CN117924500A - Heavy chain antibody against CLDN18.2, related products and uses - Google Patents

Heavy chain antibody against CLDN18.2, related products and uses Download PDF

Info

Publication number
CN117924500A
CN117924500A CN202410255907.6A CN202410255907A CN117924500A CN 117924500 A CN117924500 A CN 117924500A CN 202410255907 A CN202410255907 A CN 202410255907A CN 117924500 A CN117924500 A CN 117924500A
Authority
CN
China
Prior art keywords
seq
heavy chain
amino acid
acid sequence
cells
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
CN202410255907.6A
Other languages
Chinese (zh)
Inventor
余学军
狄升蒙
张世伟
李照润
范艳秋
马恒
陈鑫
冯冬歌
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.)
Huadao Shanghai Biopharma Co ltd
Original Assignee
Huadao Shanghai Biopharma 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 Huadao Shanghai Biopharma Co ltd filed Critical Huadao Shanghai Biopharma Co ltd
Priority to CN202410255907.6A priority Critical patent/CN117924500A/en
Publication of CN117924500A publication Critical patent/CN117924500A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • 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
    • C12N2510/00Genetically modified cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Abstract

The application discloses a heavy chain antibody against CLDN18.2, related products and uses. The heavy chain antibody consists of a heavy chain variable region, wherein the amino acid sequence of CDR1 of the heavy chain variable region comprises a sequence shown as SEQ ID No.3, the amino acid sequence of CDR2 of the heavy chain variable region comprises a sequence shown as SEQ ID No.9, and the amino acid sequence of CDR3 of the heavy chain variable region comprises a sequence shown as SEQ ID No. 15. The heavy chain antibody for resisting the CLDN18.2 only comprises a heavy chain variable region, has high affinity and specificity, can efficiently target the CLDN18.2 antigen without recognizing the CLDN18.1, has a simple structure and is easy to prepare. In addition, the CAR-T cells prepared by the method can specifically identify CLDN18.2 positive tumor cells, and simultaneously can release various cytokines to kill the tumor cells with high efficiency.

