NO:6, the amino acid sequence of LCDR2 is shown in SEQ ID NO:7, the amino acid sequence of LCDR3 is shown in SEQ ID NO:8, the amino acid sequence of the LCVR is shown as SEQ ID NO:9, the nucleotide sequence of the LCVR is shown in SEQ ID NO:10, the amino acid sequence of the linker of clone 02 is shown in SEQ ID NO:11, and the nucleotide sequence is shown as SEQ ID NO:12, nucleotide sequence of clone 02 SEQ ID NO: as shown at 14.
TABLE 1 screening results
Figure BDA0003141765580000121
Figure BDA0003141765580000131
Example 2 detection of the ability of fully human B7H3 scFv to bind target antigen
1. Detection condition setting
Experimental groups: B7H3 antigen + phage
Negative control group: BCMA antigen and phase scFv-BCMA
Negative control group 1: other biotin-free antigens (PRPS1) + phase
Negative control group 2: antigen-free + phase
2. Experimental protocol
Monoclonal phage, B7H3-02 are prepared respectively, and whether the monoclonal phage and the B7H3-02 have affinity with a target antigen is preliminarily judged through ELISA experiment color reaction and OD value.
3. Experimental procedure
Adding equal amount of antigen coating into each group of experimental wells and control wells, adding equal amount of phase, incubating, washing for multiple times to remove unbound phase, adding phase detection antibody and secondary antibody, developing TMB, and measuring OD with enzyme-labeling instrument450nmAnd (6) reading.
4. Results of the experiment
Results of the experiment were divided intoSee Table 2, FIG. 2 and FIG. 3, the results of color reaction by ELISA and OD450nmThe reading indicates that clone B7H3-02 recognizes and binds to the B7H3 target antigen.
TABLE 2B 7H3-02 statistical results
Figure BDA0003141765580000141
Example 3 validation of the ability of fully human B7H3 scFv to recognize target antigens
One, experiment one
1. scFv antibody expression purification
B7H3-02 scFv antibody expression vector was constructed using pET-22B, the identification results are shown in FIGS. 4A and B, and purified B7H3-02 scFv protein was obtained by induction expression and purification, and the purification results are shown in FIG. 5.
B7H3-02 scFv antibody: 0.456. mu.g/. mu.L.
2. Experimental protocol
Adding equal amount of antigen coating into each group of experimental wells and control wells, adding purified scFv antibody, incubating, washing for multiple times, adding His antibody and secondary antibody, developing TMB, and measuring OD with enzyme labeling instrument450nmAnd (6) reading.
3. Analysis of ELISA results
As shown in FIG. 6, FIG. 7 and Table 4, B7H3-02 has higher affinity, and B7H3-02 still has better affinity when the purified scFv antibody is diluted 1000-fold.
TABLE 4 statistical results of affinity
Figure BDA0003141765580000142
Figure BDA0003141765580000151
Second, experiment two
1. Experimental protocol
The purified B7H3-02 antibody is diluted to 1 mu g/mL by PBS buffer solution with pH 7.5 by adopting a preset coupling amount mode, a certain amount of antibody to be detected is subjected to affinity capture by a biosensor chip protein A, and unbound activated groups are blocked by ethanolamine. B7-H3 antigen was flowed over the chip surface using a series of concentration gradients, binding time 120s, dissociation time 120s, after the cycle dissociation was completed, the biochip was washed with glycine-hydrochloric acid (pH 1.5) buffer, injected at 30. mu.L/min, and regenerated for 30 s. The reaction signal was detected in real time using a Biacore instrument to obtain binding and dissociation curves.
2. Analysis of results
The data obtained by the experiment are fitted by GE software through a Langmuir model, the experimental results are shown in FIGS. 8A and B, and the results show that the affinity of the B7H3-02 antibody and the B7H3 antigen is 27.8nM, which indicates that the B7H3-02 antibody and the B7H3 antigen have better affinity.
Third, experiment three
1. Experimental protocol
The B7H3-02 scFv was constructed into a eukaryotic expression vector containing GPI anchor sequence, transfected into 293T cells, and the scFv expressed on the cell membrane surface was assayed for binding to the target antigen by flow-testing with B7H3-Fc (R & D systems,1027-B3-100) and PE-Anti-Human IgG Fc (Thermo, 12-4998-82).
2. Analysis of results
The flow detection result shows that the B7H3-02 scFv can recognize and bind to the B7H3 target antigen, wherein the B7H3-02 scFv has stronger binding capacity and is equivalent to the binding capacity of the positive control 8H9 clone scFv to the antigen (see FIG. 9A), and the average fluorescence intensity of the scFv bound to the target antigen B7H3 on the cell surface is analyzed to show that the B7H3-02 scFv is stronger (see FIG. 9B), which indicates that the scFv is bound to a larger number of target antigens, i.e. a larger number of fluorescent groups, on the surface of each cell membrane, and the results show that the B7H3-02 scFv and the B7H3 have high affinity and strong specificity.
