CN112521506A - CD123 antigen binding fragment and application thereof - Google Patents

CD123 antigen binding fragment and application thereof Download PDF

Info

Publication number
CN112521506A
CN112521506A CN202011461164.6A CN202011461164A CN112521506A CN 112521506 A CN112521506 A CN 112521506A CN 202011461164 A CN202011461164 A CN 202011461164A CN 112521506 A CN112521506 A CN 112521506A
Authority
CN
China
Prior art keywords
chimeric antigen
antigen receptor
antibody
cells
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202011461164.6A
Other languages
Chinese (zh)
Other versions
CN112521506B (en
Inventor
狄升蒙
童东阳
周建国
冯冬歌
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 Biological Medicine Co ltd
Original Assignee
Huadao Shanghai Biological Medicine 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 Biological Medicine Co ltd filed Critical Huadao Shanghai Biological Medicine Co ltd
Priority to CN202011461164.6A priority Critical patent/CN112521506B/en
Publication of CN112521506A publication Critical patent/CN112521506A/en
Application granted granted Critical
Publication of CN112521506B publication Critical patent/CN112521506B/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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • A61K39/001119Receptors for interleukins [IL]
    • 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
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion 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 CD123 antigen binding fragment and application thereof, wherein the CD123 antigen binding fragment comprises a heavy chain CDR1 shown in SEQ ID NO. 1, a heavy chain CDR2 shown in SEQ ID NO. 2 and a heavy chain CDR3 shown in SEQ ID NO. 3. The invention screens and obtains CD123 antigen binding fragments with CDR regions shown in SEQ ID NO 1-3 from a camel VHH immune library, and an antibody formed by combining the fragment and a frame region FR has strong affinity to CD123, so that the constructed chimeric antigen receptor and chimeric antigen receptor immune cells have obvious cytotoxicity to CD123 positive cells, and the application prospect in the field of tumor treatment is wide.

