CN114249836A - Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof - Google Patents

Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof Download PDF

Info

Publication number
CN114249836A
CN114249836A CN202111615701.2A CN202111615701A CN114249836A CN 114249836 A CN114249836 A CN 114249836A CN 202111615701 A CN202111615701 A CN 202111615701A CN 114249836 A CN114249836 A CN 114249836A
Authority
CN
China
Prior art keywords
virus
seq
sequence
αcd47
cell
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.)
Pending
Application number
CN202111615701.2A
Other languages
Chinese (zh)
Inventor
徐建青
张晓燕
丁相卿
廖启彬
王诗语
周俊花
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.)
Shanghai Sinobay Bio Tech Co ltd
Original Assignee
Shanghai Sinobay Bio Tech Co ltd
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 Shanghai Sinobay Bio Tech Co ltd filed Critical Shanghai Sinobay Bio Tech Co ltd
Priority to CN202111615701.2A priority Critical patent/CN114249836A/en
Publication of CN114249836A publication Critical patent/CN114249836A/en
Pending legal-status Critical Current

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
    • C07K16/2809Immunoglobulins [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 the T-cell receptor (TcR)-CD3 complex
    • 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
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • 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
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/763Herpes virus
    • 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
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/765Reovirus; Rotavirus
    • 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
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/766Rhabdovirus, e.g. vesicular stomatitis virus
    • 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
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • 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
    • 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
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24041Use of virus, viral particle or viral elements as a vector
    • C12N2710/24043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention provides bispecific T cell adaptors, recombinant oncolytic viruses thereof, and uses thereof. The invention provides an alpha CD47 and alpha CD3 bispecific T cell adaptor. The invention also provides an isolated nucleic acid molecule encoding the bispecific T cell engager. The invention also provides an expression framework of the bispecific T cell adaptor BiTE. The invention also provides a recombinant oncolytic virus operably inserted into or comprising the expression framework of the bispecific T cell engager BiTE. The present invention combines a bispecific T cell engager with an oncolytic virus that significantly enhances the ability to suppress malignant tumors relative to simple gene therapy or viral therapy.