While the preferred embodiments of the present invention have been described in detail, it will be understood by those skilled in the art that the invention is not limited thereto, and that various changes and modifications may be made without departing from the spirit of the invention, and the scope of the appended claims is to be accorded the full range of equivalents.
Sequence listing
<110> Xuzhou university of medicine
<120> novel fully human anti-human B7H3 antibody, composition comprising the same and use thereof
<141> 2021-06-30
<160> 14
<170> SIPOSequenceListing 1.0
<210> 1
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Gly Phe Thr Phe Ser Ser Tyr Ala
1 5
<210> 2
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Ile Ser Gly Ser Gly Gly Ser Thr
1 5
<210> 3
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Ala Arg Gly Val Gly Arg Gly Phe Asp Tyr
1 5 10
<210> 4
<211> 117
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Glu Val Gln Leu Phe Gln Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr
20 25 30
Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ser Ala Ile Ser Gly Ser Gly Gly Ser Thr Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Val Gly Arg Gly Phe Asp Tyr Trp Gly Gln Gly Thr Thr
100 105 110
Val Thr Val Ser Ser
115
<210> 5
<211> 351
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 5
gaggtgcagc tgttccagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60
tcctgtgcag cctctggatt cacctttagc agctatgcca tgagctgggt ccgccaggct 120
ccagggaagg ggctggagtg ggtctcagct attagtggta gtggtggtag cacatactac 180
gcagactccg tgaagggccg gttcaccatc tccagagaca attccaagaa cacgctgtat 240
ctgcaaatga acagcctgag agccgaggac acggccgtat attactgtgc caggggtgtt 300
ggccggggct ttgactactg gggccagggg accacggtca ccgtctcctc a 351
<210> 6
<211> 6
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Gln Ser Ile Ser Ile Tyr
1 5
<210> 7
<211> 3
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Ala Ala Ser
1
<210> 8
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Gln Gln Thr Tyr Ser Thr Pro Pro Trp Thr
1 5 10
<210> 9
<211> 108
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Ile Tyr
20 25 30
Leu Asn Trp Tyr Arg Gln Gln Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Phe Cys Gln Gln Thr Tyr Ser Thr Pro Pro
85 90 95
Trp Thr Phe Gly Gln Gly Thr Lys Val Asp Ile Lys
100 105
<210> 10
<211> 324
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 10
gacatccagt tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60
atcacttgcc gggcaagtca gagcattagc atctatttaa attggtatcg gcagcaacca 120
gggaaagccc ctaagctcct gatctatgct gcatccagtt tgcaaagtgg ggtcccatca 180
aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240
gaagattttg caacttactt ctgtcaacag acttacagta cccctccgtg gacgttcggc 300
caagggacca aagtggatat caaa 324
<210> 11
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 11
Ser Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15
<210> 12
<211> 45
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 12
agtggcggtg gctctggcgg tggtgggtcg ggtggcggcg gatca 45
<210> 13
<211> 240
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 13
Glu Val Gln Leu Phe Gln Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr
20 25 30
Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ser Ala Ile Ser Gly Ser Gly Gly Ser Thr Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Val Gly Arg Gly Phe Asp Tyr Trp Gly Gln Gly Thr Thr
100 105 110
Val Thr Val Ser Ser Ser Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly
115 120 125
Gly Gly Gly Ser Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser
130 135 140
Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser
145 150 155 160
Ile Ser Ile Tyr Leu Asn Trp Tyr Arg Gln Gln Pro Gly Lys Ala Pro
165 170 175
Lys Leu Leu Ile Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser
180 185 190
Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser
195 200 205
Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Phe Cys Gln Gln Thr Tyr
210 215 220
Ser Thr Pro Pro Trp Thr Phe Gly Gln Gly Thr Lys Val Asp Ile Lys
225 230 235 240
<210> 14
<211> 720
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 14
gaggtgcagc tgttccagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60
tcctgtgcag cctctggatt cacctttagc agctatgcca tgagctgggt ccgccaggct 120
ccagggaagg ggctggagtg ggtctcagct attagtggta gtggtggtag cacatactac 180
gcagactccg tgaagggccg gttcaccatc tccagagaca attccaagaa cacgctgtat 240
ctgcaaatga acagcctgag agccgaggac acggccgtat attactgtgc caggggtgtt 300
ggccggggct ttgactactg gggccagggg accacggtca ccgtctcctc aagtggcggt 360
ggctctggcg gtggtgggtc gggtggcggc ggatcagaca tccagttgac ccagtctcca 420
tcctccctgt ctgcatctgt aggagacaga gtcaccatca cttgccgggc aagtcagagc 480
attagcatct atttaaattg gtatcggcag caaccaggga aagcccctaa gctcctgatc 540
tatgctgcat ccagtttgca aagtggggtc ccatcaaggt tcagtggcag tggatctggg 600
acagatttca ctctcaccat cagcagtctg caacctgaag attttgcaac ttacttctgt 660
caacagactt acagtacccc tccgtggacg ttcggccaag ggaccaaagt ggatatcaaa 720

