CN109593137B - Construction and application of novel CD20-CAR vector of anti-CD 20 antibody - Google Patents

Construction and application of novel CD20-CAR vector of anti-CD 20 antibody Download PDF

Info

Publication number
CN109593137B
CN109593137B CN201811652891.3A CN201811652891A CN109593137B CN 109593137 B CN109593137 B CN 109593137B CN 201811652891 A CN201811652891 A CN 201811652891A CN 109593137 B CN109593137 B CN 109593137B
Authority
CN
China
Prior art keywords
seq
sequence
nucleic acid
cell
chimeric antigen
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
CN201811652891.3A
Other languages
Chinese (zh)
Other versions
CN109593137A (en
Inventor
杨林
游凤涛
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.)
Persongen Biotherapeutics Suzhou Co ltd
Original Assignee
Persongen Biotherapeutics Suzhou 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 Persongen Biotherapeutics Suzhou Co ltd filed Critical Persongen Biotherapeutics Suzhou Co ltd
Priority to CN201811652891.3A priority Critical patent/CN109593137B/en
Publication of CN109593137A publication Critical patent/CN109593137A/en
Application granted granted Critical
Publication of CN109593137B publication Critical patent/CN109593137B/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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • 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

Abstract

The invention provides the construction and application of a novel CD20-CAR vector based on a humanized CD20 antibody. Specifically, the present invention provides a chimeric antigen receptor whose antigen-binding domain (i.e., scFv) includes an antibody heavy chain variable region shown in SEQ ID No.1 and an antibody light chain variable region shown in SEQ ID No. 2. The CD20-CAR-T cells constructed by using the CD20-CAR vector constructed by the invention show very good killing effect in vitro.