Description

Heavy chain antibody against CLDN18.2, related products and uses
The application is a divisional application with the application number 2022115255188, the application date of the original application is 2022, 11 and 30, and the application is named as 'heavy chain antibody against CLDN18.2, related products and application'.
Technical Field
The application relates to the technical field of biological medicine, in particular to a heavy chain antibody for resisting CLDN18.2, a related product and application.
Background
Heavy chain antibodies (HEAVY CHAIN anti-bodies, hcAb) are found in camels and shark animals and are naturally deleted light chain antibodies, consisting of only heavy chains. Cloning of the variable region results in a single domain antibody consisting of only the heavy chain variable region, known as VHH (Variabledomain of HEAVY CHAIN of HEAVY CHAIN antibody), also known as nanobody (nanobody), which is the smallest functional antigen-binding fragment. Unlike common antibody, nanometer antibody is one peptide chain of about 110 amino acids and has molecular weight of about 1/10 that of common antibody, and has the advantages of small molecule, high stability, high solubility, easy expression, low production cost, etc. compared with common antibody and recombinant single chain antibody (SINGLE CHAIN FRAGMENT variable, scFv), the nanometer antibody has wide application foreground in immunological experiment, diagnosis and treatment.
The Claudins (CLDNs) family has been found to be expressed in a variety of epithelial tissues and to play a significant role in the tight junctions between cells. Tens of members of the CLDNs family are transmembrane proteins, with a very high degree of identity within the transmembrane region. The CLDNs family plays an important role in maintaining the osmotic pressure of epithelial cells, protecting barrier effect and the like, and participates in the immune defense process; in addition, the CLDNs family was observed to express activities different from usual in the development of many tumors, and was studied and discussed in various borders as tumor-specific marker proteins. CLDN18.2 is one member of CLDNs family, and has very low expression level in normal tissues, but research shows that it is abnormal to activate in the process of generating and developing primary malignant tumors of different organs and tissues of human body such as stomach, mammary gland and bronchus, and has over-expression level, especially high in malignant tumors of digestive system. Therefore, CLDN18.2 is expected to be a new target for first-line treatment of gastric cancer and the like due to the characteristic of high selectivity and stable expression in malignant solid tumors such as gastric cancer and the like.
One of the difficulties in CLDN18.2 targeted therapies is the presence of a protein that is very similar in structure to CLDN18.1, another cleavage entity formed by the allele of the first exon of the CLDN18 gene. CLDN18.1 has a high degree of similarity to the structural sequence of CLDN18.2, differing only by 7 amino acid residues located on the ectodomain ECL1 sequence. CLDN18.1 is highly expressed only in normal alveolar tissue, with very high specificity. Most antibodies developed by researchers and practitioners that target CLDN18.2 are also often prone to off-target recognition of CLDN18.1, thus severely compromising the targeting and safety of antibody drugs. Therefore, development of antibody drugs specifically recognizing CLDN18.2 but not CLDN18.1 is a difficulty in the development process.
In view of the above, developing efficient CAR-T therapies against digestive system malignancies is of great importance in the field of digestive system malignancies.
Disclosure of Invention
In view of the above-described drawbacks of the prior art, it is an object of the present application to provide a heavy chain antibody against CLDN18.2, related products and uses for solving the problems of the prior art.
In order to achieve the above purpose, the present invention adopts the following technical scheme.
In a first aspect, the application provides an anti-CLDN 18.2 heavy chain antibody consisting of a heavy chain variable region having CDR1 of the amino acid sequence as shown in one of SEQ ID nos. 1-6; the amino acid sequence of CDR2 of the heavy chain variable region is shown as one of SEQ ID No.7-SEQ ID No. 12; the amino acid sequence of CDR3 of the heavy chain variable region is shown as one of SEQ ID No.13-SEQ ID No. 18.
In certain embodiments, the amino acid sequence of the heavy chain variable region comprises A1) or A2):
A1 An amino acid sequence as shown in one of SEQ ID No.38-SEQ ID No. 43;
a2 An amino acid sequence having at least 80% homology with the amino acid sequence shown in A1) and having the function of the amino acid sequence defined in A1).
In certain embodiments, the heavy chain variable region further comprises framework regions FR1-FR4: the amino acid sequence of the framework region FR1 comprises a sequence shown as one of SEQ ID No.19-SEQ ID No.23, the amino acid sequence of the framework region FR2 comprises a sequence shown as one of SEQ ID No.24-SEQ ID No.29, the amino acid sequence of the framework region FR3 comprises a sequence shown as one of SEQ ID No.30-SEQ ID No.35, and the amino acid sequence of the framework region FR4 comprises a sequence shown as one of SEQ ID No.36 or SEQ ID No. 37.
In a second aspect the application provides an isolated polypeptide comprising an antigen binding domain comprising a heavy chain antibody as described above, a hinge region and a transmembrane domain.
In certain embodiments, the polypeptide is a chimeric antigen receptor.
In certain embodiments, the transmembrane domain comprises one or more of a CD 8a transmembrane region, a CD28 transmembrane region, or a DAP10 transmembrane region.
In certain embodiments, the hinge region comprises a CD8 a hinge region.
In certain embodiments, the polypeptide comprises, in order from the N-terminus to the C-terminus, the heavy chain antibody, a hinge region, and a transmembrane domain.
In a third aspect the application provides a biological material associated with a heavy chain antibody as hereinbefore described or a polypeptide as hereinbefore described, said biological material comprising one or more of the following:
1) A nucleotide encoding a heavy chain antibody as described above or a nucleotide encoding a polypeptide as described above;
2) A recombinant expression vector comprising 1) said nucleotide;
3) The bioengineering bacteria containing 1) the nucleotide or the bioengineering bacteria containing 2) the recombinant expression vector.
In certain embodiments, in 1), the sequence of the nucleotide comprises a sequence as set forth in one of SEQ ID No.44-SEQ ID No. 49.
In certain embodiments, the recombinant expression vector is selected from a lentiviral vector, a retroviral vector, or an adenoviral vector.
Preferably, the recombinant expression vector is a lentiviral vector.
In a fourth aspect the application provides a cell expressing a polypeptide as described above.
In certain embodiments, the cells are selected from one or more of T lymphocytes, B lymphocytes, NK cells, mast cells, and macrophages.
In a fifth aspect the application provides the use of a heavy chain antibody as hereinbefore described or a polypeptide as hereinbefore described or a biological material as hereinbefore described or a cell as hereinbefore described in the manufacture of an in vitro assay product, in the manufacture of a medicament for the prevention or treatment of a tumour.
In certain embodiments, the tumor is a tumor associated with expression of CLND 18.2, preferably selected from one or more of bladder cancer, liver cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, biliary tract cancer, leukemia, lymphoma, pancreatic cancer, small cell lung cancer, non-small cell lung cancer, breast cancer, urinary tract cancer, head and neck cancer, gastrointestinal cancer, gastric cancer, esophageal cancer, ovarian cancer, renal cancer, melanoma, prostate cancer, and thyroid cancer.
The sixth aspect of the application provides a test product comprising an antibody as described above or a polypeptide as described above or a biological material as described above or a cell as described above.
In certain embodiments, the product is used to detect CLND 18.2.
A seventh aspect of the application provides a pharmaceutical composition comprising an antibody as described above or a polypeptide as described above or a biological material as described above or a cell as described above, and a pharmaceutically acceptable carrier.
In certain embodiments, the pharmaceutical composition is for preventing or treating a neoplastic disease associated with expression of CLND 18.2.
Preferably, the tumor associated with expression of CLND 18.2 is selected from one or more of bladder cancer, liver cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, biliary tract cancer, leukemia, lymphoma, pancreatic cancer, small cell lung cancer, non-small cell lung cancer, breast cancer, urinary tract cancer, head and neck cancer, gastrointestinal cancer, gastric cancer, esophageal cancer, ovarian cancer, renal cancer, melanoma, prostate cancer, and thyroid cancer.
Compared with the prior art, the application has the beneficial effects that:
(1) The heavy chain antibody for resisting the CLDN18.2 only comprises a heavy chain variable region, has high affinity and specificity, can efficiently target the CLDN18.2 antigen, and has a simple structure and easy preparation.
(2) The invention constructs chimeric antigen receptor by using heavy chain antibody of anti-CLDN 18.2, and the chimeric antigen receptor can efficiently target CLDN18.2 without recognizing CLDN18.1.
(3) The chimeric antigen receptor cell of the invention can specifically identify CLDN18.2 positive tumor cells, perform high-efficiency killing, and simultaneously release various cytokines including TNF-alpha, IFN-gamma factors and the like to play a role in cell killing.
Drawings
FIG. 1A shows a graph of the affinity of the anti-CLDN 18.2-A93 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 1B shows a graph of the affinity of the anti-CLDN 18.2-A100 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 1C shows a graph of the affinity of the anti-CLDN 18.2-A102 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 1D shows a graph of the affinity of the anti-CLDN 18.2-A127 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 1E shows a graph of the affinity of the anti-CLDN 18.2-B123 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 1F shows a graph of the affinity of the anti-CLDN 18.2-B142 VHH antibody detected using Biacore in example 2 of the present invention.
FIG. 2A is a graph showing the results of FACS detection of the 6 anti-CLDN 18.2 VHH antibodies recognizing the CLDN18.2 antigen in example 3 of the present invention.
FIG. 2B is a graph showing the results of FACS detection of the 6 anti-CLDN 18.2 VHH antibodies recognizing the CLDN18.1 antigen in example 3 of the present invention.
FIG. 3A shows a plasmid map of a lentiviral vector (HD SIN03A93 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 in example 4 of the invention.
FIG. 3B shows a plasmid map of a lentiviral vector (HD SIN03A100 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 in example 4 of the invention.
FIG. 3C shows a plasmid map of a lentiviral vector (HD SIN03A102 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 in example 4 of the invention.
FIG. 3D shows a plasmid map of a lentiviral vector (HD SIN03A127 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 according to example 4 of the invention.
FIG. 3E shows a plasmid map of a lentiviral vector (HD SIN03B123 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 according to example 4 of the invention.
FIG. 3F shows a plasmid map of a lentiviral vector (HD SIN03B142 41 BBz) expressing a chimeric antigen receptor of CLDN18.2 according to example 4 of the invention.
FIG. 4 shows a schematic structural diagram of a chimeric antigen receptor expressing an anti-CLDN 18.2 antibody in example 5 of the present invention.
FIG. 5 is a graph showing the expression rate of chimeric antigen receptor of T lymphocytes in example 9 of the present invention.
Figure 6A shows a graph of the killing effect of 6 CAR-T cells on 293T cells in example 10 of the present invention.
FIG. 6B is a graph showing the killing effect of 6 CAR-T cells on HGC-27-18.2 cells in example 10 of the present invention.
