CN112812186B - Nanometer antibody for resisting B cell mature antigen and application thereof - Google Patents

Nanometer antibody for resisting B cell mature antigen and application thereof Download PDF

Info

Publication number
CN112812186B
CN112812186B CN202110183479.7A CN202110183479A CN112812186B CN 112812186 B CN112812186 B CN 112812186B CN 202110183479 A CN202110183479 A CN 202110183479A CN 112812186 B CN112812186 B CN 112812186B
Authority
CN
China
Prior art keywords
seq
cells
chimeric antigen
antigen receptor
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.)
Active
Application number
CN202110183479.7A
Other languages
Chinese (zh)
Other versions
CN112812186A (en
Inventor
狄升蒙
侯莉
余学军
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 CN202110183479.7A priority Critical patent/CN112812186B/en
Publication of CN112812186A publication Critical patent/CN112812186A/en
Application granted granted Critical
Publication of CN112812186B publication Critical patent/CN112812186B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • A61K39/001116Receptors for cytokines
    • A61K39/001117Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR] or CD30
    • 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
    • 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
    • C07K14/70517CD8
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/804Blood cells [leukemia, lymphoma]
    • 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
    • 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
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • 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
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Abstract

The invention provides a nano antibody for resisting a B cell maturation antigen and application thereof. The heavy chain variable region of the nano antibody for resisting the B cell maturation antigen comprises: complementarity determining region 1 shown in SEQ ID NO.1, complementarity determining region 2 shown in SEQ ID NO.2, and complementarity determining region 3 shown in SEQ ID NO. 3. The antibody obtained by screening the phage display nano antibody library has a specific CDR (complementary deoxyribonucleic acid) region, can be specifically combined with a BCMA (brain cell antigen) and has better affinity; the antigen is used as an antigen binding domain to construct a chimeric antigen receptor and a CAR-T cell, and has obvious killing activity on BCMA positive tumor cells. Therefore, the nano antibody has wide application prospect in the aspect of immunotherapy of multiple myeloma.