Description

Construction and application of novel CD20-CAR vector of anti-CD 20 antibody
Technical Field
The invention belongs to the technical field of biology, and particularly relates to construction and application of a novel CD20-CAR vector based on a humanized CD20 antibody.
Background
In recent years, chimeric antigen receptor T cell (CAR-T) therapy, as a powerful novel adoptive immunotherapy technology, has shown very effective therapeutic effects, and is used for treating various solid and hematological cancers, especially CD19-CAR-T therapy against B cell lymphoblastic leukemia and lymphoma, which shows very good therapeutic effects clinically, but for CD19 negative B cell tumors, CD19-CAR-T is not effective, and CD20 is also a good target of B cell tumors, so that CD20-CAR-T can be used for targeted therapy of CD20 positive B cell leukemia and lymphoma, and CD20-CAR-T cells constructed with humanized CD20 antibody sequences can reduce immunogenicity generated after entering into the body and improve therapeutic effects compared with murine CD20 sequences.
Therefore, there is an urgent need in the art to develop a novel CD20-CAR vector based on a humanized CD20 antibody.
Disclosure of Invention
The invention aims to provide construction and application of a novel CD20-CAR vector based on a humanized CD20 antibody.
In a first aspect of the invention, there is provided a Chimeric Antigen Receptor (CAR) (sequence) whose antigen-binding domain (i.e., scFv) comprises an antibody heavy chain variable region shown in SEQ ID No.1 and an antibody light chain variable region shown in SEQ ID No. 2.
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYNMHWVRQTPGQRLEWMGAIYPGNGDTSYNQKFKGRVTITADTSASTAYMELSSLRSEDTAVYYCARSTYYGGDWYFNVWGQGTTVTVS(SEQ ID No.:1)
DIQLTQSPSFLSASVGDRVTITCRASSSVSYIHWYQQKPGKAPKLLIYATSNLASGVPSRFSGSGSGTSFTLTISSLQPEDFATYYCQQWTSNPPTFGGGTKVEIKRTV(SEQ ID No.:2)
In another preferred embodiment, the antigen binding domain (scFv) of the chimeric antigen receptor is represented by formula I or formula II:
V H -V L ,(I);V L -V H ,(II)
wherein, V H Is an antibody heavy chain variable region; v L Is an antibody light chain variable region; "-" is a linker peptide or peptide bond.
In another preferred embodiment, the amino acid sequence of the connecting peptide is shown in SEQ ID NO. 3.
GGGGSGGGGSGGGGS(SEQ ID No.:3)
In another preferred embodiment, the chimeric antigen receptor has the structure shown in the following formula:
L-scFv-H-TM-C-CD3ζ
wherein the content of the first and second substances,
l is an optional Leader sequence (i.e. signal peptide sequence);
scFv is an antigen binding domain;
h is a hinge region;
TM is a transmembrane domain;
c is a costimulatory signal molecule;
CD3 ζ is a cytoplasmic signaling sequence derived from CD3 ζ;
the antigen binding domains and "-" are as described above, respectively.
In another preferred embodiment, the sequence of L is shown in SEQ ID No. 4.
MLLLVTSLLLCELPHPAFLLIP(SEQ ID No.:4)
In another preferred embodiment, the sequence of H is as shown in SEQ ID No. 5.
ESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFQSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID No.:5)
In another preferred embodiment, the sequence of TM comprises the transmembrane region from ICOS, preferably the sequence of TM is as shown in SEQ ID No. 6.
WLPIGCAAFVVVCILGCILICWL(SEQ ID No.:6)
In another preferred embodiment, the costimulatory signal molecule comprises an ICOS-derived costimulatory signal molecule, and/or a 4-1 BB-derived costimulatory signal molecule.
In another preferred embodiment, the amino acid sequence of the ICOS-derived costimulatory signal molecule is shown in SEQ ID No. 7.
TKKKYSSSVHDPNGEY MFMRAVNTAKKSRLTDVTL(SEQ ID No.:7)
In another preferred embodiment, the amino acid sequence of the co-stimulatory signaling molecule from 4-1BB source is shown in SEQ ID NO. 8.
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL(SEQ ID No.:8)
In another preferred embodiment, the sequence of CD3 ζ is as shown in SEQ ID No. 9.
RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID No.:9)
In another preferred embodiment, the sequence of the chimeric antigen receptor is as shown in SEQ ID No. 10.
MLLLVTSLLLCELPHPAFLLIPQVQLVQSGAEVKKPGASVKVSCKASGYTFTSYNMHWVRQTPGQRLEWMGAIYPGNGDTSYNQKFKGRVTITADTSASTAYMELSSLRSEDTAVYYCARSTYYGGDWYFNVWGQGTTVTVSGGGGSGGGGSGGGGSDIQLTQSPSFLSASVGDRVTITCRASSSVSYIHWYQQKPGKAPKLLIYATSNLASGVPSRFSGSGSGTSFTLTISSLQPEDFA
TYYCQQWTSNPPTFGGGTKVEIKRTVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFQSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEAL
HNHYTQKSLSLSLGKWLPIGCAAFVVVCILGCILICWLTKKKYSSSVHDPNGEYMFMRAVNTAKKSRLTDVTLKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID No.:10)
In a second aspect of the invention there is provided a nucleic acid molecule encoding a Chimeric Antigen Receptor (CAR) according to the first aspect of the invention.
In another preferred embodiment, said nucleic acid molecule comprises a nucleic acid sequence encoding said hinge region selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 5;
(b) A polynucleotide having a sequence as set forth in SEQ ID No. 11;
GAGAGCAAGTACGGACCTCCTTGTCCTCCTTGTCCAGCTCCAGAGTTTGAGGGCGGACCTAGCGTGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCAGAAGTGACTTGCGTGGTGGTGGACGTGTCTCAGGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAGGTGCACAACGCTAAGACCAAGCCCAGGGAGGAGCAGTTCCAGAGCACCTACAGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGATTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTGTCCAACAAGGGCCTGCCTAGCAGCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCTAGAGAGCCTCAGGTGTACACCCTGCCTCCTAGCCAGGAGGAGATGACCAAGAACCAGGTGTCCCTGACTTGCCTCGTGAAGGGCTTCTACCCCAGCGACATCGCAGTCGAGTGGGAAAGCAACGGCCAGCCCGAGAACAACTACAAGACCACACCTCCAGTGCTGGATAGCGACGGAAGCTTCTTCCTGTACAGCCGGCTGACCGTGGATAAAAGCCGCTGGCAGGAGGGCAACGTGTTCAGTTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCTCGGCAAG(SEQ ID NO.:11)
(c) A polynucleotide having a nucleotide sequence having 90% or more (preferably 95% or more) homology with the sequence shown in SEQ ID No. 11 and encoding the amino acid sequence shown in SEQ ID No. 5;
(d) A polynucleotide complementary to any one of the polynucleotides of (a) - (c).
In another preferred embodiment, said nucleic acid molecule comprises a nucleic acid sequence encoding the transmembrane region of ICOS selected from the group consisting of seq id no:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 6;
(b) A polynucleotide having a sequence as set forth in SEQ ID No. 12;
TGGCTGCCTATTGGTTGCGCAGCTTTCGTCGTCGTCTGCATCCTGGGTTGCATCCTGATTTGCTGGCTG(SEQ ID NO.:12)
(c) A polynucleotide having a nucleotide sequence having 90% or more (preferably 95% or more) homology to the sequence shown in SEQ ID No. 12 and encoding the amino acid sequence shown in SEQ ID No. 6;
(d) A polynucleotide complementary to any one of the polynucleotides of (a) - (c).
In another preferred embodiment, the nucleic acid molecule comprises a co-stimulatory signaling molecule coding sequence comprising an ICOS-derived co-stimulatory signaling molecule coding sequence, and/or a 4-1 BB-derived co-stimulatory signaling molecule coding sequence, wherein
The ICOS-derived costimulatory signal molecule coding sequence is selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 7;
(b) A polynucleotide having a sequence as set forth in SEQ ID No. 13;
ACCAAGAAGAAGTACAGCTCTAGCGTGCACGACCCTAACGGCGAGTACATGTTCATGCGGGCCGTCAACACCGCCAAAAAGAGCCGGCTGACCGACGTGACACTG(SEQ ID NO.:13)
(c) A polynucleotide having a nucleotide sequence having 90% or more (preferably 95% or more) homology to the sequence shown in SEQ ID No. 13 and encoding the amino acid sequence shown in SEQ ID No. 7;
(d) A polynucleotide complementary to a polynucleotide of any one of (a) - (c);
the coding sequence of the 4-1BB derived costimulatory signal molecule is selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 8;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 14;
AAGAGGGGCCGGAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGCGGCCAGTGCAGACAACACAGGAGGAAGACGGCTGCTCTTGCAGGTTCCCAGAGGAGGAGGAGGGCGGTTGCGAGCTG(SEQ ID NO.:14)
(c) A polynucleotide having a nucleotide sequence having 90% or more (preferably 95% or more) homology with the sequence shown in SEQ ID No. 14 and encoding the amino acid sequence shown in SEQ ID No. 8;
(d) A polynucleotide complementary to any one of the polynucleotides of (a) - (c).
In another preferred embodiment, said nucleic acid molecule comprises a nucleic acid sequence encoding an intracellular signaling domain of said CD3 ζ selected from the group consisting of seq id nos:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 9;
(b) A polynucleotide having a sequence as set forth in SEQ ID No. 15;
AGAGTGAAGTTCAGCAGAAGCGCCGACGCTCCAGCTTATCAGCAGGGCCAGAACCAGCTGTACAACGAGCTGAACCTGGGCAGGAGAGAGGAATACGACGTGCTGGACAAGAGGAGGGGCAGAGATCCCGAGATGGGAGGCAAGCCCAGAAGAAAGAACCCCCAGGAGGGCCTGTACAACGAGCTGCAGAAGGACAAGATGGCCGAGGCCTACAGCGAGATTGGCATGAAGGGCGAGAGAAGGAGGGGCAAAGGCCACGACGGACTGTATCAGGGCCTGTCTACCGCCACCAAGGATACCTACGACGCTCTGCACATGCAGGCTCTGCCTCCTAGA(SEQ ID NO.:15)
(c) A polynucleotide having a nucleotide sequence having 90% or more (preferably 95% or more) homology with the sequence shown in SEQ ID No. 15 and encoding the amino acid sequence shown in SEQ ID No. 9;
(d) A polynucleotide complementary to any one of the polynucleotides of (a) - (c).
In another preferred embodiment, the nucleic acid molecule comprises a nucleic acid sequence selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 10;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 16;
(c) A polynucleotide having a nucleotide sequence homology of 95% or more (preferably 98% or more) to the sequence shown in SEQ ID No. 16 and encoding an amino acid sequence shown in SEQ ID No. 10;
(d) A polynucleotide complementary to any one of the polynucleotides of (a) - (c).
In another preferred embodiment, the nucleic acid molecule is isolated.
In another preferred embodiment, the sequence of the nucleic acid molecule is as shown in SEQ ID No. 16.
ATGCTGCTGCTGGTGACCTCTCTGCTGCTCTGCGAACTGCCTCACCCAGCCTTTCTGCTGATCCCTCAGGTGCAGCTGGTGCAGTCAGGAGCCGAAGTGAAGAAGCCAGGAGCCTCAGTGAAGGTGTCTTGCAAGGCCAGCGGCTACACCTTCACCAGCTACAACATGCATTGGGTCCGGCAGACCCCAGGACAGAGACTCGAGTGGATGGGAGCCATCTACCCCGGCAACGGCGATACCAGCTACAACCAGAAGTTCAAGGGCCGCGTGACCATCACAGCCGATACAAGCGCCAGCACCGCCTACATGGAGCTGTCTAGCCTGAGGAGCGAGGACACAGCCGTGTACTATTGCGCCAGGAGCACCTACTACGGCGGCGATTGGTACTTCAACGTGTGGGGCCAGGGCACAACAGTGACAGTGAGCGGAGGAGGAGGAAGCGGAGGAGGAGGATCTGGAGGAGGCGGCAGCGATATTCAGCTGACCCAGAGCCCTTCTTTTCTGAGCGCCAGCGTGGGCGACAGAGTGACCATCACTTGCAGGGCCAGCAGCAGCGTGTCCTACATCCATTGGTACCAGCAGAAGCCAGGCAAGGCTCCTAAGCTGCTGATCTACGCCACCAGCAACCTGGCCAGCGGCGTGCCTAGCAGATTCAGCGGCAGCGGAAGCGGCACAAGCTTTACCCTGACCATCAGCTCTCTGCAGCCAGAGGACTTCGCCACCTACTACTGCCAGCAGTGGACCAGCAACCCTCCTACATTTGGCGGCGGAACCAAGGTGGAGATCAAGCGGACCGTGGAGAGCAAGTACGGACCTCCTTGTCCTCCTTGTCCAGCTCCAGAGTTTGAGGGCGGACCTAGCGTGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCAGAAGTGACTTGCGTGGTGGTGGACGTGTCTCAGGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAGGTGCACAACGCTAAGACCAAGCCCAGGGAGGAGCAGTTCCAGAGCACCTACAGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGATTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTGTCCAACAAGGGCCTGCCTAGCAGCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCTAGAGAGCCTCAGGTGTACACCCTGCCTCCTAGCCAGGAGGAGATGACCAAGAACCAGGTGTCCCTGACTTGCCTCGTGAAGGGCTTCTACCCCAGCGACATCGCAGTCGAGTGGGAAAGCAACGGCCAGCCCGAGAACAACTACAAGACCACACCTCCAGTGCTGGATAGCGACGGAAGCTTCTTCCTGTACAGCCGGCTGACCGTGGATAAAAGCCGCTGGCAGGAGGGCAACGTGTTCAGTTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCTCGGCAAGTGGCTGCCTATTGGTTGCGCCGCCTTTGTCGTCGTCTGTATCCTCGGCTGCATCCTGATTTGTTGGCTGACCAAGAAGAAGTACAGCAGCAGCGTGCACGACCCCAACGGCGAGTACATGTTCATGCGGGCCGTCAACACCGCCAAGAAGAGCAGGCTGACCGACGTGACACTGAAGAGGGGCCGGAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGCGGCCAGTGCAGACAACACAGGAGGAAGACGGCTGCTCTTGCAGGTTCCCAGAGGAGGAGGAGGGCGGTTGCGAGCTGAGAGTGAAGTTCAGCAGAAGCGCCGACGCTCCAGCTTATCAGCAGGGCCAGAACCAGCTGTACAACGAGCTGAACCTGGGCAGGAGAGAGGAATACGACGTGCTGGACAAGAGGAGGGGCAGAGATCCCGAGATGGGAGGCAAGCCCAGAAGAAAGAACCCCCAGGAGGGCCTGTACAACGAGCTGCAGAAGGACAAGATGGCCGAGGCCTACAGCGAGATTGGCATGAAGGGCGAGAGAAGGAGGGGCAAAGGCCACGACGGACTGTATCAGGGCCTGTCTACCGCCACCAAGGATACCTACGACGCTCTGCACATGCAGGCTCTGCCTCCTAGATGA(SEQ ID NO.:16)