FIG. 6C is a graph showing the killing effect of 6 CAR-T cells on N87-18.2 cells in example 10 of the present invention.
FIG. 7 is a bar graph showing the levels of TNF- α cytokine secretion by 6 CAR-T cells in example 11 of the present invention.
FIG. 8 shows a bar graph of IFN-gamma cytokine secretion levels by 6 CAR-T cells in example 11 of the present invention.
FIG. 9 shows a map of HD SIN03 CD19 CAR-Kanar plasmid in example 4 of the invention.
Detailed Description
The invention adopts CHO cells expressing CLDN18.2 protein to stimulate alpaca to generate high-titer antibodies, adopts phage display technology to construct phage display camel VHH immune library with the reservoir capacity larger than 10 9, and obtains 6 candidate antibodies by panning and amplification, including CLDN18.2-A93, CLDN18.2-A100, CLDN18.2-A102, CLDN18.2-A127, CLDN18.2-B123 and CLDN18.2-B142. The reduction molecular weight is found to be less than 40Kda, which is 1/10 of that of the common antibody; through Biacore detection, the affinities of the 6 candidate antibodies are found to reach the nM level; it was further found that the 6 candidate antibodies specifically recognize CLDN18.2 antigen on the cell surface, but not CLDN18.1 antigen. Meanwhile, CAR-T cells containing 6 candidate antibodies are constructed, and the CAR-T cells are found to have killing activity on CLDN18.2 positive tumor cells and can secrete cytokines TNF-alpha and IFN-gamma with high efficiency. The present invention has been completed on the basis of this finding.
The first aspect of the application provides a heavy chain antibody against CLDN18.2, which consists of a heavy chain variable region, wherein the amino acid sequence of CDR1 of the heavy chain variable region is as shown in one of SEQ ID nos. 1-6; the amino acid sequence of CDR2 of the heavy chain variable region is shown as one of SEQ ID No.7-SEQ ID No. 12; the amino acid sequence of CDR3 of the heavy chain variable region is shown as one of SEQ ID No.13-SEQ ID No. 18.
The heavy chain antibody provided by the application consists of a heavy chain variable region, and has the advantages of small molecular weight, high affinity, weak immunogenicity to human bodies, easier storage and transportation, easier expression, genetic engineering transformation and the like.
SEQ ID No.1:GRTFSSYH
SEQ ID No.2:GSIAHIAGINV
SEQ ID No.3:RSILSFSV
SEQ ID No.4:GRIFRINN
SEQ ID No.5:GRTFGNYN
SEQ ID No.6:GFTLDYYS
SEQ ID No.7:ISWSGGIR
SEQ ID No.8:IFYSGTT
SEQ ID No.9:IFNGGHT
SEQ ID No.10:VSLGGTT
SEQ ID No.11:ISWSGSIT
SEQ ID No.12:ISGSGVST
SEQ ID No.13:AAAGVTLSRNEGDYAY
SEQ ID No.14:YANHNIANY
SEQ ID No.15:NANDLGFLAQNY
SEQ ID No.16:NADVYETPFSSKNY
SEQ ID No.17:AAASFDLTRDASRYAY
SEQ ID No.18:IARPIGNSWYCSIAYTDFGS
In the invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises a sequence shown as SEQ ID No.1, the amino acid sequence of CDR2 comprises a sequence shown as SEQ ID No.7, and the amino acid sequence of CDR3 comprises a sequence shown as SEQ ID No. 13.
In the present invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises the sequence shown in SEQ ID No.2, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID No.8, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID No. 14.
In the invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises a sequence shown as SEQ ID No.3, the amino acid sequence of CDR2 comprises a sequence shown as SEQ ID No.9, and the amino acid sequence of CDR3 comprises a sequence shown as SEQ ID No. 15.
In the present invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises the sequence shown in SEQ ID No.4, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID No.10, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID No. 16.
In the present invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises the sequence shown in SEQ ID No.5, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID No.11, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID No. 17.
In the present invention, the amino acid sequence of CDR1 of the heavy chain variable region comprises the sequence shown in SEQ ID No.6, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID No.12, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID No. 18.
In the present invention, the heavy chain variable region comprises framework regions FR1-FR4. Preferably, the amino acid sequence of FR1 comprises the sequence shown as SEQ ID No.19 or SEQ ID No.20 or SEQ ID No.21 or SEQ ID No.22 or SEQ ID No. 23. Preferably, the amino acid sequence of FR2 comprises the sequence shown as SEQ ID No.24 or SEQ ID No.25 or SEQ ID No.26 or SEQ ID No.27 or SEQ ID No.28 or SEQ ID No. 29. Preferably, the amino acid sequence of FR3 comprises the sequence shown as SEQ ID No.30 or SEQ ID No.31 or SEQ ID No.32 or SEQ ID No.33 or SEQ ID No.34 or SEQ ID No. 35. Preferably, the amino acid sequence of FR4 comprises the sequence shown as SEQ ID No.36 or SEQ ID No. 37.
SEQ ID No.19:QVQLVESGGGLVQVGGSLRLSCAAS
SEQ ID No.20:EVQLVESGGGLVQPGGSLRLSCAAS
SEQ ID No.21:EVQVVESGGGTVQPGGSLRLSCATS
SEQ ID No.22:QVQLVESGGGSVQPGGSLRLSCAAS
SEQ ID No.23:QLQLVESGGGLVQPGGSLRLSCAAS
SEQ ID No.24:MGWFRQAPGKEREFVAA
SEQ ID No.25:MGWYRQTPGKQRDFVAH
SEQ ID No.26:MGWYRQAPGNQREWVAH
SEQ ID No.27:MYWYRQAAGKQREFVAG
SEQ ID No.28:MGWFRQAPGKEREFVAG
SEQ ID No.29:IAWFRQAPGKEREGVSC
SEQ ID No.30:DYTSSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYC
SEQ ID No.31:TYADSVKGRFIISRDSATNTMSLQMNNLKPEDTATYYC
SEQ ID No.32:NYADSVKGRATISRDNAKNTVYLQMSSLKPEDTAVYYC
SEQ ID No.33:NAADSVKGRFTVSRDNAKNTMYLQMSAMKPDDTAVYYC
SEQ ID No.34:DYGDSVKGRFTISRDNAENTLYLQMNNLKPEDTAVYFC
SEQ ID No.35:KYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAIYYC
SEQ ID No.36:WGQGTQVTSSS
SEQ ID No.37:WGQGTQVTVSS
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.19, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.24, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.30, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 36.
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.20, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.25, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.31, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 37.
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.21, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.26, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.32, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 37.
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.22, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.27, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.33, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 37.
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.23, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.28, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.34, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 37.
In the invention, the amino acid sequence of the framework region FR1 comprises a sequence shown as SEQ ID No.23, the amino acid sequence of the FR2 comprises a sequence shown as SEQ ID No.29, the amino acid sequence of the FR3 comprises a sequence shown as SEQ ID No.35, and the amino acid sequence of the FR4 comprises a sequence shown as SEQ ID No. 37.
In the present invention, the amino acid sequence of the heavy chain variable region comprises A1) or A2): a1 An amino acid sequence as shown in one of SEQ ID No.38-SEQ ID No. 43; a2 An amino acid sequence having at least 80% homology with the amino acid sequence shown in A1) and having the function of the amino acid sequence defined in A1). Specific sequences of SEQ ID No.38, SEQ ID No.39, SEQ ID No.40, SEQ ID No.41, SEQ ID No.42, SEQ ID No.43 are shown below corresponding to CLDN18.2-A93, CLDN18.2-A100, CLDN18.2-A102, CLDN18.2-A127, anti-CLDN 18.2-B123 and CLDN18.2-B142, respectively.
SEQ ID No.38 (frame region in italics, CDR region underlined):
SEQ ID No.39 (frame region in italics, CDR region underlined):
SEQ ID No.40 (frame region in italics, CDR region underlined):
SEQ ID No.41 (italics in bold represents framework regions, underlined is the CDR region):
SEQ ID No.42 (italics in bold represents framework regions, underlined is the CDR region):
SEQ ID No.43 (italics in bold represents framework regions, underlined is the CDR region):
in the invention, the preparation method of the heavy chain antibody comprises the following steps: inserting the coding gene of the heavy chain antibody into an expression vector to obtain a recombinant expression vector, introducing the recombinant expression vector into cells, culturing, and separating and purifying to obtain the heavy chain antibody.
Another aspect of the application provides an isolated polypeptide comprising an antigen binding domain comprising a heavy chain antibody as described above, a hinge region and a transmembrane domain.
In the invention, the heavy chain antibody is utilized to construct the polypeptide, and the polypeptide can efficiently target CLDN18.2.
In the present invention, the hinge region comprises a CD 8a hinge region.
In the present invention, the transmembrane domain comprises one or more of a CD8 a transmembrane region, a CD28 transmembrane region, or a DAP10 transmembrane region.
In the present invention, the polypeptide further comprises a signal peptide or a signal transduction domain. Preferably, the signal peptide comprises a CD8 a signal peptide. Preferably, the signal transduction domain comprises an immunoreceptor tyrosine activation motif. More preferably, the signal transduction domain further comprises a co-stimulatory molecule comprising any one or a combination of at least two of 4-1BB, the intracellular domain of CD28, OX40, ICOS or the intracellular domain of DAP 10. In certain embodiments, the amino acid sequence of the signal peptide comprises the sequence set forth in SEQ ID No. 50. In certain embodiments, the amino acid sequence of the immunoreceptor tyrosine activation motif comprises the sequence depicted in SEQ ID No. 52.
MALPVTALLLPLALLLHAARP(SEQ ID No.50)。
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID No.52).
In the present invention, the polypeptides include a CD8 a signal peptide, the heavy chain antibody, a CD8 a hinge region, a CD8 a transmembrane region, and an immunoreceptor tyrosine activation motif. The amino acid sequences of the CD8 alpha hinge region and the transmembrane region are
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYC(SEQ ID No.51)。
Another aspect of the application relates to a heavy chain antibody as described above or a polypeptide as described above, said biological material comprising one or more of the following:
1) A nucleotide encoding a heavy chain antibody as described above or a nucleotide encoding a polypeptide as described above;
2) A recombinant expression vector comprising 1) said nucleotide;
3) The bioengineering bacteria containing 1) the nucleotide or the bioengineering bacteria containing 2) the recombinant expression vector.
In the present invention, 1), the sequence of the nucleotide encoding the heavy chain antibody as described above comprises a sequence as shown in one of SEQ ID No.44 to SEQ ID No. 49.
SEQ ID No.44:
caggtgcagctggtggagagcggggggggcctggtgcaggtgggcgggagcctgaggctgagctgcgccgccagcggcaggacattcagcagctaccacatggggtggttccggcaggcccccggcaaggagagggagttcgtggccgccatcagctggagcgggggcatcagggactacacaagcagcgtgaagggcaggttcaccatcagccgggacaacgccaaaaacaccgtgtacctgcaaatgaacagcctcaagccagaggacaccgccgtgtactactgtgccgccgctggcgtcactctgtccaggaacgagggggattacgcttattgggggcagggcacacaggtgacaagcagcagc
SEQ ID No.45:
gaggtgcagctggtggagagcggggggggcctggtgcagcccggcggcagcctgaggctgagctgcgccgccagcgggagcatcgcccacatcgccgggatcaacgtgatggggtggtaccggcagacccccggcaagcagagggacttcgtggcccacatcttttacagcggcaccacaacatacgccgatagcgtgaagggcaggttcatcatcagccgggatagcgccaccaacaccatgagcctgcagatgaacaatctgaagcccgaggacacagccacatactactgctacgccaaccacaacatcgccaactattgggggcagggcacacaggtgaccgtgtcctcc
SEQ ID No.46:
gaagtgcaggtggtggagagcggcggcggcacagtgcagcccggcggcagcctgaggctgagctgcgccacaagccggagcatcctgagcttcagcgtgatggggtggtacaggcaggcccccggcaaccagcgggagtgggtggcccacatcttcaacggggggcacacaaactacgccgatagcgtgaaggggagggccaccatcagccgggataacgccaagaacacagtgtacctgcagatgagcagcctgaagcccgaggacaccgccgtgtactactgcaacgccaacgacctgggcttcctggcccagaactactgggggcaggggacacaggtgacagtgagcagc
SEQ ID No.47:
caggtgcagctggtggagagcggcggcgggagcgtgcagcccggcgggagcctgaggctgagctgcgccgcctccggccggatcttccgcattaacaacatgtactggtacaggcaggccgccgggaagcagcgggagttcgtggccggggtgagcctgggcggcaccaccaacgccgccgatagcgtgaagggccggttcacagtgagcagggataacgccaagaacaccatgtacctgcagatgagcgccatgaagcctgatgacaccgccgtctactactgtaacgctgacgtctatgagaccccctttagtagtaagaactactggggccagggcacacaggtgaccgtgagtagcSEQ ID No.48:
cagctgcagctggtggagagcggcgggggcctggtacagcccggcggcagcctccggctgtcctgcgccgccagcgggagaacatttgggaactacaatatggggtggtttaggcaggcccctggcaaggagcgggagttcgtcgccgggatcagctggagcggcagcatcaccgattacggcgactccgtgaaggggaggttcaccatcagccgggataacgccgagaacacactgtacctgcagatgaacaacctgaagcccgaggatacagccgtgtacttctgcgccgccgccagcttcgacctgaccagggacgccagcaggtacgcctactggggccagggcacacaggtgacagtgagcagc
SEQ ID No.49:
cagctgcagctggtggagagcgggggcggcctggtgcagcccggcgggagcctgaggctgagctgcgccgccagcgggttcaccctggactactactccatcgcctggttcaggcaggcccccggcaaggagcgggagggggtgagctgcatctccggcagcggggtgagcacaaagtacgccgatagcgtgaagggccggttcaccatcagcagggataacgccaagaacacagtgtacctgcagatgaacagcctgaagcccgaggatacagccatctactactgcatcgcccggcccatcgggaacagctggtactgcagcatcgcctacaccgacttcggcagctggggccagg ggacacaggtgacagtgagcagc
In the present invention, 2) the recombinant expression vector comprises the nucleotide described in 1). The recombinant expression vector is a viral vector or a non-viral vector. For example, non-viral vectors include: plasmids, phagemids, cosmids, artificial chromosomes such as Yeast Artificial Chromosome (YAC), bacterial Artificial Chromosome (BAC) or P1-derived artificial chromosome (PAC), phages such as lambda phage or M13 phage, animal viruses, and the like. The viral vector comprises: retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpesviruses (e.g., herpes simplex viruses), poxviruses, baculoviruses, papillomaviruses, papilloma-vacuolated viruses (e.g., SV 40). The vector may contain a variety of elements that control expression, including promoter sequences, transcription initiation sequences, enhancer sequences, selection elements, and reporter genes. In addition, the vector may also contain a replication origin. The vector may also include components that assist in its entry into the cell, including, but not limited to, viral particles, liposomes, or protein shells. Preferably, the recombinant expression vector is selected from lentiviral vectors, retroviral vectors or adenoviral vectors, more preferably lentiviral vectors.
In the invention, 3) the recombinant expression vector of the bioengineering bacterium 1). Preferably, the bioengineering bacteria are prepared by transfecting mammalian cells, such as 293T cells, with the recombinant expression vector and helper plasmid.
In another aspect the application provides a cell expressing a polypeptide as described above.
In the present invention, the cells are selected from one or more of T lymphocytes, B lymphocytes, NK cells, mast cells and macrophages.
The invention utilizes the heavy chain antibody to prepare a chimeric antigen receptor and further prepares chimeric antigen receptor immune cells (CAR-T cells), wherein the chimeric antigen receptor immune cells can specifically identify CLDN18.2 positive tumor cells and kill the tumor cells with high efficiency, and simultaneously can release various cytokines including TNF-alpha, IFN-gamma factors and the like to play a role in cell killing.
Another aspect of the application provides the use of a heavy chain antibody as described above or a polypeptide as described above or a biological material as described above or a cell as described above for the preparation of an in vitro assay product, for the preparation of a medicament for the prevention or treatment of a tumor.
In certain embodiments, the tumor is a tumor associated with expression of CLND 18.2, preferably selected from one or more of bladder cancer, liver cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, biliary tract cancer, leukemia, lymphoma, pancreatic cancer, small cell lung cancer, non-small cell lung cancer, breast cancer, urinary tract cancer, head and neck cancer, gastrointestinal cancer, gastric cancer, esophageal cancer, ovarian cancer, renal cancer, melanoma, prostate cancer, and thyroid cancer.
Another aspect of the application provides an assay product comprising a heavy chain antibody as described above or a polypeptide as described above or a biological material as described above or a cell as described above.
In the present invention, the product is used to detect CLND 18.2.
In the present invention, the detection product is selected from the group consisting of a kit, a chip and a membrane strip.
In the present invention, the detection product can be generally diagnosed against the CLND18.2 antigen serving as an action target, and CLND18.2 antigen is used as a biomarker. The product may also include a label against CLND18.2 antibodies, which may generally be used to label the anti-CLND 18.2 antibodies, optionally in the form of one or more of biotin, fluorescein, horseradish peroxidase, magnetic beads and agarose beads.
Another aspect of the application provides a pharmaceutical composition comprising an antibody as described above or a polypeptide as described above or a biological material as described above or a cell as described above, and a pharmaceutically acceptable carrier.
In the present invention, the pharmaceutical composition is used for preventing or treating a tumor disease associated with CLND 18.2 expression. Preferably, the tumor comprises a tumor expressing CLND 18.2 antigen, non-limiting examples of cancer include bladder cancer, liver cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, biliary tract cancer, leukemia, lymphoma, pancreatic cancer, small cell lung cancer, non-small cell lung cancer, breast cancer, urinary tract cancer, head and neck cancer, gastrointestinal cancer, gastric cancer, esophageal cancer, ovarian cancer, renal cancer, melanoma, prostate cancer, and thyroid cancer.
In the present invention, the pharmaceutically acceptable carrier refers to a carrier that does not cause allergic reactions or other untoward effects in the patient to whom it is administered. Pharmaceutically acceptable carriers include, for example, one or more of water, physiological saline, phosphate buffer, dextrose, glycerol, ethanol, and the like, as well as combinations of the foregoing.
In the present invention, the pharmaceutically acceptable carrier may further include a trace amount of auxiliary substances, such as a wetting or emulsifying agent, a preservative or a buffer, which can improve the shelf life or effect of the antibody.
Other advantages and effects of the present invention will become apparent to those skilled in the art from the following disclosure, which describes the embodiments of the present invention with reference to specific examples. The invention may be practiced or carried out in other embodiments that depart from the specific details, and the details of the present description may be modified or varied from the spirit and scope of the present invention.
Before the embodiments of the invention are explained in further detail, it is to be understood that the invention is not limited in its scope to the particular embodiments described below; it is also to be understood that the terminology used in the examples of the invention is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the invention; in the description and claims of the invention, the singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise.
Where numerical ranges are provided in the examples, it is understood that unless otherwise stated herein, both endpoints of each numerical range and any number between the two endpoints are significant both in the numerical range. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In addition to the specific methods, devices, materials used in the embodiments, any methods, devices, and materials of the prior art similar or equivalent to those described in the embodiments of the present invention may be used to practice the present invention according to the knowledge of one skilled in the art and the description of the present invention.
Example 1 acquisition of heavy chain antibodies against CLDN18.2
In this example, a phage display technique was used to panning a CLDN18.2 immunized alpaca VHH immune library and screening by ELISA, thereby obtaining a high affinity heavy chain antibody against CLDN 18.2. The method comprises the following steps:
1.1 construction of phage nanobody library
Immunizing alpaca by CHO-Claudin 18.2 cells expressing CLDN18.2, and extracting peripheral blood after ELISA detection of serum titer; separating lymphocytes to obtain peripheral blood mononuclear lymphocyte precipitate, extracting total RNA, performing reverse transcription by taking the RNA as a template to obtain first-strand cDNA, and amplifying VHH genes by nest PCR; inserting VHH genes into pMECS phage display vectors, electrically transforming TG1 competent cells, taking bacterial liquid for library identification, uniformly coating all the rest cultures on an LB/AMPGLU plate, collecting lawn after bacteria grow out, adding 1/3 volume of 50% glycerol, uniformly mixing and packaging, and preserving at-80 ℃, thus successfully constructing phage display camel VHH immune library with a stock capacity of more than 10 9.
1.2 Panning and amplification of phage nanobody libraries
293T cells overexpressing the CLDN 18.2 protein (293T-CLDN 18.2) were blocked with 500. Mu.L of 3% BSA.
Phage display camel VHH immune library constructed in step 1.1 was diluted with 5% serum-PBS and 3% BSA and blocked.
Finally, incubating the blocked 293T-CLDN 18.2 with a blocked phage library diluent, centrifuging to remove unbound phage, and panning; collecting specifically bound phage, and performing the next round of affinity panning; the panned phage library is amplified in culture for the next round of panning or analysis.
1.3 Identification and analysis of specific phage clones
Dripping phage panned by the library in the step 1.2 on a flat plate, randomly picking a plurality of monoclonals by using a sterilized toothpick, inoculating the monoclonals into a 2YT culture medium, and carrying out cell: phage=1: 20 was added to M13K07 phage and incubated overnight, and the supernatant was used to identify the monoclonal by cell ELISA. Positive clones were sequenced.
6 Candidate antibodies were co-screened and designated CLDN18.2-a93, CLDN18.2-a100, CLDN18.2-a102, CLDN18.2-a127, CLDN18.2-B123 and CLDN18.2-B142.
The amino acid sequences of the 6 candidate antibodies CLDN18.2-A93, CLDN18.2-A100, CLDN18.2-A102, CLDN18.2-A127, CLDN18.2-B123 and CLDN18.2-B142 are the sequences shown as SEQ ID No.38, SEQ ID No.39, SEQ ID No.40, SEQ ID No.41, SEQ ID No.42 and SEQ ID No.43 respectively.
The nucleotide sequences of the 6 candidate antibodies CLDN18.2-A93, CLDN18.2-A100, CLDN18.2-A102, CLDN18.2-A127, CLDN18.2-B123 and CLDN18.2-B142 are the sequences shown as SEQ ID No.44, SEQ ID No.45, SEQ ID No.46, SEQ ID No.47, SEQ ID No.48 and SEQ ID No.49, respectively.
Example 2
In this example, VHH Fc nanobody expression, purification, and antibody affinity measurement were performed on the 6 candidate antibodies obtained in example 1. The method comprises the following steps:
2.1VHH Fc nanobody expression and purification
The 6 candidate antibodies obtained in example 1 were cloned into eukaryotic expression plasmid PTT5-Human-FC with Human Fc tag, expressed in 293T cells and purified using Protein A/G affinity chromatography. After purification 6 anti-CLDN 18.2 VHH antibodies with human Fc tags were obtained, including anti-CLDN 18.2-a93 VHH Fc antibody, anti-CLDN 18.2-a100 VHH Fc antibody, anti-CLDN 18.2-a102 VHH Fc antibody, anti-CLDN 18.2-a127 VHH Fc antibody, anti-CLDN 18.2-B123 VHH Fc antibody, anti-CLDN 18.2-B142 VHH Fc antibody, absorbance of OD280 was detected by an enzyme-labeled instrument and concentration was calculated, purity and molecular weight were detected by SDS-PAGE gel.
The results of mass detection of the 6 human Fc tagged CLDN18.2 VHH antibodies are shown in table 1.
TABLE 1 protein expression levels and other related information
Protein name Concentration (mg/mL) Reduced molecular weight Kda Non-reducing molecular weight Kda Purity (SDS-PAGE)
CLDN18.2-A93-Fc 5.28 39Kda 78Kda >95%
CLDN18.2-A100-Fc 4.92 38Kda 76Kda >95%
CLDN18.2-A102-Fc 1.3 39Kda 78Kda >95%
CLDN18.2-A127-Fc 0.83 39Kda 78Kda >95%
CLDN18.2-B123-Fc 4.59 39Kda 78Kda >95%
CLDN18.2-B142-Fc 3.47 39Kda 78Kda >95%
2.2 Antibody affinity assay
The affinity of the 6 anti-CLDN 18.2 VHH antibodies with human Fc tag obtained in step 2.1 was determined by Biacore.