Description

CD123 antigen binding fragment and application thereof
Technical Field
The invention belongs to the technical field of biomedicine, and relates to a CD123 antigen binding fragment and application thereof.
Background
Over 30 years ago, the "3 + 7" regimen (3 days for daunorubicin chemotherapy combined with 7 days for cytarabine chemotherapy) alleviated the disease in about 60% of AML patients, becoming the standard induction regimen for the treatment of acute leukemia in children and adults. In the last 90 s of the century, clinical experts began to focus on post-remission treatment regimens and their benefits and conducted a great deal of research including high dose cytarabine chemotherapy or Hematopoietic Stem Cell Transplantation (HSCT). Although remission rates and overall survival rates for childhood acute leukemia currently reach greater than 90% and 60%, respectively, existing treatment regimens are still limited to anthracycline-based drugs, nucleoside analogs, and intensive post-remission therapy. In order to improve the prognosis of AML patients, protocols including alternatives to mitoxantrone and daunorubicin, cytarabine-based intensive therapy, etc. were used clinically, however, most clinical studies showed no significant difference in outcome for each treatment group.
The curative effect of stem cell transplantation on leukemia shows that the antitumor immunity can effectively eliminate and prevent leukemia recurrence. In fact, many studies have found that the recurrence rate of hematopoietic stem cell transplantation is significantly reduced compared to chemotherapy. However, hematopoietic stem cell transplantation in remission is controversial due to the high mortality rate of hematopoietic stem cell transplantation. In recent years, with improvements in supportive care, comprehensive HLA and NK typing contributes to reduction of side effects of hematopoietic stem cell transplantation therapy. Tyrosine Kinase Inhibitors (TKIs) have become one of the important chemotherapeutic regimens. For example, the FLT3 gene (FLT3ITD) can elicit AML in about 15% of children and 30% of adults, and individuals with a higher FLT 3-ITD/wild-type FLT3 ratio exhibit a more pronounced poor prognosis, associated with poor outcome. Sorafenib (sorafenib), sunitinib and other FLT3 inhibitors can effectively inhibit FLT3 mutation, but the drug resistance phenomenon appears after long-term use of the drugs, which is mainly related to D835 or F691 kinase region point mutation. The novel tyrosine kinase inhibitor crenolanib has a good therapeutic effect on Sorafenib (Sorafenib) resistant AML mouse models, and the inhibitor can prolong clinical benefit. Although TKIs offer different leukemia treatment regimens, they are still in the development stage and more treatment strategies are clinically needed.
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 continually optimized this technique until 2011, CAR-T cells targeted to CD19(B lymphocyte antigen CD19, CD19) achieved surprising efficacy in treating relapsed/refractory chronic B-lymphocyte leukemia, and the use of CAR-T cells in tumor therapy opened new chapters.
The chimeric antigen receptor is an artificially synthesized fusion protein with the 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 via intracellular signaling molecules, secretes perforin and granzyme B, and effects killing of the target cells. CAR-T cells recognize target cells independent of Major Histocompatibility Complex (MHC), avoiding immune escape due to down-regulation of tumor cell MHC molecules. In recent years, chimeric antigen modified T lymphocytes (CAR-T) have developed rapidly, and at present, two products are approved by FDA in the united states and are on the market, and many products are approved by clinical trials of drugs in China.
CD123, also known as CD3 receptor alpha chain, is expressed on leukemic cells and leukemic stem cells, normal hematopoietic stem cells are not or weakly expressed, and are also expressed on endothelial cells and monocytes, plasma cell DCs, but CD123 positive monocytes and plasma cells account for a small proportion of reactive lymph nodes. High expression of CD123 promotes proliferation of tumor cells. CD123 is currently the most studied AML antigen, and several monoclonal antibody-based drugs targeting CD123 have been developed, such as 7G3, CSL360 and CSL362, in preclinical and clinical trials. In addition, CAR-T therapies targeting CD123 have also completed preclinical studies and showed significant anti-tumor effects.
The anti-CD 123 chimeric antigen receptor gene is introduced into T cells through a genetic engineering method to prepare CD123CAR-T, so that the CD123CAR-T cells specifically recognize CD 123-expressing acute myeloid leukemia cells and eliminate tumor cells, thereby realizing the anti-tumor effect of the CD123 chimeric antigen receptor gene. Therefore, immunotherapeutic approaches targeting CD123 have great clinical value.
Disclosure of Invention
Aiming at the defects and practical requirements of the prior art, the invention provides a CD123 antigen binding fragment and application thereof, a high-affinity anti-CD 123 antibody containing the CD123 antigen binding fragment is screened by utilizing a phage display technology and is used as an antigen binding domain of a chimeric antigen receptor molecule to construct a CAR immune cell, and the CAR immune cell can specifically recognize and kill CD123 positive cells and has important application prospect in the field of tumor treatment.
In order to achieve the purpose, the invention adopts the following technical scheme:
in a first aspect, the invention provides a CD123 antigen-binding fragment, the CD123 antigen-binding fragment comprising the heavy chain CDR1 of SEQ ID NO. 1, the heavy chain CDR2 of SEQ ID NO. 2, and the heavy chain CDR3 of SEQ ID NO. 3;
SEQ ID NO:1:ENVYC;
SEQ ID NO:2:VSRVGWT;
SEQ ID NO:3:AADSRVCGLRSGRKYTD。
in the invention, an antibody with high affinity is screened from a CD123 immune camel VHH immune library, and the fact that a fragment with heavy chain CDRs shown in SEQ ID NO. 1-3 has specific high binding capacity to a CD123 antigen is found, and the CDR regions shown in SEQ ID NO. 1-3 have decisive effect on the affinity of the antigen binding fragment or the antibody with the CD 123.
In a second aspect, the present invention provides an anti-CD 123 antibody comprising a CD123 antigen-binding fragment according to the first aspect.
Preferably, the anti-CD 123 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO 4; 4, SEQ ID NO:
EVQLVESGGGSVQAGGSLKLSCSASENVYCMGWFRQAPGKEREGVATVSRVGWTSYADSVKGRFTISRDNAKNALYLQMNSLKPEDTAMYYCAADSRVCGLRSGRKYTDWGQGTQVTVSS。
in the invention, a phage display technology is utilized to screen a CD123 immune camel VHH library to obtain an anti-CD 123 antibody CD123-35(SEQ ID NO:4) only containing a heavy chain variable region, wherein the anti-CD 123 antibody has CDR regions shown in SEQ ID NO: 1-3, has high affinity to CD123 and has important application prospect in the aspect of constructing a chimeric antigen receptor of a target CD 123.
In a third aspect, the present invention provides a nucleic acid molecule comprising a DNA fragment encoding the CD123 antigen-binding fragment of the first aspect.
Preferably, the nucleic acid molecule comprises a DNA fragment encoding the anti-CD 123 antibody of the second aspect.
Preferably, the nucleic acid molecule comprises the nucleic acid sequence shown in SEQ ID NO. 5, which is the coding sequence of the anti-CD 123 antibody CD 123-35;
SEQ ID NO:5:
GAAGTGCAGCTGGTGGAGAGCGGCGGCGGATCTGTGCAGGCCGGAGGCTCTCTGAAACTGTCTTGTTCTGCCAGCGAGAATGTGTATTGTATGGGATGGTTTAGACAGGCCCCAGGCAAAGAGAGAGAGGGAGTGGCTACAGTGTCTAGAGTGGGATGGACATCTTATGCCGATTCTGTGAAAGGAAGATTCACAATTTCTAGAGATAATGCCAAAAATGCCCTGTATCTGCAGATGAATAGCCTGAAGCCTGAGGATACAGCCATGTATTATTGTGCCGCCGATTCTAGAGTGTGTGGACTGAGATCTGGCAGAAAATATACAGATTGGGGCCAGGGAACACAGGTGACAGTGTCTAGC。
in a fourth aspect, the present invention provides a chimeric antigen receptor comprising a signal peptide, an antigen binding domain, a hinge region, a transmembrane domain, and a signaling domain;
the antigen binding domain is an anti-CD 123 antibody according to the second aspect.
In the invention, the anti-CD 123 antibody of the second aspect is used as an extracellular region of the chimeric antigen receptor and is combined with CD123 in a targeted manner, and the constructed chimeric antigen receptor and immune cells expressing the chimeric antigen receptor have a high-efficiency targeting effect on CD123 positive cells and have important significance in the field of CD123 positive tumor treatment.
Preferably, the signal peptide comprises a CD8 a signal peptide.
Preferably, the hinge region comprises a CD8 a hinge region.
Preferably, the transmembrane domain comprises any one or a combination of at least two of a CD8 a transmembrane region, a CD28 transmembrane region or a DAP10 transmembrane region, the sequence of the CD8 a transmembrane region being referred to NM _001145873, the sequence of the CD28 transmembrane region being referred to NM _006139 and the sequence of the DAP10 transmembrane region being referred to NM _ 014266.
Preferably, the signaling domain comprises CD3 ζ, the sequence of which can be referenced to NM _ 198053.
Preferably, the signal transduction domain further comprises any one or a combination of at least two of the intracellular domains of 4-1BB, CD28, OX40, ICOS or DAP10, the sequence of 4-1BB is referred to NM _001561, the sequence of CD28 is referred to NM _006139, the sequence of OX40 is referred to NM _003327, the sequence of ICOS is referred to NM _012092, and the sequence of DAP10 is referred to NM _ 014266.
In a specific embodiment of the invention, the signaling domain comprises 4-1BB and CD3 zeta in order from N-terminus to C-terminus.
Preferably, the chimeric antigen receptor comprises, from N-terminus to C-terminus, the anti-CD 123 antibody of the second aspect, the CD8 a transmembrane region, 4-1BB and CD3 ζ in that order.
Preferably, the chimeric antigen receptor comprises, from N-terminus to C-terminus, the anti-CD 123 antibody, CD28 transmembrane region, CD28 intracellular region and CD3 ζ of the second aspect in that order.
Preferably, the chimeric antigen receptor comprises, in order from N-terminus to C-terminus, the anti-CD 123 antibody of the second aspect, a CD8 a transmembrane region, OX40, and CD3 ζ.
Preferably, the chimeric antigen receptor comprises, in order from N-terminus to C-terminus, the anti-CD 123 antibody of the second aspect, a CD8 a transmembrane region, ICOS, and CD3 ζ.
Preferably, the chimeric antigen receptor comprises, from N-terminus to C-terminus, the anti-CD 123 antibody of the second aspect, the CD28 transmembrane region, the CD28 intracellular region, OX40 and CD3 ζ in that order.
Preferably, the chimeric antigen receptor comprises an amino acid sequence shown as SEQ ID NO. 6;
6(CD 8. alpha. signal peptide-CD 123-35-CD 8. alpha. hinge-TM-41BB-CD 3. zeta):
MALPVTALLLPLALLLHAARPEVQLVESGGGSVQAGGSLKLSCSASENVYCMGWFRQAPGKEREGVATVSRVGWTSYADSVKGRFTISRDNAKNALYLQMNSLKPEDTAMYYCAADSRVCGLRSGRKYTDWGQGTQVTVSSTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR。
in a fifth aspect, the present invention provides an expression vector comprising the gene encoding the chimeric antigen receptor of the fourth 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 fourth aspect, preferably a lentiviral vector.
In the invention, a virus vector, especially a lentivirus vector, containing the encoding gene of the chimeric antigen receptor of the fourth aspect is constructed, and an anti-CD 123CAR-T cell is constructed by using a genetic engineering method, so that an anti-tumor effect is realized.
In a sixth aspect, the present invention provides a recombinant lentivirus prepared from a mammalian cell transfected with an expression vector and a helper plasmid according to the fifth aspect.
In a seventh aspect, the present invention provides a chimeric antigen receptor immune cell expressing the chimeric antigen receptor of the fourth aspect.
Preferably, the chimeric antigen receptor immune cell comprises the expression vector of the fifth aspect and/or the recombinant lentivirus of the sixth 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 an eighth aspect, the present invention provides a pharmaceutical composition comprising the chimeric antigen receptor immune cell of the seventh aspect.
Preferably, the pharmaceutical composition further comprises any one or a combination of at least two of a pharmaceutically acceptable carrier, excipient or diluent.
In a ninth aspect, the present invention provides a CD123 antigen-binding fragment according to the first aspect, an anti-CD 123 antibody according to the second aspect, a nucleic acid molecule according to the third aspect, a chimeric antigen receptor according to the fourth aspect, an expression vector according to the fifth aspect, a recombinant lentivirus according to the sixth aspect, a chimeric antigen receptor immune cell according to the seventh aspect, or a pharmaceutical composition according to the eighth aspect, for use in the preparation of a medicament for the treatment of a malignant tumor.
Preferably, the malignancy comprises a hematological tumor.
Compared with the prior art, the invention has the following beneficial effects:
(1) the invention utilizes a transient transfection mode to introduce plasmids containing CD123 protein extracellular region gene segments into eukaryotic cells, express recombinant CD123 protein for immunizing bactrian camel, construct a phage display antibody library, and screen CD123-35 antibodies which can be specifically combined with CD123 antigen;
(2) the invention further utilizes the screened antibody to transform into a chimeric antigen receptor, and utilizes a genetic engineering method to express the chimeric antigen receptor in immune cells, so that the constructed immune cells expressing the anti-CD 123 chimeric antigen receptor have a cytotoxic effect on CD123 positive tumor cells, efficiently secrete cell factors IL-2, TNF-alpha and IFN-gamma after being co-cultured with the CD123 positive cells, and have important application prospects in the aspect of treating CD123 positive acute myeloid leukemia.
Drawings
FIG. 1 is Biacore for detecting the affinity of CD123-35 antibody;
FIG. 2 shows FACS detection of CD123-35 antibody recognizing CD123 antigen on the cell surface;
FIG. 3 is a schematic diagram of the structure of a chimeric antigen receptor expressing CD 123;
FIG. 4 shows the chimeric antigen receptor expression rate of T lymphocytes;
FIG. 5 is the killing effect of CD123CAR-T cells on CD123 positive tumor cells;
FIG. 6 is the level of IL-2 secretion by CD123CAR-T cells;
FIG. 7 is the level of TNF α secretion by CD123CAR-T cells;
FIG. 8 is the level of IFN γ secretion by CD123CAR-T cells.
Detailed Description
To further illustrate the technical means adopted by the present invention and the effects thereof, the present invention is further described below with reference to the embodiments and the accompanying drawings. It is to be understood that the specific embodiments described herein are merely illustrative of the invention and are not limiting of the invention.
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 construction of phage antibody library, panning and ELISA Primary screening
(1) Construction of phage antibody libraries
In the embodiment, firstly, CD123 antigen extracellular region is adopted to immunize bactrian camel, and 200mL of peripheral blood is extracted after the titer is verified by ELISA; separating lymphocytes from peripheral blood to obtain peripheral blood mononuclear lymphocyte sediment, and extracting RNA;
using the extracted RNA as a template
Figure BDA0002831851970000041
III reverse transcriptase synthesizing first strand cDNA, and subsequently amplifying VHH gene by using nested PCR; inserting the amplified VHH gene into a pMECS phage display carrier, electrically converting TG1 competent cells, taking a proper amount of bacterial liquid for library identification, and uniformly coating all the rest cultures on an LB/AMPGLU flat plate;
collecting thallus Porphyrae after bacteria growth, adding 1/3 vol 50% glycerol, mixing, packaging, and storing at-80 deg.C to obtain product with storage capacity greater than 109The phage display camelid VHH immune library of (a).
(2) Panning of phage antibody libraries
Performing 3 rounds of solid phase screening on the immune library, and enriching to obtain phage clones with binding activity; after prokaryotic induction expression is carried out on the monoclonal phage, phage clones which can be combined with the extracellular region of the CD123 antigen are further screened by ELISA;
diluting the purified CD123 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) of CD123 recombinant protein diluent into each well, coating overnight at 4 ℃, and setting a PBS negative control group; discarding the coating solution, adding 150 mu L of 2% skimmed milk powder into each hole, sealing at room temperature for 1h, and preparing an ELISA plate coated with the CD123 recombinant protein;
washing enzyme labeling plate with PBST for 4 times, diluting the prepared phage solution with 2% skimmed milk powder to 5 × 1011pfu/mL, then adding 100 mu L/hole into the enzyme label plate, and incubating for 2h at room temperature; discarding a 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 into each well, and standing at room temperature for 10 min; the eluate was immediately neutralized with an equal volume of 1M Tris-HCl (pH 7.4) by aspiration;
taking part of the eluent to determine the titer of the phage; infecting 4mL of log-phase TG1 bacterial solution (OD600 about 0.6) with 400 μ L of eluate, and incubating at 37 deg.C for 30 min; then adding 16mL of 2 XYT/ampicillin/glucose (2 XYT/AMP-GLU), and continuing to culture at 37 ℃ and 200r/min until OD600 reaches 0.6-0.8;
taking 100 mu L of bacterial suspension, performing gradient dilution, uniformly smearing the bacterial suspension on a 2 XYT/ampicillin/glucose agar plate, and performing library capacity and diversity determination; inoculating 100 mu L of bacterial suspension into a 2 XYT/AMP-GLU culture medium, culturing to a logarithmic phase, adding an auxiliary phage, performing library rescue, obtaining phage particles, performing phage titer detection, and concentrating and purifying to obtain phage particles for the next round of screening; the screening operation was repeated 3 times;
after centrifuging the residual bacterial liquid, re-suspending the residual bacterial liquid by using 2 XYT culture solution with proper volume, and smearing the re-suspended bacterial liquid on a flat plate with screening resistance for overnight culture; the bacteria were scraped from the plate using the appropriate amount of liquid culture medium, resuspended in 2 XYT medium containing 1/3 vol 50% glycerol, aliquoted, and stored at-80 ℃.
(3) Phage packaging
Adding 100 mu L of the bacterium solution elutriated in the step (2) into 100mL of 2 XYT/AMPGL culture solution, and carrying out shaking culture at 37 ℃ and 200rpm until the logarithmic phase (the OD600 value is 0.6-0.8); add 90. mu.L of the helper phage M13K07 (1.7X 10)13PFU/mL), standing at 37 ℃ for 30min, centrifuging at 2800g for 10min to collect the thallus, resuspending with 200mL 2 XYT/AMP-KAN culture medium, and culturing at 37 ℃ for 12h under shaking at 200 rpm;
centrifuging at 4 deg.C and 3800g for 30min, collecting supernatant, adding 1/5 volume of precooled PEG/NaCl, mixing, and precipitating bacteriophage for 2 h; centrifuging at 4 ℃ and 3800g for 30min, collecting phage, resuspending the thallus with PBS solution with the final volume of 2mL, 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, shaking and incubating overnight at 4 deg.C 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.
(4) ELISA Primary screening
As 3 rounds of panning are carried out on the phage library by adopting the CD123 antigen in the step (2), in order to avoid losing sequence diversity, ELISA primary screening is carried out on panning products of the 2 nd round and the 3 rd round, positive clones are randomly selected from the panning products and are induced to express, an expression supernatant is a crude extraction VHH antibody, and the sequence SEQ ID NO. 4 and SEQ ID NO. 5 of the VHH antibody CD123-35 sequence of a monoclonal strain are determined by sequencing.
Example 2 FACS screening of candidate clones
This example performs cell culture according to standard cell culture protocols:
digesting the cells with pancreatin to prepare a suspension of CD 123-positive cells or CD 123-negative cells, centrifuging at 300g for 5min to remove the culture medium, and resuspending the cells with a Flow Buffer to a cell concentration of 2X 106Per mL;
adding 2X 10 to each well of a V-bottom 96-well plate5Each cell was centrifuged at 300g for 5min to remove the supernatant, and VHH antibody crude was addedThe extract is used for resuspending the cells and is incubated for 1h at 4 ℃;
centrifuging at 300g for 5min to remove supernatant, resuspending cells with Flow Buffer, adding 100. mu.L of Flow Buffer diluted APC anti-his antibody (2. mu.g/mL), and incubating at 4 ℃ for 1 h;
after the cells were washed 3 times with the Flow Buffer, the cells were resuspended using 200. mu.L of the Flow Buffer and subjected to Flow assay.
Example 3 expression, purification and affinity determination of VHH-mIgG2a Fc antibodies
To further identify the antibodies screened, the example constructed vector C-4pcp. stuffer VHH-mCg2a-FC (with mouse FC tag) expressing VHH, as follows:
the anti-CD 123 heavy chain variable region encoding gene is amplified by PCR, wherein an upstream primer of CD123-35(SEQ ID NO:5) is HD-CD 35-F, a downstream primer is HD-B8-R1, the sequence is shown in Table 2, a PCR reaction system is shown in Table 3, the reaction conditions are pre-denaturation at 95 ℃ for 1min, denaturation at 95 ℃ for 10s, annealing at 55 ℃ for 10s, extension at 72 ℃ for 10s, 30 cycles, extension at 72 ℃ for 5min and storage at 4 ℃.
TABLE 2
Figure BDA0002831851970000051
TABLE 3
Figure BDA0002831851970000052
Subjecting the empty vector to enzyme digestion treatment at 37 deg.C for 6h, wherein the system is shown in Table 4, and the enzyme digested vector is used
Figure BDA0002831851970000053
The PCR purification kit was recovered and purified, and dissolved in 45. mu.L of water to detect the DNA concentration.
TABLE 4
Reagent Dosage of
C-4pCP.Stuffer-mCg2a-FC 5μg
10 Xdigestion Buffer (10 Xreaction Buffer) 5μL
FspA I 2μL
PfI 23II 2μL
ddH2O Make up to 50 μ L
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
2×Exnase II buffer 2
Linearized vector 4
Amplification product (Insert fragment) 3
The entire homologous recombination reaction was added to DH 5. alpha. competent cells under the transformation conditions shown in Table 6.
TABLE 6
Procedure Temperature (. degree.C.) Time
Ice bath
0 5min
Heating of 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 (16 to 18h)
Selecting a single clone from a transformation plate to carry out PCR pre-identification, wherein the system is 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 the amplification product is sent to a sequencing company for sequencing identification. Sequencing results were found to be in line with expectations, indicating the successful construction of expression vector C-4pCP. Stuffer VHH-mCg2a-FC with mouse Fc-tagged VHH (scFV-mIgG 1).
TABLE 7
Reagent Volume (μ L)
pEF1A 1
PSV40 1
2×Fast Taq Mix 15
ddH2O Is supplemented to 30
Approximately 24h before plasmid transfection, 293E cells were subcultured to fineThe cell density was about 0.6X 106Per mL; when the cell density is (1.0-1.2) × 106unit/mL, rate of activity>At 95%, 0.15 μ g C-4pCP.Stuffer VHH-mCg2a-FC was transfected into 100mL 293E cells with PEI, the ratio of plasmid DNA to PEI being 1: 2;
293E cells transfected with plasmids at 37 ℃ at 130rpm in 8% CO2Culturing in a shaking table for 5-7 days, centrifuging at 3000rpm for 30min, collecting supernatant, sterile filtering with Millex-GP Filter Unit 0.45 μm, and selecting with MabSelectTMSuReTMConcentrating by centrifugation, washing the column with 1 × PBS, eluting the protein with 0.1M Gly-HCl, and neutralizing with 1/10 volumes of Tris-HCl at pH 8.5 to obtain antibody protein; the resulting antibody protein was dialyzed overnight at 4 ℃ and the antibody concentration was quantitatively determined by NanoDrop 2000 and the antibody purity was determined by SEC-HPLC.
The purified anti-CD 123 VHH antibody CD123-35(SEQ ID NO:4) was subjected to affinity assay using Biacore, a bioanalytical sensing technique developed based on Surface Plasmon Resonance (SPR), which can detect binding of molecules in a tracking solution to molecules on the chip surface, the course of dissociation changes, recorded as sensorgrams, 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
Fixability of Mobile phase Ka(1/Ms) Kd(1/s) KD(M)
0.5μg/mL CD123-35 Human IL-3R alpha/CD 123 protein 1.68E+05 2.76E-04 1.64E-09
Example 4 flow assay of anti-CD 123 VHH antibodies
After mixing K562(CD123-), KG-1 alpha (CD123+), THP-1(CD123+) tumor cells with purified recombinant CD123-35 antibody, ice-cooling for 30min, then adding APC labeled goat anti-mouse IgG antibody, incubating for 30min, and detecting with flow cytometry.
The results are shown in FIG. 2, indicating that the anti-CD 123-35 antibody recognizes CD123 antigen on the cell surface.
EXAMPLE 5 design and construction of expression vectors for chimeric antigen receptors
This example constructs a lentiviral vector expressing a CAR molecule, the CAR molecule is schematically depicted in FIG. 3 and comprises a CD8 α signal peptide (SEQ ID NO:9), anti CD123 VHH (SEQ ID NO:4), a CD8 α hinge and transmembrane region (SEQ ID NO:10), 4-1BB (SEQ ID NO:11), CD3 ζ (SEQ ID NO:12), and an amino acid sequence depicted in SEQ ID NO: 6;
SEQ ID NO:9:
MALPVTALLLPLALLLHAARP;
SEQ ID NO:10:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYC;
SEQ ID NO:11:
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL;
SEQ ID NO:12:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR。
(1) the gene coding for the anti CD123 VHH is amplified by PCR, the primers are shown in Table 9, the system is shown in Table 10 (the reagents are from TOYOBO Inc.), and the PCR program is shown in Table 11;
TABLE 9
Figure BDA0002831851970000071
Watch 10
Figure BDA0002831851970000072
Figure BDA0002831851970000081
TABLE 11
Figure BDA0002831851970000082
(2) The CD8 α signal peptide was added to the scFv fragment by PCR, the primers are shown in Table 12, the PCR reaction system is shown in Table 13, and the PCR reaction was carried out according to the PCR program shown in Table 11; after the reaction is finished, detecting the PCR product by using 1% agarose gel electrophoresis, recovering fragments of about 460bp, and quantitatively detecting by using an ultraviolet absorption method;
TABLE 12
Figure BDA0002831851970000083
Watch 13
Figure BDA0002831851970000084
(3) The CD8 α hinge region-transmembrane region-41 BB-CD3 ζ (CD8 α hinge-TM-41BB-CD3Z) was amplified by PCR using the following primers:
CD8αH-F(SEQ ID NO:16):accacgacgccagcgccgcgac;
Vector-R(SEQ ID NO:17):tcgataagcttgatatcg;
the PCR reaction system is shown in table 14 (reagents derived from TOYOBO Inc.) and PCR reactions were performed according to the PCR program in table 11; after the reaction is finished, detecting the PCR product by using 1% agarose gel electrophoresis, recovering fragments of about 780bp, and quantitatively detecting by using an ultraviolet absorption method;
TABLE 14
Figure BDA0002831851970000085
Figure BDA0002831851970000091
(4) Mu.g of HD CD19 CAR plasmid was digested with BamHI and EcoRI in a water bath at 37 ℃ for 2h to recover the vector.
The fragments were ligated with recombinase in 37 ℃ water bath for 0.5h in the reaction system shown in Table 15, and the ligation products were transformed into E.coli stbl3 competent cells according to the conventional method. And selecting a single clone from a solid culture medium for overnight culture, performing PCR identification, wherein the system is shown in table 16, the program is shown in table 17, selecting a positive clone after PCR is finished, further performing sequencing identification, and the sequencing result is in line with expectation.
Watch 15
Reagent Dosage of
HD CD19 CAR 184.54ng
CD8αsignal CD123 VHH 31.32ng
CD8αhinge-TM-41BB-CD3Z 29.72ng
5×CE buffer 2μL
ExnaseTM II 1μL
PCR grade water Make up to 10 mu L
TABLE 16
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
PCR grade water 7
TABLE 17
Figure BDA0002831851970000092
Example 6 Lentiviral packaging, concentration and Titer assay
(1) Lentiviral packaging
Mixing 1.6X 107293T cells were plated in 15cm dishes in DMEM containing 10% Fetal Bovine Serum (FBS) at 37 ℃ with 5% CO2Culturing overnight; adding 30 μ g pRRL. EF1 α -CD123 CAR-WPRE lentiviral vector, 12.5 μ g gag/pol helper plasmid and 10 μ g VSVg envelope plasmid into 2000 μ L serum-free DMEM medium, 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 gently mixing uniformly, and incubating for 20min at room temperature; dripping 4mL of the transfection compound into 293T cells, and replacing a fresh culture medium after 4-5 h; after 48h, the viral 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-volume PEG-NaCl virus concentrated solution, turning upside down, mixing uniformly, and standing overnight at 4 ℃; centrifuging at 4 deg.C and 3500rpm for 30min, removing supernatant, adding appropriate amount of RPMI 1640 culture medium (containing 10% FBS), and resuspending virus precipitate; the concentrated lentivirus suspension was divided into 50 μ L portions and stored at-80 ℃.
(3) Lentiviral titer detection
500 μ L K562 cells (1X 10)5Individual cells) were inoculated in a 24-well cell culture plate, 1. mu.L, 0.2. mu.L and 0.04. mu.L of the concentrated lentivirus, respectively, and polybrene was added to a final concentration of 5. mu.g/mL, 37 ℃ and 5% CO2Culturing overnight, and replacing fresh culture medium;
after 72h of infection, 400g of the cells were collected by centrifugation for 5min, 100. mu.L of PBS + 2% FBS was added to resuspend the cells, 1. mu.g of hCD123-EcD-Fc antibody was added, and incubation was performed on ice for 30 min; after washing with PBS + 2% FBS for 1 time, adding 100. mu.L PBS + 2% FBS for resuspension of cells, adding APCanti-human IgG Fc antibody, and incubating on ice for 30 min; after washing with PBS + 2% FBS for 2 times, adding 300 μ L of PBS + 2% FBS for resuspension of cells, detecting infection efficiency by using a flow cytometer, preferably taking a cell sample with a positive rate of 5-20%, and calculating the lentivirus titer according to the following formula.
Titer (TU/mL) is cell number (10)5) X Positive Rate/Virus volume (mL)
Example 7 Lentivirally transduced T lymphocytes
Diluting anti-human CD3 antibody and anti-human CD28 antibody with PBS to final concentration of 1 μ g/mL and 0.5 μ g/mL respectively, coating the well plate, and standing overnight at 4 deg.C; discarding the antibody coating solution in the pore plate, and washing twice with 1mL of PBS;
X-VIVO + 10% FBS + IL-2(300U/mL) T cell culture medium was added to human PBMC to adjust the cell density to 1X 106mL, then inoculated into CD3 and CD28 antibody coated well plates, activated for 48 h;
collecting activated T cells, adjusting cell density to 1 × 106Per mL, lentivirus was added at a multiplicity of infection (MOI) of 10, polybrene was added to a final concentration of 5. mu.g/mL, and 5% CO was added at 37 ℃2After overnight culture in the environment, replacing a fresh culture medium, and carrying out passage every 2-3 days;
after 5 days of T cell infection with lentivirus, 3X 10 cells were taken5Centrifuging T cells at 4 ℃ for 5min at 400g, discarding the supernatant, and washing the cells once with PBS and 2% FBS; adding 100 μ L PBS + 2% FBS to resuspend the cells, adding 0.25 μ g Human CD123 protein antibody, incubating on ice for 30min, and washing with PBS + 2% FBS for 1 time; adding 100 μ L PBS + 2% FBS to resuspend cells, adding APC anti-human IgG Fc antibody, and incubating on ice for 30 min; after washing 2 times with PBS + 2% FBS, 300. mu.L of PBS + 2% FBS was added to resuspend the cells.
The lentiviral infection efficiency was measured by flow cytometry, and the results are shown in fig. 4, where the expression efficiency of the CAR molecule was 79.2%.
Example 8 in vitro toxicity assay of CAR-T cells
293T (CD123-), 293T-CD123(CD123+), BxPC3-CD123(CD123+) were adjusted to a cell concentration of 1X 105mL, 100. mu.L of the target cells were inoculatedSeeding in 96-well plates; adding CD123CAR-T cells and control T cells to a 96-well plate at an effective target ratio of 0.3:1, 1:1, and 3: 1; each set was provided with 3 multiple wells. The experimental and control groups were 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;
after the effector cells and the target cells are co-cultured for 18h, the killing effect of the effector cells on the target cells is detected by using a CytoTox 96 nonradioactive cytotoxicity detection kit (Promega). The method is 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 after cell lysis in a manner similar to that of51Cr is released in the radioactive assay in essentially the same manner, and the released LDH can be detected by a coupled enzymatic reaction in which LDH converts a tetrazolium salt (INT) to red formazan (formazan), with the amount of red product produced being proportional to the number of cells lysed. Reference is made in particular to the instructions of the CytoTox 96 nonradioactive cytotoxicity detection kit.
The killing ability of effector cells to target cells was calculated according to the following cytotoxicity calculation formula:
percent cytotoxicity ═ 100% (experimental-control 2-control 3)/(control 1-control 2) ×
The results are shown in figure 5, where CD123CAR-T cells have killing activity against CD123 positive tumor cells, but no killing effect on CD123 negative cells.
Example 9 CAR-T cytokine secretion
Centrifuging 400g of a cell culture with an effective target ratio of 1:1 for 10min, removing precipitates, placing 100 mu L of supernatant into a sample hole of an enzyme label plate, adding 50 mu L of detection antibody diluent (1:100 dilution), and performing shaking incubation for 2 hours at room temperature at 300rpm/min by using a plate sealing membrane sealing plate; discarding liquid, adding 300 mu L of washing liquid into each hole to wash the plate for 6 times, and drying the ELISA plate on absorbent paper after washing the plate each time;
adding 100 μ L of horse radish peroxidase labeled streptavidin diluent (1:100 dilution) into each well, sealing with a new sealing plate membrane, and incubating at room temperature at 300rpm/min under shaking for 45 min; discarding liquid, adding 300 mu L of washing liquid into each hole to wash the plate for 6 times, and drying the ELISA plate on absorbent paper after washing the plate each time;
adding 100 mu L of chromogenic substrate TMB into each hole, incubating for 5-30 min at room temperature in the dark, then adding 100 mu L of stop solution into each hole, changing the color from blue to yellow, measuring the OD value at the maximum absorption wavelength of 450nm and the reference wavelength of 570nm or 630nm by using an enzyme labeling instrument, and subtracting the measured value of 570nm or 630nm from the measured value of 450nm by using the calibrated OD value.
The secretion results of IL-2, TNF-alpha and IFN-gamma factors are respectively shown in figure 6, figure 7 and figure 8, and the constructed CAR-T cells release cytokines after being incubated with CD123 positive tumor cells, but have no obvious cytokine secretion after being incubated with CD123 negative cells.
In conclusion, the invention utilizes phage display technology to screen camel VHH immune libraries immunized by CD123 to obtain anti-CD 123 antibody with high affinity; the anti-CD 123 antibody is used as an antigen binding structural domain to construct a chimeric antigen receptor, a chimeric antigen receptor gene is introduced into immune cells through a genetic engineering method, and the prepared CD123 chimeric antigen receptor immune cells can specifically recognize and kill tumor cells expressing CD123, so that the anti-tumor effect is realized.
The applicant states that the present invention is illustrated in detail by the above examples, but the present invention is not limited to the above detailed methods, i.e. it is not meant that the present invention must rely on the above detailed methods for its implementation. It should be understood by those skilled in the art that any modification of the present invention, equivalent substitutions of the raw materials of the product of the present invention, addition of auxiliary components, selection of specific modes, etc., are within the scope and disclosure of the present invention.
Sequence listing
<110> Huadao (Shanghai) biopharmaceutical Co., Ltd
<120> CD123 antigen binding fragment and application thereof
<130> 20201210
<160> 17
<170> SIPOSequenceListing 1.0
<210> 1
<211> 5
<212> PRT
<213> Artificial sequence ()
<400> 1
Glu Asn Val Tyr Cys
1 5
<210> 2
<211> 7
<212> PRT
<213> Artificial sequence ()
<400> 2
Val Ser Arg Val Gly Trp Thr
1 5
<210> 3
<211> 17
<212> PRT
<213> Artificial sequence ()
<400> 3
Ala Ala Asp Ser Arg Val Cys Gly Leu Arg Ser Gly Arg Lys Tyr Thr
1 5 10 15
Asp
<210> 4
<211> 120
<212> PRT
<213> Artificial sequence ()
<400> 4
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Ser Val Gln Ala Gly Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ser Ala Ser Glu Asn Val Tyr Cys Met Gly
20 25 30
Trp Phe Arg Gln Ala Pro Gly Lys Glu Arg Glu Gly Val Ala Thr Val
35 40 45
Ser Arg Val Gly Trp Thr Ser Tyr Ala Asp Ser Val Lys Gly Arg Phe
50 55 60
Thr Ile Ser Arg Asp Asn Ala Lys Asn Ala Leu Tyr Leu Gln Met Asn
65 70 75 80
Ser Leu Lys Pro Glu Asp Thr Ala Met Tyr Tyr Cys Ala Ala Asp Ser
85 90 95
Arg Val Cys Gly Leu Arg Ser Gly Arg Lys Tyr Thr Asp Trp Gly Gln
100 105 110
Gly Thr Gln Val Thr Val Ser Ser
115 120
<210> 5
<211> 360
<212> DNA
<213> Artificial sequence ()
<400> 5
gaagtgcagc tggtggagag cggcggcgga tctgtgcagg ccggaggctc tctgaaactg 60
tcttgttctg ccagcgagaa tgtgtattgt atgggatggt ttagacaggc cccaggcaaa 120
gagagagagg gagtggctac agtgtctaga gtgggatgga catcttatgc cgattctgtg 180
aaaggaagat tcacaatttc tagagataat gccaaaaatg ccctgtatct gcagatgaat 240
agcctgaagc ctgaggatac agccatgtat tattgtgccg ccgattctag agtgtgtgga 300
ctgagatctg gcagaaaata tacagattgg ggccagggaa cacaggtgac agtgtctagc 360
<210> 6
<211> 364
<212> PRT
<213> Artificial sequence ()
<400> 6
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 Glu Val Gln Leu Val Glu Ser Gly Gly Gly Ser
20 25 30
Val Gln Ala Gly Gly Ser Leu Lys Leu Ser Cys Ser Ala Ser Glu Asn
35 40 45
Val Tyr Cys Met Gly Trp Phe Arg Gln Ala Pro Gly Lys Glu Arg Glu
50 55 60
Gly Val Ala Thr Val Ser Arg Val Gly Trp Thr Ser Tyr Ala Asp Ser
65 70 75 80
Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Ala Leu
85 90 95
Tyr Leu Gln Met Asn Ser Leu Lys Pro Glu Asp Thr Ala Met Tyr Tyr
100 105 110
Cys Ala Ala Asp Ser Arg Val Cys Gly Leu Arg Ser Gly Arg Lys Tyr
115 120 125
Thr Asp Trp Gly Gln Gly Thr Gln Val Thr Val Ser Ser Thr Thr Thr
130 135 140
Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala Ser Gln Pro
145 150 155 160
Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly Gly Ala Val
165 170 175
His Thr Arg Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile Trp Ala Pro
180 185 190
Leu Ala Gly Thr Cys Gly Val Leu Leu Leu Ser Leu Val Ile Thr Leu
195 200 205
Tyr Cys Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro
210 215 220
Phe Met Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys
225 230 235 240
Arg Phe Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu Arg Val Lys Phe
245 250 255
Ser Arg Ser Ala Asp Ala Pro Ala Tyr Lys Gln Gly Gln Asn Gln Leu
260 265 270
Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr Asp Val Leu Asp
275 280 285
Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys Pro Arg Arg Lys
290 295 300
Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys Asp Lys Met Ala
305 310 315 320
Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg Arg Arg Gly Lys
325 330 335
Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala Thr Lys Asp Thr
340 345 350
Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
355 360
<210> 7
<211> 42
<212> DNA
<213> Artificial sequence ()
<400> 7
cgcgattctt aagggtgtcc agtgcgaagt gcagctggtg ga 42
<210> 8
<211> 45
<212> DNA
<213> Artificial sequence ()
<400> 8
gcatggagga cagggcttga ttgtggggct agacactgtc acctg 45
<210> 9
<211> 21
<212> PRT
<213> Artificial sequence ()
<400> 9
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> 10
<211> 69
<212> PRT
<213> Artificial sequence ()
<400> 10
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> 11
<211> 42
<212> PRT
<213> Artificial sequence ()
<400> 11
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> 12
<211> 112
<212> PRT
<213> Artificial sequence ()
<400> 12
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> 13
<211> 67
<212> DNA
<213> Artificial sequence ()
<400> 13
ctgccgctgg ccttgctgct ccacgccgcc aggccggaag tgcagctggt ggagagcggc 60
ggcggat 67
<210> 14
<211> 32
<212> DNA
<213> Artificial sequence ()
<400> 14
gcgctggcgt cgtggtgcta gacactgtca cc 32
<210> 15
<211> 75
<212> DNA
<213> Artificial sequence ()
<400> 15
gctgcaggtc gactctagag gatcccgcca ccatggcctt accagtgacc gccttgctcc 60
tgccgctggc cttgc 75
<210> 16
<211> 22
<212> DNA
<213> Artificial sequence ()
<400> 16
accacgacgc cagcgccgcg ac 22
<210> 17
<211> 18
<212> DNA
<213> Artificial sequence ()
<400> 17
tcgataagct tgatatcg 18

Claims (10)

1. A CD123 antigen binding fragment, wherein said CD123 antigen binding fragment comprises the heavy chain CDR1 of SEQ ID NO. 1, the heavy chain CDR2 of SEQ ID NO. 2, and the heavy chain CDR3 of SEQ ID NO. 3.
2. An anti-CD 123 antibody, wherein said anti-CD 123 antibody comprises the CD123 antigen-binding fragment of claim 1;
preferably, the anti-CD 123 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO. 4.
3. A nucleic acid molecule comprising a DNA segment encoding the CD123 antigen-binding fragment of claim 1;
preferably, the nucleic acid molecule comprises a DNA fragment encoding the anti-CD 123 antibody of claim 2;
preferably, the nucleic acid molecule comprises the nucleic acid sequence shown in SEQ ID NO. 5.
4. A chimeric antigen receptor comprising a signal peptide, an antigen binding domain, a hinge region, a transmembrane domain, and a signaling domain;
the antigen binding domain is the anti-CD 123 antibody of claim 2.
5. The chimeric antigen receptor according to claim 4, wherein the signal peptide comprises a CD8 a signal peptide;
preferably, the hinge region comprises a CD8 a hinge region;
preferably, the transmembrane domain comprises any one or a combination of at least two of the CD8 a transmembrane region, CD28 transmembrane region, or DAP10 transmembrane region;
preferably, the signaling domain comprises CD3 ζ;
preferably, the signaling domain further comprises any one of or a combination of at least two of the 4-1BB, CD28 intracellular domains, OX40, ICOS or DAP10 intracellular domains;
preferably, the chimeric antigen receptor comprises the anti-CD 123 antibody of claim 2, a CD8 a transmembrane region, 4-1BB, and CD3 ζ;
preferably, the chimeric antigen receptor comprises the anti-CD 123 antibody of claim 2, a CD28 transmembrane region, a CD28 intracellular region, and a CD3 ζ;
preferably, the chimeric antigen receptor comprises the anti-CD 123 antibody of claim 2, a CD8 a transmembrane region, OX40, and CD3 ζ;
preferably, the chimeric antigen receptor comprises the anti-CD 123 antibody of claim 2, a CD8 a transmembrane region, ICOS, and CD3 ζ;
preferably, the chimeric antigen receptor comprises the anti-CD 123 antibody of claim 2, a CD28 transmembrane region, a CD28 intracellular region, OX40, and CD3 ζ;
preferably, the chimeric antigen receptor comprises the amino acid sequence shown in SEQ ID NO. 6.
6. An expression vector comprising a gene encoding the chimeric antigen receptor of claim 4 or 5;
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 claim 4 or 5, preferably a lentiviral vector.
7. A recombinant lentivirus prepared from a mammalian cell transfected with the expression vector of claim 6 and a helper plasmid.
8. A chimeric antigen receptor immune cell, wherein said chimeric antigen receptor immune cell expresses the chimeric antigen receptor of claim 4 or 5;
preferably, the chimeric antigen receptor immune cell comprises the expression vector of claim 6 and/or the recombinant lentivirus of claim 7;
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.
9. A pharmaceutical composition comprising the chimeric antigen receptor immune cell of claim 8;
preferably, the pharmaceutical composition further comprises any one or a combination of at least two of a pharmaceutically acceptable carrier, excipient or diluent.
10. Use of the CD123 antigen-binding fragment of claim 1, the anti-CD 123 antibody of claim 2, the nucleic acid molecule of claim 3, the chimeric antigen receptor of claim 4 or 5, the expression vector of claim 6, the recombinant lentivirus of claim 7, the chimeric antigen receptor immune cell of claim 8, or the pharmaceutical composition of claim 9 in the preparation of a medicament for the treatment of a malignant tumor;
preferably, the malignancy comprises a hematological tumor.
CN202011461164.6A 2020-12-11 2020-12-11 CD123 antigen binding fragment and application thereof Active CN112521506B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011461164.6A CN112521506B (en) 2020-12-11 2020-12-11 CD123 antigen binding fragment and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011461164.6A CN112521506B (en) 2020-12-11 2020-12-11 CD123 antigen binding fragment and application thereof

Publications (2)

Publication Number Publication Date
CN112521506A true CN112521506A (en) 2021-03-19
CN112521506B CN112521506B (en) 2022-09-09

Family

ID=74999237

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011461164.6A Active CN112521506B (en) 2020-12-11 2020-12-11 CD123 antigen binding fragment and application thereof

Country Status (1)

Country Link
CN (1) CN112521506B (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160297882A1 (en) * 2015-04-08 2016-10-13 Sorrento Therapeutics, Inc. Antibody Therapeutics That Bind CD123
CN107312097A (en) * 2017-07-18 2017-11-03 深圳市免疫基因治疗研究院 A kind of Chimeric antigen receptor and its application based on CD30
CN107312098A (en) * 2017-07-18 2017-11-03 深圳市免疫基因治疗研究院 A kind of Chimeric antigen receptor and its application based on CD22
CN110804096A (en) * 2019-11-21 2020-02-18 深圳普瑞金生物药业有限公司 CD123 single domain antibody, nucleotide sequence, expression vector and kit
CN111944062A (en) * 2019-12-09 2020-11-17 深圳市体内生物医药科技有限公司 Chimeric antigen receptor for recognizing Fc fragment and application thereof
CN112028996A (en) * 2020-10-30 2020-12-04 南京北恒生物科技有限公司 Single domain antibodies targeting BCMA and uses thereof
CN112048481A (en) * 2020-09-09 2020-12-08 广东昭泰体内生物医药科技有限公司 Chimeric antigen receptor NK (natural killer) cell targeting CD19 and application thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160297882A1 (en) * 2015-04-08 2016-10-13 Sorrento Therapeutics, Inc. Antibody Therapeutics That Bind CD123
CN107312097A (en) * 2017-07-18 2017-11-03 深圳市免疫基因治疗研究院 A kind of Chimeric antigen receptor and its application based on CD30
CN107312098A (en) * 2017-07-18 2017-11-03 深圳市免疫基因治疗研究院 A kind of Chimeric antigen receptor and its application based on CD22
CN110804096A (en) * 2019-11-21 2020-02-18 深圳普瑞金生物药业有限公司 CD123 single domain antibody, nucleotide sequence, expression vector and kit
CN111944062A (en) * 2019-12-09 2020-11-17 深圳市体内生物医药科技有限公司 Chimeric antigen receptor for recognizing Fc fragment and application thereof
CN112048481A (en) * 2020-09-09 2020-12-08 广东昭泰体内生物医药科技有限公司 Chimeric antigen receptor NK (natural killer) cell targeting CD19 and application thereof
CN112028996A (en) * 2020-10-30 2020-12-04 南京北恒生物科技有限公司 Single domain antibodies targeting BCMA and uses thereof

Also Published As

Publication number Publication date
CN112521506B (en) 2022-09-09

Similar Documents

Publication Publication Date Title
CN112480258B (en) anti-CD 123 nano antibody and application thereof
RU2748281C2 (en) Fully human mesothelin antibodies and immune effector cells targeting mesothelin
CN112673025B (en) Chimeric antigen receptor containing third signal receptor and application thereof
CN112812186B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN112500487B (en) anti-CD 123 monoclonal antibody and application thereof
CN111944062A (en) Chimeric antigen receptor for recognizing Fc fragment and application thereof
CN112029729B (en) CD19 and CD22 double-target chimeric antigen receptor NK (natural killer) cell and application thereof
CN111944054A (en) anti-BCMA CAR and expression vector and application thereof
CN116234911A (en) Chimeric antigen receptor specifically binding MSLN and application thereof
CN113621073A (en) Novel chimeric receptor composition, recombinant vector, cell and application thereof
CN110862456B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN112812185B (en) Monoclonal antibody for resisting B cell maturation antigen and application thereof
CN112830971B (en) anti-BCMA antigen binding fragment and application thereof
CN110713539B (en) Anti-carcinoembryonic antigen antibody and preparation method and application thereof
CN113265001B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN112521505B (en) anti-CD 123 antibodies and uses thereof
CN110194803A (en) A kind of Chimeric antigen receptor and its application targeting EpCAM
CN110396131B (en) ErbB2 single-chain antibody, chimeric antigen receptor targeting human ErbB2, recombinant vector, recombinant cell and application
CN112521506B (en) CD123 antigen binding fragment and application thereof
CN114149506B (en) anti-CD 22 antibody and application thereof
CN112979808B (en) Antibody for resisting B cell mature antigen and application thereof
US11590168B2 (en) Chimeric antigen receptor comprising co-stimulatory receptor and application thereof
CN114805596A (en) Chimeric antigen receptor taking glypican 3 as target spot and application thereof
WO2023246578A1 (en) Chimeric antigen receptor specifically binding to gpc3 and use thereof
CN116675773A (en) anti-MXR 7 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