Description

Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof
Technical Field
The invention belongs to the technical field of biomedicine, and relates to a novel bispecific T cell adaptor (BiTE) containing specific binding fragments of human CD47 and human CD 3. The invention also relates to a recombinant oncolytic virus comprising the bispecific T cell adaptor (BiTE), and also provides the preparation method of the bispecific T cell adaptor and the oncolytic virus and application thereof in the aspect of tumor resistance.
Background
Cancer is the second leading cause of death worldwide, with 960 million estimated to die of cancer in 2018. Around one sixth of deaths worldwide are caused by cancer. Related cancer therapies, particularly tumor immunotherapy via the human autoimmune system, are becoming a focus of increasing attention.
Human CD47, an integrin-associated protein, is a cell membrane surface glycoprotein and belongs to the immunoglobulin superfamily. CD47 is widely expressed on the surface of various cancer cells, releases an 'other eating me' signal by connecting with a signaling regulatory protein alpha (SIRP alpha) on the surface of a tumor phagocyte, and prevents phagocytosis of the macrophage. In recent years, many companies have invested a lot of manpower and material resources to develop CD47 antibody, but the effect after clinical treatment is very small, and the best level of CD47 antibody can be put into combination drug at present. The main reason for the poor therapeutic effect is the difficulty in controlling the side effects due to the fatal weakness of CD47, i.e., its expression on erythrocytes. After the CD47 antibody drug or the SIRP alpha-Fc fusion protein is combined with red blood cells, the agglutination of the red blood cells can be caused, and then the rupture of the red blood cells is triggered; the CD47 antibody drug with Fc of IgG1 can also activate phagocytosis of erythrocytes by macrophages or antibody-dependent cell-mediated cytotoxicity (ADCC), further induce lysis of erythrocytes and finally cause anemia.
How to maximize the advantages of the CD47 target on killing tumor cells and simultaneously reducing damage to red blood cells is always the key point for successfully developing CD47 medicaments. The bispecific T-cell engager BiTE (bispecific T-cell engage) represents a bispecific antibody with a remarkable anti-tumor effect, and can be used for targeting activation of self T cells to kill tumor cells. BiTE consists of two single-chain variable fragments (scFv) connected in series by a flexible linker. One scFv recognizes the T cell surface protein CD3 epsilon H, while the other scFv recognizes a specific tumor cell surface antigen. This structure of BiTE and the ability to specifically bind proteins allows it to physically bridge T cells to tumor cells to form a T cell-BiTE-tumor cell complex, induce immune synapse formation, stimulate T cell activation, and produce tumor-killing cytokines. In recent years, BiTE has made remarkable progress in antitumor studies, and has achieved clinically desirable therapeutic effects.
Oncolytic viruses targeting tumors have attracted considerable attention in recent decades due to the limited therapeutic efficacy of standard tumor treatment regimens in the treatment of advanced tumors and severe side effects. Oncolytic viruses, which benefit from their own ability to selectively infect and lyse tumor cells locally in tumors, are becoming increasingly the first choice for anti-tumor. The tumor selectivity of oncolytic viruses can be the tumor tropism itself or the result of genetic modification. Oncolytic viruses act on multiple cellular pathways, thereby reducing tumor resistance and also inducing different forms of cell death. In addition, oncolytic viruses can break immune tolerance of tumor microenvironment and induce long-term tumor specific immune response. Oncolytic viruses can specifically transport therapeutic proteins into tumor tissue, with further replication of the virus increasing the level of expression in malignant cells. Alternatively, the oncolytic virus may be used in combination with chemotherapy, radiotherapy and immunotherapy.
In 2006, oncolytic adenovirus product (oncorine) has been used in china for clinical treatment of nasopharyngeal carcinoma and the like. The oncolytic virus deletes the E1B-55kD gene of the human adenovirus type 5, can replicate and proliferate in cancer cells with p53 gene mutation and kill host cells to generate an oncolytic therapeutic effect; the deletion of the E3 region allows tumor antigen information to be transmitted by dendritic cells to activate T cell immunity. However, clinical data show that oncolytic virus oncorine in combination with chemotherapy is less effective in treating patients with nasopharyngeal carcinoma than radiotherapy.
In 2015, the american FDA approval for melanoma was obtained by the oncolytic herpes simplex virus (T-VEC) of the ann company, and again in 12 months of the year, the european union was approved for local treatment of unresectable skin, subcutaneous and lymph node lesions in melanoma patients who had relapsed after the first surgery. The results of clinical studies on T-VEC have greatly promoted the development of oncolytic viruses in the field of tumor therapy. However, the treatment type is limited by the intratumoral administration mode, the method can only be applied to tumor types which are close to the body surface and are convenient for operation, the problems of difficult administration and incomplete treatment exist in the treatment of a plurality of non-superficial solid tumors and metastatic tumors, and if the method can be proved to have good tumor treatment effect by intravenous administration, the clinical application value of the method can be greatly improved; because of the limited oncolytic effect of herpes viruses, incomplete clearance of bulky and/or metastatic tumors, there is a need for additional therapeutic approaches to enhance the anti-neoplastic effect.
The biological properties and pathogenic mechanism of Vaccinia Virus (VV) are relatively clear, plays a key role in eliminating smallpox, and the safety of the Vaccinia virus in human bodies is fully proved. Vaccinia viruses are classified into WR (Western Reserve) strain, Wyeth strain, Copenhagen strain, Lister strain, Tian Tan strain, etc., according to pathogenicity, host range, etc. Because of its wide host range, high conservation, good safety and large foreign gene capacity, vaccinia virus is used as a vector for a plurality of recombinant vaccines such as influenza virus and human immunodeficiency virus. Most of the oncolytic viruses are currently in preclinical research phase as research and development of oncolytic viruses, and only a few enter clinical research phase.
The research on the use of vaccinia virus as an oncolytic virus for tumor therapy is currently the most rapidly progressing Pexa-Vec (JX-594) developed by Jennerex, USA. JX-594 is based on Wyeth strain virus, hGM-CSF and LacZ genes are inserted into the TK region, and JX-594 can express and replicate in cancer cells expressing high level of thymokinase due to deletion of the thymokinase gene, but does not affect normal cells. Meanwhile, the JX-594 can express GM-CSF in tumor cells due to the insertion of GM-CSF gene, and can activate the anti-tumor immune response of the body. The results of clinical trials of JX-594 tumor types prove that the drug combination has good tolerance after intratumoral administration or intravenous drip administration, and the effect of the combination of the Pexa-Vec and the sorafenib is better than that of a single drug combination. The interim analysis results indicate that it is not highly likely to prolong patient survival. Pexa-Vec originally planned to be marketed 2020, the clinical phase III trial was terminated early.
Oncolytic virus (GL-ONC1, also called GLV-1h68) developed by Genelux corporation, USA, is based on vaccinia virus (Lister strain), whose genes F14.5L, TK (encoding thymidine kinase) and HA (encoding hemagglutinin) are deleted to enhance targeting of tumor, and luciferase-GFP fusion protein, beta-galactosyltransferase and beta-glucuronidase are inserted respectively for screening and production preparation of poxvirus. The already completed clinical trial of i.v. administration of GL-ONC1 showed good safety and efficacy, no dose limiting toxicity, no maximum tolerated dose reached, and all patients had a neutralization response against GL-ONC 1; GL-ONC1 is currently undergoing extensive experimentation with intravenous administration.
The life cycle of vaccinia virus is strictly carried out in the cytoplasm of host cells, and the thymidine kinase gene of vaccinia virus is required for the genome of progeny virus to be successfully replicated. In the normal tissue cell cycle, however, synthesis of Thymidine Kinase (TK) occurs in the S phase of the cell division cycle, and after cell division is complete, Thymidine Kinase degrades inside the cell, so the concentration of Thymidine Kinase in the cytoplasm is low. However, tumor cells divide actively and thymidine kinase continues to be synthesized. By utilizing the characteristics, the thymidine kinase gene of the vaccinia virus is deleted, so that the vaccinia virus is specifically amplified in tumor cells to play a role in dissolving tumor.
Clinical trial evidence shows that vaccinia virus has shown some initial advantages in tumor treatment. Most of the existing vaccinia viruses used as oncolytic viruses are high in safety and clinical effectiveness to be further observed, but further optimization is needed in immune regulation and accurate tumor targeting.
Based on the above, there is no bispecific T cell engager in the prior art that is particularly suitable for recombinant oncolytic viruses, and there is currently a need for safer, more targeted bispecific T cell engagers and oncolytic viruses comprising the same.
Disclosure of Invention
Accordingly, it is an object of the present invention to address the deficiencies of the prior art by providing a bispecific T cell engager. The invention also provides recombinant oncolytic viruses expressing the bispecific T cell engagers (bites). The bispecific T cell adaptor (BiTE) provided by the invention overcomes the limitation of tumor immune editing (immunosuppression and immune evasion) of solid tumor cells on human T cells, can directly recruit the human T cells into the solid tumor to kill the tumor cells, and has wide application prospect.
The purpose of the invention is realized by the following technical scheme:
in one aspect, the invention provides an α CD47 and α CD3 bispecific T cell engager α CD47- α CD3 BiTE comprising a fusion protein represented by any one of the following formulae:
VLαCD47-L-VHαCD3-L-VLαCD3-L-VHαCD47(ii) a Or
VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
Wherein said VHαCD47Is the heavy chain variable region of the CD47 antibody, which comprises the following 3 complementarity determining regions:
(i) a VH CDR1, consisting of the sequence: 1, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto,
(ii) a VH CDR2, consisting of the sequence: 2, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto, and
(iii) a VH CDR3, consisting of the sequence: 3, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto;
preferably, the substitution recited in any one of (i) - (iii) is a conservative substitution;
preferably, said VHαCD47Comprises the following steps: VH CDR1 shown in SEQ ID NO. 1, VH CDR2 shown in SEQ ID NO. 2, VH CDR3 shown in SEQ ID NO. 3;
wherein, the VL isαCD47Is the light chain variable region of the CD47 antibody, which comprises the following 3 complementarity determining regions:
(iv) a VL CDR1, consisting of the sequence: any one of SEQ ID NO 4, SEQ ID NO 7, SEQ ID NO 10, SEQ ID NO 13 or SEQ ID NO 16, or a sequence having substitution, deletion or addition of one or more amino acids compared thereto,
(v) a VL CDR2, consisting of the sequence: a sequence shown in any one of SEQ ID NO 5, SEQ ID NO 8, SEQ ID NO 11, SEQ ID NO 14 or SEQ ID NO 17, or having substitution, deletion or addition of one or more amino acids compared thereto, and
(vi) a VL CDR3, consisting of the sequence: any one of SEQ ID NO 6, SEQ ID NO 9, SEQ ID NO 12, SEQ ID NO 15 or SEQ ID NO 18, or a sequence having substitution, deletion or addition of one or more amino acids compared thereto;
preferably, the substitution recited in any one of (iv) - (vi) is a conservative substitution;
preferably, said VLαCD47Comprises the following steps:VL CDR1 shown in SEQ ID NO. 4, VL CDR2 shown in SEQ ID NO. 5, VL CDR3 shown in SEQ ID NO. 6;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 7, VL CDR2 shown in SEQ ID NO. 8, VL CDR3 shown in SEQ ID NO. 9;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 10, VL CDR2 shown in SEQ ID NO. 11, VL CDR3 shown in SEQ ID NO. 12;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 13, VL CDR2 shown in SEQ ID NO. 14, VL CDR3 shown in SEQ ID NO. 15;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 16, VL CDR2 shown in SEQ ID NO. 17, and VL CDR3 shown in SEQ ID NO. 18.
The bispecific T cell engager according to the present invention, wherein the VHαCD47Comprises 3 CDRs contained in the heavy chain variable region shown in SEQ ID NO. 19;
preferably, said VLαCD47Comprises 3 CDRs contained in a light chain variable region represented by any one of SEQ ID NOS 20-24;
preferably, the 3 CDRs contained in the heavy chain variable region, and/or the 3 CDRs contained in the light chain variable region, are defined by the Kabat, Chothia or IMGT numbering system.
The bispecific T cell engager according to the present invention, wherein the VHαCD47Comprising an amino acid sequence selected from the group consisting of:
(i) 19 in SEQ ID NO;