Claims (58)

1. An isolated fully human monoclonal antibody or antigen-binding fragment thereof, which specifically binds to B7H 3;
the antibody or antigen-binding fragment thereof comprises a HCVR, LCVR;
the HCVR comprises HCDR1, HCDR2, HCDR 3;
the LCVR comprises LCDR1, LCDR2, LCDR 3;
the amino acid sequence of the HCDR1 is shown in SEQ ID NO: 1;
the amino acid sequence of the HCDR2 is shown in SEQ ID NO. 2;
the amino acid sequence of the HCDR3 is shown in SEQ ID NO. 3;
the amino acid sequence of the LCDR1 is shown in SEQ ID NO. 6;
the amino acid sequence of LCDR2 is shown in SEQ ID NO. 7;
the amino acid sequence of LCDR3 is shown in SEQ ID NO. 8.
2. The antibody or antigen-binding fragment thereof of claim 1, wherein the amino acid sequence of the antibody or antigen-binding fragment thereof HCVR is set forth in SEQ ID No. 4.
3. The antibody or antigen-binding fragment thereof of claim 1, wherein the amino acid sequence of the LCVR is as set forth in SEQ ID No. 9.
4. The antibody or antigen-binding fragment thereof of claim 1, wherein the amino acid sequence of the antibody or antigen-binding fragment thereof is set forth in SEQ ID No. 13.
5. A pharmaceutical composition comprising an effective amount of the antibody or antigen-binding fragment thereof of any one of claims 1-4.
6. The pharmaceutical composition of claim 5, further comprising one or more pharmaceutically acceptable excipients.
7. Pharmaceutical composition according to claim 5, characterized in that it is used for the treatment of ovarian cancer, and/or renal cancer, and/or lung cancer, and/or breast cancer, and/or colorectal cancer, and/or esophageal cancer, and/or prostate cancer, and/or oral cancer, and/or gastric cancer, and/or pancreatic cancer, and/or endometrial cancer, and/or liver cancer, and/or bladder cancer, and/or osteosarcoma, and/or acute myeloid leukemia.
8. The pharmaceutical composition of claim 5, wherein the pharmaceutical composition is in combination with a second therapeutic agent.
9. The pharmaceutical composition of claim 8, wherein the pharmaceutical composition is used in combination with a second therapeutic agent to enhance the therapeutic effect.
10. The pharmaceutical composition according to claim 9, characterized in that the second therapeutic agent comprises regorafenib, and/or sorafenib tosylate, and/or trastuzumab, and/or apatinib mesylate, and/or imatinib mesylate, and/or vemurafenib, and/or everolimus, and/or pezopanib, and/or cidam, and/or thalidomide, and/or ruckertinib, and/or bortezomib, and/or cetuximab, and/or bevacizumab, and/or dasatinib, and/or nilotinib, and/or rituximab, and/or ibrutinib, and/or lapatinib tosylate, and/or rituximab.
11. An antibody-drug conjugate comprising the antibody or antigen-binding fragment thereof of any one of claims 1-4.
12. The antibody-drug conjugate of claim 11, further comprising a small molecule drug.
13. The antibody-drug conjugate of claim 12, which is formed by covalently attaching the antibody or antigen-binding fragment thereof of any one of claims 1 to 4 to a small molecule drug.
14. The antibody-drug conjugate of claim 13, wherein the small molecule drug comprises an alkylating agent, and/or an antimetabolite, and/or an antitumor antibiotic, and/or a mitotic inhibitor, and/or an inhibitor of chromatin function, and/or an antiangiogenic agent, and/or an antiestrogen, and/or an antiandrogen, and/or an immunomodulator.