Description

Nanometer antibody for resisting B cell mature antigen and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a nano antibody for resisting a B cell maturation antigen and application thereof.
Background
Multiple Myeloma (MM) is a malignant proliferative disease of plasma cells characterized by abnormal proliferation of clonal plasma cells in the bone marrow, secretion of monoclonal immunoglobulins or fragments thereof (M protein), and resulting in damage to associated organs or tissues. The common clinical manifestations are bone pain, anemia, renal insufficiency and infection. MM accounts for approximately 10% of hematological malignancies that are the second most prevalent in many countries. China has no exact epidemiological survey data of the incidence rate of MM, generally, the number of the data is estimated to be about 1/10 ten thousand, and the median survival time of the multiple myeloma is 3.5.
Current induction therapy for MM, while showing some efficacy, eventually almost all patients relapse and die. Allogeneic hematopoietic stem cell transplantation can eliminate myeloma cells, but the side effects of this therapy are very significant and only a few benefit. In addition, nanobodies also show some potential in the treatment of myeloma, but there is currently insufficient clinical data to justify. Therefore, it is of great interest to develop new methods for the effective treatment of multiple myeloma.
The concept of Chimeric antigen receptor modified T lymphocytes (CAR-T) was developed as early as 1989, but the ideal effect has not been achieved in clinical trials. Over the next two decades scientists have continually tried and optimized this until 2011, CD19(B lymphocyte antigen CD19, CD19) targeted CAR-T cells were used in clinical studies of relapsed/refractory chronic B-lymphocyte leukemia and achieved dramatic efficacy in the cure. To this end, the use of CAR-T cells for tumor therapy opens new sections.
The chimeric antigen receptor is an artificially synthesized fusion protein with a function similar to that of a T cell receptor, and mainly comprises a signal peptide, an antigen recognition region, a hinge region, a transmembrane region and an intracellular signal region. Upon binding to the target antigen, the chimeric antigen receptor forms a dimer, activates T cells by intracellular signaling molecules, secretes perforin and granzyme B, etc., to achieve killing of the target cells. Thus, CAR-T cells recognize target cells independently of MHC (MHC), thereby avoiding immune escape due to down-regulation of tumor cell MHC molecules.
In recent years, chimeric antigen receptor T lymphocytes (CAR-T) are rapidly developed, two products are approved by FDA in the United states and are on the market, and a plurality of products are approved by drug clinical trials in China.
The B cell maturation antigen (BCMA, also known as CD269), which is expressed only on the surface of mature B cells, is an important B cell biomarker. The gene encoding BCMA belongs to a member of the TNF receptor superfamily, which is preferentially expressed in mature B lymphocytes and plays an important role in B cell development and autoimmune response. The receptor specifically binds to tumor necrosis factor (ligand) superfamily member 13B (TNFSF13B/TALL-1/BAFF) and causes activation of NF-. kappa.B and MAPK 8/JNK. This receptor also binds to various members of the TRAF family, mediating signals for cell survival and proliferation. In multiple myeloma, BCMA expression increases, a proliferation promoting signal increases, and finally canceration occurs.
In multiple myeloma cells, BCMA is expressed at levels significantly higher than healthy plasma cells and is an ideal target. Therefore, the BCMA CAR-T prepared by introducing the BCMA-resistant chimeric antigen receptor gene into the T cell by a genetic engineering method can ensure that the BCMA CAR-T cell specifically recognizes and kills the BCMA-expressing multiple myeloma cells, thereby realizing the anti-tumor effect of the BCMA-expressing multiple myeloma cells, and having great clinical transformation value on a BCMA-targeted immunotherapy method.
Disclosure of Invention
In view of the problems in the prior art, the present invention aims to provide a nanobody against B cell maturation antigen and its application. The anti-BCMA monoclonal antibody has high affinity, can be used as an antigen binding domain of a chimeric antigen receptor molecule to construct CAR-T cells, and the obtained CAR-T cells have good application prospects in the aspect of tumors.
In order to achieve the purpose, the invention adopts the following technical scheme:
in a first aspect, the present invention provides a nanobody against B Cell Maturation Antigen (BCMA), the heavy chain variable region of which comprises:
(1) CDR1 comprising an amino acid sequence shown in SEQ ID No.1, wherein the sequence of SEQ ID No.1 is GYSDSNYC;
(2) CDR2 comprising the amino acid sequence shown in SEQ ID No.2, wherein the sequence of SEQ ID No.2 is INGDGVI;
(3) CDR3 comprising the amino acid sequence shown in SEQ ID No.3, the sequence of SEQ ID No.3 being AALTAGCVRYAA.
In the invention, the plasmid containing BCMA protein extracellular region gene segment is introduced into eukaryotic cell Freestyle by a transient transfection mode TM 293F cells are expressed, and the recombinant protein is used for immunizing an unimmunized bactrian camel to construct a phage display nano antibody library. Screening anti-BCMA antibodies according to the phage display nano antibody library, and when CDR regions of heavy chain variable regions of the nano antibodies are respectively amino acid sequences shown in SEQ ID No. 1-3, the obtained monoclonal antibodies can be specifically combined with the BCMA antibodies And (6) originally.
As a preferred technical scheme of the invention, the heavy chain variable region of the nano antibody comprises any one or the combination of at least two of the amino acid sequences described by SEQ ID NO.4, SEQ ID NO.5, SEQ ID NO.6 or SEQ ID NO. 7.
Preferably, the heavy chain variable region of the nanobody further comprises: framework region 1(FR1) shown by SEQ ID NO.4, framework region 2(FR2) shown by SEQ ID NO.5, framework region 3(FR3) shown by SEQ ID NO.6, and framework region 4(FR4) shown by SEQ ID NO. 7.
Preferably, the heavy chain variable region of the nanobody comprises the amino acid sequence shown in SEQ ID NO. 8.
In the invention, a BCMA immune camel VHH library is screened by utilizing a phage display technology to obtain the nano antibody shown in SEQ ID NO.8, the affinity of the nano antibody to BCMA is high, and the nano antibody has an important application prospect in the aspect of constructing a chimeric antigen receptor targeting BCMA.
Wherein, the sequences of SEQ ID No. 4-7 are shown in the following table 1:
TABLE 1
Figure BDA0002942099300000031
SEQ ID No.8:
EVQLVESGGGPVQAGGSLRLSCTASGYSDSNYCMAWFRQAPGKARQGVAFINGDGVITYTDSVKGRFTISKDNAQKTLDLQMNSLKPEDTAMYYCAALTAGCVRYAAWGQGTQVTVSS
In a second aspect, the present invention provides a nucleic acid molecule encoding the nanobody of the first aspect.
Preferably, the nucleic acid molecule comprises the nucleotide sequence shown in SEQ ID NO. 9.
SEQ ID No.9:
GAGGTGCAGCTGGTGGAAAGCGGCGGCGGCCCCGTGCAGGCCGGCGGCAGCCTGAGACTGAGCTGCACCGCCAGCGGCTACAGCGACAGCAACTACTGCATGGCCTGGTTCAGACAGGCCCCCGGCAAGGCCAGACAGGGCGTGGCCTTCATCAACGGCGACGGCGTGATCACCTACACCGACAGCGTGAAGGGCAGATTCACCATTAGCAAAGATAATGCCCAGAAAACACTGGATCTGCAGATGAATAGCCTGAAACCTGAAGATACAGCCATGTATTATTGTGCCGCCCTGACAGCCGGATGTGTGAGATATGCCGCCTGGGGACAGGGCACACAGGTGACAGTGTCTAGC。
In a third aspect, the present invention also provides a chimeric antigen receptor comprising a signal peptide, an antigen binding domain, a hinge region, a transmembrane region, and a signaling domain; the antigen binding domain is a nanobody according to the first aspect.
Preferably, the signal peptide comprises a CD8 a signal peptide.
Preferably, the hinge region comprises a CD8 a hinge region.
Preferably, the transmembrane region comprises any one of or a combination of at least two of a CD8 a transmembrane region, a CD28 transmembrane region, or a DAP10 transmembrane region.
Preferably, the signaling domain comprises an immunoreceptor tyrosine activation motif (CD3 ζ).
Preferably, the signaling domain further comprises a co-stimulatory molecule comprising any one of the 4-1BB, CD28 intracellular region, OX40, ICOS or DAP10 intracellular region or a combination of at least two thereof.
Preferably, the chimeric antigen receptor comprises a CD8 a signal peptide, the nanobody of the first aspect, a CD8 a hinge region, a CD8 a transmembrane region, and an immunoreceptor tyrosine-activation motif.
In a fourth aspect, the present invention also provides an expression vector comprising a gene encoding the chimeric antigen receptor of the third aspect.
Preferably, the expression vector is any one of a lentiviral vector, a retroviral vector or an adeno-associated viral vector containing the gene encoding the chimeric antigen receptor according to the third aspect, preferably a lentiviral vector.
In a fifth aspect, the present invention provides a recombinant lentivirus prepared from a mammalian cell transfected with the expression vector and helper plasmid of the fourth aspect.