(ii) a sequence having substitution, deletion or addition of one or several amino acids as compared with the sequence shown in SEQ ID NO. 19; or
(iii) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the sequence set forth in SEQ ID NO. 19;
and/or the presence of a gas in the gas,
the VLαCD47Comprising an amino acid sequence selected from the group consisting of:
(iv) a sequence set forth in any one of SEQ ID NOs 20-24;
(v) a sequence having substitution, deletion or addition of one or several amino acids compared to the sequence shown in any one of SEQ ID NOs 20 to 24; or
(vi) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in any one of SEQ ID NOS 20-24;
preferably, the substitutions described in (ii) or (v) are conservative substitutions.
The bispecific T cell engager according to the present invention, wherein the VHαCD3Comprising an amino acid sequence selected from the group consisting of:
(i) the sequence shown as SEQ ID NO 25 or 27;
(ii) a sequence having substitution, deletion or addition of one or several amino acids compared to the sequence shown in SEQ ID NO 25 or 27; or
(iii) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in SEQ ID NO 25 or 27;
and/or the presence of a gas in the gas,
the VLαCD3Comprising an amino acid sequence selected from the group consisting of:
(iv) 26 or 28;
(v) a sequence having substitution, deletion or addition of one or several amino acids compared with the sequence shown in SEQ ID NO. 26 or 28; or
(vi) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in SEQ ID NO 26 or 28;
preferably, the substitutions described in (ii) or (v) are conservative substitutions.
The bispecific T cell engager according to the present invention, wherein the VHαCD47、VHαCD3、 VLαCD47And VLαCD3Linked by a linker peptide;
preferably, said VHαCD47、VHαCD3、VLαCD47And VLαCD3Optionally connected by one, two or three connecting peptides;
preferably, the bispecific T cell engager comprises a fusion protein according to any one of the following formulae:
VLαCD47-L-VHαCD3-L-VLαCD3-L-VHαCD47(ii) a Or
VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
More preferably, L is KESGSVSSEQLAQFRSLD, EGKSSGSGSESKST, GGGGGG, GGGGGGGG or (GGGGS)n(ii) a Further preferably, n is an integer from 1 to 5, most preferably, n is 3;
preferably, the bispecific T cell engager comprises a fusion protein represented by the formula:
VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
wherein said VHαCD47As shown in SEQ ID NO 19, the VLαCD4721 or 24, the VHαCD3As shown in SEQ ID NO. 25, VLαCD326, and the L is GGGGSGGGGSGGGGS.
The invention also provides an isolated nucleic acid molecule encoding the bispecific T cell adaptor;
preferably, the nucleic acid molecule comprises SEQ ID NO: 31-35, said nucleic acid sequence being a VLαCD47The coding sequence of (a);
preferably, the nucleic acid molecule comprises the nucleotide sequence as set forth in SEQ ID NO: 36 positionThe nucleic acid sequence shown is VHαCD47The coding sequence of (a).
Expression framework for the bispecific T cell engager according to the present invention:
5’-E1-E2-E3-E4-E5-E6-E7-3’
wherein:
e1 is a CMV enhancer or/and other cis-acting elements; preferably comprises SEQ ID NO: 37;
e2 is a promoter for recombinant expression, preferably a CMV promoter, more preferably a promoter comprising SEQ ID NO: 38;
e3 is a 5' untranslated region, optionally containing no or one intron sequence, optionally containing one or more restriction enzyme sites; preferably comprises SEQ ID NO: 39;
e4 is the coding nucleotide sequence of the signal peptide of the BiTE protein; preferably, the signal peptide is derived from a human or mouse signal peptide; preferably comprises SEQ ID NO: 40;
e5 is the coding nucleotide sequence of the bispecific T cell engager;
e6 is a 3' untranslated region, optionally comprising one or more restriction enzyme sites; preferably comprises SEQ ID NO: 41;
e7 is the SV40 transcriptional stop signal region; preferably comprises SEQ ID NO: 42, or a nucleotide sequence shown in the specification.
In a further aspect, the present invention provides a recombinant oncolytic virus operably inserted into or comprising the expression framework of the bispecific T cell engager BiTE.
Preferably, the expression cassette is located in the Thymidine Kinase (TK) region of the recombinant oncolytic virus.
Preferably, the expression frame can be expressed alone or in fusion with other genes or fragments.
Preferably, the recombinant oncolytic virus further comprises gene coding sequences for other immunomodulatory factors, more preferably, the other immunomodulatory factors include, but are not limited to, IL-1, IL-2, IL-3, IL-7, IL-11, IL-12, IL-15, IL-17, IL-18, IL-21, IL-33, IL-35, IL-37, GM-CSF, IFN- α, IFN- β, IFN- γ, anti-PD-1/PD-L1 antibodies, anti-CTLA-4 antibodies, anti-Lag-3 antibodies, anti-TIGIT antibodies, or anti-Tim-3 antibodies; or
The recombinant oncolytic virus further comprises gene coding sequences for apoptosis and apoptosis-related proteins including, but not limited to, apoptosis-related factor 1(Apaf-1), interleukin-1 beta converting enzyme (ICE), Bcl-2 protein, Fas/APO-1, p53, myc, ataxia telangiectasia mutated gene (ATM), corticotropin d (gasdermin d), corticotropin e (gasdermin e), and the like; or
The recombinant oncolytic virus also comprises small RNA of a targeted immune regulation gene and an apoptosis gene.
The recombinant oncolytic Virus of the invention, wherein the viral backbone of the oncolytic Virus is derived from a modified or modified vaccinia Virus Tiantan strain, vaccinia Virus New York strain, vaccinia Virus Copenhagen strain, vaccinia Virus canary strain, vaccinia Virus Ankara strain, adenovirus, adeno-associated Virus, herpes simplex Virus, varicella-zoster Virus (VZV), Respiratory Syncytial Virus (RSV), Shenglikichen Virus (SFV), EB Virus, cytomegalovirus, human herpesvirus type 6, smallpox Virus, infectious molluscum Virus, orf Virus, reovirus, rotavirus, enterovirus, seneca Virus, poliovirus, coxsackie Virus, rhinovirus, hepatitis A Virus, foot and mouth disease Virus, Togavirus, alphavirus, semliki forest virus, eastern equine encephalitis virus, sindbis virus, rubella virus, coronavirus, flavivirus, hepatitis c virus, japanese encephalitis virus, st louis encephalitis virus, murray valley fever virus, yellow fever virus, west nile virus, zika virus, dengue virus, ebola virus, marburg virus, arenavirus, lassa fever virus, lymphocytic choriomeningitis virus, pichinde virus, hunin virus, marjo virus, hantaan virus, rift valley fever virus, paramyxovirus, human parainfluenza virus, mumps virus, monkey virus 5, measles virus, vesicular stomatitis virus, rabies virus, orthomyxovirus, influenza a virus, influenza b virus, influenza c virus, hepatitis d virus, monkey immunodeficiency virus, human immunodeficiency virus type 1 and human immunodeficiency virus type 2, Rous sarcoma virus, human T cell leukemia virus type 1, simian foamy virus, hepatitis B virus, hepatitis E virus, human papilloma virus or polyoma virus.
Preferably, the oncolytic viral scaffold is an intracellular maturation virus, an intracellular packaging virus, a cell-associated packaging virus or an extracellular packaging virus.
The recombinant oncolytic virus is a recombinant vaccinia virus Tiantan strain containing a coding gene of a bispecific T cell adaptor alpha CD 47-alpha CD3-BITE, named as rTV-alpha CD 47-alpha CD3-BITE, and has a preservation number of: CCTCC NO of V202081, the preservation date is 2021 year, 1 month and 2 days, and the preservation address is China center for type culture Collection.
In still another aspect, the present invention provides a method for preparing the recombinant oncolytic virus, comprising the steps of:
1) synthesizing an expression framework for the bispecific T cell engager BiTE;
2) subcloning the expression framework obtained in the step 1) into a shuttle plasmid of an oncolytic virus to construct a recombinant plasmid vector;
3) transfecting the recombinant plasmid vector obtained in the step 2) into the oncolytic virus, and screening to obtain the recombinant oncolytic virus.
Optionally, the obtained recombinant oncolytic virus is cultured.
In a specific embodiment, the invention provides a method for preparing a recombinant vaccinia virus Tiantan strain, comprising the following steps:
1) an expression framework for the synthetic bispecific T cell engager α CD47- α CD3-BITE comprising the amino acid sequence as set forth in SEQ ID NO: 31-42;
2) subcloning the synthesized expression framework into TK region of vaccinia virus shuttle plasmid (pSC65) to construct recombinant plasmid pSC 65-alpha CD 47-alpha CD 3-BITE;
3) by means of gene homologous recombination, pSC 65-alpha CD 47-alpha CD3-BITE plasmid is transfected into TK143 infected by wild type vaccinia virus-In cells, the two are subjected to homologous recombination to generate recombinant vaccinia virus rTV-alpha CD 47-alpha CD 3-BITE; screening to obtain the recombinant oncolytic vaccinia virus of which the TK region comprises the coding sequence of alpha CD 47-alpha CD 3-BITE.
Wherein the expression frame of the bispecific T cell adaptor alpha CD 47-alpha CD3-BITE is controlled by the early/late promoter p7.5 of vaccinia virus.
Preferably, the specific steps of expanding the recombinant vaccinia virus using VERO cells comprise: culturing VERO cells until the density is close to 100%, replacing low-concentration fetal calf serum to maintain the culture medium, adding oncolytic vaccinia virus (the inoculum size is about 0.02MOI per 10cm of culture plate), culturing in an incubator, collecting virus liquid after the recombinant poxvirus is amplified, repeatedly freezing and thawing, and performing density gradient centrifugation purification by using sucrose solution.
In still another aspect, the present invention also provides the use of the bispecific T cell engager or the recombinant oncolytic virus for the preparation of an anti-tumor medicament; wherein the tumor is selected from the group consisting of a B cell lymphoma, T cell lymphoma, melanoma, prostate cancer, renal cell carcinoma, sarcoma, glioma such as high grade glioma, parent cell tumor such as neuroblastoma, osteosarcoma, plasmacytoma, histiocytoma, pancreatic cancer, breast cancer, lung cancer such as small cell lung cancer and non-small cell lung cancer, gastric cancer, liver cancer, colon cancer, rectal cancer, esophageal cancer, large intestine cancer, hematopoietic cancer, testicular cancer, cervical cancer, ovarian cancer, bladder cancer, squamous cell cancer, adenocarcinoma, AIDS-related lymphoma, bladder cancer, brain cancer, nervous system cancer, head and neck squamous cell cancer, hodgkin's lymphoma, non-hodgkin's lymphoma, or hematologic malignancy.
In another aspect, the invention provides a method of treating a tumor, the method comprising administering to a subject in need thereof a therapeutically effective amount of a bispecific T cell engager or a recombinant oncolytic virus. The method according to the present invention, wherein the method further comprises administering to a subject in need thereof additional chemotherapeutic drugs, radiotherapeutic techniques, surgical treatments, immunocytotic drugs (including but not limited to CAR-T, NK, NKT, iNKT, CAR-NK, CAR-NKT, CAR-iNKT, etc.), other oncolytic viruses; preferably, the method is intravenous injection or intratumoral injection.
The invention concept of the invention is as follows:
the invention provides a novel alpha CD 47-alpha CD3-BITE bispecific T cell adaptor which has good antitumor activity in vitro. Furthermore, the inventor provides a vaccinia virus Tiantan strain carrying the bispecific T cell adaptor of the invention by taking the vaccinia virus Tiantan strain as a research model. Then, the inventor finds that the vaccinia virus Tiantan strain has a strong anti-tumor effect; the bispecific T cell adaptor carrying the alpha CD 47-alpha CD3-BITE can avoid the combination of the CD47 antibody and red blood cells of peripheral blood, recognize tumor cells by utilizing the broad-spectrum expression characteristic of CD47 and activate T cells by utilizing the CD3 antibody, thereby playing a role in killing the tumor cells, improving the tumor immunotherapy effect and simultaneously increasing the safety.
The invention has the beneficial effects that:
1. the bispecific T cell adaptor (BiTE) provided by the invention has small molecular weight, can penetrate through the cell gap to reach the Tumor Microenvironment (TME) inside the solid tumor to play a role, and can efficiently mediate the killing of T cells on cancer cells.
2. The invention combines the bispecific T cell adapter with the oncolytic virus and provides a vaccinia virus Tiantan strain capable of efficiently expressing the bispecific T cell adapter alpha CD 47-alpha CD3-BITE gene. When the oncolytic virus is administrated to tumor focus, the oncolytic virus can crack tumor cells to exert an oncolytic effect, and simultaneously, a high-killing antibody of a tumor-targeting gene anti-human CD47 is carried to block the combination of CD47 and SIRP alpha, so that the phagocytic function of macrophages in vivo is improved, and the anti-tumor effect is enhanced. The oncolytic virus significantly enhances the suppression of malignant tumors relative to gene therapy or viral therapy alone.
3. According to the in vitro experimental study, the vaccinia virus Tiantan strain disclosed by the invention is proved to be capable of treating liver cancer and malignant lung cancer, realizing good targeting and anti-tumor effects on tumors, having a relatively complete virus amplification and quality control system and laying a foundation for further industrialization, so that the vaccinia virus Tiantan strain has a good application prospect.
Drawings
Embodiments of the invention are described in detail below with reference to the attached drawing figures, wherein:
FIG. 1 shows the construction of the shuttle plasmid vector α CD47- α CD3 BiTE hIgG1 WT and the expression purification of 5 corresponding BiTE proteins of the present invention; wherein FIG. 1A is an expression map of shuttle plasmid hIgG1 WT integrated with α CD47- α CD3-BITE expression framework; FIG. 1B shows the result of 293T culture supernatant transiently expressing α CD47- α CD3-BITE after protein purification using a nickel column.
FIG. 2 shows the effect of α CD47- α CD3-BITE on anti-human ovarian adenocarcinoma cells in vitro; among them, the CD47 antibody cloned No. 68 and BiTE composed of alpha CD3 were both able to mediate the killing of T cells to SK-OV3 well relative to the T cell control group. The alpha CD 47-alpha CD3-BITE can well recognize the tumor cells of CD47 and activate T cells, so that the specific killing of the T cells to the tumor cells is mediated; bispecific T cell engagers (BITE) comprising specific binding of human CD47 and human CD3 were shown to mediate well the anti-tumor activity of T cells in vitro and to have a broad spectrum.
FIG. 3 shows the effect of α CD47- α CD3-BITE on in vitro anti-human lung cancer cells; wherein, compared with a T cell control group, the BiTE consisting of the CD47 antibody cloned in No. 101 and the alpha CD3 can mediate the T cells to kill the NCI-H292 tumor cells, and compared with a single T cell group, the BiTE group has obvious effect of killing the NCI-H292 cells; it was shown that bispecific T cell engagers (BITE) comprising specific binding of human CD47 and human CD3 mediate the antitumor activity of T cells well in vitro and with a broad spectrum.
FIG. 4 shows the construction of a shuttle plasmid vector (α CD47- α CD3-BITE) of vaccinia virus Tiantan strain and the expression of α CD47- α CD3-BITE protein. FIG. 4A is a diagram of the expression map of shuttle plasmid pSC65 of vaccinia virus incorporating the gene α CD47- α CD 3-BITE; FIG. 4B shows the result of PCR identification of the recombinant vaccinia virus rTV- α CD47- α CD3-BITE inserted with the fragment α CD47- α CD 3-BITE; FIG. 4C shows expression of CD47- α CD3-BITE protein in the supernatant of VERO cells infected with recombinant vaccinia virus. As shown in FIG. 4, the recombinant vaccinia virus rTV- α CD47- α CD3-BITE carried α CD47- α CD3-BITE was successfully expressed.
FIG. 5 shows that recombinant vaccinia virus rTV- α CD47- α CD 3-BITE-loaded α CD47- α CD3-BITE has high affinity for CD47 protein; FIG. 5 shows the flow-through results of affinity assay of cell supernatants collected from recombinant vaccinia virus rTV- α CD47- α CD3-BITE infected VERO cells and human CD47 expressing lung cancer cell A549 cells; the result shows that compared with the wild type vaccinia virus, the binding rate of the recombinant vaccinia virus rTV-alpha CD 47-alpha CD3-BITE expression protein and CD47 is more than 85%.
FIG. 6 shows the in vitro killing of A549 cells by recombinant vaccinia virus, i.e., vaccinia virus Tiantan strain rTV- α CD47- α CD 3-BITE. The experiment was divided into four groups, including the control group of supernatants infected with wild vaccinia virus, the group of supernatants + T cells infected with wild vaccinia virus, the control group of supernatants infected with recombinant vaccinia virus, and the group of supernatants + T cells infected with recombinant vaccinia virus. From the figure, it can be seen that the supernatant + T cell group infected by the recombinant vaccinia virus has a significant killing effect on the lung cancer a549 cells compared with the blank control group.
FIG. 7 shows the anti-cancer effect of α CD47- α CD3-BITE protein of different structures.
Preservation information
The vaccinia virus Tiantan strain is named as rTV-alpha CD 47-alpha CD3-BITE, and the preservation number is as follows: CCTCC NO of V202081, the preservation date is 2021 year 1 month 2 days, the preservation address is China center for type culture Collection, the address is: wuhan university in Wuhan, China.
Detailed Description
Example 1: construction and expression verification of 293T recombinant expression plasmid of alpha CD 47-alpha CD3-BITE
1.1 hIgG1 WT vector construction with alpha CD 47-alpha CD3-BITE target gene
First, high affinity 5 anti-human CD were screened47 antibody, the heavy chain variable region and the light chain variable region are selected, and the fusion protein is constructed according to the following formula: VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
Wherein said VHαCD47As shown in SEQ ID NO 19, the VLαCD4720-24, said VHαCD3As shown in SEQ ID NO. 25, VLαCD326, and the L is GGGGSGGGGSGGGGS.
And according to VLαCD47In contrast, Hu004-67 shows a VL comprising SEQ ID NO:20αCD47α CD47- α CD3 BiTE; hu004-68 shows a VL comprising SEQ ID NO:21αCD47α CD47- α CD3 BiTE; hu004-73 represents a polypeptide comprising the VL shown in SEQ ID NO:22αCD47α CD47- α CD3 BiTE; hu004-100 shows a VL comprising SEQ ID NO:23αCD47α CD47- α CD3 BiTE; hu004-101 represents a polypeptide comprising the VL shown in SEQ ID NO:24αCD47In alpha CD 47-alpha CD3 BiTE, wherein WT Cloning vector AbVec-hIgG1(GenBank: FJ 475055.1).
The DNA sequence of alpha CD 47-alpha CD3-BITE was artificially synthesized according to the structural formula, and the plasmid construction map is shown in FIG. 1a, wherein VL isαCD47The coding sequence of (A) is shown in SEQ ID NO: 31-35 any of claims VHαCD47The coding sequence of (A) is shown in SEQ ID NO: shown at 36.
The artificially synthesized hIgG1 WT vector containing the target gene of alpha CD 47-alpha CD3-BITE was transformed into E.coli TOP10 (Dimocha, cat # DL1010S) and grown overnight on a culture plate containing ampicillin. On day 2, single colonies were randomly picked for overnight culture at 37 ℃ and plasmids were extracted using a plasmid extraction kit (Qiagen, cat. No. 27106) for transfection of 293T cells.
1.2 expression and purification of 293T cells with alpha CD 47-alpha CD3-BITE target genes
1.293T cell preparation cells grown in T175 flasks with confluency of more than 90% were removed from the supernatant, added with 2ml of 2.5% Trypsin-EDTA (GIBCO Co., Ltd., cat # 25200072) to digest the cells until the cells were completely detached, and added to 10ml of complete medium (DMEM medium + 10% FBS + 1% PS)Stopping digestion, centrifuging at 150g for 5 min to collect cells, discarding supernatant, and reselecting cells to a cell concentration of about 8X 10 using an appropriate amount of complete medium6~10×106One per ml. The cell suspension was counted and 1.3X 10 cells were taken7The cells were plated on 150mm cell culture dishes (Thermo Scientific, cat # 150468) with a medium volume of 25mL and cultured the next day for subsequent plasmid transfection.
2. hIgG1 WT shuttle plasmid of α CD47- α CD3-BITE transfected 293T cells: a1 mL DMEM-based culture medium was taken out of an EP tube, and 20. mu.g of hIgG1 WT shuttle plasmid Cloning vector AbVec-hIgG1(GenBank: FJ475055.1) of. alpha. CD 47-. alpha. CD3-BITE was added thereto, and after mixing, 30. mu.l of FectoPro transfection reagent (Polyplus, cat. 116-001) was added thereto, and immediately thereafter, the mixture was shaken vigorously on a vortex shaker for 15 seconds and allowed to stand at room temperature for 20 minutes. Taking out 293T cells to be transfected in an incubator, sucking and discarding 10mL of culture medium, adding the immobilized transfection reagent while shaking, and placing at 37 ℃ in 5% CO2Cultured in an incubator for 4-6 hours. The medium was completely removed, Expi293 expression medium (GIBCO Co., Ltd., Cat. A1435102) was added, and the mixture was left at 37 ℃ with 5% CO2Cultured in an incubator for 4 days.
3. Collecting the alpha CD 47-alpha CD3-BITE expression supernatant: the culture medium in the expression dish was collected in a 50mL centrifuge tube, centrifuged at 4000g for 5 minutes, and the expression supernatant was transferred to a new 50mL centrifuge tube.
4. The expression supernatant was subjected to His-tag protein purification: the purification was performed using a gravity chromatography column packed with nickel beads as Ni-NTAAgarose (QIAGEN, cat # 30210); after completion of column packing, 5mM, 10mM imidazole (dissolved in 50mM NaH) was used2PO4·2H2O, pH 8.0, 300mM NaCl) to elute the non-specifically adsorbed protein, eluting at a natural flow rate, and collecting the filtrate; then using 20mM, 100mM (collecting two tubes in sequence), 200mM (collecting two tubes in sequence), 500mM imidazole (dissolved in 50mM NaH)2PO4·2H2O, pH 8.0, 300mM NaCl) to elute the target protein, and collecting the corresponding filtrate.
5. mu.L of the filtrate was sampled and examined by 10% SDS-PAGE. As shown, the sample was sequentially loaded from 5mM imidazole eluent to 500mM imidazole eluent. The target protein appears in the 100mM eluate and 200mM eluate shown in FIG. 1B, and the band size is about 54 kD.
Example 2: alpha CD 47-alpha CD3-BITE mediated T cell in vitro anti-human ovarian adenocarcinoma cell effect
SK-OV3 cell plating: SK-OV3 (deposited at Shanghai Xinwan Biotech, Inc.) is a Luciferase expressing cell line. The cells were cultured at 1X 104The cells/well are plated in 96-well plates and incubated overnight at 37 ℃ until they adhere.
After 2.24 hours, the medium was removed, and 7 groups were set, to which T cells, B6H12- α CD3-BITE (the heavy chain variable region and the light chain variable region of B6H12 are shown in SEQ ID NOS: 29 and 30, respectively, and the remaining structures are the same as those of BITE of example 1 of the present invention), T cells, Hu004-67- α CD3-BITE and T cells, Hu004-68- α CD3-BITE and T cells, Hu004-73- α CD3-BITE and T cells, Hu004-100- α CD3-BITE and T cells, Hu004-101- α CD3-BITE and T cells, respectively, were added.
Killing was performed by overnight incubation at 3.37 ℃. After 24 hours, the supernatant was removed, 50. mu.L of cell lysate luciferase cell lysate (Promega, cat. No. E1531) was added to each well, and incubated at room temperature with shaking for 30 minutes, and 30. mu.L of luciferase substrate (Promega, cat. No. E151A) was added to each well. On-line assay (GloMax Navigator Microplate Luminometer, Promega, Steady-Glo protocol).
The results are shown in FIG. 2. Wherein B6H12 is a known CD47 antibody clone (US 9017675B2) as a positive control. Hu004-68 and Hu004-101 are selected antibody clones with high affinity to CD47 protein (obtained by selecting antibodies from Hexagrammos otakii wipe Biotech). As can be seen from FIG. 2, for this tumor cell line, the CD47 antibody (scFv) - α CD3-BITE of clone No. 68 has a 3.5-fold increase in tumor killing activity compared to the T cell control group, a 2-fold increase compared to clone No. 100 with a lower killing ability, and a 1.5-fold increase compared to the positive control antibody, and can well mediate the effect of the T cell on anti-ovarian adenocarcinoma cells in vitro.
Example 3: in-vitro anti-human lung cancer cell effect of alpha CD 47-alpha CD3-BITE mediated T cells
NCI-H292 cell plating: NCI-H292 (deposited at Shanghai Xinwan Biotech, Inc.) is a Luciferase expressing cell line. The cells were cultured at 1X 104The density of each well was plated in 96-well plates and incubated overnight at 37 ℃ until they adhered.
After 2.24 hours the medium was removed and 7 groups were added T cells, B6H12- α CD3-BITE and T cells, Hu004-67- α CD3-BITE and T cells, Hu004-68- α CD3-BITE and T cells, Hu004-73- α CD3-BITE and T cells, Hu004-100- α CD3-BITE and T cells, Hu004-101- α CD3-BITE and T cells, respectively.
Killing was performed by overnight incubation at 3.37 ℃. After 24 hours, the supernatant was removed, 50. mu.L of cell lysate (Promega, cat. No. E1531) was added to each well, and incubated at room temperature for 30 minutes with shaking, and 30. mu.L of luciferase substrate (Promega, cat. No. E151A) was added to each well. Detection on a machine (GloMax Navigator Microplate Luminometer, Promega, Steady-Glo protocol).
The measured results are shown in fig. 3, CD47 antibody- α CD3-BITE of clone No. 101 can well mediate the in vitro anti-lung cancer cell effect of T cells, CD47 antibody (scFv) - α CD3-BITE of clone No. 101 has a 4-fold increase in the killing tumor activity compared with a T cell control group, has a 1.2-fold increase compared with clone No. 100 with low killing ability, and has a 1.2-fold increase compared with a positive control antibody, and can well mediate the in vitro anti-ovarian adenocarcinoma cell effect of T cells. Together, the results of fig. 2 and 3 indicate that bispecific T cell engagers (BITE) comprising specific binding of human CD47 and human CD3 mediate the anti-tumor activity of T cells well in vitro and with a broad spectrum.
Example 4: construction and expression verification of recombinant vaccinia virus of alpha CD 47-alpha CD3-BITE
4.1 construction of pSC65 vector with alpha CD 47-alpha CD3-BITE target Gene
The DNA sequence of alpha CD 47-alpha CD3-BITE was artificially synthesized, and PCR amplification was performed using the synthesized DNA sequence as a template and the following primers.
The primers for amplification were:
αCD47-αCD3-BITE-F:SEQ ID NO:43
GTACCAGGCCTAGTACTATGGAGAGGACCCTTGTCTG
αCD47-αCD3-BITE-R:SEQ ID NO:44
AATAAGCTCGAAGTCGAC CTAGGAGAGATGCTGATG
PCR reaction procedure: pre-denaturation at 94 ℃ for 5 min; denaturation at 98 ℃ for 10 seconds, 58 ℃: annealing for 30 seconds, extending for 1 minute at 72 ℃, and reacting for 30 cycles; a further 10 minutes of extension at 72 ℃ was carried out, terminating at 25 ℃.