15. The antibody-drug conjugate according to claim 14, wherein the alkylating agent comprises mechlorethamine, and/or chlorambucil, and/or melphalan, and/or bromopropylpiperazine, and/or prednimustine, and/or estramustine phosphate, and/or cyclophosphamide, and/or hexamethylmelamine, and/or chloroacetcyclophosphamide, and/or ifosfamide, and/or thiotepa, and/or carmustine, and/or streptozocin, and/or fotemustine, and/or lomustine, and/or busulfan, and/or improsulfan, and/or dacarbazine, and/or cisplatin, and/or oxaliplatin, and/or carboplatin.
16. The antibody-drug conjugate according to claim 14, wherein the antimetabolite comprises methotrexate, and/or 5-fluorouracil, and/or a fluoroglycoside, and/or 5-fluorodeoxyuracil, and/or capecitabine, and/or cytarabine, and/or fludarabine, and/or 6-mercaptopurine (6-MP), and/or 6-mercaptoguanine (6-TG), and/or 2-chlorodeoxyadenosine, and/or 5-azacytidine, and/or 2, 2-difluorodeoxycytidine, and/or cladribine, and/or deoxycoformycin, and/or pentostatin.
17. The antibody-drug conjugate according to claim 14, wherein the antitumor antibiotic comprises doxorubicin, and/or daunorubicin, and/or nornornornorubicin, and/or valrubicin, and/or mitoxantrone hydrochloride, and/or dactinomycin, and/or mithramycin, and/or mitomycin C, and/or bleomycin, and/or procarbazine.
18. The antibody-drug conjugate according to claim 14, wherein the mitotic inhibitor comprises paclitaxel, and/or docetaxel, and/or vinblastine, and/or vincristine, and/or vindesine, and/or vinorelbine.
19. The antibody-drug conjugate according to claim 14, wherein the chromatin function inhibitor comprises topotecan, and/or irinotecan, and/or etoposide phosphate, and/or etoposide.
20. The antibody-drug conjugate according to claim 14, wherein the anti-angiogenic agent comprises propylenimine, and/or marimastat, and/or batimastat, and/or prinomastat, and/or tanostat, and/or ilomastat, and/or CGS-27023A, and/or bromoclopidogrel, and/or COL-3, and/or neovastat, and/or BMS-275291, and/or thalidomide.
21. The antibody-drug conjugate according to claim 14, wherein the antiestrogen comprises tamoxifen, and/or toremifene, and/or raloxifene, and/or droloxifene, and/or oxifene, and/or anastrozole, and/or letrozole, and/or exemestane.
22. The antibody-drug conjugate according to claim 14, wherein the antiandrogen comprises flutamide, and/or nilutamide, and/or bicalutamide, and/or spironolactone, and/or cyproterone acetate, and/or finasteride, and/or cimetidine.
23. The antibody-drug conjugate of claim 14, wherein the immunomodulator comprises an interferon, and/or an interleukin, and/or a tumor necrosis factor, and/or lentinan, and/or a tezomib, and/or a loquimod, and/or pidotene, and/or methoxypolyethylene glycol succinamide adenosine deaminase, and/or a thymosin agent.
24. An isolated polynucleotide molecule comprising a nucleotide sequence encoding a HCVR of an antibody or antigen-binding fragment thereof as set forth in any one of claims 1-4 and a nucleotide sequence encoding a LCVR of an antibody or antigen-binding fragment thereof as set forth in any one of claims 1-4.
25. The polynucleotide molecule of claim 24, wherein said HCVR has a nucleotide sequence as set forth in SEQ ID No. 5.
26. The polynucleotide molecule of claim 24, wherein the LCVR has the nucleotide sequence set forth in SEQ ID No. 10.
27. The polynucleotide molecule of claim 24, wherein the nucleotide sequence of said polynucleotide molecule is set forth in SEQ ID No. 14.
28. An expression vector comprising the polynucleotide molecule of claim 24.
29. The expression vector of claim 28, wherein the vector comprises a plasmid, and/or a vector of viral origin.
30. The expression vector of claim 29, wherein the virally-derived vector comprises a lentiviral vector, and/or a retroviral vector, and/or an adenoviral vector, and/or an adeno-associated viral vector.