In a sixth aspect, the present invention provides a chimeric antigen receptor immune cell expressing the chimeric antigen receptor of the third aspect.
Preferably, the chimeric antigen receptor immune cell comprises the expression vector of the fourth aspect and/or the recombinant lentivirus of the fifth aspect.
Preferably, the immune cells comprise any one of T cells, B cells, NK cells, mast cells or macrophages or a combination of at least two thereof.
In a seventh aspect, the present invention also provides a pharmaceutical composition comprising the chimeric antigen receptor immune cell of the sixth aspect.
Preferably, the pharmaceutical composition further comprises pharmaceutically acceptable excipients.
In an eighth aspect, the nanobody of the first aspect, the nucleic acid molecule of the second aspect, the chimeric antigen receptor of the third aspect, the expression vector of the fourth aspect, the recombinant lentivirus of the fifth aspect, the chimeric antigen receptor immune cell of the sixth aspect, or the pharmaceutical composition of the seventh aspect is used for preparing a tumor therapeutic drug.
Preferably, the neoplasm comprises multiple myeloma.
Compared with the prior art, the invention has at least the following beneficial effects:
(1) according to the invention, a BCMA recombinant protein is used for immunizing unimmunized bactrian camel to construct a phage display nano antibody library in a transient transfection manner, anti-BCMA antibodies are screened according to the phage display nano antibody library, when CDR regions of heavy chain variable regions of the nano antibodies are respectively amino acid sequences shown in SEQ ID No. 1-3, the obtained monoclonal antibodies can be specifically combined with BCMA antigens, the affinity is good, and the determination of the antibody affinity shows that the antibody has the advantages that the Ka (1/Ms) is 2.70E +06, the Kd (1/s) is 2.43E-04, and the KD (M) is 9.01E-11;
(2) the anti-BCMA nano antibody provided by the invention has better affinity, is used as an antigen binding structural domain to construct a chimeric antigen receptor, and utilizes the chimeric antigen receptor to prepare a T cell, and experiments verify that the constructed CAR-T cell has killing activity on BCMA positive tumor cells, and efficiently secretes cell factors IL-2, TNF-alpha and IFN-gamma after being co-cultured with the BCMA positive cells, so that the nano antibody, the chimeric antigen receptor constructed by the nano antibody and the CAR-T cell can improve the killing capacity on tumor cells, particularly multiple myeloma.
Drawings
FIG. 1 is a graph of the affinity of Biacore for detecting BCMA nanobodies in example 3.
FIG. 2 is a graph showing the results of FACS detection of BCMA antigen recognition by BCMA nanobodies in example 4.
FIG. 3 is a plasmid map of pSIN-HD BCMACAR 8# lentiviral vector in example 5.
FIG. 4 is a schematic diagram of the structure of a chimeric antigen receptor expressing BCMA in example 5.
FIG. 5 is a graph showing the results of flow-based assay of the expression rate of chimeric antigen receptor of T lymphocytes in example 7, wherein the graph I is a blank control and the graph II is a BCMA-8-containing CAR T cells.
FIG. 6 is a graph of the results obtained by FACS detection of T cells and BCMA CAR-T cell phenotypes in example 7.
Figure 7A is a graph of the killing effect of BCMA CAR-T cells on K562 cells in example 8.
Figure 7B is a graph of the killing effect of BCMA CAR-T cells on RPMI 8226 cells in example 8.
Figure 7C is a graph of the killing effect of BCMA CAR-T cells on U266 cells in example 8.
FIG. 8 is a histogram of IL-2 secretion by BCMACAR-T cells in example 9.
FIG. 9 is a histogram of TNF α secretion by BCMA CAR-T cells of example 9.
Figure 10 is a histogram of the secretion of IFN γ by BCMA CAR-T cells in example 9.
Detailed Description
The technical solutions of the present invention are further described in the following embodiments with reference to the drawings, but the following examples are only simple examples of the present invention and do not represent or limit the scope of the present invention, which is defined by the claims.
The examples do not show the specific techniques or conditions, according to the technical or conditions described in the literature in the field, or according to the product specifications. The reagents or apparatus used are conventional products commercially available from normal sources, not indicated by the manufacturer.
Example 1
This example was used for phage nanobody library construction and panning, and for primary screening using ELISA. The method comprises the following specific steps:
(1) construction of phage Nanobody libraries
Adopting BCMA-Fc expressing the extracellular region to immunize bactrian camel, and extracting 200mL of peripheral blood after the titer is verified by ELISA; sorting lymphocytes to obtain peripheral blood mononuclear lymphocyte sediment and extracting RNA; by using
Figure BDA0002942099300000061
III, synthesizing first strand cDNA by using reverse transcriptase with RNA as a template, and then amplifying a VHH gene by using nested PCR; inserting VHH gene into pMECS phage display carrier, after electrotransformation of TG1 competent cell, taking appropriate amount of bacterial liquid for library identification, uniformly coating the rest culture on LB/AMPGLU flat plate, collecting bacterial lawn after bacterial growth, adding 1/3 volume of 50% glycerol, uniformly mixing and subpackaging, and storing at-80 deg.C to obtain the final product with storage capacity greater than 10 9 The phage display camelid VHH immune library of (a).
(2) Panning of phage Nanobody libraries
3 rounds of solid-phase screening are carried out on the immune nano antibody library in vitro, and phage clones with binding activity are enriched; after prokaryotic induction expression is carried out on the monoclonal phage, phage clones capable of combining with BCMA antigen extracellular regions are further screened out through ELISA. The method comprises the following specific steps:
firstly, diluting the purified BCMA-His recombinant protein to 4 mu g/mL by using PBS buffer solution, taking a 96-well enzyme label plate, selecting 3 wells, adding 100 mu L (400 ng/well) into each well, coating overnight at 4 ℃, and using PBS as negative control; discarding the coating solution, adding 150 mu L of 2% skimmed milk powder into each hole, sealing at room temperature for 1h to obtain an ELISA plate coated with BCMA-His recombinant protein;
then, the enzyme-linked plate was washed 4 times with PBST, and the prepared phage solution was diluted to 5X 10 with 2% skim milk powder 11 PFU/mL, at 1Adding the enzyme label plate with the amount of 00 mu L/hole, and incubating for 2h at room temperature;
then, discarding the phage sample, washing with PBST for 10 times, then washing with PBS for 5 times, adding 100 μ L of freshly prepared 0.1M triethylamine to each well, standing at room temperature for 10min, sucking out the eluate, and rapidly neutralizing with an equal volume of 1M Tris-HCl (pH 7.4);
measuring bacteriophage titer with part of eluate, and infecting with 4mL fresh culture log-phase TG1 bacterial solution (OD) with 400 μ L eluate 600 About 0.6), incubating at 37 deg.C for 30min, adding 16mL 2 XYT/ampicillin/glucose (2 XYT/AMP-GLU), culturing at 37 deg.C for 200r/min to OD 600 0.6 to 0.8.
Taking 100 mu L of bacterial suspension, performing gradient dilution, and uniformly smearing the bacterial suspension on a 2 XYT/AMP-GLU agar plate so as to perform library capacity and diversity determination; inoculating 100 μ L bacterial suspension, namely phage display carrier library, into a 2 XYT/AMP-GLU culture medium, culturing to logarithmic phase, adding auxiliary phage, performing library rescue, obtaining phage titer to be measured of phage particles, and concentrating and purifying to obtain phage particles for next round of screening; the screening operation was repeated 3 times;
the remaining bacterial liquid was centrifuged and resuspended in 2 XYT medium of appropriate volume, spread on a plate with screening resistance for overnight culture, scraped from the plate with appropriate volume of liquid medium, resuspended in 2 XYT medium containing 1/3 volume of 50% glycerol, and then aliquoted and stored at-80 ℃.
(3) Phage packaging
100 μ L of the frozen stock was added to 100mL of 2 XYT/AMP-GLU medium and cultured at 37 ℃ with shaking (200rpm) until logarithmic phase (OD) 600 The value is 0.6 to 0.8); add 90. mu.L of the helper phage M13K07 (1.7X 10) 13 PFU/mL), standing at 37 ℃ for 30min, centrifuging at 2800g for 10min to collect the thallus, resuspending with 200mL of 2 XYT/AMP-KAN medium, and culturing at 37 ℃ for 12h with shaking (200 rpm);
centrifuging at 4 deg.C for 30min at 3800g for 30min to remove thallus and collect supernatant, adding 1/5 volume of precooled PEG/NaCl, mixing, and precipitating bacteriophage for 2 h; centrifuging at 4 ℃ and 3800g for 30min, collecting the phage, resuspending with a final volume of 2mL PBS solution and transferring to a 15mL centrifuge tube; centrifuging at 4 ℃ and 12000g for 15min, collecting supernatant, adding 1/5 volumes of precooled PEG/NaCl solution, turning upside down and mixing uniformly, and standing on ice for 2 h; centrifuging at 4 deg.C and 10000g for 10min, discarding supernatant, resuspending phage precipitate with 1mL PBS, incubating overnight at 4 deg.C with shaking table to dissolve phage particles completely, mixing phage solution with equal volume of 60% glycerol, packaging into 1.5mL EP tube, and storing at-80 deg.C.