In a third aspect of the invention, there is provided a vector comprising a nucleic acid molecule according to the second aspect of the invention.
In another preferred embodiment, the vector is a lentiviral vector.
In a fourth aspect of the invention, there is provided a host cell comprising the vector or chromosome of the third aspect of the invention integrated with an exogenous nucleic acid molecule of the second aspect of the invention.
In another preferred embodiment, the cell is an isolated cell, and/or the cell is a genetically engineered cell.
In another preferred embodiment, the cell is a mammalian cell.
In another preferred embodiment, the cell is a T cell.
In a fifth aspect of the invention, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a chimeric antigen receptor according to the first aspect of the invention, a nucleic acid molecule according to the second aspect of the invention, a vector according to the third aspect of the invention, or a cell according to the fourth aspect of the invention.
In a sixth aspect of the invention, there is provided a use of the chimeric antigen receptor of the first aspect of the invention, the nucleic acid molecule of the second aspect of the invention, the vector of the third aspect of the invention, or the cell of the fourth aspect of the invention, for the manufacture of a medicament or formulation for the treatment of a tumour.
In another preferred embodiment, the tumor comprises a CD20 positive tumor.
In a seventh aspect of the invention, there is provided a method of treating a disease, comprising administering to a subject in need thereof an amount of the chimeric antigen receptor of the first aspect of the invention, the nucleic acid molecule of the second aspect of the invention, the vector of the third aspect of the invention, or the cell of the fourth aspect of the invention, or the pharmaceutical composition of the fifth aspect of the invention.
In another preferred embodiment, the disease is a tumor.
In an eighth aspect of the invention there is provided a method of making a CAR-T cell (CAR-modified T cell) expressing a chimeric antigen receptor according to the first aspect of the invention,
the method comprises the following steps: (ii) transfecting the nucleic acid molecule of the second aspect of the invention or the vector of the third aspect of the invention into a T cell, thereby obtaining the CAR-T cell.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1 shows the structure of the humanized CD20-CAR vector.
Figure 2 shows a flow chart of transfection efficiency of humanized CD20-CAR-T cells.
Figure 3 shows the cytotoxicity of humanized CD20-CAR-T cells against K562 cells overexpressing CD 20.
FIG. 4 shows the cytotoxicity of humanized CD20-CAR-T cells against B-lymphoid leukemia cells (Nalm-6) and lymphoma cells (Raji) naturally expressing CD 20.
FIG. 5 shows cytokine secretion following incubation of CD20-CAR-T cells with Raji and Nalm-6 cells.
Detailed Description
The inventor of the invention has extensively and deeply studied and constructed a novel CD20-CAR vector targeting CD20 malignant tumor for the first time (third generation CAR vector). The CAR-T cell constructed by the CAR vector constructed by the humanized CD20 antibody sequence can clinically target B cell malignant tumors (B cell leukemia and lymphoma) of a CD20 antigen, can avoid the immunogenicity of the CD20-CAR-T cell constructed by the murine CD20scFv in a human body, improves the clinical curative effect, and lays a foundation for better targeted treatment of the B cell derived leukemia and lymphoma. In addition, the CD20-CAR-T cells constructed by using the CD20-CAR vector constructed by the invention show very good killing effect in vitro. The present invention has been completed based on this finding.
Chimeric antigen receptors
The invention provides Chimeric Antigen Receptors (CARs) comprising an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain includes a target-specific binding member (also referred to as an antigen-binding domain). The intracellular domain includes a costimulatory signaling region and a zeta chain moiety. The costimulatory signaling region refers to a portion of the intracellular domain that includes the costimulatory molecule. Costimulatory molecules are cell surface molecules required for efficient response of lymphocytes to antigens, rather than antigen receptors or their ligands.
A linker can be incorporated between the extracellular domain and the transmembrane domain of the CAR, or between the cytoplasmic domain and the transmembrane domain of the CAR. As used herein, the term "linker" generally refers to any oligopeptide or polypeptide that functions to connect a transmembrane domain to an ectodomain or a cytoplasmic domain of a polypeptide chain. The linker may comprise 0-300 amino acids, preferably 2 to 100 amino acids and most preferably 3 to 50 amino acids.
In a preferred embodiment of the invention, the extracellular domain of the CAR provided by the invention comprises an antigen binding domain that targets CD 20. The CARs of the invention, when expressed in T cells, are capable of antigen recognition based on antigen binding specificity. When it binds its associated antigen, it affects the tumor cells, causing the tumor cells to not grow, to be driven to death, or to otherwise be affected, and causing the patient's tumor burden to shrink or be eliminated. The antigen binding domain is preferably fused to an intracellular domain from one or more of the costimulatory molecule and the zeta chain. Preferably, the antigen binding domain is fused to the intracellular domain of the combination of the 4-1BB signaling domain, and the CD3 zeta signaling domain.
In one embodiment, a CD20 targeting CAR of the invention comprises the specific signaling domains of the invention (transmembrane region of ICOS, intracellular signaling domains of 4-1BB and CD3 ζ in tandem). The signaling domains of the invention significantly increase the anti-tumor activity and in vivo persistence of CAR-T cells compared to other means of CD 20-targeted CARs.
In a preferred embodiment of the invention, the amino acid sequence of the Chimeric Antigen Receptor (CAR) provided by the invention is as follows:
MLLLVTSLLLCELPHPAFLLIPQVQLVQSGAEVKKPGASVKVSCKASGYTFTSYNMHWVRQTPGQRLEWMGAIYPGNGDTSYNQKFKGRVTITADTSASTAYMELSSLRSEDTAVYYCARSTYYGGDWYFNVWGQGTTVTVSGGGGSGGGGSGGGGSDIQLTQSPSFLSASVGDRVTITCRASSSVSYIHWYQQKPGKAPKLLIYATSNLASGVPSRFSGSGSGTSFTLTISSLQPEDFA
TYYCQQWTSNPPTFGGGTKVEIKRTVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFQSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEAL
HNHYTQKSLSLSLGKWLPIGCAAFVVVCILGCILICWLTKKKYSSSVHDPNGEYMFMRAVNTAKKSRLTDVTLKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID NO.:10)
the coding DNA sequence is as follows:
ATGCTGCTGCTGGTGACCTCTCTGCTGCTCTGCGAACTGCCTCACCCAGCCTTTCTGCTGATCCCTCAGGTGCAGCTGGTGCAGTCAGGAGCCGAAGTGAAGAAGCCAGGAGCCTCAGTGAAGGTGTCTTGCAAGGCCAGCGGCTACACCTTCACCAGCTACAACATGCATTGGGTCCGGCAGACCCCAGGACAGAGACTCGAGTGGATGGGAGCCATCTACCCCGGCAACGGCGATACCAGCTACAACCAGAAGTTCAAGGGCCGCGTGACCATCACAGCCGATACAAGCGCCAGCACCGCCTACATGGAGCTGTCTAGCCTGAGGAGCGAGGACACAGCCGTGTACTATTGCGCCAGGAGCACCTACTACGGCGGCGATTGGTACTTCAACGTGTGGGGCCAGGGCACAACAGTGACAGTGAGCGGAGGAGGAGGAAGCGGAGGAGGAGGATCTGGAGGAGGCGGCAGCGATATTCAGCTGACCCAGAGCCCTTCTTTTCTGAGCGCCAGCGTGGGCGACAGAGTGACCATCACTTGCAGGGCCAGCAGCAGCGTGTCCTACATCCATTGGTACCAGCAGAAGCCAGGCAAGGCTCCTAAGCTGCTGATCTACGCCACCAGCAACCTGGCCAGCGGCGTGCCTAGCAGATTCAGCGGCAGCGGAAGCGGCACAAGCTTTACCCTGACCATCAGCTCTCTGCAGCCAGAGGACTTCGCCACCTACTACTGCCAGCAGTGGACCAGCAACCCTCCTACATTTGGCGGCGGAACCAAGGTGGAGATCAAGCGGACCGTGGAGAGCAAGTACGGACCTCCTTGTCCTCCTTGTCCAGCTCCAGAGTTTGAGGGCGGACCTAGCGTGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCAGAAGTGACTTGCGTGGTGGTGGACGTGTCTCAGGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAGGTGCACAACGCTAAGACCAAGCCCAGGGAGGAGCAGTTCCAGAGCACCTACAGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGATTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTGTCCAACAAGGGCCTGCCTAGCAGCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCTAGAGAGCCTCAGGTGTACACCCTGCCTCCTAGCCAGGAGGAGATGACCAAGAACCAGGTGTCCCTGACTTGCCTCGTGAAGGGCTTCTACCCCAGCGACATCGCAGTCGAGTGGGAAAGCAACGGCCAGCCCGAGAACAACTACAAGACCACACCTCCAGTGCTGGATAGCGACGGAAGCTTCTTCCTGTACAGCCGGCTGACCGTGGATAAAAGCCGCTGGCAGGAGGGCAACGTGTTCAGTTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCTCGGCAAGTGGCTGCCTATTGGTTGCGCCGCCTTTGTCGTCGTCTGTATCCTCGGCTGCATCCTGATTTGTTGGCTGACCAAGAAGAAGTACAGCAGCAGCGTGCACGACCCCAACGGCGAGTACATGTTCATGCGGGCCGTCAACACCGCCAAGAAGAGCAGGCTGACCGACGTGACACTGAAGAGGGGCCGGAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGCGGCCAGTGCAGACAACACAGGAGGAAGACGGCTGCTCTTGCAGGTTCCCAGAGGAGGAGGAGGGCGGTTGCGAGCTGAGAGTGAAGTTCAGCAGAAGCGCCGACGCTCCAGCTTATCAGCAGGGCCAGAACCAGCTGTACAACGAGCTGAACCTGGGCAGGAGAGAGGAATACGACGTGCTGGACAAGAGGAGGGGCAGAGATCCCGAGATGGGAGGCAAGCCCAGAAGAAAGAACCCCCAGGAGGGCCTGTACAACGAGCTGCAGAAGGACAAGATGGCCGAGGCCTACAGCGAGATTGGCATGAAGGGCGAGAGAAGGAGGGGCAAAGGCCACGACGGACTGTATCAGGGCCTGTCTACCGCCACCAAGGATACCTACGACGCTCTGCACATGCAGGCTCTGCCTCCTAGATGA(SEQ ID NO.:16)
antigen binding domains
In one embodiment, the CAR of the invention comprises a target-specific binding element referred to as an antigen-binding domain. The antigen binding domain of the CAR of the invention is a specific binding element that targets CD 20.
In a preferred embodiment of the invention, the antigen binding domain comprises the heavy chain variable region and the light chain variable region of an anti-CD 20 antibody.
In another preferred embodiment, the amino acid sequence of the antibody heavy chain variable region is as follows:
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYNMHWVRQTPGQRLEWMGAIYPGNGDTSYNQKFKGRVTITADTSASTAYMELSSLRSEDTAVYYCARSTYYGGDWYFNVWGQGTTVTVS(SEQ ID NO.:1);
in another preferred embodiment, the amino acid sequence of the variable region of the antibody light chain is as follows:
DIQLTQSPSFLSASVGDRVTITCRASSSVSYIHWYQQKPGKAPKLLIYATSNLASGVPSRFSGSGSGTSFTLTISSLQPEDFATYYCQQWTSNPPTFGGGTKVEIKRTV(SEQ ID NO.:2);
in a preferred embodiment of the present invention, the amino acid sequence of the linkage between the heavy chain variable region and the light chain variable region is as follows:
GGGGSGGGGSGGGGS(SEQ ID NO.:3)
hinge region and transmembrane region
For the hinge region and transmembrane region (transmembrane domain), the CAR can be designed to include a transmembrane domain fused to the extracellular domain of the CAR. In one embodiment, a transmembrane domain that is naturally associated with one of the domains in the CAR is used. In some examples, the transmembrane domains may be selected or modified by amino acid substitutions to avoid binding such domains to the transmembrane domains of the same or different surface membrane proteins, thereby minimizing interaction with other members of the receptor complex.
In a preferred embodiment of the invention, the hinge region comprises the following amino acid sequence:
ESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFQSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID No.:5)
the coding DNA sequence is as follows:
GAGAGCAAGTACGGACCTCCTTGTCCTCCTTGTCCAGCTCCAGAGTTTGAGGGCGGACCTAGCGTGTTCCTGTTCCCTCCCAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCAGAAGTGACTTGCGTGGTGGTGGACGTGTCTCAGGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAGGTGCACAACGCTAAGACCAAGCCCAGGGAGGAGCAGTTCCAGAGCACCTACAGGGTGGTGTCCGTGCTGACAGTGCTGCACCAGGATTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTGTCCAACAAGGGCCTGCCTAGCAGCATCGAGAAGACCATCAGCAAGGCCAAGGGCCAGCCTAGAGAGCCTCAGGTGTACACCCTGCCTCCTAGCCAGGAGGAGATGACCAAGAACCAGGTGTCCCTGACTTGCCTCGTGAAGGGCTTCTACCCCAGCGACATCGCAGTCGAGTGGGAAAGCAACGGCCAGCCCGAGAACAACTACAAGACCACACCTCCAGTGCTGGATAGCGACGGAAGCTTCTTCCTGTACAGCCGGCTGACCGTGGATAAAAGCCGCTGGCAGGAGGGCAACGTGTTCAGTTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGAGCCTCGGCAAG(SEQ ID NO.:11)
in a preferred embodiment of the invention, the transmembrane region of the CAR of the invention is an ICOS-derived transmembrane region.
In a preferred embodiment of the invention, the amino acid sequence of the ICOS-derived transmembrane region is as follows:
WLPIGCAAFVVVCILGCILICWL(SEQ ID NO.:6)
the coding DNA sequence is as follows:
TGGCTGCCTATTGGTTGCGCAGCTTTCGTCGTCGTCTGCATCCTGGGTTGCATCCTGATTTGCTGGCTG(SEQ ID NO.:12)
intracellular domains
The intracellular domains in the CAR of the invention include the signaling domain of ICOS, the signaling domain of 4-1BB, and the signaling domain of CD3 zeta.
Preferably, the intracellular signaling domain of ICOS comprises the amino acid sequence:
TKKKYSSSVHDPNGEYMFMRAVNTAKKSRLTDVTL(SEQ ID NO.:7)
the coding DNA sequence is as follows:
ACCAAGAAGAAGTACAGCTCTAGCGTGCACGACCCTAACGGCGAGTACATGTTCATGCGGGCCGTCAACACCGCCAAAAAGAGCCGGCTGACCGACGTGACACTG(SEQ ID NO.:13)
preferably, the intracellular signaling domain of 4-1BB comprises the amino acid sequence:
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL(SEQ ID No.:8)
the coding DNA sequence is as follows:
AAGAGGGGCCGGAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGCGGCCAGTGCAGACAACACAGGAGGAAGACGGCTGCTCTTGCAGGTTCCCAGAGGAGGAGGAGGGCGGTTGCGAGCTG(SEQ ID NO.:14)
preferably, the intracellular signaling domain of CD3 ζ comprises the amino acid sequence:
RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR(SEQ ID No.:9)
the coding DNA sequence is as follows:
AGAGTGAAGTTCAGCAGAAGCGCCGACGCTCCAGCTTATCAGCAGGGCCAGAACCAGCTGTACAACGAGCTGAACCTGGGCAGGAGAGAGGAATACGACGTGCTGGACAAGAGGAGGGGCAGAGATCCCGAGATGGGAGGCAAGCCCAGAAGAAAGAACCCCCAGGAGGGCCTGTACAACGAGCTGCAGAAGGACAAGATGGCCGAGGCCTACAGCGAGATTGGCATGAAGGGCGAGAGAAGGAGGGGCAAAGGCCACGACGGACTGTATCAGGGCCTGTCTACCGCCACCAAGGATACCTACGACGCTCTGCACATGCAGGCTCTGCCTCCTAGA(SEQ ID NO.:15)
carrier
The invention also provides DNA constructs encoding the CAR sequences of the invention.
Nucleic acid sequences encoding the desired molecule can be obtained using recombinant methods known in the art, such as, for example, by screening libraries from cells expressing the gene, by obtaining the gene from vectors known to include the gene, or by direct isolation from cells and tissues containing the gene using standard techniques. Alternatively, the gene of interest may be produced synthetically.
The present invention also provides a vector into which the DNA construct of the present invention is inserted. Vectors derived from retroviruses such as lentiviruses are suitable tools for achieving long-term gene transfer, since they allow long-term, stable integration of the transgene and its propagation in daughter cells. Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses such as murine leukemia virus, in that they can transduce non-proliferating cells such as hepatocytes. They also have the advantage of low immunogenicity.
In brief summary, expression of a natural or synthetic nucleic acid encoding a CAR is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or a portion thereof to a promoter and incorporating the construct into an expression vector. The vector is suitable for replication and integration into eukaryotic cells. Typical cloning vectors contain transcriptional and translational terminators, initiation sequences, and promoters that may be used to regulate the expression of the desired nucleic acid sequence.
The expression constructs of the invention may also be used for nucleic acid immunization and gene therapy using standard gene delivery protocols. Methods of gene delivery are known in the art. See, for example, U.S. Pat. nos. 5,399,346, 5,580,859, 5,589,466, which are incorporated herein by reference in their entirety. In another embodiment, the invention provides a gene therapy vector.
The nucleic acid can be cloned into many types of vectors. For example, the nucleic acid can be cloned into vectors including, but not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. Specific vectors of interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
Further, the expression vector may be provided to the cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Sambrook et al (2001, molecular cloning. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. Generally, suitable vectors comprise an origin of replication, a promoter sequence, a convenient restriction enzyme site, and one or more selectable markers that function in at least one organism (e.g., WO01/96584, WO01/29058; and U.S. Pat. No. 6,326,193).
Many virus-based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. The selected gene can be inserted into a vector and packaged into a retroviral particle using techniques known in the art. The recombinant virus can then be isolated and delivered to the subject cells in vivo or ex vivo. Many retroviral systems are known in the art. In some embodiments, an adenoviral vector is used. Many adenoviral vectors are known in the art. In one embodiment, a lentiviral vector is used.
Additional promoter elements, such as enhancers, may regulate the frequency of transcription initiation. Typically, these are located in the 30-110bp region upstream of the start site, although many promoters have recently been shown to also contain functional elements downstream of the start site. The spacing between promoter elements is often flexible so that promoter function is maintained when the elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased by 50bp apart, and activity begins to decline. Depending on the promoter, it appears that the individual elements may function cooperatively or independently to initiate transcription.
An example of a suitable promoter is the immediate early Cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto. Another example of a suitable promoter is elongation growth factor-1 α (EF-1 α). However, other constitutive promoter sequences may also be used, including, but not limited to, the simian virus 40 (SV 40) early promoter, the mouse mammary cancer virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the Epstein-Barr (Epstein-Barr) virus immediate early promoter, the rous sarcoma virus promoter, and human gene promoters, such as, but not limited to, the actin promoter, myosin promoter, heme promoter, and creatine kinase promoter. Further, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch that is capable of turning on expression of a polynucleotide sequence operably linked to the inducible promoter when such expression is desired, or turning off expression when expression is not desired. Examples of inducible promoters include, but are not limited to, the metallothionein promoter, the glucocorticoid promoter, the progesterone promoter, and the tetracycline promoter.
To assess the expression of the CAR polypeptide or portion thereof, the expression vector introduced into the cells can also comprise either or both of a selectable marker gene or a reporter gene to facilitate identification and selection of expressing cells from a population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both the selectable marker and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in a host cell. Useful selectable markers include, for example, antibiotic resistance genes, such as neo and the like.
Reporter genes are used to identify potentially transfected cells and to evaluate the functionality of regulatory sequences. Typically, the reporter gene is the following: which is not present in or expressed by the recipient organism or tissue and which encodes a polypeptide whose expression is clearly indicated by some readily detectable property, such as enzymatic activity. After the DNA has been introduced into the recipient cell, the expression of the reporter gene is assayed at an appropriate time. Suitable reporter genes may include genes encoding luciferase, β -galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein genes (e.g., ui-Tei et al, 2000febs letters 479. Suitable expression systems are well known and can be prepared using known techniques or obtained commercially. Generally, the construct with the minimum of 5 flanking regions that showed the highest level of reporter gene expression was identified as the promoter. Such promoter regions can be linked to reporter genes and used to evaluate the ability of an agent to modulate promoter-driven transcription.
Methods for introducing and expressing genes into cells are known in the art. In the context of expression vectors, the vector may be readily introduced into a host cell by any method known in the art, e.g., mammalian, bacterial, yeast or insect cells. For example, the expression vector may be transferred into a host cell by physical, chemical or biological means.
Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well known in the art. See, e.g., sambrook et al (2001, molecular cloning. A preferred method for introducing the polynucleotide into a host cell is calcium phosphate transfection.
Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors. Viral vectors, particularly retroviral vectors, have become the most widely used method for inserting genes into mammalian, e.g., human, cells. Other viral vectors may be derived from lentiviruses, poxviruses, herpes simplex virus I, adenoviruses, adeno-associated viruses, and the like. See, for example, U.S. Pat. nos. 5,350,674 and 5,585,362.
Chemical means of introducing polynucleotides into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Exemplary colloidal systems for use as delivery vehicles in vitro and in vivo are liposomes (e.g., artificial membrane vesicles).
In the case of non-viral delivery systems, an exemplary delivery vehicle is a liposome. Lipid formulations are contemplated for use to introduce nucleic acids into host cells (ex vivo or in vivo). In another aspect, the nucleic acid can be associated with a lipid. The nucleic acid associated with the lipid may be encapsulated in the aqueous interior of the liposome, dispersed within the lipid bilayer of the liposome, attached to the liposome via a linker molecule associated with both the liposome and the oligonucleotide, entrapped in the liposome, complexed with the liposome, dispersed in a solution comprising the lipid, mixed with the lipid, associated with the lipid, contained as a suspension in the lipid, contained in or complexed with a micelle, or otherwise associated with the lipid. The lipid, lipid/DNA or lipid/expression vector associated with the composition is not limited to any particular structure in solution. For example, they may be present in bilayer structures, either as micelles or with a "collapsed" structure. They may also simply be dispersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances, which may be naturally occurring or synthetic lipids. For example, lipids include fatty droplets that occur naturally in the cytoplasm as well as such compounds that contain long-chain aliphatic hydrocarbons and their derivatives such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
Therapeutic applications
The invention includes cells (e.g., T cells) transduced with a Lentiviral Vector (LV) encoding a CAR of the invention. The transduced T cells can elicit a CAR-mediated T-cell response.
Accordingly, the present invention also provides a method of stimulating a T cell-mediated immune response to a target cell population or tissue of a mammal comprising the steps of: administering to a mammal a T cell expressing a CAR of the invention.
In one embodiment, the invention includes a class of cell therapies in which T cells are genetically modified to express a CAR of the invention, and the CAR-T cells are injected into a recipient in need thereof. The injected cells are capable of killing tumor cells of the recipient. Unlike antibody therapy, CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control.
In one embodiment, the CAR-T cells of the invention can undergo robust in vivo T cell expansion and can last for an extended amount of time. In addition, the CAR-mediated immune response can be part of an adoptive immunotherapy step, wherein the CAR-modified T cell induces an immune response specific to the antigen binding domain in the CAR. For example, anti-CD 20 CAR-T cells elicit a specific immune response against CD 20-expressing cells.
Although the data disclosed herein specifically disclose lentiviral vectors comprising an anti-CD 20scFv, a human Fc hinge region, an ICOS transmembrane and intracellular region, and 4-1BB and CD3 zeta signaling domains, the invention should be construed to include any number of variations on each of the construct components.
Treatable cancers include tumors that are not vascularized or have not substantially vascularized, as well as vascularized tumors. The cancer may comprise a non-solid tumor (such as a hematological tumor, e.g., leukemia and lymphoma) or may comprise a solid tumor. The types of cancer treated with the CAR of the invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemias or lymphoid malignancies, benign and malignant tumors, e.g., sarcomas, carcinomas, and melanomas. Adult tumors/cancers and pediatric tumors/cancers are also included.
Hematologic cancers are cancers of the blood or bone marrow. Examples of hematologic (or hematological) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelogenous leukemia and myeloblastic, promyelocytic, granulo-monocytic, monocytic and erythrocytic leukemias), chronic leukemias (such as chronic myelogenous (granulocytic) leukemia, chronic myelogenous leukemia and chronic lymphocytic leukemia), polycythemia vera, lymphoma, hodgkin's disease, non-hodgkin's lymphoma (indolent and higher forms), multiple myeloma, waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia, and myelodysplasia.
A solid tumor is an abnormal mass of tissue that generally does not contain cysts or fluid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the cell types that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include fibrosarcoma, myxosarcoma, liposarcoma mesothelioma, lymphoid malignancies, pancreatic cancer, ovarian cancer.
The CAR-modified T cells of the invention may also be used as a type of vaccine for ex vivo immunization and/or in vivo therapy of mammals. Preferably, the mammal is a human.
For ex vivo immunization, at least one of the following occurs in vitro prior to administration of the cells into a mammal: i) Expanding the cell, ii) introducing a nucleic acid encoding the CAR into the cell, and/or iii) cryopreserving the cell.
Ex vivo procedures are well known in the art and are discussed more fully below. Briefly, cells are isolated from a mammal (preferably a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. The CAR-modified cells can be administered to a mammalian recipient to provide a therapeutic benefit. The mammalian recipient can be a human, and the CAR-modified cells can be autologous with respect to the recipient. Alternatively, the cell may be allogeneic, syngeneic (syngeneic) or xenogeneic with respect to the recipient.
In addition to using cell-based vaccines for ex vivo immunization, the present invention also provides compositions and methods of in vivo immunization to elicit an immune response against an antigen in a patient.
Generally, cells activated and expanded as described herein are useful for the treatment and prevention of diseases arising in immunocompromised individuals. In particular, the CAR-modified T cells of the invention are used to treat CCL. In certain embodiments, the cells of the invention are used to treat a patient at risk of developing CCL. Accordingly, the invention provides a method of treating or preventing CCL, comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-modified T cell of the invention.
The CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2, IL-17 or other cytokines or cell populations. Briefly, a pharmaceutical composition of the invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients. Such compositions may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids such as glycine; an antioxidant; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The compositions of the present invention are preferably formulated for intravenous administration.
The pharmaceutical compositions of the present invention may be administered in a manner suitable for the disease to be treated (or prevented). The number and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease-although the appropriate dosage may be determined by clinical trials.
When referring to an "immunologically effective amount", "anti-tumor effective amount", "tumor-inhibiting effective amount", or "therapeutic amount", the precise amount of the composition of the invention to be administered can be determined by a physician, taking into account the age, weight, tumor size, extent of infection or metastasis and individual variability of the condition of the patient (subject). It is possible to generally point out: pharmaceutical compositions comprising T cells described herein can be in the range of 10 4 To 10 9 Dosage of individual cells/kg body weight, preferably 10 5 To 10 6 Doses of individual cells per kg body weight (including all integer values within those ranges) are administered. T cell compositionsMultiple administrations at these doses are also possible. Cells can be administered by using infusion techniques well known in immunotherapy (see, e.g., rosenberg et al, new Eng.J.of Med.319:1676, 1988). Optimal dosages and treatment regimens for a particular patient can be readily determined by those skilled in the medical arts by monitoring the patient for signs of disease and adjusting the treatment accordingly.
Administration of the subject composition may be carried out in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or transplantation. The compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodal, intraspinally, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one embodiment, the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell composition of the invention is preferably administered by i.v. injection. The composition of T cells can be injected directly into the tumor, lymph node or site of infection.
In certain embodiments of the invention, cells activated and expanded using the methods described herein or other methods known in the art for expanding T cells to therapeutic levels are administered to a patient in conjunction with (e.g., prior to, concurrently with, or subsequent to) any number of relevant treatment modalities, including but not limited to treatment with: such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as ARA-C) or natalizumab treatment for MS patients or efavirenz treatment for psoriasis patients or other treatment for PML patients. In further embodiments, the T cells of the invention may be used in combination with: chemotherapy, radiation, immunosuppressive agents such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil, and FK506, antibodies, or other immunotherapeutic agents. In a further embodiment, the cell composition of the invention is administered to the patient in conjunction with (e.g., prior to, concurrently with, or subsequent to) bone marrow transplantation with a chemotherapeutic agent such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide. For example, in one embodiment, the subject may undergo standard treatment with high-dose chemotherapy followed by peripheral blood stem cell transplantation. In some embodiments, after transplantation, the subject receives an injection of the expanded immune cells of the invention. In an additional embodiment, the expanded cells are administered pre-or post-surgery.
The dosage of the above treatments administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The dosage rates for human administration can be effected in accordance with accepted practice in the art. Typically, 1X 10 may be administered per treatment or per course of treatment 6 1 to 10 10 The modified T cells of the invention (e.g., CAR-T19 cells) are administered to a patient, for example, by intravenous infusion.
The main advantages of the invention include:
1) The novel CD20-CAR vectors of the humanized CD20 antibodies of the present invention can target B cell malignancies of the CD20 antigen;
2) The novel CD20-CAR vector of the humanized CD20 antibody can avoid the immunogenicity generated in a human body by CD20-CAR-T cells constructed by murine CD20scFv, and improve the clinical curative effect;
3) The CD20-CAR-T cells constructed by using the CD20-CAR vector constructed by the invention show very good killing effect in vitro.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally followed by conventional conditions, such as Sambrook et al, molecular cloning: the conditions described in the Laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer's recommendations. Unless otherwise indicated, percentages and parts are percentages and parts by weight.
Example 1.
Humanized CD20-CAR-T transfection efficiency
The human Fc antibody of the invention detects the CAR positive rate of CD20-CAR-T cells, and the result is shown in figure 2, and the positive rate of humanized CD20-CAR-T is 28.49%.
Example 2.
Cytotoxicity of humanized CD20-CAR-T cells on CD20 overexpressing K562 cells
The inventors constructed CD 20-overexpressing K562 (K562-CD 20) cells as target cells and evaluated the killing function of humanized CD20-CAR-T cells in vitro with a potency to target ratio of 0.5, 1, 5, 1, 10. K562 cells were negative target cells and T cells were control effector cells.
The results are shown in fig. 3, compared with the control T cells, the CD20-CAR-T cells have significant specific killing effect on the K562-CD20 cells (CD 20 positive) but no specific killing effect on the K562 cells (CD 20 negative), which indicates that the constructed humanized CD20-CAR-T cells can generate specific cytotoxicity on the overexpressed CD20 positive K562-CD20 cells in vitro but have no specific cytotoxicity on the control K562 cells.
Example 3.
Cytotoxicity of humanized CD20-CAR-T cells against B-lymphoblastic leukemia cells (Nalm-6) and lymphoma cells (Raji) naturally expressing CD20
We performed in vitro killing experiments on B-lymphocytic leukemia (Nalm-6) and lymphoma (Raji) cells with the constructed humanized CD20-CAR-T cells at a potency to target ratio of 0.5, 1, 5. T cells were control effector cells.
The results are shown in fig. 4, and compared with the control T cells, the CD20-CAR-T cells have obvious specific killing effect on both Raji and Nalm-6, which indicates that the constructed humanized CD20-CAR-T cells have obvious cytotoxicity on the Raji and Nalm-6 cells which are positive for CD20 in vitro.
Example 4.
Cytokine secretion after incubation of CD20-CAR-T cells with Raji and Nalm-6 cells
The inventors further analyzed the release of cytokines after incubation of CD20-CAR-T cells with Raji and Nalm-6 cells, with the effective target ratio of CD20-CAR-T cells to target cells being 1 and 5 for the 1,t cell group as control group, and the cytokines tested were IL-2, IFN- γ, TNF- α, granzyme B.
Results as shown in fig. 5, T cells produced substantially no or very low cytokine production after co-incubation with target cells, whereas CD20-CAR-T cells produced significant cytokine release after incubation with Raji and Nalm-6 cells, demonstrating that CD20-CAR-T cells are specifically activated by CD 20-positive tumor cells.
All documents mentioned in this application are incorporated by reference in this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.
Sequence listing
<110> Bosheng Ji medicine technology (Suzhou) Co., ltd
<120> construction and application of novel CD20-CAR vector of anti-CD 20 antibody
<130> P2018-2464
<160> 16
<170> PatentIn version 3.5
<210> 1
<211> 120
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Arg Gln Thr Pro Gly Gln Arg Leu Glu Trp Met
35 40 45
Gly Ala Ile Tyr Pro Gly Asn Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Thr Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Ser Thr Tyr Tyr Gly Gly Asp Trp Tyr Phe Asn Val Trp Gly
100 105 110
Gln Gly Thr Thr Val Thr Val Ser
115 120
<210> 2
<211> 109
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Asp Ile Gln Leu Thr Gln Ser Pro Ser Phe Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Ser Ser Val Ser Tyr Ile
20 25 30
His Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr
35 40 45
Ala Thr Ser Asn Leu Ala Ser Gly Val Pro Ser Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu
65 70 75 80
Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Trp Thr Ser Asn Pro Pro Thr
85 90 95
Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val
100 105
<210> 3
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15
<210> 4
<211> 22
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Met Leu Leu Leu Val Thr Ser Leu Leu Leu Cys Glu Leu Pro His Pro
1 5 10 15
Ala Phe Leu Leu Ile Pro
20
<210> 5
<211> 229
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe
1 5 10 15
Glu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr
20 25 30
Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val
35 40 45
Ser Gln Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val
50 55 60
Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Gln Ser
65 70 75 80
Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu
85 90 95
Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser
100 105 110
Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro
115 120 125
Gln Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln
130 135 140
Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala
145 150 155 160
Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr
165 170 175
Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu
180 185 190
Thr Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser
195 200 205
Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser
210 215 220
Leu Ser Leu Gly Lys
225
<210> 6
<211> 23
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Trp Leu Pro Ile Gly Cys Ala Ala Phe Val Val Val Cys Ile Leu Gly
1 5 10 15
Cys Ile Leu Ile Cys Trp Leu
20
<210> 7
<211> 35
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Thr Lys Lys Lys Tyr Ser Ser Ser Val His Asp Pro Asn Gly Glu Tyr
1 5 10 15
Met Phe Met Arg Ala Val Asn Thr Ala Lys Lys Ser Arg Leu Thr Asp
20 25 30
Val Thr Leu
35
<210> 8
<211> 42
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
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> 9
<211> 112
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Gln 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> 10
<211> 707
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 10
Met Leu Leu Leu Val Thr Ser Leu Leu Leu Cys Glu Leu Pro His Pro
1 5 10 15
Ala Phe Leu Leu Ile Pro Gln Val Gln Leu Val Gln Ser Gly Ala Glu
20 25 30
Val Lys Lys Pro Gly Ala Ser Val Lys Val Ser Cys Lys Ala Ser Gly
35 40 45
Tyr Thr Phe Thr Ser Tyr Asn Met His Trp Val Arg Gln Thr Pro Gly
50 55 60
Gln Arg Leu Glu Trp Met Gly Ala Ile Tyr Pro Gly Asn Gly Asp Thr
65 70 75 80
Ser Tyr Asn Gln Lys Phe Lys Gly Arg Val Thr Ile Thr Ala Asp Thr
85 90 95
Ser Ala Ser Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp
100 105 110
Thr Ala Val Tyr Tyr Cys Ala Arg Ser Thr Tyr Tyr Gly Gly Asp Trp
115 120 125
Tyr Phe Asn Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Gly Gly
130 135 140
Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ile Gln
145 150 155 160
Leu Thr Gln Ser Pro Ser Phe Leu Ser Ala Ser Val Gly Asp Arg Val
165 170 175
Thr Ile Thr Cys Arg Ala Ser Ser Ser Val Ser Tyr Ile His Trp Tyr
180 185 190
Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Ala Thr Ser
195 200 205
Asn Leu Ala Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly
210 215 220
Thr Ser Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala
225 230 235 240
Thr Tyr Tyr Cys Gln Gln Trp Thr Ser Asn Pro Pro Thr Phe Gly Gly
245 250 255
Gly Thr Lys Val Glu Ile Lys Arg Thr Val Glu Ser Lys Tyr Gly Pro
260 265 270
Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Glu Gly Gly Pro Ser Val
275 280 285
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
290 295 300
Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu
305 310 315 320
Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
325 330 335
Thr Lys Pro Arg Glu Glu Gln Phe Gln Ser Thr Tyr Arg Val Val Ser
340 345 350
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
355 360 365
Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile
370 375 380
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
385 390 395 400
Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
405 410 415
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
420 425 430
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
435 440 445
Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg
450 455 460
Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
465 470 475 480
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys Trp
485 490 495
Leu Pro Ile Gly Cys Ala Ala Phe Val Val Val Cys Ile Leu Gly Cys
500 505 510
Ile Leu Ile Cys Trp Leu Thr Lys Lys Lys Tyr Ser Ser Ser Val His
515 520 525
Asp Pro Asn Gly Glu Tyr Met Phe Met Arg Ala Val Asn Thr Ala Lys
530 535 540
Lys Ser Arg Leu Thr Asp Val Thr Leu Lys Arg Gly Arg Lys Lys Leu
545 550 555 560
Leu Tyr Ile Phe Lys Gln Pro Phe Met Arg Pro Val Gln Thr Thr Gln
565 570 575
Glu Glu Asp Gly Cys Ser Cys Arg Phe Pro Glu Glu Glu Glu Gly Gly
580 585 590
Cys Glu Leu Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr
595 600 605
Gln Gln Gly Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg
610 615 620
Glu Glu Tyr Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met
625 630 635 640
Gly Gly Lys Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu
645 650 655
Leu Gln Lys Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys
660 665 670
Gly Glu Arg Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu
675 680 685
Ser Thr Ala Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu
690 695 700
Pro Pro Arg
705
<210> 11
<211> 687
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 11
gagagcaagt acggacctcc ttgtcctcct tgtccagctc cagagtttga gggcggacct 60
agcgtgttcc tgttccctcc caagcccaag gacaccctga tgatcagccg gaccccagaa 120
gtgacttgcg tggtggtgga cgtgtctcag gaggaccccg aggtgcagtt caattggtac 180
gtggacggcg tggaggtgca caacgctaag accaagccca gggaggagca gttccagagc 240
acctacaggg tggtgtccgt gctgacagtg ctgcaccagg attggctgaa cggcaaggag 300
tacaagtgca aggtgtccaa caagggcctg cctagcagca tcgagaagac catcagcaag 360
gccaagggcc agcctagaga gcctcaggtg tacaccctgc ctcctagcca ggaggagatg 420
accaagaacc aggtgtccct gacttgcctc gtgaagggct tctaccccag cgacatcgca 480
gtcgagtggg aaagcaacgg ccagcccgag aacaactaca agaccacacc tccagtgctg 540
gatagcgacg gaagcttctt cctgtacagc cggctgaccg tggataaaag ccgctggcag 600
gagggcaacg tgttcagttg cagcgtgatg cacgaggccc tgcacaacca ctacacccag 660
aagagcctga gcctgagcct cggcaag 687
<210> 12
<211> 69
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 12
tggctgccta ttggttgcgc agctttcgtc gtcgtctgca tcctgggttg catcctgatt 60
tgctggctg 69
<210> 13
<211> 105
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 13
accaagaaga agtacagctc tagcgtgcac gaccctaacg gcgagtacat gttcatgcgg 60
gccgtcaaca ccgccaaaaa gagccggctg accgacgtga cactg 105
<210> 14
<211> 126
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 14
aagaggggcc ggaagaagct gctgtacatc ttcaagcagc ccttcatgcg gccagtgcag 60
acaacacagg aggaagacgg ctgctcttgc aggttcccag aggaggagga gggcggttgc 120
gagctg 126
<210> 15
<211> 336
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 15
agagtgaagt tcagcagaag cgccgacgct ccagcttatc agcagggcca gaaccagctg 60
tacaacgagc tgaacctggg caggagagag gaatacgacg tgctggacaa gaggaggggc 120
agagatcccg agatgggagg caagcccaga agaaagaacc cccaggaggg cctgtacaac 180
gagctgcaga aggacaagat ggccgaggcc tacagcgaga ttggcatgaa gggcgagaga 240
aggaggggca aaggccacga cggactgtat cagggcctgt ctaccgccac caaggatacc 300
tacgacgctc tgcacatgca ggctctgcct cctaga 336
<210> 16
<211> 2124
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 16
atgctgctgc tggtgacctc tctgctgctc tgcgaactgc ctcacccagc ctttctgctg 60
atccctcagg tgcagctggt gcagtcagga gccgaagtga agaagccagg agcctcagtg 120
aaggtgtctt gcaaggccag cggctacacc ttcaccagct acaacatgca ttgggtccgg 180
cagaccccag gacagagact cgagtggatg ggagccatct accccggcaa cggcgatacc 240
agctacaacc agaagttcaa gggccgcgtg accatcacag ccgatacaag cgccagcacc 300
gcctacatgg agctgtctag cctgaggagc gaggacacag ccgtgtacta ttgcgccagg 360
agcacctact acggcggcga ttggtacttc aacgtgtggg gccagggcac aacagtgaca 420
gtgagcggag gaggaggaag cggaggagga ggatctggag gaggcggcag cgatattcag 480
ctgacccaga gcccttcttt tctgagcgcc agcgtgggcg acagagtgac catcacttgc 540
agggccagca gcagcgtgtc ctacatccat tggtaccagc agaagccagg caaggctcct 600
aagctgctga tctacgccac cagcaacctg gccagcggcg tgcctagcag attcagcggc 660
agcggaagcg gcacaagctt taccctgacc atcagctctc tgcagccaga ggacttcgcc 720
acctactact gccagcagtg gaccagcaac cctcctacat ttggcggcgg aaccaaggtg 780
gagatcaagc ggaccgtgga gagcaagtac ggacctcctt gtcctccttg tccagctcca 840
gagtttgagg gcggacctag cgtgttcctg ttccctccca agcccaagga caccctgatg 900
atcagccgga ccccagaagt gacttgcgtg gtggtggacg tgtctcagga ggaccccgag 960
gtgcagttca attggtacgt ggacggcgtg gaggtgcaca acgctaagac caagcccagg 1020
gaggagcagt tccagagcac ctacagggtg gtgtccgtgc tgacagtgct gcaccaggat 1080
tggctgaacg gcaaggagta caagtgcaag gtgtccaaca agggcctgcc tagcagcatc 1140
gagaagacca tcagcaaggc caagggccag cctagagagc ctcaggtgta caccctgcct 1200
cctagccagg aggagatgac caagaaccag gtgtccctga cttgcctcgt gaagggcttc 1260
taccccagcg acatcgcagt cgagtgggaa agcaacggcc agcccgagaa caactacaag 1320
accacacctc cagtgctgga tagcgacgga agcttcttcc tgtacagccg gctgaccgtg 1380
gataaaagcc gctggcagga gggcaacgtg ttcagttgca gcgtgatgca cgaggccctg 1440
cacaaccact acacccagaa gagcctgagc ctgagcctcg gcaagtggct gcctattggt 1500
tgcgccgcct ttgtcgtcgt ctgtatcctc ggctgcatcc tgatttgttg gctgaccaag 1560
aagaagtaca gcagcagcgt gcacgacccc aacggcgagt acatgttcat gcgggccgtc 1620
aacaccgcca agaagagcag gctgaccgac gtgacactga agaggggccg gaagaagctg 1680
ctgtacatct tcaagcagcc cttcatgcgg ccagtgcaga caacacagga ggaagacggc 1740
tgctcttgca ggttcccaga ggaggaggag ggcggttgcg agctgagagt gaagttcagc 1800
agaagcgccg acgctccagc ttatcagcag ggccagaacc agctgtacaa cgagctgaac 1860
ctgggcagga gagaggaata cgacgtgctg gacaagagga ggggcagaga tcccgagatg 1920
ggaggcaagc ccagaagaaa gaacccccag gagggcctgt acaacgagct gcagaaggac 1980
aagatggccg aggcctacag cgagattggc atgaagggcg agagaaggag gggcaaaggc 2040
cacgacggac tgtatcaggg cctgtctacc gccaccaagg atacctacga cgctctgcac 2100
atgcaggctc tgcctcctag atga 2124

Claims (28)

1. An anti-CD 20 chimeric antigen receptor, wherein the antigen binding domain of the chimeric antigen receptor, i.e., the scFv comprises an antibody heavy chain variable region as set forth in SEQ ID No.1 and an antibody light chain variable region as set forth in SEQ ID No. 2;
the structure of the chimeric antigen receptor is shown as the following formula:
L-scFv-H-TM-C-CD3ζ
wherein the content of the first and second substances,
l is an optional leader sequence, i.e. a signal peptide sequence;
scFv is an antigen binding domain;
h is a hinge region;
TM is a transmembrane domain;
c is a costimulatory signal molecule;
CD3 ζ is a cytoplasmic signaling sequence derived from CD3 ζ;
"-" is a linker peptide or peptide bond.
2. The chimeric antigen receptor of claim 1, wherein the amino acid sequence of the linker peptide is set forth in SEQ ID No. 3.
3. The chimeric antigen receptor of claim 1, wherein the sequence of L is as set forth in SEQ ID No. 4.
4. The chimeric antigen receptor of claim 1, wherein the sequence of H is as set forth in SEQ ID No. 5.
5. The chimeric antigen receptor according to claim 1, wherein the sequence of the TM comprises a transmembrane region derived from ICOS.
6. The chimeric antigen receptor of claim 5, wherein the sequence of TM is as shown in SEQ ID No. 6.
7. The chimeric antigen receptor according to claim 1, wherein the costimulatory signal molecule comprises an ICOS-derived costimulatory signal molecule, and/or a 4-1 BB-derived costimulatory signal molecule.
8. The chimeric antigen receptor of claim 7, wherein the amino acid sequence of the ICOS-derived costimulatory signal molecule is as set forth in SEQ ID No. 7.
9. The chimeric antigen receptor of claim 7, wherein the amino acid sequence of the 4-1 BB-derived costimulatory signal molecule is set forth in SEQ ID No. 8.
10. The chimeric antigen receptor of claim 1, wherein the sequence of CD3 ζ is as set forth in SEQ ID No. 9.
11. The chimeric antigen receptor of claim 1, wherein the sequence of the chimeric antigen receptor is set forth in SEQ ID No. 10.
12. A nucleic acid molecule encoding the chimeric antigen receptor of claim 1.
13. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule comprises a nucleic acid sequence encoding a hinge region selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 5;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 11.
14. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule comprises a nucleic acid sequence encoding a transmembrane domain selected from the group consisting of seq id nos:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 6;
(b) The sequence is shown in SEQ ID NO. 12.
15. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule comprises a costimulatory signal molecule coding sequence, said costimulatory signal molecule coding sequence comprising an ICOS-derived costimulatory signal molecule coding sequence, and/or a 4-1 BB-derived costimulatory signal molecule coding sequence, wherein
The ICOS-derived costimulatory signal molecule coding sequence is selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 7;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 13;
the coding sequence of the 4-1BB derived costimulatory signal molecule is selected from the group consisting of:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 8;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 14.
16. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule comprises a nucleic acid sequence encoding an intracellular signaling domain of CD3 ζ selected from the group consisting of seq id nos:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 9;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 15.
17. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule comprises a nucleic acid sequence selected from the group consisting of seq id no:
(a) A polynucleotide encoding a polypeptide as set forth in SEQ ID No. 10;
(b) A polynucleotide having a sequence as shown in SEQ ID No. 16.
18. The nucleic acid molecule of claim 12, wherein said nucleic acid molecule is isolated.
19. The nucleic acid molecule of claim 12, wherein the sequence of the nucleic acid molecule is set forth in SEQ ID No. 16.
20. A vector comprising the nucleic acid molecule of claim 12.
21. The vector of claim 20, wherein said vector is a lentiviral vector.
22. A host cell comprising the vector of claim 20 or having integrated into its chromosome an exogenous nucleic acid molecule of claim 12.
23. The host cell of claim 22, wherein the cell is an isolated cell, and/or the cell is a genetically engineered cell.
24. The host cell of claim 22, wherein the cell is a mammalian cell.
25. The host cell of claim 22, wherein the cell is a T cell.
26. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and the chimeric antigen receptor of claim 1, the nucleic acid molecule of claim 12, the vector of claim 20, or the host cell of claim 22.
27. Use of the chimeric antigen receptor of claim 1, the nucleic acid molecule of claim 12, the vector of claim 20, or the host cell of claim 22 for the preparation of a medicament or formulation for the treatment of a CD 20-positive tumor;
the CD20 positive tumor is a B lymphocyte leukemia or lymphoma.
28. A method of making a CAR-T cell expressing the chimeric antigen receptor of claim 1,
the method comprises the following steps: transferring the nucleic acid molecule of claim 12 or the vector of claim 20 into a T cell, thereby obtaining the CAR-T cell.
CN201811652891.3A 2018-12-29 2018-12-29 Construction and application of novel CD20-CAR vector of anti-CD 20 antibody Active CN109593137B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811652891.3A CN109593137B (en) 2018-12-29 2018-12-29 Construction and application of novel CD20-CAR vector of anti-CD 20 antibody

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811652891.3A CN109593137B (en) 2018-12-29 2018-12-29 Construction and application of novel CD20-CAR vector of anti-CD 20 antibody

Publications (2)

Publication Number Publication Date
CN109593137A CN109593137A (en) 2019-04-09
CN109593137B true CN109593137B (en) 2023-04-14

Family

ID=65965695

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811652891.3A Active CN109593137B (en) 2018-12-29 2018-12-29 Construction and application of novel CD20-CAR vector of anti-CD 20 antibody

Country Status (1)

Country Link
CN (1) CN109593137B (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016115482A1 (en) * 2015-01-16 2016-07-21 Novartis Pharma Ag Phosphoglycerate kinase 1 (pgk) promoters and methods of use for expressing chimeric antigen receptor
CN108395482A (en) * 2017-02-08 2018-08-14 西比曼生物科技(上海)有限公司 A kind of activity identification of targeting CD20 antigen Chimeric antigen receptors built and its be engineered T cell
CN108395481A (en) * 2017-02-08 2018-08-14 西比曼生物科技(上海)有限公司 A kind of activity identification of the CAR of targeting CD20 built and its be engineered T cell
CN109651509A (en) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 The humanization monoclonal antibody and its preparation of anti-CD20

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016115482A1 (en) * 2015-01-16 2016-07-21 Novartis Pharma Ag Phosphoglycerate kinase 1 (pgk) promoters and methods of use for expressing chimeric antigen receptor
CN108395482A (en) * 2017-02-08 2018-08-14 西比曼生物科技(上海)有限公司 A kind of activity identification of targeting CD20 antigen Chimeric antigen receptors built and its be engineered T cell
CN108395481A (en) * 2017-02-08 2018-08-14 西比曼生物科技(上海)有限公司 A kind of activity identification of the CAR of targeting CD20 built and its be engineered T cell
CN109651509A (en) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 The humanization monoclonal antibody and its preparation of anti-CD20

Also Published As

Publication number Publication date
CN109593137A (en) 2019-04-09

Similar Documents

Publication Publication Date Title
CN109575143B (en) Bispecific CD20-CD19-CAR and application thereof
CN110760007B (en) CD7-CAR-T cell and preparation and application thereof
JP7148611B2 (en) CHIMERIC ANTIGEN RECEPTOR TARGETING BCMA AND METHOD FOR PRODUCING AND USE THEREOF
EP3599251B1 (en) Construction of chimeric antigen receptor targeting cd20 antigen and activity identification of engineered t cells thereof
US20110044953A1 (en) Methods and Compositions for Localized Secretion of Anti-CTLA-4 Antibodies
CN115141279A (en) Monoclonal antibodies against B7-H3 and their use in cell therapy
CN112778427B (en) Bispecific CS1-BCMA CAR-T cells and uses thereof
CN108395481B (en) Construction of CD 20-targeted CAR and activity identification of engineered T cell thereof
JP7178568B2 (en) CAR immune cells targeting carcinoembryonic antigen-related cell adhesion molecule 6 for treating cancer
CN111378625A (en) Preparation and application of CXCL13 chemotactic CAR-T cell
CN110054698B (en) Construction and application of novel CD19-CAR vector of anti-CD 19 antibody
CN107936120B (en) CD19 targeted chimeric antigen receptor and preparation method and application thereof
CN113087806A (en) Novel CAR-T cells targeting multiple tumors, and preparation and methods thereof
WO2023016524A1 (en) Combined her2 and meso dual-target car-t vector, construction method therefor and application thereof in cancer
US20210395362A1 (en) Car-t cells with humanized cd19 scfv with mutation in cdr 1 region
CN109593137B (en) Construction and application of novel CD20-CAR vector of anti-CD 20 antibody
JP2023521218A (en) CD22-targeted chimeric antigen receptor, its preparation method, and its application
CN114685683A (en) GD 2-targeted CAR-T cells and preparation and application thereof
WO2022151959A1 (en) Car-t cell targeting b7-h3 and application thereof in treatment of acute myeloid leukemia
CN116462770B (en) Humanized antibody of CD19, CAR-T cell expressing bispecific chimeric antigen receptor and application thereof
CN116444669B (en) Humanized antibodies targeting BCMA CAR-T cells
WO2024051641A1 (en) Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof
CN115975056A (en) Construction of TSHR-targeted CAR-T cells Using native protein TSH as antigen binding site
CN114685684A (en) MUC1-Tn chimeric antigen receptor modified V gamma 9V delta 2T cell and application thereof
CN116178562A (en) Preparation and application of chimeric antigen receptor immune cells constructed based on EFNA1

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