31. A host cell comprising the expression vector of claim 28.
32. The host cell of claim 31, wherein the host cell comprises a prokaryotic cell, and/or a eukaryotic cell.
33. The host cell of claim 32, wherein the prokaryotic cell comprises a bacterium, and/or an actinomycete, and/or a cyanobacterium, and/or a mycoplasma, and/or a chlamydia, and/or a rickettsia.
34. The host cell of claim 33, wherein the bacteria comprise escherichia coli, and/or bacillus subtilis, and/or salmonella typhimurium, and/or pseudomonas, and/or streptomyces, and/or staphylococcus.
35. The host cell of claim 32, wherein the eukaryotic cell comprises a mammalian cell, and/or an insect cell, and/or a plant cell, and/or a yeast cell.
36. A population of host cells, wherein said population of host cells comprises the host cell of claim 31.
37. The population of host cells of claim 36, further comprising host cells that do not comprise the expression vector of claim 28.
38. The population of host cells of claim 37, wherein the host cells comprise prokaryotic cells, and/or eukaryotic cells.
39. The population of host cells of claim 38, wherein the prokaryotic cells comprise bacteria, and/or actinomycetes, and/or cyanobacteria, and/or mycoplasma, and/or chlamydia, and/or rickettsia.
40. The population of host cells of claim 39, wherein the bacteria comprise Escherichia coli, and/or Bacillus subtilis, and/or Salmonella typhimurium, and/or Pseudomonas, and/or Streptomyces, and/or Staphylococcus.
41. The population of host cells of claim 38, wherein the eukaryotic cells comprise mammalian cells, and/or insect cells, and/or plant cells, and/or yeast cells.
42. A kit comprising the antibody or antigen-binding fragment thereof of any one of claims 1 to 4, and/or the polynucleotide molecule of claim 24, and/or the expression vector of claim 28, and/or the host cell of claim 31, and/or the population of host cells of claim 36.
43. A method of producing the antibody or antigen-binding fragment thereof of any one of claims 1-4, comprising the steps of: culturing the host cell of claim 31 or the population of host cells of claim 36, and isolating the antibody or antigen-binding fragment thereof of any one of claims 1-4 from the culture.
44. A method for detecting B7H3 in a test sample in a non-diagnostic and non-therapeutic destination, said method comprising the steps of: contacting a test sample with the antibody or antigen-binding fragment thereof of any one of claims 1-4, and detecting the formation of a complex between the antibody or antigen-binding fragment thereof and B7H 3.
45. The method of claim 44, wherein the antibody or antigen-binding fragment thereof is an antibody or antigen-binding fragment thereof labeled with a detectable label.
46. The method of claim 45, wherein the detectable label comprises a fluorochrome, and/or an avidin, and/or a paramagnetic atom, and/or a radioisotope.
47. The method of claim 46, wherein said fluorochrome is fluorescein, and/or rhodamine, and/or Texas Red, and/or phycoerythrin, and/or phycocyanin, and/or allophycocyanin, and/or polymetaxanthin-chlorophyll protein.
48. The method according to claim 46, wherein the avidin is biotin, and/or avidin, and/or streptavidin, and/or vitelline avidin, and/or avidin-like.
49. The method of claim 46, wherein the radioisotope is radioactive iodine, and/or radioactive cesium, and/or radioactive iridium, and/or radioactive cobalt.
50. A method of making the host cell of claim 31, comprising the steps of: introducing the expression vector of claim 28 into a host cell.