As the BCMA antigen is adopted to carry out 3 rounds of panning on the phage library in the step (2), in order to avoid losing the diversity of sequences, the preliminary ELISA screening is carried out on products of panning of the 2 nd round and the 3 rd round, positive clones are randomly selected from the panning products and are induced to express, the expression supernatant is a crude VHH antibody, the VHH antibody sequence of the monoclonal strain is determined by sequencing, the antibody sequence is marked as BCMA-8, and the sequence is shown as SEQ ID NO. 8.
Example 2
Candidate clones were screened using flow cytometric activated Cell Sorting (FACS) in this example.
Performing cell culture according to a standard cell culture scheme, and preparing BCMA positive and negative cell suspensions by using pancreatin digestive cells; the culture medium was removed by centrifugation at 300g for 5min, and the cells were resuspended to 2X 10 with a Flow Buffer 6 cell/mL; adding 2X 10 to each well in a V-bottom 96 well plate 5 Centrifuging 300g of each cell for 5min, removing supernatant, adding a VHH antibody crude extract to resuspend the cells, and incubating at 4 ℃ for 1 h;
centrifuging 300g for 5min, removing supernatant, resuspending cells by using Flow Buffer, diluting the APC anti-his antibody to 2 μ g/mL by using the Flow Buffer, resuspending cells by 100 μ L per well, and incubating for 1h at 4 ℃; after 3 times of washing the cells with Flow Buffer, 200. mu.L of Flow Buffer was used to resuspend the cells and examined by Flow cytometry.
Example 3
In this example, VHH-mIgG2a Fc nanobody was expressed and purified, and the affinity of the antibody was determined. To further identify the antibodies screened, it is necessary to express the antibodies by mammalian cells. Therefore, a plasmid vector for expressing VHH with a mouse Fc tag is firstly constructed and is marked as C-4 pCP.Stuffer-mCg2a-FC, and the specific steps are as follows:
1. BCMA VHH B8 was amplified using PCR with primers:
HDB8-F(SEQ ID NO.10):
CGCGATTCTTAAGGGTGTCCAGTGCGAGGTGCAGCTGGTGGA;
HD-B8-R(SEQ ID NO.11):
GCATGGAGGACAGGGCTTGATTGTGGGGCTAGACACTGTCACCTG
the reaction system is shown in Table 2, and the amplification procedure is shown in Table 3 below:
TABLE 2
Figure BDA0002942099300000081
TABLE 3
Figure BDA0002942099300000082
2. The enzyme digestion system is shown in Table 4, the enzyme digestion temperature is 37 ℃, the time is 6h, the carrier after enzyme digestion is used
Figure BDA0002942099300000083
The PCR purification kit was purified, and the recovered DNA was dissolved in 45. mu.L of water to detect the concentration of the DNA.
TABLE 4
Reagent Amount of the composition used
C-4pCP.Stuffer-mCg2a-FC 5μg
10 Xdigestion Buffer (10 Xreaction Buffer) 5μL
FspA I 2μL
PfI 23II 2μL
ddH 2 O Make up to 50. mu.L
3. The PCR amplification product was ligated into the enzyme-digested linearized vector by homologous recombination, as shown in Table 5, in a 37 ℃ water bath for 30 min.
TABLE 5
Reagent Volume (μ L)
Exnase II 1
Exnase II buffer 2
Linearized vector 4
Insert (Insert fragment) 3
4. All the homologous recombination reaction systems are added into DH5 alpha competent cells, and DH5 alpha competent cells are transformed, and the transformation conditions are shown in Table 6.
TABLE 6
Procedure Temperature of Time
Ice bath
0℃ 5min
Heat shock 42℃ 1min
Ice bath 0℃ 3min
Adding 500. mu.L LB medium, shaking at 220rpm 37℃ 1.5h
Pipette
200. mu.L of the solution and spread on LB/Amp plates 37℃ Overnight
5. Selecting monoclonal PCR for pre-identification by the transformation plate, wherein the conditions of a PCR identification system are shown in Table 7; the conditions are pre-denaturation at 95 ℃ for 3min, denaturation at 95 ℃ for 30s, annealing at 55 ℃ for 30s, extension at 72 ℃ for 30s, 35 cycles, extension at 72 ℃ for 5min, storage at 4 ℃, and sending to a sequencing company for sequencing and identification. The sequencing result is in line with the expectation, and the plasmid vector with the mouse Fc tag for expressing VHH is successfully constructed in the example of the specification.
TABLE 7
Figure BDA0002942099300000091
Figure BDA0002942099300000101
293E cells were passaged to a cell density of about 0.6X 10 approximately 24h prior to plasmid transfection 6 cells/mL; when the cell density is (1.0-1.2) × 10 6 cells/mL, viability>At 95%, 293E cells were transfected with PEI at a ratio of 0.15. mu.g scFV-mIgG1/100mL 293E, wherein the ratio of plasmid DNA to PEI was 1: 2;
37℃、130rpm、8%CO 2 the cell culture supernatant was collected by shaking for 5 days at 3000rpm for 30min, filtered through a Millex-GP Filter Unit 0.45 μm Sterile, and subjected to MabSelect TM SuRe TM Concentrate by centrifugation, wash the column with 1 × PBS, elute protein with 0.1M Gly-HCl, and neutralize with 1/10 volumes of Tris-HCl at pH 8.5.
Protein dialysis overnight at 4 ℃ A was measured using NanoDrop 2000 280 The method of (3) is quantitative, and the SEC-HPLC is used for determining the purity of the antibody.
In addition, in this example, affinity of purified BCMA VHH antibody was measured by Biacore.
Biacore is a bioanalytical sensing technology developed based on Surface Plasmon Resonance (SPR), and can detect the whole process of change of binding and dissociation of molecules in a tracking solution and molecules fixed on the surface of a chip, record the change in the form of a sensorgram, and provide kinetic and affinity data.
In the measurement process, the antibody is immobilized on the surface of the chip, and the mobile phase is a solution containing the antigen. The measurement results are shown in table 8 and fig. 1.
TABLE 8
Figure BDA0002942099300000102
Example 4
In this example, single chain antibodies against BCMA VHH were subjected to flow assay.
Three tumor cells, K562(BCMA-), RPMI 8226(BCMA +), and U266(BCMA +), were mixed with purified recombinant anti-BCMA VHH-mIgG2 antibody, incubated for 30min under ice bath, then incubated for 30min with APC-labeled goat anti-mouse IgG antibody, and detected by flow cytometry.
As shown in FIG. 2, the single-chain antibody recognized BCMA antigen on the cell surface, which was 84.9% and 95.2% in RPMI 8226(BCMA +) and U266(BCMA +), respectively.
Example 5
This example was used to prepare lentiviral vectors expressing chimeric antigen receptors for BCMA VHH.
First, a lentiviral vector pSIN-HD BCMA CAR 8# carrying a BCMA VHH chimeric antigen receptor was constructed, the vector map is shown in FIG. 3, and the schematic representation of the chimeric antigen receptor is shown in FIG. 4, comprising the CD8 α signal peptide, BCMA VHH, CD8 α hinge region, transmembrane region, and immunoreceptor tyrosine activation motif (CD3 ζ).
Wherein the amino acid sequence of the signal peptide (SEQ ID No.12) is:
MALPVTALLLPLALLLHAARP
the amino acid sequence of BCMA VHH is shown in SEQ ID No. 8;
The amino acid sequence of the CD8 α hinge and transmembrane regions (SEQ ID No.13) is:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYC
the amino acid sequence of the 4-1BB intracellular domain (SEQ ID No.14) is:
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL
the CD3 ζ amino acid sequence (SEQ ID No.15) is:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR。
the preparation method comprises the following steps:
1. a PCR reaction system was prepared according to table 9 to amplify BCMA VHH fragments using primers:
B8(op)-FR1F-F(E)(SEQ ID NO.16):
CTGCCGCTGGCCTTGCTGCTCCACGCCGCCAGGCCGGAGGTGCAGCTGGTGGAAAGCGGCGGCGGCC
B8(op)-R(SEQ ID NO.17):
GCGCTGGCGTCGTGGTGCTAGACACTGTCACC
TABLE 9
Figure BDA0002942099300000111
Figure BDA0002942099300000121
The above reagent is from TOYOBO Inc.
After preparation, the reaction was performed according to the PCR procedure shown in Table 10.
Watch 10
Figure BDA0002942099300000122
2. PCR reaction systems were prepared according to table 11, and the CD8 α signal peptide was added before the amplification product obtained, using the primers:
BamH-CD8αsig-F(SEQ ID NO.18):
GCTGCAGGTCGACTCTAGAGGATCCCGCCACCATGGCCTTACCAGTGACCGCCTTGCTCCTGCCGCTGGCCTTGC
B8(op)-R(SEQ ID NO.17);
TABLE 11
Reagent Volume (μ L)
10×buffer 5
2mM dNTP 5
25mM MgSO 4 3
10 μ M upstream primer (BamH-CD8 α sig-F) 1
10 μ M downstream primer 1
Template DNA (VHH fragment PCR reaction solution) 4
Sterile deionized water (PCR grade water) 30
KOD-Plus-Neo high fidelity PCR enzyme 1
After preparation, PCR reactions were performed according to the PCR procedure shown in Table 10.
After the reaction is finished, carrying out 1% agarose gel electrophoresis on the PCR product, recovering fragments of about 500bp, and quantifying by an ultraviolet absorption method.
3. The PCR reaction system was prepared in accordance with Table 12, and after the preparation, the PCR reaction was carried out in accordance with the PCR procedure shown in Table 10. The CD8 α hinge-TM-41BB-CD3 ζ fragment was amplified using the following primers:
CD8αH-F(SEQ ID NO.19):ACCACGACGCCAGCGCCGCGAC
Vector-R(SEQ ID NO.20):TCGATAAGCTTGATATCG
TABLE 12
Reagent Volume (μ L)
10×buffer 5