Recovery and cloning construction of PCR products: after completion of amplification, the desired gene was isolated on 2% agarose gel, and the vector pSC65 was linearized by digestion with Sal I (Thermo Scientific, cat. No. ER0642), and recovered by gel cutting, and the PCR fragment and the vector digested fragment were recovered using a Sanprep column DNA gel recovery kit (Promega, cat. No. A9282). The gene recovery product was ligated with the enzyme-digested linearized vector by homologous recombination (Novozak, cat. c 112-02). The ligation products were transformed into E.coli TOP10 and grown overnight on ampicillin-containing plates. On day 2, single colonies were randomly picked for sequencing, mutation site correction was performed, and after the complete sequence was verified to be correct, the shuttle plasmid pSC65 of α CD47- α CD3-BITE was successfully cloned, and the plasmid construction map is shown in FIG. 4A.
4.2 construction of recombinant vaccinia Virus α CD47- α CD3-BITE
1. Cell preparation: will be 143TK-Cells were plated in 6-well plates at approximately 1X 10 per well6And (4) respectively. The culture was carried out for about 24 hours, and when the cells adhered to the wall and spread over the entire bottom surface, the next operation was carried out. 2. Incubation of the vaccinia virus: the cells were infected with 0.0125/3PFU (PFU: plaque Forming Unit, virus titer)/wild type vaccinia virus Tiantan strain of the cells, incubated at 37 ℃ for 1 hour in an incubator, removed, the supernatant aspirated, washed once with 1mL of PBS and then added with 1mL of complete medium.
3. Plasmid transfection: the shuttle plasmid PSC 65-alpha CD 47-alpha CD3-BITE was transfected into 143TK-A cell. Culturing in 37 ℃ incubator for about 48 hours, wherein the specific time is determined according to the cytopathic condition.
4.2 XDMEM maintenance medium (containing 2% PS and 4% FBS) for virus spotting was prepared, 2% pre-warmed low melting agarose was added, and X-gal (final concentration of 200. mu.g/mL) was added.
5. The supernatant in the 6-well plate was aspirated off, and the spotting mixture was added to the 6-well plate at 1mL per well. Then carefully placing the mixture into a refrigerator at 4 ℃ to promote solidification, and transferring the mixture into a incubator at 37 ℃ to perform inverted culture after the low-melting-point agarose is solidified until clear blue spots appear.
6. Bluespots (recombinant vaccinia virus rTV- α CD47- α CD3-BITE) were picked to 500 μ L of complete medium. Repeatedly freezing and thawing at-80 deg.C for more than three times to release virus as much as possible.
7. Will be 143TK-Cells were plated in 6-well plates at approximately 1X 10 per well6And (4) respectively. Culture for about 24 hours until the cells adhere to the wall and spread over the entire floor.
8. Repeatedly blow the blue spots in the EP tube to completely disperse the blue spots.
9. The complete medium was changed to the maintenance medium and then the viral fluid containing the blue spots was added and incubated in an incubator at 37 ℃ for 3-4 hours.
10. Adding screening pressure: the BrdU working concentration is 50 mug/mL, and the BrdU working concentration is placed into an incubator at 37 ℃ for incubation for about 48 hours, and spot paving is carried out according to the virus spot forming condition. The purification process needs to be performed at least 5 times.
11. Then performing small sample amplification of the recombinant vaccinia virus, and paving 143TK-Cells were plated in six well plates, 1X 10 per well6And (3) cells, wherein the cells are about 100% of the bottom area of the pore plate when in use.
12. The medium in the wells was changed to 2mL of maintenance medium before inoculation. Repeatedly blowing and beating the virus liquid containing the blue spots obtained by purification until the blue spots are scattered. Approximately 100. mu.L of virus solution was added to each well. Incubating in an incubator at 37 ℃ for about 48 hours, and collecting samples according to the virus spot forming condition.
13. Collecting a sample: the culture supernatant in the wells was carefully aspirated off 1 mL. The cells were blown down well with the remaining 1mL of medium, harvested in EP tubes and used for subsequent genome extraction and amplification as a seed virus.
4.3 amplification and purification of recombinant vaccinia Virus of α CD47- α CD3-BITE
VERO cell plating: 10cm dishAbout 5X 10 per dish6Ensuring that the cell density reaches 100 percent when the vaccinia virus is inoculated on the next day;
2. before virus inoculation, the complete medium was replaced with 8mL of maintenance medium (DMEM medium + 2% FBS + 1% PS), and the virus was inoculated into cells in the maintenance medium in an amount of about 0.02MOI (MOI ═ virus PFU/cell count). Continuing at 37 ℃ with 5% CO2Culturing in an incubator for about 48 hours, and collecting samples according to the virus spot forming condition;
3. collecting vaccinia virus: discarding 8mL of culture medium in the dish, blowing down the rest cells by using 2mL of maintenance culture medium, and collecting in a 15mL centrifuge tube;
4. after being frozen and stored for 24 hours, the collected virus liquid is repeatedly frozen and thawed for 2 times, the density gradient centrifugation is carried out by 36 percent of sucrose solution, the centrifugation is carried out for 90 minutes at 16000g and 4 ℃, the supernatant is carefully poured off, the virus precipitate in a centrifugal tube is dissolved by PBS buffer solution, and the virus precipitate is subpackaged and stored at-80 ℃ to be tested for the virus titer.
4.4 titer determination of recombinant vaccinia Virus of α CD47- α CD3-BITE
1.143TK-Preparation of cells: will be 143TK-Cells were plated in 24-well plates at approximately 2X 10 per well5The cell density is 100 percent of the bottom area of the 24-pore plate when the cell is used;
2. diluting the virus, namely diluting vaccinia virus solution by using a maintenance medium, and diluting the solution by 10 times from 1:100 to a final volume of 1100 mu L;
3. the complete medium in the 24-well plate was discarded, and 500. mu.L of diluted virus solution was added to the wells to make two duplicate wells. 5% CO at 37 ℃2Incubating in the incubator for about 48 hours, and determining the plaque paving time according to the virus plaque forming condition;
4. the spot paving method comprises the following steps: preparing 8mL of spot-paving culture medium containing 2 XDMEM culture medium, 4% FBS and 2% PS and 8mL of low-melting-point agarose which is placed in a water bath kettle at 37 ℃ after being melted in a boiling water bath, mixing the two, and adding X-gal into the mixture to obtain the final concentration of 200 mu g/mL for later use;
5. the supernatant in the 24-well plate was aspirated off. The spotting mixture of step 4 was immediately added to a 24-well plate at 500. mu.L per well. Carefully placing the agarose gel into a refrigerator at 4 ℃ to promote solidification, transferring the agarose gel into an incubator at 37 ℃ after the agarose gel with low melting point is solidified, and performing inverted culture until clear blue spots appear;
6. and (3) counting virus plaques: firstly, observing whether the number of virus plaques is decreased in a ten-fold ratio trend, then counting the number of single-digit blue plaques in two multiple virus wells, and multiplying the sum of the obtained blue plaque values in the two wells by the reciprocal value of the corresponding dilution of the well to obtain the titer of the virus in 1 mL.
4.5 expression verification of recombinant vaccinia virus rTV- α CD47- α CD3-BITE
1. Collecting virus supernatant: inoculating 5X 10cm of culture dish6VERO cells/dish, ensuring that the cell density reaches 100% when vaccinating vaccinia virus the next day. Before virus inoculation, the complete medium needs to be changed into 8mL of maintenance medium (DMEM medium + 2% FBS + 1% PS); the virus was then added at approximately 0.02MOI (MOI ═ virus PFU/cell number). 5% CO at 37 ℃2Culturing in an incubator for 48 hours; collecting cell supernatant according to virus spot formation condition, centrifuging for 5 min at 10000g, and taking supernatant to a new centrifuge tube.
3. Viral supernatants were purified for His-tag protein: purification was performed manually using a syringe, and the packed nickel beads were Ni-NTAAgarose (QIAGEN, cat # 30210); after completion of column packing, 5nM, 10nM imidazole (dissolved in 50mM NaH) was used2PO4·2H2O, pH 8.0, 300mM NaCl) to elute the non-specifically adsorbed protein, eluting at a natural flow rate, and collecting the filtrate; then 20nM, 100nM, 200nM, 500nM imidazole (dissolved in 50mM NaH)2PO4·2H2O, pH 8.0, 300mM NaCl) to elute the protein of interest, and collecting the corresponding filtrate.
4. 30 μ L of the filtrate was sampled and examined by 10% SDS-PAGE, and the results are shown in FIG. 4B. The loading was carried out from 5mM imidazole eluent to 500mM imidazole eluent in sequence. The target protein appears in 200nM eluate as shown in FIG. 4C, with a band size of about 54 kD.
Example 5: affinity detection of recombinant vaccinia virus rTV-alpha CD 47-alpha CD3-BITE
1. Collecting virus supernatant: inoculating 5X 10cm of culture dish6VERO cells/dish, ensuring that the cell density reaches 100% when vaccinating vaccinia virus the next day. Before virus inoculation, the complete medium needs to be changed into 8mL of maintenance medium (DMEM medium + 2% FBS + 1% PS); then virus was added at approximately 0.02MOI (MOI ═ virus PFU/cell number) while wild-type virus controls were set; 5% CO at 37 ℃2Culturing in an incubator for 48 hours; collecting cell supernatant according to virus spot formation condition, centrifuging at 10000g for 5 min, and collecting supernatant to a new centrifuge tube for His tag protein purification.
His-tagged protein purification was performed as described above.
Preparation of CD47-A549 cells: take 2X 106The cells were split into two EP tubes, centrifuged at 800g for 3 min and the supernatant discarded.
4. The cells were washed 2 times with 1mL of a pre-cooled wash solution (1 XPBS + 2% FBS), centrifuged at 800g for 3 minutes, and the supernatant discarded.
5. A suitable amount of purified protein was incubated with CD47-A549 cells at room temperature for 15 minutes.
6. Washed 2 times with 1mL of precooled wash, centrifuged at 800g for 3 minutes, and the supernatant discarded.
7. 0.1. mu.L of PE-tagged anti-His antibody was added to each sample for 15 minutes at room temperature.
8. Washed 2 times with 1mL of precooled wash, centrifuged at 800g for 3 minutes, and the supernatant discarded.
9. Each sample was resuspended by adding 200. mu.L of wash solution and the affinity was measured by flow cytometry.
The results in FIG. 5 show that the cell supernatants expressed by vaccinia virus recombinant with α CD47- α CD3-BITE bound to CD47 with a positive rate of 85.9% compared to wild-type vaccinia virus. Selecting positive clone to carry out passage stability test, selecting a strain with strong passage stability and high expression target protein to preserve, and naming the strain as rTV-alpha CD 47-alpha CD3-BITE, wherein the preservation number is as follows: CCTCC NO:
v202081, preservation date 1/2/2021, preservation address is chinese type culture collection center, address: wuhan university in Wuhan, China.
Example 6: recombinant vaccinia virus of alpha CD 47-alpha CD3-BITE with in vitro anti-lung cancer effect
1.143TK-Cell plating: will be 143TK-Cells were plated in 6-well plates at approximately 1X 10 per well6The cell density is 100% of the bottom area of the 6-pore plate when the cell is used;
2. complete medium was replaced with 2mL of maintenance medium per well (DMEM medium + 2% FBS + 1% PS) before virus inoculation, and virus was inoculated into the cells at an inoculum size of 0.02 MOI. 5% CO at 37 ℃2The incubation time in the incubator is about 48 hours, and the supernatant collection time is determined according to the virus plaque formation condition. Meanwhile, wild vaccinia virus was used as a control;
3. respectively taking 100 mu L of supernatant to carry out subsequent A549 in vitro killing experiments;
an A549 cell in vitro killing experiment performed according to the kit instructions (Dojindo, K17); pipette 100. mu.L of resuspended A549 cells into a 96-well plate at 1X 10 cells per well437 ℃ and 5% CO2The culture was carried out overnight.
5. The next day, the supernatant was added at 37 ℃ with 5% CO2After 1 hour of incubation, the ratio of effective target was 5: 1 addition of T cells, continued at 37 ℃ with 5% CO2The culture was carried out for 4 hours.
6. After 20. mu.L lysis buffer was added to the high control wells, 20. mu.L medium was added to the low control and background wells at 37 ℃ with 5% CO2Incubate for 30 minutes.
7. Pipette 100 μ L of supernatant from each well into a new 96 well plate.
8. After adding 100. mu.L of a developing solution to each well, the reaction was carried out for 5 minutes in the dark at room temperature.
9. Finally, 50. mu.L of stop buffer was added to each well, and the absorbance at 490nm was immediately measured by a microplate reader.
The result is shown in figure 6, compared with a blank control group, the recombinant vaccinia virus loaded with the alpha CD 47-alpha CD3-BITE can remarkably improve the killing effect on A549 cells in vitro, and has the improvement effect of 4 times.
In conclusion, the alpha CD 47-alpha CD3-BITE protein combined with T cells has very strong killing activity on tumor cells, and the prepared recombinant vaccinia virus rTV-alpha CD 47-alpha CD3-BITE as an oncolytic virus can also obviously control various solid tumors such as human lung cancer and the like, has very high application value for treating tumors, is simple to prepare, and is convenient for large-scale preparation and popularization.
Example 7: t cell mediated in vitro anti-human ovarian adenocarcinoma cell of different structural formulas alpha CD 47-alpha CD3-BITE Effect