51. The method of claim 50, wherein the means of introduction comprises calcium phosphate transfection, and/or DEAE, and/or dextrose-mediated transfection, and/or electroporation, and/or phage infection.
52. A method of specifically inhibiting B7H3 activity, comprising the steps of: introducing the polynucleotide molecule of claim 24 into a cell of an organism, and inhibiting the activity of B7H3 by expressing the antibody or antigen-binding fragment thereof of any one of claims 1 to 4.
53. Use of the antibody or antigen-binding fragment thereof of any one of claims 1-4, and/or the polynucleotide molecule of claim 24, and/or the expression vector of claim 28, and/or the host cell of claim 31, and/or the population of host cells of claim 36, in the manufacture of a medicament for the prevention and/or treatment of a tumor that expresses B7H 3.
54. The use of claim 53, wherein said B7H 3-expressing tumor comprises ovarian cancer, and/or renal cancer, and/or lung cancer, and/or breast cancer, and/or colorectal cancer, and/or esophageal cancer, and/or prostate cancer, and/or oral cancer, and/or gastric cancer, and/or pancreatic cancer, and/or endometrial cancer, and/or liver cancer, and/or bladder cancer, and/or osteosarcoma, and/or acute myeloid leukemia.
55. Use of the antibody or antigen-binding fragment thereof of any one of claims 1-4, and/or the polynucleotide molecule of claim 24, and/or the expression vector of claim 28, and/or the host cell of claim 31, and/or the population of host cells of claim 36, in the preparation of an antibody-drug conjugate for the prevention and/or treatment of a tumor that expresses B7H 3.
56. The use of claim 55, wherein said B7H 3-expressing tumor comprises ovarian cancer, and/or renal cancer, and/or lung cancer, and/or breast cancer, and/or colorectal cancer, and/or esophageal cancer, and/or prostate cancer, and/or oral cancer, and/or gastric cancer, and/or pancreatic cancer, and/or endometrial cancer, and/or liver cancer, and/or bladder cancer, and/or osteosarcoma, and/or acute myeloid leukemia.
57. Use of the antibody or antigen-binding fragment thereof of any one of claims 1-4, and/or the polynucleotide molecule of claim 24, and/or the expression vector of claim 28, and/or the host cell of claim 31, and/or the population of host cells of claim 36, in the preparation of a kit for detecting B7H3 in a test sample, and/or diagnosing whether a subject has a tumor that expresses B7H 3.
58. The use of claim 57, wherein said B7H 3-expressing tumor comprises ovarian cancer, and/or renal cancer, and/or lung cancer, and/or breast cancer, and/or colorectal cancer, and/or esophageal cancer, and/or prostate cancer, and/or oral cancer, and/or gastric cancer, and/or pancreatic cancer, and/or endometrial cancer, and/or liver cancer, and/or bladder cancer, and/or osteosarcoma, and/or acute myeloid leukemia.
CN202110736498.8A 2021-06-30 2021-06-30 Novel fully human anti-human B7H3 antibody, composition containing same and application thereof Active CN113336851B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110736498.8A CN113336851B (en) 2021-06-30 2021-06-30 Novel fully human anti-human B7H3 antibody, composition containing same and application thereof
PCT/CN2021/115806 WO2023272924A1 (en) 2021-06-30 2021-08-31 Novel fully human antibody for human b7h3, chimeric antigen receptor and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110736498.8A CN113336851B (en) 2021-06-30 2021-06-30 Novel fully human anti-human B7H3 antibody, composition containing same and application thereof