2mM dNTP 5
25mM MgSO 4 3
10 μ M of the forward primer CD8 α H-F 1
10 mu M downstream primer Vector-R 1
Template DNA (HD CD19CAR) 1
Sterile deionized water (PCR grade water) 33
KOD-Plus-Neo high fidelity PCR enzyme 1
After the PCR is finished, 1% agarose gel electrophoresis is carried out, fragments of about 780bp are recovered, and the quantification is carried out by an ultraviolet absorption method.
4. Mu.g of the laboratory-constructed HD CD19CAR plasmid was digested with BamHI and EcoRI, reacted in a water bath at 37 ℃ for 2 hours, and then the vector was recovered.
The 3 fragments were ligated with a recombinase, and the recombination reaction system was as shown in Table 13, followed by a reaction in water bath at 37 ℃ for 0.5 hour after the preparation, and transformation into Escherichia coli stbl3 competent cells by a conventional method.
Watch 13
Reagent Amount of the composition used
HD CD19 CAR 184.54ng
CD8αsingal BCMA VHH 31.32ng
CD8αhinge-TM-41BB-CD3ζ 29.72ng
5×CE buffer 2μL
Exnase TM II 1μL
Sterile deionizationWater (PCR grade water) Make up to 10. mu.L
Single clones were selected from the solid medium, cultured overnight, and identified by PCR, the PCR reaction formulations are shown in Table 14, and the PCR procedures are shown in Table 15. After the PCR is finished, the positive clone is selected for further sequencing identification, and the sequencing result is in line with expectation.
TABLE 14
Reagent Volume (μ L)
Taq PCR Master Mix 10
10μM F Seq-trEF1a-F 1
10μM R Vector-R 1
Template DNA bacterial liquid 1
Sterile deionized water (PCR grade water) 7
Watch 15
Figure BDA0002942099300000141
Separating the two gene fragments of the obtained signal peptide-VHH, CD8 alpha hinge region, transmembrane region and immunoreceptor tyrosine activation motif by agarose gel electrophoresis, and then recovering, purifying and quantifying by using an agarose gel DNA fragment recovery kit;
The lentiviral expression vector pRRL. EF1. alpha. -BCMA CAR-WPRE was cut with the restriction enzymes BamHI and EcoRI (from NEB) and manipulated as described. Separating the enzyme digestion product by agarose gel electrophoresis, and then recovering, purifying and quantifying by using an agarose gel DNA fragment recovery kit;
then, the two target fragments and the vector are cloned into a lentiviral vector by using a recombinase, sequencing verification is carried out, and the sequencing result is in accordance with expectation.
Example 6
In this example, lentiviral vector pSIN-HD BCMA CAR 8# prepared in example 5 was subjected to lentiviral packaging, concentration and titer detection.
(1) Lentiviral packaging
At 1.6X 10 7 Cell number 293T cells were plated in 15cm dishes at 37 ℃ with 5% CO 2 Culturing overnight to prepare packaged virus, wherein the culture medium contains DMEM and 10% Fetal Bovine Serum (FBS); dissolving lentiviral vector 30 μ g pSIN-HD BCMA CAR 8#, helper plasmid gag/pol 12.5 μ g and envelope plasmid VSVg 10 μ g into 2000 μ L serum-free DMEM culture solution, and mixing;
mu.g PEI (1. mu.g/. mu.L) was dissolved in 2000. mu.L serum free DMEM medium, gently mixed (or vortexed at 1000rpm for 5 seconds) and incubated at room temperature for 5 min; adding the PEI mixed solution into the DNA mixed solution, immediately mixing by vortex or mixing lightly, and incubating for 20min at room temperature to form a transfection compound; dripping 4mL of the transfection compound into 25mL of DMEM medium containing 293T cells, and replacing the fresh medium after 4-5 h; after 48h, the virus supernatant was collected.
(2) Lentiviral concentration
Filtering the virus supernatant with a 0.45-micron filter membrane, collecting the filtrate in a 50mL centrifuge tube, adding 1/4 PEG-NaCl virus concentrated solution, turning upside down, mixing uniformly, and standing at 4 ℃ overnight; centrifuging at 4 deg.C and 3500rpm for 30 min; removing supernatant, adding an appropriate amount of RPMI 1640 medium (containing 10% of FBS by mass), and dissolving the heavy suspension virus precipitate; subpackaging the concentrated lentivirus suspension into 50 μ L portions, storing in a finished product tube, and storing at-80 deg.C;
(3) lentiviral titer detection
500 μ L K562 cells (1X 10) 5 Individual cells) were seeded in 24-well culture plates, and the concentrated lentivirus was added to the cell suspension in volumes of 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, 37 ℃, 5% CO 2 After overnight culture, the fresh medium was replaced;
after infection for 72h, centrifuging for 5min at 400g, discarding the supernatant to collect cells, adding 100 μ L PBS + 2% FBS to resuspend the cells, adding 1 μ g hBCMA-EcD-Fc antibody, and incubating on ice for 30 min; washing with PBS (containing FBS with mass fraction of 2%) buffer solution for 1 time, adding 100 μ L buffer solution to resuspend cells, adding APCanti-human IgG Fc antibody, and incubating on ice for 30 min; after being washed for 2 times by PBS (containing FBS with the mass fraction of 2%), 300 mu L of buffer solution is added to resuspend cells, and a flow cytometer is adopted to detect the infection efficiency; preferably, a cell sample with the positive rate of 5-20% is taken, the titer is calculated, and the titer calculation formula is as follows: titer (TU/mL) is cell number (10) 5 ) X positive rate/virus volume (mL).
Example 7
In this example, T lymphocytes were transduced with the lentiviruses prepared in example 6.
(1) Diluting anti-human CD3 antibody and anti-human CD28 antibody with PBS to final concentrations of 1 μ g/mL and 0.5 μ g/mL respectively, coating the well plate, and standing overnight in a refrigerator at 4 deg.C; discarding the antibody coating solution in the pore plate, and washing twice with 1mL of PBS;
(2) human PBMC were adjusted to a density of 1X 10 with T cell culture medium (X-VIVO + 10% FBS +300U/mL IL-2) 6 Activation for 48 h/mL, inoculated into CD3 and CD28 antibody coated well plates; collecting activated T cells, adjusting cell density to 1 × 10 6 (iv)/mL, lentivirus was added at a multiplicity of infection (MOI) of 10, polybrene was added to a final concentration of 5. mu.g/mL; at 37 ℃ with 5% CO 2 After overnight culture in the environment, replacing a fresh culture medium, and carrying out passage every 2-3 days;
(3) after 5 days of T cell infection, 3X 10 cells were taken 5 Centrifuging the T cells at 4 ℃ for 5min at 400g, discarding the supernatant, and washing the cells once with PBS (containing FBS with the mass fraction of 2%); adding 100. mu.L buffer solution to resuspend the cells, adding 1. mu.g of hBCMA-EcD-Fc antibody, and incubating on ice for 30 min; washing with buffer solution for 1 time, adding 100 μ L buffer solution to resuspend cells, adding APCanti-human IgG Fc antibody, and incubating on ice for 30 min; after washing for 2 times by the buffer solution, adding 300 mu L of the buffer solution to resuspend the cells;
The expression rate of the chimeric antigen receptor of the T lymphocytes was measured by flow cytometry, and the result is shown in FIG. 5, in which FIG. I is a blank control and FIG. II is an experimental group to which BCMA-8 was added, the expression rate of the chimeric antigen receptor was 59.9%.
In addition, flow cytometry was also used to detect lymphocyte phenotypes in this example.
(1) After 5 days of T cell infection, 3X 10 cells were taken 5 Centrifuging the T cells at 4 ℃ for 5min at 400g, discarding the supernatant, and washing the cells once with PBS (containing FBS with the mass fraction of 2%);
(2) add 50 u L buffer heavy suspension cells, add 1 u L FITC labeled Anti-CD3 Ab, Percp-Cy5.5 labeled Anti-CD4 Ab and PE-Cy7 labeled Anti-CD8 Ab, ice incubation for 30 min; after the buffer solution is washed twice, 300 mu L of buffer solution is added to resuspend the cells, and a flow cytometer is adopted to detect the cell phenotype;
the results are shown in FIG. 6, where the proportion of CD3+ CD4+ cells was 8.66% and the proportion of CD3+ CD8+ cells was 90.6% in the untransfected T cells; the proportion of T lymphocytes CD3+ CD4+ cells in the experimental group was 29.1%, and the proportion of CD3+ CD8+ cells was 68.5%.
Example 8
In vitro toxicity experiments were performed on CAR-T cells in this example.
1. Target cell seeding
The target cell concentrations were adjusted to 1X 10 using K562(BCMA-), 8266(BCMA +), and U266(BCMA +) as target cells 5 mL, 100. mu.L of the suspension was inoculated into a 96-well plate;
2. effector cell inoculation:
the BCMA CAR-T and the control T cells are effector cells, and the CAR-T cells and the control T cells are added into a 96-well plate according to an effective target ratio of 0.3:1, 1:1 and 3: 1;
3. each group was set with 3 replicate wells, and the average of the 3 replicate wells was taken. Wherein each experimental group and each control group are as follows:
experimental groups: each target cell + CAR-T;
control group 1: maximal release of LDH by target cells;
control group 2: target cells spontaneously release LDH;
control group 3: the effector cells spontaneously release LDH;
4. the detection method comprises the following steps:
after the effector cells and the target cells were co-cultured for 18 hours, they were subjected to CytoTox 96 nonradioactive cytotoxicity assay kit (Promega).