To demonstrate the anticancer effects of the different structures of α CD47- α CD3-BITE protein, the applicants designed the following BITE proteins, respectively
BITE1:VLαCD47-L-VHαCD3-L-VLαCD3-L-VHαCD47
Said VHαCD47As shown in SEQ ID NO 19, the VLαCD4721, the VH shown as SEQ ID NOαCD3As shown in SEQ ID NO. 25, VLαCD326, and the L is GGGGSGGGGSGGGGS.
BITE2:VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
Said VHαCD47As shown in SEQ ID NO 19, the VLαCD4721, the VH shown as SEQ ID NOαCD3As shown in SEQ ID NO. 25, VLαCD326, and the L is GGGGSGGGGSGGGGS.
BITE3:VHαCD47-L-VLαCD47-L-VHαCD3-L-VLαCD3
Said VHαCD47As shown in SEQ ID NO 19, the VLαCD4721, the VH shown as SEQ ID NOαCD3As shown in SEQ ID NO. 25, VLαCD3As shown in SEQ ID NO. 26; one linker peptide is GGGGSGGGGSGGS.
BITE 4: like BITE1, only the linker peptide was changed to GGGG;
BITE 5: as with BITE1, the VHαCD3As shown in SEQ ID NO 27, VLαCD3As shown in SEQ ID NO 28.
Practice ofThe results are shown in FIG. 7, which are the same as in examples 2 and 3. VLαCD47-L-VHαCD3-L-VLαCD3-L -VHαCD47Wherein L is (G)4S)3The method is the optimal design of alpha CD 47-alpha CD3-BITE, can mediate strong tumor cell killing function, changes the antibody clone species of CD47 or CD3 (such as clone number SP34), and still has strong killing function;
VH α CD47-L-VH α CD3-L-VL α CD3-L-VL α CD47 wherein L is (G)4S)3It is a suboptimal design for α CD47- α CD3-BITE, but still has similar tumor killing activity as the control antibody.
The above-described embodiments are illustrative, and should not be construed as limiting the invention, and variations, modifications, substitutions, and alterations may be made therein by those of ordinary skill in the art without departing from the scope of the invention.
Sequence listing
<110> Shanghai Xinwan Biotech Co., Ltd
<120> bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof
<130> DIC21110049
<160> 44
<170> SIPOSequenceListing 1.0
<210> 1
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Gly Tyr Gly Met Ser
1 5
<210> 2
<211> 17
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Thr Ile Thr Ser Gly Gly Thr Tyr Thr Tyr Tyr Pro Asp Ser Val Lys
1 5 10 15
Gly
<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Ser Leu Ala Gly Asn Ala Met Asp Tyr
1 5
<210> 4
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Arg Ala Ser Gln Ser Ile Ser Ser Trp Leu Ala
1 5 10
<210> 5
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Lys Ala Ser Ser Leu Glu Ser Gly
1 5
<210> 6
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Gln His Arg Arg Ala
1 5
<210> 7
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Arg Ala Ser Gln Gly Val Thr Ser Asn Leu Ala
1 5 10
<210> 8
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Gly Ala Ser Thr Arg Ala Thr
1 5
<210> 9
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Gln His Arg Gly Thr
1 5
<210> 10
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 10
Arg Ala Ser Gln Gly Ile Ser Ser Arg Leu Ala
1 5 10
<210> 11
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 11
Ala Ala Ser Ser Leu Gln Ser
1 5
<210> 12
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 12
Gln Gln Ala Asn Ser
1 5
<210> 13
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 13
Arg Ala Ser Gln Ser Ile Ser Asn Trp Val Ala
1 5 10
<210> 14
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 14
Lys Ala Ser Thr Leu Ala Asn
1 5
<210> 15
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 15
Gln His Arg Val Thr
1 5
<210> 16
<211> 11
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 16
Arg Ala Ser Gln Ser Ile Ser Asn Trp Leu Ala
1 5 10
<210> 17
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 17
Lys Ala Ser Asn Leu Glu Ser
1 5
<210> 18
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 18
Gln Gln Phe Gly Thr
1 5
<210> 19
<211> 118
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 19
Glu Val Gln Leu Val Glu 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 Gly Tyr
20 25 30
Gly Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Thr Ile Thr Ser Gly Gly Thr Tyr Thr Tyr Tyr Pro Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Ser 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 Ser Leu Ala Gly Asn Ala Met Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 20
<211> 103
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 20
Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Ser Trp
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Lys Ala Ser Ser Leu Glu Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Asp Asp Phe Ala Thr Tyr Tyr Cys Gln His Arg Arg Ala Phe Gly Gln
85 90 95
Gly Thr Lys Val Glu Ile Lys
100
<210> 21
<211> 103
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 21
Glu Ile Val Leu Thr Gln Ser Pro Val Thr Leu Ser Val Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Gly Val Thr Ser Asn
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Tyr Gly Ala Ser Thr Arg Ala Thr Gly Val Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Ser
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln His Arg Gly Thr Phe Gly Gln
85 90 95
Gly Thr Lys Val Asp Ile Lys
100
<210> 22
<211> 103
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 22
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Gly Ile Ser Ser Arg
20 25 30
Leu Ala Trp Tyr Gln Gln Lys 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 Tyr Cys Gln Gln Ala Asn Ser Phe Gly Gly
85 90 95
Gly Thr Lys Val Glu Ile Lys
100
<210> 23
<211> 103
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 23
Asp Ile Gln Met Thr Gln Ser Pro Thr Thr Leu Ser Ala Ser Val Gly
1 5 10 15
His Arg Val Thr Ile Asn Cys Arg Ala Ser Gln Ser Ile Ser Asn Trp
20 25 30
Val Ala Trp Tyr Gln Gln Lys Ala Gly Ser Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Lys Ala Ser Thr Leu Ala Asn Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Ile Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Val Ala Thr Tyr Tyr Cys Gln His Arg Val Thr Phe Gly Pro
85 90 95
Gly Thr Lys Val Asp Leu Lys
100
<210> 24
<211> 103
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 24
Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Asn Trp
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Glu Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Lys Ala Ser Asn Leu Glu Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Asp Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Phe Gly Thr Phe Gly Gln
85 90 95
Gly Thr Lys Val Glu Ile Lys
100
<210> 25
<211> 119
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 25
Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Arg Tyr
20 25 30
Thr Met His Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Met
35 40 45
Gly Tyr Ile Asn Pro Ser Arg Gly Tyr Thr Asn Tyr Asn Gln Lys Phe
50 55 60
Lys Asp Arg Val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Tyr Tyr Asp Asp His Tyr Cys Leu Asp Tyr Trp Gly Gln Gly
100 105 110
Thr Thr Val Thr Val Ser Ser
115
<210> 26
<211> 106
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 26
Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Ser Ala Ser Ser Ser Val Ser Tyr Met
20 25 30
Asn Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Arg Leu Ile Tyr
35 40 45
Asp Thr Ser Lys Leu Ala Ser Gly Val Pro Ser Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Asn Ser Leu Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Trp Ser Ser Asn Pro Phe Thr
85 90 95
Phe Gly Gln Gly Thr Lys Val Glu Ile Lys
100 105
<210> 27
<211> 122
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 27
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ser Lys Tyr Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Ile Leu Tyr
65 70 75 80
Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Val Arg His Gly Asn Phe Asn Ser Tyr Val Ser Trp Phe Ala Tyr Trp
100 105 110
Gly Gln Gly Thr Leu Val Thr Val Ser Ser
115 120
<210> 28
<211> 110
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 28
Gly Ser Gln Ala Val Val Thr Gln Glu Ser Ala Leu Thr Thr Ser Pro
1 5 10 15
Gly Glu Thr Val Thr Leu Thr Cys Arg Ser Ser Thr Gly Ala Val Thr
20 25 30
Thr Ser Asn Tyr Ala Asn Trp Val Gln Glu Lys Pro Asp His Leu Phe
35 40 45
Thr Gly Leu Ile Gly Gly Thr Asn Lys Arg Ala Pro Gly Val Pro Ala
50 55 60
Arg Phe Ser Gly Ser Leu Ile Gly Lys Ala Ala Leu Thr Ile Thr Gly
65 70 75 80
Ala Gln Thr Glu Asp Glu Ala Ile Tyr Phe Cys Ala Leu Trp Tyr Ser
85 90 95
Asn Leu Trp Val Phe Gly Gly Gly Thr Lys Leu Thr Val Leu
100 105 110
<210> 29
<211> 118
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 29
Glu Val Gln Leu Val Glu 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 Gly Tyr
20 25 30
Gly Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Thr Ile Thr Ser Gly Gly Thr Tyr Thr Tyr Tyr Pro Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Ser 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 Ser Leu Ala Gly Asn Ala Met Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 30
<211> 107
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 30
Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Thr Ile Ser Asp Tyr
20 25 30
Leu His Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Lys Phe Ala Ser Gln Ser Ile Ser Gly Ile Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Glu Pro
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Asn Gly His Gly Phe Pro Arg
85 90 95
Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys
100 105
<210> 31
<211> 309
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 31
gacattcaga tgacacagtc tccatctaca ctgtccgcct ccgtgggcga ccgggtgacc 60
attacatgcc gcgcctctca gtctattagc tcttggctcg cctggtatca gcagaagccc 120
ggcaaggccc ctaagctgct gatttacaag gcctccagtc tggaatctgg cgtgccatct 180
aggttcagcg gctctggcag cggcaccgag ttcacactca caatttcttc cttacagccc 240
gacgacttcg ccacatacta ctgccagcac cggagagcct tcggccaggg cacaaaggtg 300
gagatcaag 309
<210> 32
<211> 309
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 32
gagatcgtgc tgacccagtc tccagtgaca ctgagcgtgt cccctggcga gagagccacc 60
ctgtcttgca gagcctctca gggcgtgaca tctaacctcg cctggtatca gcagaagcct 120
ggccaggccc caagactcct gatttacggc gcctctacac gcgccacagg cgtgcccgcc 180
cggttcagcg gctctggctc cggcaccgag ttcacactca caattagctc tcttcagtcc 240
gaggacttcg ccgtgtacta ctgccagcac cgcggcacat tcggccaggg cacaaaggtg 300
gacattaag 309
<210> 33
<211> 309
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 33
gacattcaga tgacacagtc cccatctagc gtgtctgcct ccgtgggcga ccgggtgaca 60
attacatgcc gcgcctctca gggcattagc tcacggctcg cctggtatca gcagaagcct 120
ggcaaggccc ctaagctgct gatttacgcc gcctcttccc tccagagcgg cgtgccatct 180
aggttctctg gctccggctc tggcaccgac ttcacactga ccatttcttc ccttcaaccc 240
gaggacttcg ccacatacta ctgccagcag gccaactcct tcggcggcgg cacaaaggtg 300
gagatcaag 309
<210> 34
<211> 309
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 34
gacatccaga tgacacagtc tcctacaaca ctgagcgcct ccgtgggcca cagagtgaca 60
attaactgcc gcgcctctca gtctatctct aactgggtgg cctggtatca gcagaaggcc 120
ggctccgccc caaagctgct gatttacaag gcctctacac tcgccaacgg cgtgcctagc 180
cggttcagcg gctccggctc tggcaccgag ttcattctca ccatttcttc cttgcaaccc 240
gaggacgtgg ccacatacta ctgccagcac agggtgacct tcggcccagg caccaaggtg 300
gacctcaag 309
<210> 35
<211> 309
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 35
gacattcaga tgacacagtc cccatccacc ctgagcgcct ccgtgggcga cagagtgacc 60
attacatgcc gcgcctctca gtctatttcc aactggctcg cctggtatca gcagaagcca 120
ggcgaggccc ctaagctgct gatttacaag gcctctaact tggagtctgg cgtgccatct 180
aggttctccg gcagcggctc cggcaccgag ttcacactca caattagctc tttacaaccc 240
gacgacttcg ccacatacta ctgccagcag ttcggcacct tcggccaggg cacaaaggtg 300
gagatcaag 309
<210> 36
<211> 354
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 36
gaggtgcagc tggtggagtc tggcggcggc ctcgtgcagc ctggcggctc cctgagactg 60
tcctgcgccg cctctggctt cacattctcc ggctacggca tgtcttgggt gagacaggcc 120
ccaggcaagg gcctagagtg ggtggccaca attacatccg gcggcacata cacctactac 180
cccgacagcg tgaagggcag attcaccatt tcccgcgaca acgccaagaa ctctctgtac 240
ctccagatga acagcctgag agccgaggac accgccgtgt actactgcgc ccggtcactc 300
gccggcaacg ctatggacta ctggggccag ggcacactcg tgaccgtgtc tagc 354
<210> 37
<211> 380
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 37
gacattgatt attgactagt tattaatagt aatcaattac ggggtcatta gttcatagcc 60
catatatgga gttccgcgtt acataactta cggtaaatgg cccgcctggc tgaccgccca 120
acgacccccg cccattgacg tcaataatga cgtatgttcc catagtaacg ccaataggga 180
ctttccattg acgtcaatgg gtggagtatt tacggtaaac tgcccacttg gcagtacatc 240
aagtgtatca tatgccaagt acgcccccta ttgacgtcaa tgacggtaaa tggcccgcct 300
ggcattatgc ccagtacatg accttatggg actttcctac ttggcagtac atctacgtat 360
tagtcatcgc tattaccatg 380
<210> 38
<211> 204
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 38
gtgatgcggt tttggcagta catcaatggg cgtggatagc ggtttgactc acggggattt 60
ccaagtctcc accccattga cgtcaatggg agtttgtttt ggcaccaaaa tcaacgggac 120
tttccaaaat gtcgtaacaa ctccgcccca ttgacgcaaa tgggcggtag gcgtgtacgg 180
tgggaggtct atataagcag agct 204
<210> 39
<211> 331
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 39
cgtttagtga accgtcagat cgcctggaga cgccatccac gctgttttga cctccataga 60
agacaccggg accgatccag cctccgcggc cgggaacggt gcattggaac gcggattccc 120
cgtgccaaga gtgacgtaag taccgcctat agagtctata ggcccacccc cttggcttcg 180
ttagaacgcg gctacaatta atacataacc ttatgtatca tacacatacg atttaggtga 240
cactatagaa taacatccac tttgcctttc tctccacagg tgtccactcc caggtccaac 300
tgcacctcgg ttctatcgat tgaattccac c 331
<210> 40
<211> 45
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 40
atgggatggt catgtatcat cctttttcta gtagcaactg caacc 45
<210> 41
<211> 20
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 41
aagcttggcc gccatggccc 20
<210> 42
<211> 135
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 42
aacttgttta ttgcagctta taatggttac aaataaagca atagcatcac aaatttcaca 60
aataaagcat ttttttcact gcattctagt tgtggtttgt ccaaactcat caatgtatct 120
tatcatgtct ggatc 135
<210> 43
<211> 37
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 43
gtaccaggcc tagtactatg gagaggaccc ttgtctg 37
<210> 44
<211> 36
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 44
aataagctcg aagtcgacct aggagagatg ctgatg 36