Publications (2)

Publication Number Publication Date
CN113336851A CN113336851A (en) 2021-09-03
CN113336851B true CN113336851B (en) 2021-12-24

Family

ID=77481903

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110736498.8A Active CN113336851B (en) 2021-06-30 2021-06-30 Novel fully human anti-human B7H3 antibody, composition containing same and application thereof

Country Status (1)

Country Link
CN (1) CN113336851B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113621068B (en) * 2021-10-11 2022-01-07 上海恒润达生生物科技股份有限公司 Antibody or antigen binding fragment thereof specifically binding to CD276, and preparation method and application thereof
TW202402808A (en) * 2022-05-18 2024-01-16 中國大陸商上海藥明生物技術有限公司 Anti-b7h3 antibody and uses thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105308069A (en) * 2013-04-03 2016-02-03 罗氏格莱卡特有限公司 Bispecific antibodies specific for FAP and DR5, antibodies specific for DR5 and methods of use
WO2016044383A1 (en) * 2014-09-17 2016-03-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-cd276 antibodies (b7h3)
CN108137669A (en) * 2015-05-18 2018-06-08 蓝鸟生物公司 Anti- ROR1 Chimeric antigen receptors
CN109971711A (en) * 2017-12-27 2019-07-05 张曼 The application of killing human bladder cancer cell is oriented about CD3 × B7H3 bispecific antibody
CN110325860A (en) * 2017-01-31 2019-10-11 免疫媒介有限公司 Method, array and its purposes
CN113402619A (en) * 2021-06-30 2021-09-17 徐州医科大学 Targeting B7H3 co-expression IL-21 fully human chimeric antigen receptor, iNKT cell and application thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105308069A (en) * 2013-04-03 2016-02-03 罗氏格莱卡特有限公司 Bispecific antibodies specific for FAP and DR5, antibodies specific for DR5 and methods of use
WO2016044383A1 (en) * 2014-09-17 2016-03-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-cd276 antibodies (b7h3)
CN108137669A (en) * 2015-05-18 2018-06-08 蓝鸟生物公司 Anti- ROR1 Chimeric antigen receptors
CN110325860A (en) * 2017-01-31 2019-10-11 免疫媒介有限公司 Method, array and its purposes
CN109971711A (en) * 2017-12-27 2019-07-05 张曼 The application of killing human bladder cancer cell is oriented about CD3 × B7H3 bispecific antibody
CN113402619A (en) * 2021-06-30 2021-09-17 徐州医科大学 Targeting B7H3 co-expression IL-21 fully human chimeric antigen receptor, iNKT cell and application thereof
CN113501884A (en) * 2021-06-30 2021-10-15 徐州医科大学 Fully human chimeric antigen receptor targeting B7H3, iNKT cell and application thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
B7-H3单抗交联作用对Mo-DC体外生物学功能的影响;朱一蓓等;《现代免疫学》;20081231;全文 *
Development of an Fc-Enhanced Anti–B7-H3 Monoclonal Antibody with Potent Antitumor Activity;Deryk Loo等;《CLINICAL CANCER RESEARCH》;20120731;全文 *
Monoclonal antibody-based immunotherapy of ovarian cancer: Targeting ovarian cancer cells with the B7-H3-specific mAb 376.96;J.Fauci等;《Gynecologic Oncology》;20130731;全文 *