The method is a detection method based on a colorimetric method, and reflects the cracking degree of cells by detecting the content of Lactate Dehydrogenase (LDH). LDH is a stable cytosolic enzyme that is released upon cell lysis in a manner substantially identical to that of 51Cr in a radioactive assay. The released LDH medium supernatant can be detected by a coupled enzymatic reaction in which LDH converts a tetrazolium salt (INT) to red formazan (formazan). The amount of red product produced is proportional to the number of cells lysed; reference is made in particular to the instructions of the CytoTox 96 nonradioactive cytotoxicity detection kit.
5. The cytotoxicity was calculated as:
Figure BDA0002942099300000171
the results are shown in fig. 7A, the constructed CAR-T cells did not have a killing effect on BCMA negative cells, and as shown in fig. 7B and 7C, BCMA positive tumor cells had killing activity.
Example 9
The secretion of CAR-T cytokines was examined in this example.
1. Cell culture supernatant
400g of cell culture with an effective target ratio of 1:1 is centrifuged for 10 minutes to remove precipitates, and the supernatant is taken and stored at-80 ℃ for detection.
2. Reagent preparation
Before detection, all reagents and samples are returned to room temperature, and if the concentrated reagents are crystallized, the temperature of the concentrated reagents is warmed at 37 ℃ until all crystals are dissolved. The 1 × wash solution, 1 × detection buffer, detection antibody were prepared according to the instructions and diluted detection antibody was used within 30 minutes.
3. Standard and sample preparation
And (3) standard substance: the standard stock was diluted 2-fold using 5% 1640 medium for a total of 8 dilution gradients, including zero concentration.
Sample preparation: samples were diluted proportionally using 5% 1640 medium.
4. Detection step
(1) Soaking the enzyme label plate: add 300. mu.L of 1 Xlotion and allow to stand for 30 seconds. Soaking is necessary to obtain the desired experimental results. After discarding the wash solution, the microplate was patted dry on absorbent paper. After the plate wash was complete, please use the plate immediately without allowing the plate to dry.
(2) Adding a standard substance: the standard was added to 100. mu.L of 2 fold diluted standard. The blank wells were filled with 100 μ L of standard dilutions (serum/plasma samples) or culture medium (cell culture supernatant samples).
(3) Adding a sample: cell culture supernatant: 100 μ L of cell culture supernatant was added to the wells.
(4) Adding a detection antibody: add 50. mu.L of diluted detection antibody per well (1:100 dilution). Ensuring continuous sample adding in the steps 4, 5 and 6 without interruption. The loading process was completed in 15 minutes.
(5) And (3) incubation: a closure plate membrane is used to close the plate. Shaking at 300 rpm, and incubating at room temperature for 2 hours.
(6) Washing: the liquid was discarded and the plate washed 6 times by adding 300. mu.L of wash solution per well. The plate was washed each time and patted dry on absorbent paper. To obtain the desired experimental performance, the residual liquid must be thoroughly removed.
(7) Adding enzyme for incubation: mu.L of diluted horseradish peroxidase-labeled streptavidin (1:100 dilution) was added to each well.
(8) And (3) incubation: the plates were sealed with a new sealing plate, shaken at 300 rpm, incubated at room temperature for 45 minutes and washed.
(9) Adding a substrate for color development: add 100. mu.L chromogenic substrate TMB to each well, protect from light, incubate for 20min at room temperature.
(10) Adding a stop solution: add 100. mu.L of stop buffer to each well. The color changed from blue to yellow. If the color is green or the color change is obviously uneven, the plate frame is tapped lightly and the mixture is mixed well.
(11) And (3) detection reading: within 30 minutes, using an enzyme-labeling instrument to carry out dual-wavelength detection, and measuring OD values under the maximum absorption wavelength of 450nm and the reference wavelength; the OD value after calibration was the value measured at 450nm minus the value measured at the reference wavelength.
The secretion results of IL-2, TNF-alpha and IFN-gamma factors are respectively shown in figure 8, figure 9 and figure 10, and the constructed CAR-T cells release cytokines for BCMA positive tumor cells, but do not have obvious cytokine secretion for BCMA negative cells.
In conclusion, the anti-BCMA antibody is obtained by screening a phage display nano antibody library, has better antibody affinity, is used as an antigen binding domain to construct a chimeric antigen receptor and a CAR-T cell, has the proportion of CD3+ CD4+ cells of 68.5% and CD3+ CD8+ cells of 66.4%, and has killing activity on BCMA positive tumor cells through cytotoxicity test detection.
The applicant declares that the above description is only a specific embodiment of the present invention, but the scope of the present invention is not limited thereto, and it should be understood by those skilled in the art that any changes or substitutions that can be easily conceived by those skilled in the art within the technical scope of the present invention are within the scope and disclosure of the present invention.
SEQUENCE LISTING
<110> Huadao (Shanghai) biopharmaceutical Co., Ltd
<120> nanometer antibody for resisting B cell maturation antigen and application thereof
<130> 20210127
<160> 20
<170> PatentIn version 3.3
<210> 1
<211> 8
<212> PRT
<213> Artificial Synthesis
<400> 1
Gly Tyr Ser Asp Ser Asn Tyr Cys
1 5
<210> 2
<211> 7
<212> PRT
<213> Artificial Synthesis
<400> 2
Ile Asn Gly Asp Gly Val Ile
1 5
<210> 3
<211> 12
<212> PRT
<213> Artificial Synthesis
<400> 3
Ala Ala Leu Thr Ala Gly Cys Val Arg Tyr Ala Ala
1 5 10
<210> 4
<211> 25
<212> PRT
<213> Artificial Synthesis
<400> 4
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Pro Val Gln Ala Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ala Ser
20 25
<210> 5
<211> 17
<212> PRT
<213> Artificial Synthesis
<400> 5
Met Ala Trp Phe Arg Gln Ala Pro Gly Lys Ala Arg Gln Gly Val Ala
1 5 10 15
Phe
<210> 6
<211> 38
<212> PRT
<213> Artificial Synthesis
<400> 6
Thr Tyr Thr Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Lys Asp Asn
1 5 10 15
Ala Gln Lys Thr Leu Asp Leu Gln Met Asn Ser Leu Lys Pro Glu Asp
20 25 30
Thr Ala Met Tyr Tyr Cys
35
<210> 7
<211> 11
<212> PRT
<213> Artificial Synthesis
<400> 7
Trp Gly Gln Gly Thr Gln Val Thr Val Ser Ser
1 5 10
<210> 8
<211> 118
<212> PRT
<213> Artificial Synthesis
<400> 8
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Pro Val Gln Ala Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ala Ser Gly Tyr Ser Asp Ser Asn Tyr
20 25 30
Cys Met Ala Trp Phe Arg Gln Ala Pro Gly Lys Ala Arg Gln Gly Val
35 40 45
Ala Phe Ile Asn Gly Asp Gly Val Ile Thr Tyr Thr Asp Ser Val Lys
50 55 60
Gly Arg Phe Thr Ile Ser Lys Asp Asn Ala Gln Lys Thr Leu Asp Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Pro Glu Asp Thr Ala Met Tyr Tyr Cys Ala
85 90 95
Ala Leu Thr Ala Gly Cys Val Arg Tyr Ala Ala Trp Gly Gln Gly Thr
100 105 110
Gln Val Thr Val Ser Ser
115
<210> 9
<211> 354
<212> DNA
<213> Artificial Synthesis
<400> 9
gaggtgcagc tggtggaaag cggcggcggc cccgtgcagg ccggcggcag cctgagactg 60
agctgcaccg ccagcggcta cagcgacagc aactactgca tggcctggtt cagacaggcc 120
cccggcaagg ccagacaggg cgtggccttc atcaacggcg acggcgtgat cacctacacc 180
gacagcgtga agggcagatt caccattagc aaagataatg cccagaaaac actggatctg 240
cagatgaata gcctgaaacc tgaagataca gccatgtatt attgtgccgc cctgacagcc 300
ggatgtgtga gatatgccgc ctggggacag ggcacacagg tgacagtgtc tagc 354
<210> 10
<211> 42
<212> DNA
<213> Artificial Synthesis
<400> 10
cgcgattctt aagggtgtcc agtgcgaggt gcagctggtg ga 42
<210> 11
<211> 45
<212> DNA
<213> Artificial Synthesis
<400> 11
gcatggagga cagggcttga ttgtggggct agacactgtc acctg 45
<210> 12
<211> 21
<212> PRT
<213> Artificial Synthesis
<400> 12
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro
20
<210> 13
<211> 69
<212> PRT
<213> Artificial Synthesis
<400> 13
Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala
1 5 10 15
Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly
20 25 30
Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile
35 40 45
Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu Leu Ser Leu Val
50 55 60
Ile Thr Leu Tyr Cys
65
<210> 14
<211> 42
<212> PRT
<213> Artificial Synthesis
<400> 14
Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met
1 5 10 15
Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe
20 25 30
Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
35 40
<210> 15
<211> 112
<212> PRT
<213> Artificial Synthesis
<400> 15
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Lys Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 16
<211> 67
<212> DNA
<213> Artificial Synthesis
<400> 16
ctgccgctgg ccttgctgct ccacgccgcc aggccggagg tgcagctggt ggaaagcggc 60
ggcggcc 67
<210> 17
<211> 32
<212> DNA
<213> Artificial Synthesis
<400> 17
gcgctggcgt cgtggtgcta gacactgtca cc 32
<210> 18
<211> 75
<212> DNA
<213> Artificial Synthesis
<400> 18
gctgcaggtc gactctagag gatcccgcca ccatggcctt accagtgacc gccttgctcc 60
tgccgctggc cttgc 75
<210> 19
<211> 22
<212> DNA
<213> Artificial Synthesis
<400> 19
accacgacgc cagcgccgcg ac 22
<210> 20
<211> 18
<212> DNA
<213> Artificial Synthesis
<400> 20
tcgataagct tgatatcg 18