Claims (12)

1. An α CD47 and α CD3 bispecific T cell engager α CD47- α CD3 BiTE comprising a fusion protein of any one of the following formulae:
VLαCD47-L-VHαCD3-L-VLαCD3-L-VHαCD47(ii) a Or
VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
Wherein L represents a linker peptide;
wherein said VHαCD47Is the heavy chain variable region of the CD47 antibody, which comprises the following 3 complementarity determining regions:
(i) a VH CDR1, consisting of the sequence: 1, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto,
(ii) a VH CDR2, consisting of the sequence: 2, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto, and
(iii) a VH CDR3, consisting of the sequence: 3, or a sequence having substitution, deletion or addition of one or several amino acids compared thereto;
preferably, the substitution recited in any one of (i) - (iii) is a conservative substitution;
preferably, said VHαCD47Comprises the following steps: VH CDR1 shown in SEQ ID NO. 1, VH CDR2 shown in SEQ ID NO. 2, VH CDR3 shown in SEQ ID NO. 3;
wherein, the VL isαCD47Is the light chain variable region of the CD47 antibody, which comprises the following 3 complementarity determining regions:
(iv) a VL CDR1, consisting of the sequence: any one of SEQ ID NO 4, SEQ ID NO 7, SEQ ID NO 10, SEQ ID NO 13 or SEQ ID NO 16, or a sequence having substitution, deletion or addition of one or more amino acids compared thereto,
(v) a VL CDR2, consisting of the sequence: a sequence shown in any one of SEQ ID NO 5, SEQ ID NO 8, SEQ ID NO 11, SEQ ID NO 14 or SEQ ID NO 17, or having substitution, deletion or addition of one or more amino acids compared thereto, and
(vi) a VL CDR3, consisting of the sequence: any one of SEQ ID NO 6, SEQ ID NO 9, SEQ ID NO 12, SEQ ID NO 15 or SEQ ID NO 18, or a sequence having substitution, deletion or addition of one or more amino acids compared thereto;
preferably, the substitution recited in any one of (iv) - (vi) is a conservative substitution;
preferably, said VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 4, VL CDR2 shown in SEQ ID NO. 5, VL CDR3 shown in SEQ ID NO. 6;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 7, VL CDR2 shown in SEQ ID NO. 8, VL CDR3 shown in SEQ ID NO. 9;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 10, VL CDR2 shown in SEQ ID NO. 11, VL CDR3 shown in SEQ ID NO. 12;
or the VLαCD47Comprises the following steps: VL CDR1 shown in SEQ ID NO. 13, VL CDR2 shown in SEQ ID NO. 14, VL CDR3 shown in SEQ ID NO. 15;
or the VLαCD47Comprises the following steps: VL CDR1 as shown in SEQ ID NO. 16, VL CD as shown in SEQ ID NO. 17R2, VL CDR3 as shown in SEQ ID NO: 18.
2. The bispecific T cell engager of claim 1, wherein the VHαCD47Comprises 3 CDRs contained in the heavy chain variable region shown in SEQ ID NO. 19;
preferably, said VLαCD47Comprises 3 CDRs contained in the light chain variable region set forth in any one of SEQ ID NOs 20-24;
preferably, the 3 CDRs contained in the heavy chain variable region, and/or the 3 CDRs contained in the light chain variable region, are defined by the Kabat, Chothia or IMGT numbering system.
3. The bispecific T-cell engager of claim 1 or 2, wherein the VHαCD47Comprising an amino acid sequence selected from the group consisting of:
(i) 19 in SEQ ID NO;
(ii) a sequence having substitution, deletion or addition of one or several amino acids as compared with the sequence shown in SEQ ID NO. 19; or
(iii) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the sequence set forth in SEQ ID NO. 19;
and/or the presence of a gas in the gas,
the VLαCD47Comprising an amino acid sequence selected from the group consisting of:
(iv) a sequence set forth in any one of SEQ ID NOs 20-24;
(v) a sequence having substitution, deletion or addition of one or several amino acids compared to the sequence shown in any one of SEQ ID NOs 20 to 24; or
(vi) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in any one of SEQ ID NOS 20-24;
preferably, the substitutions described in (ii) or (v) are conservative substitutions.
4. The bispecific T-cell engager of any one of claims 1 to 3, wherein said VHαCD3Comprising an amino acid sequence selected from the group consisting of:
(i) the sequence shown as SEQ ID NO 25 or 27;
(ii) a sequence having substitution, deletion or addition of one or several amino acids compared with the sequence shown in SEQ ID NO. 25 or 27; or
(iii) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in SEQ ID NO 25 or 27;
and/or the presence of a gas in the gas,
the VLαCD3Comprising an amino acid sequence selected from the group consisting of:
(iv) 26 or 28;
(v) a sequence having substitution, deletion or addition of one or several amino acids compared with the sequence shown in SEQ ID NO. 26 or 28; or
(vi) A sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to a sequence set forth in SEQ ID NO 26 or 28;
preferably, the substitutions described in (ii) or (v) are conservative substitutions.
5. The bispecific T-cell engager of any one of claims 1 to 4, wherein said VHαCD47、VHαCD3、VLαCD47And VLαCD3Linked by a linker peptide;
preferably, said VHαCD47、VHαCD3、VLαCD47And VLαCD3Connected with each other through one, two or three connecting peptides;
more preferably, L is KESGSVSSEQLAQFRSLD, EGKSSGSGSESKST, GGGGGG, GGGGGGGG or (GGGGS)n(ii) a Further preferably, n is an integer from 1 to 5, most preferably, n is 3;
preferably, the bispecific T cell engager comprises a fusion protein represented by the formula:
VHαCD47-L-VHαCD3-L-VLαCD3-L-VLαCD47
wherein said VHαCD47As shown in SEQ ID NO 19, the VLαCD4721 or 24, the VHαCD3As shown in SEQ ID NO. 25, VLαCD326, and the L is GGGGSGGGGSGGGGS.
6. An isolated nucleic acid molecule encoding the bispecific T-cell engager of any one of claims 1-5;
preferably, the nucleic acid molecule comprises SEQ ID NO: 31-35, said nucleic acid sequence being a VLαCD47The coding sequence of (a);
preferably, the nucleic acid molecule comprises the nucleotide sequence as set forth in SEQ ID NO: 36, said nucleic acid sequence being VHαCD47The coding sequence of (a).
7. The expression framework of the bispecific T-cell engager of any one of claims 1-5:
5’-E1-E2-E3-E4-E5-E6-E7-3’
wherein:
e1 is a CMV enhancer or/and other cis-acting elements; preferably comprises SEQ ID NO: 37;
e2 is a promoter for recombinant expression, preferably a CMV promoter, more preferably a promoter comprising SEQ ID NO: 38;
e3 is a 5' untranslated region, optionally containing no or one intron sequence, optionally containing one or more restriction enzyme sites; preferably comprises SEQ ID NO: 39;
e4 is the coding nucleotide sequence of the signal peptide of the BiTE protein; preferably, the signal peptide is derived from a human or mouse signal peptide; preferably comprises SEQ ID NO: 40;
e5 is a nucleotide sequence encoding the bispecific T cell engager of any one of claims 1 to 5;
e6 is a 3' untranslated region, optionally comprising one or more restriction enzyme sites; preferably comprises SEQ ID NO: 41;
e7 is the SV40 transcriptional stop signal region; preferably comprises SEQ ID NO: 42.
8. A recombinant oncolytic virus operably inserted or comprising the expression framework of claim 7;
preferably, the expression cassette is located in the Thymidine Kinase (TK) region of the recombinant oncolytic virus;
preferably, the expression frame can be expressed alone, or in fusion with other genes or fragments;
preferably, the recombinant oncolytic virus further comprises gene coding sequences for other immunomodulatory factors, more preferably, the other immunomodulatory factors include, but are not limited to, IL-1, IL-2, IL-3, IL-7, IL-11, IL-12, IL-15, IL-17, IL-18, IL-21, IL-33, IL-35, IL-37, GM-CSF, IFN- α, IFN- β, IFN- γ, anti-PD-1/PD-L1 antibodies, anti-CTLA-4 antibodies, anti-Lag-3 antibodies, anti-TIGIT antibodies, or anti-Tim-3 antibodies; or
The recombinant oncolytic virus further comprises gene coding sequences for proteins associated with apoptosis and apoptosis of cells, preferably selected from apoptosis-related factor 1, interleukin-1 beta converting enzyme, Bcl-2 protein, Fas/APO-1, p53, myc, ataxia telangiectasia mutated gene, corticotropin D or corticotropin E; or
The recombinant oncolytic virus also comprises small RNA of a targeted immune regulation gene and an apoptosis gene.
9. The recombinant oncolytic virus of claim 8, wherein the viral backbone of the oncolytic virus is derived from a modified or engineered vaccinia virus Tiantan strain, vaccinia virus New York strain, vaccinia virus Copenhagen strain, vaccinia virus canary strain, vaccinia virus Ankara strain, adenovirus, adeno-associated virus, herpes simplex virus, varicella-zoster virus, respiratory syncytial virus, Sendzein virus, EB virus, cytomegalovirus, human herpesvirus 6, variola virus, vaccinia virus, molluscum contagiosum virus, orf virus, reovirus, rotavirus, enterovirus, Seneca virus, polio virus, Coxsackie virus, rhinovirus, hepatitis A virus, foot and mouth disease virus, togavirus, alphavirus, Selim virus, eastern equine encephalitis virus, Sindbis virus, Simmondsi virus, Va virus, Coxsackie virus, rhinovirus, hepatitis A virus, foot and mouth disease virus, Tokyo virus, Cinders virus, and Addison virus, Rubella virus, coronavirus, flavivirus, hepatitis C virus, Japanese encephalitis virus, St.Louis encephalitis virus, Murray Valley fever virus, yellow fever virus, West Nile virus, Zika virus, dengue virus, Ebola virus, Marburg virus, arenavirus, Lassa virus, lymphocytic choriomeningitis virus, Pichimede virus, Hunning virus, Martha virus, Hantaan virus, rift Valley fever virus, paramyxovirus, human parainfluenza virus, mumps virus, monkey virus 5, measles virus, vesicular stomatitis virus, rabies virus, orthomyxovirus, influenza A virus, influenza B virus, influenza C virus, hepatitis D virus, monkey immunodeficiency virus, human immunodeficiency virus type 1 and human immunodeficiency virus type 2, Rous sarcoma virus, human T cell leukemia virus type 1, simian foamy virus, herpes virus, Rous sarcoma virus, Rous virus, Ro, Hepatitis b virus, hepatitis e virus, human papilloma virus or polyoma virus;
preferably, the oncolytic viral scaffold is an intracellular maturation virus, an intracellular packaging virus, a cell-associated packaging virus or an extracellular packaging virus.
10. A recombinant vaccinia virus Tiantan strain rTV-alpha CD 47-alpha CD3-BITE, which has a preservation number of: CCTCC NO: V202081.
11. A method of producing a recombinant oncolytic virus according to any one of claims 8 to 10, comprising the steps of:
1) synthesizing an expression framework for the bispecific T cell engager BiTE of claim 7;
2) subcloning the expression framework obtained in the step 1) into a shuttle plasmid of an oncolytic virus to construct a recombinant plasmid vector;
3) transfecting the recombinant plasmid vector obtained in the step 2) into an oncolytic virus, and screening to obtain a recombinant oncolytic virus;
optionally, culturing the obtained recombinant oncolytic virus;
preferably, the method comprises the steps of:
1) an expression framework for the synthetic bispecific T cell engager α CD47- α CD3-BITE comprising the amino acid sequence as set forth in SEQ ID NO: 31-42;
2) subcloning the synthesized expression framework into TK region of vaccinia virus shuttle plasmid (pSC65) to construct recombinant plasmid pSC 65-alpha CD 47-alpha CD 3-BITE;
3) by means of gene homologous recombination, pSC 65-alpha CD 47-alpha CD3-BITE plasmid is transfected into TK143 infected by wild type vaccinia virus-In cells, the two are subjected to homologous recombination to generate recombinant vaccinia virus rTV-alpha CD 47-alpha CD 3-BITE; screening to obtain the recombinant oncolytic vaccinia virus of which the TK region comprises the coding sequence of alpha CD 47-alpha CD 3-BITE;
preferably, the expression frame of the bispecific T-cell engager α CD47- α CD3-BITE is controlled by the early/late promoter p7.5 of vaccinia virus.
12. Use of the bispecific T-cell engager of any one of claims 1-5 or the recombinant oncolytic virus of any one of claims 8-10 for the preparation of an anti-tumor medicament;
preferably, the tumour is selected from B-cell lymphoma, T-cell lymphoma, melanoma, prostate cancer, renal cell carcinoma, sarcoma, gliomas such as high grade glioma, blastoma such as neuroblastoma, osteosarcoma, plasmacytoma, histiocytoma, pancreatic cancer, breast cancer, lung cancer such as small cell lung cancer and non-small cell lung cancer, gastric cancer, liver cancer, colon cancer, rectal cancer, oesophageal cancer, large bowel cancer, hematopoietic cancer, testicular cancer, cervical cancer, ovarian cancer, bladder cancer, squamous cell cancer, adenocarcinoma, AIDS-related lymphoma, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell cancer of the head and neck, hodgkin's lymphoma, non-hodgkin's lymphoma or hematological tumour disease.
CN202111615701.2A 2021-12-27 2021-12-27 Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof Pending CN114249836A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111615701.2A CN114249836A (en) 2021-12-27 2021-12-27 Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111615701.2A CN114249836A (en) 2021-12-27 2021-12-27 Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof

Publications (1)

Publication Number Publication Date
CN114249836A true CN114249836A (en) 2022-03-29

Family

ID=80795257

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111615701.2A Pending CN114249836A (en) 2021-12-27 2021-12-27 Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof

Country Status (1)

Country Link
CN (1) CN114249836A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116284448A (en) * 2023-02-14 2023-06-23 浙江大学 Super antigen-participated three-function T cell adapter and application thereof
WO2023122882A1 (en) * 2021-12-27 2023-07-06 上海鑫湾生物科技有限公司 Bispecific t-cell engager, recombinant oncolytic virus thereof, and use thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103665165A (en) * 2013-08-28 2014-03-26 江苏匡亚生物医药科技有限公司 Bispecific antibody targeting human CD47-SIRP alpha signal passage and preparation method and application thereof
WO2018049261A1 (en) * 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus expressing immune checkpoint modulators
CN109153728A (en) * 2016-03-21 2019-01-04 埃尔斯塔治疗公司 Polyspecific and polyfunctional molecule and application thereof
CN109983121A (en) * 2016-06-30 2019-07-05 昂克诺斯公司 The pseudotyping oncolytic virus of therapeutical peptide delivers
CN110300603A (en) * 2017-02-14 2019-10-01 亘喜生物科技(上海)有限公司 CD47-CAR-T cell
CN113185611A (en) * 2019-12-05 2021-07-30 启愈生物技术(上海)有限公司 Three-function fusion protein containing tumor-associated antigen TAA antibody and application thereof
CN113481165A (en) * 2020-07-16 2021-10-08 山东博安生物技术股份有限公司 CAR-T secreting bispecific T cell adaptors and uses for treating solid tumors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103665165A (en) * 2013-08-28 2014-03-26 江苏匡亚生物医药科技有限公司 Bispecific antibody targeting human CD47-SIRP alpha signal passage and preparation method and application thereof
CN109153728A (en) * 2016-03-21 2019-01-04 埃尔斯塔治疗公司 Polyspecific and polyfunctional molecule and application thereof
CN109983121A (en) * 2016-06-30 2019-07-05 昂克诺斯公司 The pseudotyping oncolytic virus of therapeutical peptide delivers
WO2018049261A1 (en) * 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus expressing immune checkpoint modulators
CN110300603A (en) * 2017-02-14 2019-10-01 亘喜生物科技(上海)有限公司 CD47-CAR-T cell
CN113185611A (en) * 2019-12-05 2021-07-30 启愈生物技术(上海)有限公司 Three-function fusion protein containing tumor-associated antigen TAA antibody and application thereof
CN113481165A (en) * 2020-07-16 2021-10-08 山东博安生物技术股份有限公司 CAR-T secreting bispecific T cell adaptors and uses for treating solid tumors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GOEBELER ME等: "T cell-engaging therapies - BiTEs and beyond", NAT REV CLIN ONCOL *
吴丹青等: "双特异性抗体技术及其临床应用进展", 药学进展 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023122882A1 (en) * 2021-12-27 2023-07-06 上海鑫湾生物科技有限公司 Bispecific t-cell engager, recombinant oncolytic virus thereof, and use thereof
CN116284448A (en) * 2023-02-14 2023-06-23 浙江大学 Super antigen-participated three-function T cell adapter and application thereof

Similar Documents

Publication Publication Date Title
CN107827990B (en) Polypeptide, nucleic acid for encoding polypeptide, T lymphocyte modified by polypeptide and application of T lymphocyte
CN109824781B (en) Specific chimeric antigen receptor of anti-human HER2 antigen, encoding gene, expression vector and application
US10934357B2 (en) Malignant glioma CAR-T therapeutic vector based on OCTS technology, and construction method and application thereof
CN109862912A (en) Carry the adenovirus of bispecific T cell adapter (BiTE)
CN110709425A (en) Chimeric Antigen Receptor (CAR) combined with BCMA and application thereof
US11098123B2 (en) OCTS technology-based pancreatic cancer and malignant mesothelioma CAR-T therapeutic vector, construction method therefor, and use thereof
CN114249836A (en) Bispecific T cell engagers, recombinant oncolytic viruses thereof and uses thereof
JP7420751B2 (en) Oncolytic virus- or antigen-presenting cell-mediated cancer therapy using type I interferon and CD40-ligand
CN110386987B (en) Inhibitory synthetic Notch and double-target system and preparation method and application thereof
CN107033248A (en) Recognize Chimeric antigen receptor and its application of carcinomebryonic antigen
WO2021093251A1 (en) Fgfr4- and dr5-targeted chimeric antigen receptor t cell, preparation method therefor, and application thereof
CN112522208A (en) Transgenic tumor infiltrating lymphocyte and application thereof
CN110204619B (en) Chimeric antigen receptor comprising Fc gamma RI and uses thereof
CN105950662B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD22 and its construction method and application
WO2020019983A1 (en) Genetically engineered cell used for treating tumour
WO2023122882A1 (en) Bispecific t-cell engager, recombinant oncolytic virus thereof, and use thereof
CN115125272A (en) CAR-T therapeutic vector and construction method and application thereof
CN108101994B (en) anti-CD 19 antibodies and uses thereof
WO2023024084A1 (en) Chimeric antigen receptor and use thereof
WO2018218875A1 (en) Octs technology-based prostate cancer car-t therapeutic vector, construction method therefor, and use thereof
CN103882057B (en) Carry structure and the application thereof of p21ras single-chain antibody gene tomour specific adenoviral vectors
WO2023025187A1 (en) Antibody specifically binding to cd47, recombinant oncolytic virus thereof and use thereof
JP2020099326A (en) Nucleic acid for coding chimeric antigen receptor protein and t lymphocyte for expression of chimeric antigen receptor protein
CN117024605B (en) Chimeric antigen receptor, microglia expressing chimeric antigen receptor and application thereof
CN116478929B (en) Bispecific CAR-T cells targeting BCMA and CD19

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