Biacore is a bioanalytical sensing technology developed based on surface plasmon resonance (surface Plasmon resonance, SPR), and can detect and track the whole change process of the combination and dissociation of molecules in a solution and molecules fixed on the surface of a chip, record the whole change process in the form of a sensor graph, provide kinetic and affinity data, solidify an anti-CLDN 18.2 VHH antibody purified in the step 2.1 on the surface of the chip in the measuring process, and obtain a mobile phase which is a solution containing human Claudin-18.2 protein, wherein the measuring results are shown in Table 2 and FIGS. 1A to 1F.
Table 26 results of affinity test of human Fc tagged anti-CLDN18.2VHH antibodies with human Claudin-18.2 protein
Antibodies to ka(1/Ms) kd(1/s) KD(M)
CLDN18.2-A93-Fc 9.59E+05 4.99E-04 5.21E-10
CLDN18.2-A100-Fc 4.97E+05 6.41E-04 1.29E-09
CLDN18.2-A102-Fc 8.30E+05 4.86E-04 5.86E-10
CLDN18.2-A127-Fc 1.29E+06 5.82E-04 4.53E-10
CLDN18.2-B123-Fc 4.69E+05 6.39E-04 1.36E-09
CLDN18.2-B142-Fc 1.28E+06 4.45E-04 3.48E-10
K a is the binding constant, in contrast to Kd, the greater the value the stronger the affinity.
K d is a dissociation constant, indicating how fast the dissociation is, with larger K d representing faster dissociation and smaller K d representing slower dissociation.
KD is an equilibrium dissociation constant, KD represents the dissociation degree in an equilibrium state, and the larger KD indicates more dissociation and represents weaker affinity; smaller KD indicates less dissociation, representing stronger affinity.
Equilibrium dissociation constant K D =kd/Ka
As can be seen from table 2 and figures 1A to 1F, the affinity of these 6 anti-CLDN 18.2VHH antibodies with human Fc tags all reached nM level.
Example 3
In this example 3, a flow assay was performed against the heavy chain antibody of CLDN 18.2.
293T cells (293T-claudin 18.2-GFP, preparation details see example 7) or 293T cells (293T-claudin 18.1-GFP, preparation details see example 7) stably overexpressing the CLDN18.2 protein were incubated with the purified 6 anti-CLDN 18.2 VHH antibodies (anti-CLDN 18.2-A93 VHH Fc antibody, anti-CLDN 18.2-A100 VHH Fc antibody, anti-CLDN 18.2-A102VHH Fc antibody, anti-CLDN 18.2-A127 VHH Fc antibody, anti-CLDN 18.2-B123 VHH Fc antibody and anti-CLDN 18.2-B142 VHH Fc antibody) obtained in step 2.1 of example 2, respectively, for 30min under an ice bath, and then incubated with the APC-labeled goat anti-mouse IgG antibody, and the results were detected by flow cytometry (FACS) as shown in FIGS. 2A and 2B.
A blank group was also established, with wild-type 293T cells as the blank group.
As can be seen from fig. 2A and 2B, it is shown that the heavy chain antibody against CLDN18.2 of the present application is capable of specifically recognizing CLDN18.2 antigen on the cell surface, but not recognizing CLDN18.1 antigen on the cell surface.
Example 4
In this example 4, lentiviral vectors were prepared that expressed chimeric antigen receptor of heavy chain antibody against CLDN18.2 VHH.
A schematic representation of the chimeric antigen receptor is shown in FIG. 4, comprising a promoter, signal peptide, anti-CLDN18.2 VHH, CD8 alpha hinge region, transmembrane region and immune receptor tyrosine activation motif.
The amino acid sequence of the CD8 a signal peptide is: MALPVTALLLPLALLLHAARP (SEQ ID No. 50).
The amino acid sequences of the CD 8a hinge region and the transmembrane region are:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLL LSLVITLYC(SEQ ID No.51)。
the amino acid sequence of the immunoreceptor tyrosine activation motif is:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQE GLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID No.52).
4.1 construction of lentiviral vectors
The anti-CLDN 18.2-a93 VHH Fc antibody, anti-CLDN 18.2-a100 VHH Fc antibody, anti-CLDN 18.2-a102 VHH Fc antibody, anti-CLDN 18.2-a127 VHH Fc antibody, anti-CLDN 18.2-B123 VHH Fc antibody and anti-CLDN 18.2-B142 VHH Fc antibody obtained in example 2 were used, respectively, 6 anti-CLDN 18.2 VHH Fc antibody fragments (abbreviated as antibody fragments) containing CD8a signal peptide were synthesized using primers corresponding to table 3, and then the antibody fragments were ligated with the CD8a range-TM-41 BB-CD3Z fragment and the digested sinhd 03 CD19 CAR-KanaR plasmid, respectively, to obtain 6 chronic viral vectors including HD SIN03 A93 41BBz、SIN03 A100 41BBz、SIN03 A102 41BBz、SIN03 A12741BBz、SIN03 B123 41BBz、SIN03 B142 41BBz.
The maps of lentiviral vector HD SIN03 A93 41BBz、SIN03 A100 41BBz、SIN03 A102 41BBz、SIN03 A12741BBz、SIN03 B123 41BBz、SIN03 B142 41BBz are shown in FIGS. 3A, 3B, 3C, 3D, 3E, and 3F, respectively.
TABLE 3 Table 3
The specific construction method comprises the following steps:
4.1.1 preparation of fragment CD8a Signal-VHH
The primers corresponding to the antibodies in Table 3 (see Table 3) were used to obtain 6 PCR amplified fragments by PCR amplification using the nucleotide sequences of the 6 human Fc tagged anti-CLDN18.2VHH antibodies synthesized in example 2 as templates. A PCR reaction system was prepared according to Table 4, and a PCR reaction was performed according to the procedure of Table 7.
And (3) 1% agarose electrophoresis, and respectively recovering 6 PCR amplified fragments by ZymocleanTM Gel DNA Recovery kit, wherein the length of each PCR amplified fragment is about 450bp, so as to obtain 6 anti-CLDN 18.2 VHH antibody fragments containing CD8 alpha signal peptide.
TABLE 4PCR reaction System
4.1.2 Preparation of fragment CD8a range-TM-41 BB-CD3Z
PCR amplification was performed using two primers, CD8aH-F and Vector-R (see Table 5), and the HD SIN03 CD19 CAR-Kanar plasmid (plasmid map see FIG. 9) as a template. A PCR reaction system was prepared according to Table 6, and a PCR reaction was performed according to the procedure of Table 7. Wherein the HD SIN03 CD19 CAR-Kanar plasmid is based on the 3G11-BBz plasmid (patent application No. 201710357213.3), the ampicillin resistance gene is replaced by the kana resistance gene, and the 5' LTR fragment is truncated to become a viral vector containing a terminal self-inactivating (SIN) structure (plasmid map is shown in FIG. 9).
1% Agarose electrophoresis, recovering fragment with ZymocleanTM Gel DNA Recovery kit bp length, obtaining CD8a range-TM-41 BB-CD3Z fragment.
TABLE 5
TABLE 6PCR reaction System
TABLE 7
4.1.3 Preparation of the digestion vector
The HD SIN03 CD19 CAR-Kanar plasmid (plasmid map, see FIG. 9) was digested with BamHI-HF and EcoRI-HF, and the vector was recovered by 1% agarose electrophoresis ZymocleanTM Gel DNA Recovery kit to a length of 7710bp. The cleavage reaction system is shown in Table 8.
TABLE 8
4.2 Recombination and transformation
4.2.1 The recovery vector of step 4.1.3 in this example and the CD8a signal-VHH fragment of step 4.1.1 in this example and the CD8a range-TM-41 BB-CD3Z fragment of step 4.1.2 in this example were subjected to a recombination reaction with ClonExpress II one step cloning kit, reaction conditions: the recombinant product is obtained at 37 ℃ for 30 min. The recombination reaction system is shown in Table 9.
TABLE 9
Contents Volume(μL)
Step 4.1.3 HD SIN03 CD19 CAR-Kanar 200ng
Step 4.1.1 CD8a Signal-VHH fragment 18ng
Step 4.1.2 CD8a range-TM-41 BB-CD3Z fragment 28ng
5×Buffer 4μL
Recombinant enzyme 2μL
Total volume Add ddH2O to 20μL
4.2.2 Using 10. Mu.L of the recombinant product obtained in step 4.2.1 of the present example, the recombinant product was thermally transformed into E.coli stbl3 competent (100. Mu.L), and positive monoclonal strains were selected by kana resistance plate for the next sequencing.
4.3PCR identification and sequencing identification
LV-F2 and LV-R are used as primers (see table 12 for details) and are prepared according to the reaction system of table 10, then PCR reaction is carried out according to the procedure of table 11 to obtain PCR products, and then 1% agarose electrophoresis and EB staining identification are carried out, wherein the length of the PCR products is 1200-1300bp.
Table 10
After preparing a PCR reaction system according to Table 10, PCR was performed according to the procedure of Table 11.
TABLE 11
After the PCR is finished, selecting and identifying correct clones, sending the clones to a company for sequencing (sequencing primers are LV-F2 and LV-R, and the primers are shown in table 12), and finally selecting correct clones, extracting plasmids by using a plasmid large extraction kit, and positive clones are constructed lentiviral vectors.
Table 12
Primer(s) Sequence(s) Sequence numbering
LV-F2 tcttggttcattctcaagcctc SEQ ID No.67
LV-R gcaacatagttaagaatacc SEQ ID No.68
Example 5
In this example, the lentiviral vector obtained in example 4 was subjected to lentiviral packaging comprising the steps of:
(1) 293T cells were inoculated in a 15cm dish at 1.6X10 7 cells and cultured overnight at 37℃in 5% CO 2 to prepare packaging virus, the medium was DMEM-containing, 10% fetal bovine serum (fetal bovine serum, FBS) was added;
(2) Respectively dissolving 14.5 mug of the lentiviral vector obtained in example 4, 16.7 mug of helper plasmid pMDlg-RRE, 16.7 mug of helper plasmid pRSV-REV and 6.5 mug of envelope plasmid VSVg into 2mL of serum-free DMEM culture solution, and uniformly mixing;
(3) 163.2. Mu.g PEI (1. Mu.g/. Mu.L) was dissolved in 2mL serum-free DMEM medium, gently mixed (or vortexed at 1000rpm for 5 seconds), and incubated at room temperature for 5min to give a PEI mixture;
(4) Formation of transfection complexes: adding the PEI mixed solution obtained in the step (3) into the DNA mixed solution, immediately mixing by vortex or gently mixing, and incubating for 20min at room temperature to obtain a transfection complex;
(5) 4mL of the transfection complex obtained in the step (4) was added dropwise to a 15cm dish containing 25mL of DMEM medium, after 4-5 hours, the DMEM medium containing 2% FCS was changed, and after 48 hours of culture, the virus supernatant was collected.
Example 6
In this example, the packaged lentivirus obtained in example 5 was subjected to lentivirus concentration comprising the steps of:
Filtering the virus supernatant obtained in the step (5) in the example 5 by using a 0.45 mu m filter membrane, collecting the filtered virus supernatant into a 50mL centrifuge tube, adding 1/4 PEG-NaCl virus concentrate, mixing the mixture upside down, and standing the mixture at 4 ℃ overnight; then centrifuging at 4 ℃ and 3500rpm for 30min; removing the supernatant, adding RPMI 1640 medium (containing 10% FBS) into the virus pellet to dissolve the resuspended virus pellet; the concentrated lentiviral suspension was split into 50. Mu.L portions each, stored in finished tubes and stored at-80 ℃.
Example 7
In this example, the construction of the overexpressing cell line includes the following:
Claudin 18.2-GFP lentivirus acquisition: 14.5. Mu.g of claudin 18.2-GFP plasmid (purchased from Crohn's organism), 16.7. Mu.g of helper plasmid pMDlg-RRE, 16.7. Mu.g of helper plasmid pRSV-REV and 6.5. Mu.g of envelope plasmid VSVg were co-transfected and obtained in the same manner as in example 5.
Claudin 18.1-GFP lentivirus acquisition: 14.5. Mu.g of claudin 18.1-GFP plasmid (purchased from Crohn's organism), 16.7. Mu.g of helper plasmid pMDlg-RRE, 16.7. Mu.g of helper plasmid pRSV-REV and 6.5. Mu.g of envelope plasmid VSVg were co-transfected and obtained in the same manner as in example 5.
Obtaining 293T cells overexpressing the CLDN18.2 protein (abbreviated as 293T-CLDN 18.2): 293T cells were seeded at 1X 10 6 cells/well in 6-well plates and 1mL of claudin 18.2-GFP lentivirus was added to obtain.
Obtaining 293T cells overexpressing the CLDN18.1 protein (abbreviated as 293T-CLDN 18.1): 293T cells were seeded at 1X 10 6 cells/well in 6-well plates and 1mL of claudin 18.1-GFP lentivirus was added to obtain.
Acquisition of HGC27 cells overexpressing the CLDN18.2 protein (HGC-27-18.2-GFP cells): HGC-27 cells were seeded at 1X 10 6 cells/well in 6-well plates, and 1mL of claudin 18.2-GFP lentivirus was added to obtain.
Obtaining of N87 cells (N87-18.2-GFP) overexpressing the CLDN18.2 protein: NCI-N87 cells were seeded at 1X 10 6 cells/well in 6-well plates and 1mL of claudin 18.2-GFP lentivirus was added to obtain.
Example 8
In this example, lentivirus titer detection was performed on the concentrated virus obtained in example 6, comprising the steps of:
Cells were seeded at 500 μ L Junkat cells (1×10 5 cells) in 24 well plates to form a cell suspension; the concentrated lentiviruses obtained in example 6 were added to the cell suspension at 1. Mu.L, 0.2. Mu.L and 0.04. Mu.L, respectively, and polybrene was added to a final concentration of 5. Mu.g/mL; then after overnight incubation at 37 ℃ with 5% co 2, fresh medium was changed; after 72h of infection, cells were collected, centrifuged at 400g for 5min, the supernatant was discarded, and the flow buffer (PBS+2% neonatal calf serum) was washed once; adding MonoRab TM Rabbit Anti-CAMELID VHH Cocktail [ iFluor 488] antibody according to a dilution ratio of 1:100, and incubating on ice for 30min; then, after washing twice with 1mL of flow buffer, detecting by using a flow cytometer; taking a cell sample with a positive rate of 5-20%, and calculating the titer.
Titer (TU/mL) =cell number (10 5) ×positive rate/virus volume (mL).
Example 9
In this example, T lymphocytes were transduced with lentiviruses of example 8 titers satisfactory.
9.1 Resuscitation of T lymphocytes
Resuscitated PBMC cells were centrifuged at 200g for 9min, the supernatant discarded, the pellet resuspended in T cell medium, counted, density adjusted to 1.5-2X 10 6 cells/mL, and inoculated into 48 well cell culture plates, allowed to stand for 2h for activation.
9.2 Activation of T lymphocytes
PBMC were density-adjusted to 2X 10 6 cells/mL with T cell medium (X-VIVO+10% FBS+IL-2 (300U/mL), T CELL TRANSACT was added at 1:100, and inoculated into a suitable culture vessel for activation for 24-48h while satisfying the requirement of maintaining the cell density at 2X 10 6 cells/mL, to obtain activated T cells.
9.3T cell infection
Collecting the activated T cells obtained in the step 9.2, adjusting the cell density to 1X 10 6/mL, adding the slow virus obtained in the example 8 according to the multiplicity of infection MOI=10, and adding polybrene to a final concentration of 5 mug/mL; after overnight incubation at 37℃in 5% CO 2, fresh medium was changed. Passaging was performed every 2-3 days.
9.4 Chimeric antigen receptor expression
In this example, after 5 days of infection in step 9.3, 3X 10 5 T cells were collected, centrifuged at 400g at 4℃for 5min, and the supernatant was discarded and washed once with streaming buffer (PBS+2% rabbit serum); monoRab TM Rabbit Anti-CAMELID VHH Cocktail (iFluor 488) antibody was added at a dilution ratio of 1:100 and incubated on ice for 30min; then, after washing twice with 1mL of the flow buffer, the T lymphocyte chimeric antigen receptor infection efficiency was examined by flow cytometry, and the results are shown in FIG. 5.
As can be seen from fig. 5, the CAR-T cells after infection had a significant positive population, indicating that 6 CAR-T cells expressing chimeric antigen receptors of different structures were successfully constructed, the antigen binding domains of which contained CLDN18.2-a93, CLDN18.2-a100, CLDN18.2-a102, CLDN18.2-a127, CLDN18.2-B123 and CLDN18.2-B142 antibodies, respectively.
The CAR-T cell markers corresponding to CLDN18.2-a93, CLDN18.2-a100, CLDN18.2-a102, CLDN18.2-a127, CLDN18.2-B123 and CLDN18.2-B142 antibodies were CLDN18.2 (a 93), CLDN18.2 (a 100), CLDN18.2 (a 102), CLDN18.2 (a 127), CLDN18.2 (B123) and CLDN18.2 (B142), respectively.
Example 10
In this example, an in vitro toxicity assay was performed using the 6 CAR-T cells prepared in example 9, comprising the steps of:
10.1 target cell seeding
293T cells (claudin 18.2-), HGC-27-18.2-GFP cells (claudin 18.2+), N87-18.2-GFP cells (claudin 18.2+) were taken as target cells, centrifuged at 400g for 5min, resuspended in 1640 medium of 5% FBS, counted and the target cell concentration was adjusted to 1X 10 5/mL.
Among them, HGC-27-18.2-GFP cells (claudin 18.2+) and N87-18.2-GFP cells (claudin 18.2+) were prepared as described in example 7.
10.2 Effector cell seeding
6 CAR-T cells (CLDN 18.2 (a 93), CLDN18.2 (a 100), CLDN18.2 (a 102), CLDN18.2 (a 127), CLDN18.2 (B123) and CLDN18.2 (B142)) prepared in example 9 and control T cells (not lentivirally infected) were used as effector cells; effector cells 400g were centrifuged for 5min, resuspended in 1640 medium of 5% FBS, counted, and cell density adjusted to the desired concentration a (modulated CAR-T density = 3 x 10 5 cells/mL/infection efficiency);
10.3 dilution of effector cell ratios
3-Fold dilution of CAR-T cells at a concentration to car+ T cells at B concentration, 3-fold dilution of CAR-T cells at B concentration to CAR-T cells at C concentration;
10.4 effector cells and target cells were added to 96-well plates containing medium at an effective target ratio of 0.3:1, 1:1 and 3:1 (appropriate 96-well plates were selected according to target cells), 3 multiple wells per group, total volume per well 200 μl;
After incubation at 37 ℃ for 18h, 20 μl of lysate (10×) was added to the wells; the cell lysis condition is not observed regularly, so that the cell lysis is ensured to be complete;
10.6 preparing a substrate, adding 12mL of assay buffer (lactic acid) into a substrate mix (tetrazolium salt)/bottle, uniformly mixing and keeping away from light (about the dosage of two 96-well plates) to obtain a substrate solution;
10.7 all wells treated in step 10.5 were sucked 6-8 times with a row gun and centrifuged at 500g for 5min, and 50. Mu.L/well supernatant was taken into a new ELISA plate (flat bottom 96 well plate).
10.8 Adding the substrate solution prepared in the step 10.6 into the new ELISA plate prepared in the step 10.7 at a concentration of 50 mu L/hole, covering the plate, and incubating for about 30min at room temperature in a dark place;
10.9 add 50. Mu.L stop solution (acetic acid) per well and measure absorbance at 490nm over 30 min.
Data were derived and plotted analytically. The cytotoxicity calculation formula is (all minus blank medium control):
FIGS. 6A, 6B and 6C are graphs showing the killing effect of constructed CAR-T cells containing anti-CLDN 18.2 heavy chain antibody on 293T cells, HGC-27-18.2-GFP cells and N87-18.2-GFP cells, respectively.
From fig. 6A, 6B and 6C, it can be seen that the CAR-T cells constructed according to the present application (CLDN 18.2 (a 93), CLDN18.2 (a 100), CLDN18.2 (a 102), CLDN18.2 (a 127), CLDN18.2 (B123) and CLDN18.2 (B142)) have high killing activity against CLDN18.2 positive tumor cells (including HGC-27-18.2-GFP (claudin 18.2+) and N87-18.2-GFP (claudin 18.2+)), and have no killing effect against CLDN18.2 negative cells (293T (claudin 18.2-)), indicating that the constructed CAR-T cells have high specificity.
Example 11
In this example, the secretion of CAR-T cytokines was detected, including the following:
11.1 cell culture supernatant
The experiments were divided into three groups, one group of effector cells from example 9 were cultured alone at 37℃for 18h, one group of effector cells from example 9 and 293T cells were co-cultured at 37℃for 18h, one group of effector cells from example 9 and CLDN 18.2 positive 293T cells (293T-CLDN 18.2) were co-cultured at 37℃for 18h, with an effective target ratio of 1:1. While setting up Control T group (no lentiviral infection). CLDN 18.2 positive 293T cells (293T-CLDN 18.2) were obtained as detailed in example 7.
After the completion of the culture, the cell culture was centrifuged at 400 Xg for 10min to remove the precipitate, and the supernatant was stored at-80℃for examination.
11.2 Detection of IFN- α and IFN- γ
11.2.1 Reagents: the detection was carried out using a Union ELISA kit (product numbers: human gamma interferon ELISA kit: EK180-96; human tumor necrosis factor alpha (TNF-alpha) ELISA kit: EK 182-96), all reagents and samples were returned to 25℃before detection, and 1 Xwash solution, 1 Xdetection buffer solution, and detection antibody were prepared according to the instructions of use.
11.2.2 Standard and sample preparation standard: standard stock was 2-fold diluted with 5%1640 medium for a total of 8 dilution gradients, including zero concentration.
Sample: samples were diluted in ratio using 5%1640 medium.
11.2.3 Detection step
(1) Soaking the ELISA plate: adding 300 mu L of 1 Xwashing liquid, standing and soaking for 30s, removing the washing liquid, and then beating the micro-porous plate on water-absorbing paper;
(2) Adding a standard substance: standard wells were filled with 100 μl of 2-fold diluted standard and blank wells were filled with 100 μl of 5%1640 medium;
(3) Adding a sample: sample wells were added with 100 μl of the cell culture supernatant obtained in step 11.1 of this example;
(4) Adding a detection antibody: 50. Mu.L of diluted detection antibody (1:100 dilution) was added to each well;
(5) Incubation: sealing plates by using sealing plates, vibrating at 300rpm, and incubating at 25 ℃ for 2 hours;
(6) Washing: liquid was discarded, and 300. Mu.L of wash solution was added to wash the plate 6 times per well;
(7) And (3) enzyme adding and incubation: mu.L of diluted horseradish peroxidase-labeled streptavidin (1:100 dilution) was added to each well;
(8) Incubation: using a new sealing plate membrane sealing plate, oscillating at 300rpm, and incubating at 25 ℃ for 45min;
(9) Washing: repeating step (6);
(10) And (3) color development of the substrate: 100 mu L of chromogenic substrate TMB is added into each hole, and incubated for 15min at 25 ℃ in the dark;
(11) Adding a stop solution: adding 100 mu L of stop solution into each hole, and fully and uniformly mixing;
(12) Detecting and reading: the dual wavelength detection was performed using an enzyme-labeled instrument, and the OD at the 450nm maximum absorption wavelength and the 630nm reference wavelength was measured, and the OD after calibration was measured at 450nm minus the measured at 630 nm. The results are shown in FIGS. 7 and 8.
The IFN-alpha secretion results are shown in FIG. 7, where spontaneous is CAR-T cell culture alone, trace IFN-alpha was detected in CAR-T cell culture alone, as was trace IFN-alpha detected in CAR-T cell co-culture with 18.2 negative 293T cells, and higher levels of IFN-alpha were detected in CAR-T cell co-culture with 18.2 positive 293T-CLDN 18.2 cells (claudin 18.2+).
The results of FN-gamma factor secretion are shown in FIG. 8, where spontaneous CAR-T cell cultures alone, where almost no IFN-gamma factor was detected, as was the case in co-cultures of CAR-T cells and 18.2 negative 293T cells, whereas higher levels of IFN-gamma factor were detected in co-cultures of CAR-T cells and 18.2 positive 293T-CLDN 18.2 cells (claudin 18.2+).
From the comprehensive results of fig. 7 and 8, it is shown that the CAR-T cell constructed by the application can release cytokines to CLDN 18.2 positive tumor cells to perform a killing function, has high specificity, and has no cytokine secretion to CLDN 18.2 negative cells.
In summary, according to the application, by means of transient transfection, a llama is immunized by using a CLDN18.2 recombinant protein, a phage display nanobody library is constructed, and screening is carried out on anti-CLDN 18.2 antibodies according to the phage display nanobody library, so that the heavy chain antibody of the anti-CLDN 18.2 with the CDR of the heavy chain variable region as shown in SEQ ID NO. 38-43 is obtained, and the determination of Ka, kd and KD of antibody affinity proves that the heavy chain antibody of the anti-CLDN 18.2 can specifically bind to the CLDN18.2 antigen and has high affinity. The heavy chain antibody against CLDN18.2 of the present application specifically recognizes CLDN18.2 but does not recognize CLDN18.1. The heavy chain antibody for resisting CLDN18.2 provided by the application has better affinity, a chimeric antigen receptor is constructed by taking the heavy chain antibody as an antigen binding domain, the chimeric antigen receptor is utilized to prepare CAR-T cells, the constructed CAR-T cells have killing activity on CLDN18.2 positive tumor cells, and after the constructed CAR-T cells are co-cultured with the CLDN18.2 positive cells, cytokines TNF-alpha and IFN-gamma are secreted efficiently, so that the nano antibody, the chimeric antigen receptor constructed by the nano antibody and the CAR-T cells can improve the killing capacity on tumor cells, especially stomach cancer.
The above embodiments are merely illustrative of the principles of the present application and its effectiveness, and are not intended to limit the application. Modifications and variations may be made to the above-described embodiments by those skilled in the art without departing from the spirit and scope of the application. Accordingly, it is intended that all equivalent modifications and variations of the application be covered by the claims, which are within the ordinary skill of the art, be within the spirit and scope of the present disclosure.