Claims (16)

1. A nanobody against B cell maturation antigen, wherein the heavy chain variable region of the nanobody comprises:
(1) CDR1 of an amino acid sequence shown in SEQ ID NO.1, wherein the sequence of SEQ ID NO.1 is GYSDSNYC;
(2) CDR2 of the amino acid sequence shown in SEQ ID NO.2, wherein the sequence of SEQ ID NO.2 is INGDGVI;
(3) CDR3 of the amino acid sequence shown in SEQ ID NO.3, wherein the sequence of SEQ ID NO.3 is AALTAGCVRYAA.
2. The nanobody of claim 1, wherein the amino acid sequence of the heavy chain variable region of the nanobody is represented by SEQ ID No.8, and the sequence of SEQ ID No.8 is:
EVQLVESGGGPVQAGGSLRLSCTASGYSDSNYCMAWFRQAPGKARQGVAFINGDGVITYTDSVKGRFTISKDNAQKTLDLQMNSLKPEDTAMYYCAALTAGCVRYAAWGQGTQVTVSS。
3. A nucleic acid molecule encoding the nanobody of any one of claims 1-2.
4. The nucleic acid molecule of claim 3, wherein the nucleotide sequence of said nucleic acid molecule is set forth as SEQ ID No. 9.
5. A chimeric antigen receptor comprising a signal peptide, an antigen binding domain, a hinge region, a transmembrane region, and a signaling domain;
the antigen binding domain is a nanobody according to any one of claims 1-2.
6. The chimeric antigen receptor according to claim 5, wherein the signal peptide comprises a CD8 a signal peptide;
the hinge region comprises a CD8 a hinge region;
the transmembrane region comprises any one of or a combination of at least two of a CD8 alpha transmembrane region, a CD28 transmembrane region or a DAP10 transmembrane region;
the signaling domain comprises an immunoreceptor tyrosine activation motif;
the signaling domains also include co-stimulatory molecules comprising any one of the 4-1BB, CD28 intracellular region, OX40, ICOS or DAP10 intracellular region, or a combination of at least two thereof.
7. The chimeric antigen receptor according to claim 6, wherein the chimeric antigen receptor comprises a CD8 a signal peptide, the nanobody of claim 1 or 2, a CD8 a hinge region, a CD8 a transmembrane region, and an immunoreceptor tyrosine activation motif.
8. An expression vector comprising a gene encoding the chimeric antigen receptor of any one of claims 5-7.
9. The expression vector according to claim 8, wherein the expression vector is any one of a lentiviral vector, a retroviral vector or an adenoviral vector comprising the gene encoding the chimeric antigen receptor according to any one of claims 5 to 7.
10. The expression vector of claim 9, wherein the expression vector is a lentiviral vector.
11. A recombinant lentivirus produced from a mammalian cell transfected with the expression vector of any one of claims 8 to 10 and a helper plasmid.
12. A chimeric antigen receptor immune cell, wherein said chimeric antigen receptor immune cell expresses the chimeric antigen receptor of any one of claims 5-7.
13. The chimeric antigen receptor immune cell according to claim 12, wherein the chimeric antigen receptor immune cell comprises the expression vector of any one of claims 8-10 and/or the recombinant lentivirus of claim 11;
the immune cells include any one of T cells, B cells, NK cells, mast cells or macrophages or a combination of at least two of the same.
14. A pharmaceutical composition comprising the chimeric antigen receptor immune cell of claim 12 or 13.
15. The pharmaceutical composition of claim 14, further comprising a pharmaceutically acceptable excipient.
16. Use of a nanobody according to any one of claims 1 to 2, a nucleic acid molecule according to claim 3 or 4, a chimeric antigen receptor according to any one of claims 5 to 7, an expression vector according to any one of claims 8 to 10, a recombinant lentivirus according to claim 11, a chimeric antigen receptor immune cell according to claim 12 or 13 or a pharmaceutical composition according to claim 14 or 15 for the preparation of a medicament for the treatment of tumors;
the tumor is multiple myeloma.
CN202110183479.7A 2021-02-08 2021-02-08 Nanometer antibody for resisting B cell mature antigen and application thereof Active CN112812186B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110183479.7A CN112812186B (en) 2021-02-08 2021-02-08 Nanometer antibody for resisting B cell mature antigen and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110183479.7A CN112812186B (en) 2021-02-08 2021-02-08 Nanometer antibody for resisting B cell mature antigen and application thereof