Also Published As

Publication number Publication date
CN113336851A (en) 2021-09-03

Similar Documents

Publication Publication Date Title
CN108699146B (en) anti-PD-L1 antibodies and uses thereof
CN107405397B (en) anti-TIM-3 antibodies
KR101418606B1 (en) Antibodies directed to her-3 and uses thereof
CN109762066B (en) 4-1BB antibody and preparation method and application thereof
KR20180132751A (en) BCMA binding molecules and methods for their use
CN107921127A (en) There is specific φt cell receptor sample antibody for PRAME peptides
CN110272490B (en) Targeted CTLA-4 antibody, preparation method and application thereof
CN113336851B (en) Novel fully human anti-human B7H3 antibody, composition containing same and application thereof
CN108337890A (en) Novel PD-1 immunomodulators
KR20080090393A (en) Biological materials and uses thereof
CH701730B1 (en) VCAM-1 specific monoclonal antibodies.
CN115768793A (en) Novel LILRB2 antibodies and uses thereof
CN113508139B (en) Antibodies that bind human LAG-3, methods of making, and uses thereof
KR20180122456A (en) Antibodies that bind to the outer membrane glycoprotein of severe hyperthermia and thrombocytopenic syndrome viruses and uses thereof
CN113402602B (en) Novel coronavirus SARS-CoV-2 neutralizing antibody and application thereof
CN109219446A (en) ALK7 binding protein and application thereof
CN108699152A (en) Chimeric dog anti-CD 20 antibodies
CN114685652A (en) Total human broad-spectrum cross-neutralizing antibody aiming at SARS-CoV-2 and SARS-CoV and application thereof
CN113480650B (en) Preparation method and application of fully human-derived targeting CD276 CAR-T cell
CN112851814B (en) Fully human single-chain antibody targeting BCMA and preparation method and application thereof
CN110713539A (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
KR20200135785A (en) Bispecific antibodies against human PD-L1 and PD-L2 and methods of use thereof
CN109293774B (en) Fully humanized antibody specifically binding to CD19 and application thereof
CN108546297B (en) For the monoclonal antibody and its application of PD-1
CN109265551B (en) CD38 antibodies, chimeric antigen receptors, and drugs

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right

Effective date of registration: 20230907

Address after: Room 501, 5th Floor, Building C, Building 1, Innovation Port, Donghu Pharmaceutical Industrial Park, Xuzhou Economic and Technological Development Zone, Xuzhou City, Jiangsu Province, 221132

Patentee after: Jiangsu Jicui Juchuang Biotechnology Research Institute Co.,Ltd.

Address before: 221004 No. 209 Copper Mountain Road, Jiangsu, Xuzhou

Patentee before: XUZHOU MEDICAL University

TR01 Transfer of patent right