Claims (12)

1. An anti-CLDN 18.2 heavy chain antibody comprising a heavy chain variable region, wherein the amino acid sequence of CDR1 of said heavy chain variable region comprises the sequence set forth in SEQ ID No.3, the amino acid sequence of CDR2 of said heavy chain variable region comprises the sequence set forth in SEQ ID No.9, and the amino acid sequence of CDR3 of said heavy chain variable region comprises the sequence set forth in SEQ ID No. 15.
2. The heavy chain antibody of claim 1, wherein the heavy chain variable region further comprises framework regions FR1-FR4, the amino acid sequence of FR1 of the framework regions comprises the sequence set forth in SEQ ID No.21, the amino acid sequence of FR2 of the framework regions comprises the sequence set forth in SEQ ID No.26, the amino acid sequence of FR3 of the framework regions comprises the sequence set forth in SEQ ID No.32, and the amino acid sequence of FR4 of the framework regions comprises the sequence set forth in SEQ ID No. 37.
3. The heavy chain antibody of claim 1, wherein the amino acid sequence of the heavy chain variable region comprises the amino acid sequence set forth in SEQ id No. 40.
4. An isolated polypeptide, wherein the polypeptide is a chimeric antigen receptor, and wherein the polypeptide comprises, in order from N-terminus to C-terminus, a CD8 a signal peptide, an antigen binding domain comprising the heavy chain antibody of any one of claims 1-3, a CD8 a hinge region, a CD8 a transmembrane domain, and an immunoreceptor tyrosine activation motif.
5. A biological material associated with the heavy chain antibody of any one of claims 1-3 or the polypeptide of claim 4, wherein the biological material comprises one or more of the following:
1) A nucleotide encoding the heavy chain antibody of any one of claims 1-3 or a nucleotide encoding the polypeptide of claim 4;
2) A recombinant expression vector comprising 1) said nucleotide;
3) The bioengineering bacteria containing 1) the nucleotide or the bioengineering bacteria containing 2) the recombinant expression vector.
6. The biomaterial of claim 5, wherein in 1) the sequence of nucleotides comprises a sequence as set forth in SEQ ID No. 46;
and/or 2) the recombinant expression vector is selected from a lentiviral vector, a retroviral vector, or an adenoviral vector.
7. The biomaterial of claim 6, wherein the recombinant expression vector is a lentiviral vector.
8. A cell expressing the polypeptide of claim 4.
9. The cell of claim 8, wherein the cell is selected from one or more of T lymphocytes, B lymphocytes, NK cells, and macrophages.
10. Use of the heavy chain antibody of any one of claims 1-3 or the polypeptide of claim 4 or the biological material of claims 5-7 or the cell of claim 8 or 9 for the preparation of an in vitro test product, a kit, chip or membrane strip for the detection of CLND 18.2, for the preparation of a medicament for the prevention or treatment of a tumor, which tumor is gastric cancer.
11. An assay product for the in vitro assay of CLND 18.2 comprising a heavy chain antibody according to any one of claims 1 to 3 or a polypeptide according to claim 4 or a biological material according to claims 5 to 7 or a cell according to claim 8 or 9.
12. A pharmaceutical composition for preventing or treating a tumor disease associated with expression of CLND 18.2, characterized in that,
Comprising the heavy chain antibody of any one of claims 1-3 or the polypeptide of claim 4 or the biological material of claims 5-7 or the cell of claim 8 or 9, and a pharmaceutically acceptable carrier, wherein the neoplastic disease associated with expression of CLND 18.2 is gastric cancer.
CN202410255907.6A 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses Pending CN117924500A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410255907.6A CN117924500A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211525518.8A CN116253800B (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses
CN202410255907.6A CN117924500A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202211525518.8A Division CN116253800B (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Publications (1)