Publications (2)

Publication Number Publication Date
CN112812186A CN112812186A (en) 2021-05-18
CN112812186B true CN112812186B (en) 2022-07-29

Family

ID=75865111

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110183479.7A Active CN112812186B (en) 2021-02-08 2021-02-08 Nanometer antibody for resisting B cell mature antigen and application thereof

Country Status (1)

Country Link
CN (1) CN112812186B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116063530B (en) * 2021-12-29 2023-09-29 华道(上海)生物医药有限公司 Antibody for resisting mesothelin and application thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114149506B (en) * 2021-12-16 2023-01-10 华道(上海)生物医药有限公司 anti-CD 22 antibody and application thereof
CN116396385A (en) * 2022-09-23 2023-07-07 四川大学 anti-BCMA nano antibody and preparation method and application thereof
CN116120465B (en) * 2022-11-01 2023-10-13 邦恩泰(山东)生物医药科技集团股份有限公司 Chimeric antigen receptor targeting BCMA and/or FCRH5 and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020018825A1 (en) * 2018-07-19 2020-01-23 Regeneron Pharmaceuticals, Inc. Chimeric antigen receptors with bcma specificity and uses thereof
CN111333729A (en) * 2020-03-17 2020-06-26 深圳市南科生物工程有限公司 Nano antibody for resisting B cell mature antigen and application
CN111848798A (en) * 2020-07-27 2020-10-30 南京安锐生物科技有限公司 Nano antibody capable of being combined with BCMA (brain cell activating antigen) and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020018825A1 (en) * 2018-07-19 2020-01-23 Regeneron Pharmaceuticals, Inc. Chimeric antigen receptors with bcma specificity and uses thereof
CN111333729A (en) * 2020-03-17 2020-06-26 深圳市南科生物工程有限公司 Nano antibody for resisting B cell mature antigen and application
CN111848798A (en) * 2020-07-27 2020-10-30 南京安锐生物科技有限公司 Nano antibody capable of being combined with BCMA (brain cell activating antigen) and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116063530B (en) * 2021-12-29 2023-09-29 华道(上海)生物医药有限公司 Antibody for resisting mesothelin and application thereof

Also Published As

Publication number Publication date
CN112812186A (en) 2021-05-18

Similar Documents

Publication Publication Date Title
CN112812186B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN112480258B (en) anti-CD 123 nano antibody and application thereof
CN112029729B (en) CD19 and CD22 double-target chimeric antigen receptor NK (natural killer) cell and application thereof
CN113265001B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN112830971B (en) anti-BCMA antigen binding fragment and application thereof
CN112812185B (en) Monoclonal antibody for resisting B cell maturation antigen and application thereof
CN110862456B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN110713539B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN110655581B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN112500487B (en) anti-CD 123 monoclonal antibody and application thereof
CN111944053A (en) anti-BCMA CAR and expression vector and application thereof
CN112979808B (en) Antibody for resisting B cell mature antigen and application thereof
CN114276454B (en) Anti-mesothelin nano antibody and application thereof
CN115925948B (en) anti-CD 22 nano antibody and application thereof
CN112521505B (en) anti-CD 123 antibodies and uses thereof
CN114409782B (en) anti-IL13Ra2 nano antibody and application thereof
CN112521506B (en) CD123 antigen binding fragment and application thereof
CN116023500B (en) Chimeric antigen receptor targeting fully humanized CD70 and application thereof
CN116253800B (en) Heavy chain antibody against CLDN18.2, related products and uses
CN116675773A (en) anti-MXR 7 antibody and application thereof
CN116693688A (en) anti-Intestinal protein OCI-5 antibody and application thereof
CN116410323A (en) anti-GPC 3 antibody and application thereof
CN116640217A (en) anti-GTR 2-2 antibody and application thereof
CN116574181A (en) anti-Glypican 3 antibody and application thereof
CN117843787A (en) anti-CD7 antibody and application thereof

Legal Events

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