Publication Number Publication Date
CN117924500A true CN117924500A (en) 2024-04-26

Family

ID=86686941

Family Applications (4)

Application Number Title Priority Date Filing Date
CN202410255922.0A Pending CN117924501A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses
CN202410255849.7A Pending CN117903310A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses
CN202410255907.6A Pending CN117924500A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses
CN202211525518.8A Active CN116253800B (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN202410255922.0A Pending CN117924501A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses
CN202410255849.7A Pending CN117903310A (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202211525518.8A Active CN116253800B (en) 2022-11-30 2022-11-30 Heavy chain antibody against CLDN18.2, related products and uses

Country Status (1)

Country Link
CN (4) CN117924501A (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109762067B (en) * 2019-01-17 2020-02-28 北京天广实生物技术股份有限公司 Antibodies that bind human Claudin18.2 and uses thereof
CN114761433A (en) * 2019-12-11 2022-07-15 上海复宏汉霖生物技术股份有限公司 anti-Claudin18.2 monoclonal antibody, preparation method and application thereof
CN114685670A (en) * 2020-12-31 2022-07-01 上海莱馥医疗科技有限公司 CLDN18.2 antibodies and uses thereof
CN113788894B (en) * 2021-09-03 2022-08-05 深圳市先康达生命科学有限公司 Monoclonal antibody targeting human Claudin18.2 protein and application thereof

Also Published As

Publication number Publication date
CN116253800A (en) 2023-06-13
CN116253800B (en) 2024-04-12
CN117924501A (en) 2024-04-26
CN117903310A (en) 2024-04-19

Similar Documents

Publication Publication Date Title
CN109942709B (en) anti-BCMA single domain antibody and application thereof
CN112812186B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
KR20210050535A (en) Anti-BCMA single domain antibody and its application
CN110655581B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN110713539B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN114276454B (en) Anti-mesothelin nano antibody and application thereof
CN112812185B (en) Monoclonal antibody for resisting B cell maturation antigen and application thereof
CN113265001B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN114957484A (en) CAR vector targeting solid tumor cell B7-H3 protein, CAR-T cell and construction method and application thereof
CN110862456A (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN112830971B (en) anti-BCMA antigen binding fragment and application thereof
CN116751303B (en) Nanometer antibody for resisting guanylate cyclase 2C and application thereof
CN114149506B (en) anti-CD 22 antibody and application thereof
CN116253800B (en) Heavy chain antibody against CLDN18.2, related products and uses
CN112979808B (en) Antibody for resisting B cell mature antigen and application thereof
CN118005794A (en) Heavy chain antibody against CLDN18.2, related products and uses
CN107216395A (en) A kind of TM4SF1 specific chimerics antigen receptor and its application
WO2023125813A1 (en) Anti-mesothelin nanobody chimeric antigen receptor and use thereof
CN117567627A (en) anti-PSMA heavy chain antibody, related products and uses
CN114805581B (en) Antibodies targeting IL13RA2, chimeric antigen receptors and uses thereof
CN116410323A (en) anti-GPC 3 antibody and application thereof
CN116675773A (en) anti-MXR 7 antibody and application thereof
CN116693688A (en) anti-Intestinal protein OCI-5 antibody and application thereof
CN116640217A (en) anti-GTR 2-2 antibody and application thereof
CN117024594A (en) anti-CD 20 nano antibody and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination