CN111620955B - Multi-target-site composite antigen and application thereof - Google Patents

Multi-target-site composite antigen and application thereof Download PDF

Info

Publication number
CN111620955B
CN111620955B CN202010627451.3A CN202010627451A CN111620955B CN 111620955 B CN111620955 B CN 111620955B CN 202010627451 A CN202010627451 A CN 202010627451A CN 111620955 B CN111620955 B CN 111620955B
Authority
CN
China
Prior art keywords
antigen
polypeptide
seq
ctl
vaccine
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
CN202010627451.3A
Other languages
Chinese (zh)
Other versions
CN111620955A (en
Inventor
卢戌
刘静维
王跃
刘雪松
李君阳
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.)
Beijing Biohealthcare Cell Technology Co ltd
Original Assignee
Beijing Biohealthcare Cell Technology 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 Beijing Biohealthcare Cell Technology Co ltd filed Critical Beijing Biohealthcare Cell Technology Co ltd
Priority to CN202010627451.3A priority Critical patent/CN111620955B/en
Publication of CN111620955A publication Critical patent/CN111620955A/en
Application granted granted Critical
Publication of CN111620955B publication Critical patent/CN111620955B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • 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
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • 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
    • A61K2039/5158Antigen-pulsed cells, e.g. T-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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2304Interleukin-4 (IL-4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/00022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16211Lymphocryptovirus, e.g. human herpesvirus 4, Epstein-Barr Virus
    • C12N2710/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Abstract

The invention relates to a multi-target composite antigen and application thereof, and aims to provide a multi-target composite antigen, namely a combined CTL epitope peptide. Another objective of the invention is to provide a multivalent vaccine against multiple target sites on the surface of tumor cells. It is still another object of the present invention to provide a multi-target complex antigen loaded CD8 + Cytotoxic T lymphocytes and a preparation method thereof. It is still another object of the present invention to provide an epitope-induced specific CTL effector cell.

Description

Multi-target-site composite antigen and application thereof
Technical Field
The invention relates to the fields of biology and medicine, in particular to a multi-target composite antigen and application thereof.
Background
In recent years, the important role of the immune system in tumor prevention and treatment has been widely agreed, and immunotherapy based on anti-tumor specific immune reconstitution is an important means which is internationally acknowledged at present and hopefully and completely eliminates tumor cells in vivo after surgery, radiotherapy and chemotherapy so as to radically treat tumors, and becomes a main direction of future tumor treatment. The immunotherapy mainly activates and mobilizes cells with cytotoxic activity and cytokines by supplementing, inducing and activating the inherent biological response regulation system of an organism in vitro, improves the functions of the autoimmune system of a patient, enhances the differentiation capacity of specific tumor killer cells, kills the tumor cells on the cellular level, effectively inhibits the metastasis, diffusion and recurrence of the tumor cells, has slight side reaction and good clinical treatment effect, and overcomes the defects of incomplete treatment, easy metastasis, great side effect and the like of the traditional tumor treatment mode. With the intensive research on tumor immunity at cellular and molecular level, tumor antigen-loaded Dendritic Cell (DCs) -sensitized tumor-specific Cytotoxic T Lymphocytes (CTLs) as an important part of immunotherapy can specifically recognize tumors, expand in cloning, form immunological memory in vivo, and mediate persistent anti-tumor specific immune effects. Activation of CTL requires the co-stimulation of a tumor antigen priming and an Antigen Presenting Cell (APC) provided co-stimulatory signals as well as a third class of signal molecules provided by cytokines. The existence of tumor-associated and specific antigens lays a theoretical and material foundation for specific immunotherapy. Only tumor antigens with strong immunogenicity and high specificity can effectively activate T lymphocytes with tumor specificity. In recent years, tumor polypeptide therapeutic vaccines developed aiming at tumor-associated antigens (TAA) or tumor-specific antigens (TSA) expressed by tumors are expected to overcome the defects of the therapy by virtue of the characteristics of strong specificity, safety, low price, easiness in storage and application and the like, so as to achieve the aim of effectively treating the tumors.
Tumor antigens must be degraded into short peptides and form peptide-MHC-TCR complexes in Antigen Presenting Cells (APC) to be recognized by T cells, triggering corresponding cytotoxic T lymphocyte responses. Since the polypeptide vaccine aims to deliver a high dose of tumor antigen polypeptides to MHC (major histocompatibility complex) molecules on the surface of APC, suitable and effective tumor antigen polypeptides are very important for the preparation of tumor vaccines. And the tumor antigen polypeptides are restricted by MHC, only patients with the same MHC class I molecule can use the same peptide, and some tumor antigen peptides may induce immune tolerance rather than activate immune response due to tumor heterogeneity. In addition, the weak immunogenicity is another weakness of polypeptide vaccines, and the immunogenicity can be effectively improved and stronger CTL activity can be induced by modifying epitope polypeptides (substitution of single amino acid, PMRI modification of polypeptide, lipopeptide and the like) or adopting multivalent vaccines.
Enzyme-linked immunospot assay (ELISPOT) is a cellular immunological detection technique that can detect cytokine-secreting T cells from the single cell level. The principle of using ELISPOT to detect CTL secreting IFN-gamma is that the monoclonal antibody of IFN-gamma is coated on a 96-well plate attached with PVDF membrane, then T cells stimulated by immunogenic epitope polypeptide are added into micropores to be cultured, and the secreted IFN-gamma is combined with the coated antibody. After washing the cells and unbound components away, an enzyme-labeled IFN-gamma detection monoclonal antibody is added to bind with the captured IFN-gamma. Upon addition of the substrate, colored spots can be generated where there is IFN- γ, each spot representing one IFN- γ secreting cell. The number of the spots is counted by using a computer imaging analysis system, and the number of the IFN-gamma-secreting CTLs in the sample can be determined. The ELISPOT technology is highly sensitive, and due to the high concentration of cytokines in the vicinity of the cells, cells secreting 100 cytokines can also be found, which is sufficient to detect 1/105 IFN-. Gamma.secreting cells. And the ELISPOT technique is highly specific because two monoclonal antibodies recognizing two different epitopes of cytokines or antibodies are used.
The invention is based on the follow-up research of the Chinese invention patent with the application number of 201510270656. The documents cited in the present invention are as follows, which are incorporated by reference into the present patent application. Patent document 1: publication No.: CN101302537A; patent document 2: publication No.: CN 101854945A; patent document 3: publication No.: CN 102625832A.
Disclosure of Invention
It is an object of the present invention to provide a multi-target complex antigen, i.e., a combined CTL antigenAn epitope peptide. It is another object of the present invention to provide a multivalent vaccine against multiple targets on the surface of tumor cells. It is still another object of the present invention to provide a multi-target complex antigen loaded CD8 + Cytotoxic T lymphocytes and a preparation method thereof. It is still another object of the present invention to provide an epitope-induced specific CTL effector cell.
The invention provides a polypeptide, which consists of EBV, HPV, HSV and CMV, wherein:
the amino acid sequence of the EBV is SEQ ID NO: 1. SEQ ID NO: 7. SEQ ID NO: 8. SEQ ID NO: 9. SEQ ID NO: 10. SEQ ID NO: 11. SEQ ID NO:12 or SEQ ID NO:13;
the amino acid sequence of the HPV is SEQ ID NO: 2. the amino acid sequence of SEQ ID NO: 14. SEQ ID NO: 15. the amino acid sequence of SEQ ID NO: 16. SEQ ID NO: 17. the amino acid sequence of SEQ ID NO: 18. SEQ ID NO: 19. the amino acid sequence of SEQ ID NO: 20. the amino acid sequence of SEQ ID NO:21 or SEQ ID NO:22;
the HSV has an amino acid sequence shown as SEQ ID NO:3;
the amino acid sequence of CMV is SEQ ID NO:4 or SEQ ID NO:23.
wherein the amino acid sequence of the polypeptide is SEQ ID NO:5.
the invention also provides a nucleic acid molecule encoding the polypeptide of claim 2.
The nucleic acid is a DNA molecule, and the DNA molecule is a DNA molecule shown as a sequence 6 in a sequence table.
Recombinant expression vectors, expression cassettes, transgenic cell lines, recombinant bacteria or recombinant viral vectors containing said nucleic acid molecules should also be within the scope of the present invention.
The invention also provides a vaccine, wherein the active ingredients of the vaccine are any one of the following substances:
1) The polypeptide;
2) The nucleic acid molecule;
3) The recombinant expression vector;
4) The expression cassette;
5) The transgenic cell line;
6) The recombinant bacterium;
7) The recombinant viral vector.
The vaccine is a DC vaccine.
The invention also provides a multi-target composite antigen loaded CD8 + Cytotoxic T lymphocytes, the multi-target complex antigen loaded with CD8 + An epitope of a cytotoxic T lymphocyte comprises the polypeptide.
The invention also provides an epitope-induced specific CTL effector cell, which is characterized in that an antigenic epitope comprises the polypeptide.
The application of any one of the following substances in preparing the medicine for treating cancer also falls within the protection scope of the present invention:
1) The polypeptide;
2) The nucleic acid molecule;
3) The recombinant expression vector;
4) The expression cassette;
5) The transgenic cell line;
6) The recombinant bacterium;
7) The recombinant viral vector;
8) The vaccine as described;
9) The multi-target composite antigen loaded with CD8 + Cytotoxic T lymphocytes.
The multi-Target complex antigen (MTCA) provided by the invention is a mode of combining a plurality of epitopes, the combination mode is that at least four polypeptide combinations matched with human leukocyte antigens are selected from a series of candidate polypeptides according to genetic gene structure and function difference to prepare tumor vaccines, so that specific immune response of a patient body to tumors is stimulated, the survival time of the tumor vaccines is prolonged, and the MTCA has the unique advantages of bypassing immune diversity and tumor heterogeneity compared with other tumor vaccines, and can effectively induce stronger CTL killing activity.
In an embodiment of the present invention, CTLs for a plurality of tumor antigens and methods of preparing the same are provided. The present invention can be used for any tumor, including but not limited to: lung cancer, breast cancer, kidney cancer, stomach cancer, colorectal cancer, pancreatic cancer, liver cancer, cervical cancer, bladder cancer, prostate cancer, melanoma, head and neck tumor, etc., and can also be used for cancer related to blood and bone marrow.
In a preferred embodiment of the invention, the CTL product of the invention is expressed as an antigen that specifically corresponds to that exhibited by the tumor. Thus, co-infection of DC cells with a combination of multiple epitope polypeptides in combination with lentivirus Lenti-hGM-CSF provides a means to generate a single CTL product with specific tumor independent antigenic characteristics.
The invention has important significance in that: provides a brand-new cell immunotherapy concept, namely an autoimmune cell therapy technology on the basis of breaking tumor immune tolerance, and establishes a new technology and a new method for implementing efficient specific tumor killing on the basis of breaking tumor immune tolerance. The technology solves the bottleneck problems of short survival time of the effector cells in vivo, low tumor killing efficiency, poor clinical curative effect and the like in the prior tumor immunotherapy, greatly improves the clinical curative effect of the cell immunotherapy, and has very wide application prospect.
In an embodiment of the invention, a lentiviral vector is constructed by the following method: providing PCR primers aiming at CDS region sequence of human peripheral blood hGM-CSF gene (GeneID: NM-000758.3), adding enzyme cutting sites at 5 'end and 3' end of the gene through PCR amplification, and amplifying hGM-CSF gene fragment by PCR method with plasmid pORFhGM-CSF as template; the hGM-CSF gene is directionally cloned into a lentivirus linked vector system through a PacI restriction endonuclease site to construct a recombinant vector. And mixing the recombinant vector, the envelope plasmid and the packaging structure plasmid to form a lentivirus vector system. The lentivirus of the invention is preferably Lenti-hGM-CSF.
In an embodiment of the invention, the DC vaccine is prepared by the following method: separating out monocyte, preferably separating out monocyte from peripheral blood, inducing rhGM-CSF, rhIL-4 and TNF-alpha to obtain DC cell, and infecting DC cell with the combined antigen epitope polypeptide (preferably SEQ ID NO: 5) and the lentivirus of the present invention to obtain DC vaccine.
In an embodiment of the present invention, theThe DC vaccine obtained after the DC cell is infected by the combined antigen epitope polypeptide (preferably SEQ ID NO: 5) and the lentivirus of the invention at the same time is incubated with the T lymphocyte to obtain the sensitized cytotoxic T lymphocyte. Preferably, the MTCA-DC/APC-CTL immune cell therapy technique is to express CD3 + CD8 + Predominantly cytotoxic T cells are the primary effector cells.
In an embodiment of the present invention, antigen-specific CTLs are prepared as follows: the DC cell loaded with the tumor-associated antigen obtained by the method is mixed with T lymphocyte, GM-CSF, IL-4 and IL-2 are added into a culture medium for continuous culture, and the cell is collected to obtain the epitope-induced specific CTL effector cell.
The specific CTL of the invention has obviously improved IFN-gamma secretion capability. In the test of the killing effect of MTCA-multi-target complex antigen induced antigen specific CTL on specific tumor cells, the antigen specific CTL induced by the MTCA-multi-target complex antigen can form specific lysis on target cells in the co-culture process of effective target cells, namely the CTL induced by the MTCA-multi-target complex antigen can kill target cells loaded with control peptides at the same time, and the application range of experimental polypeptide is expanded.
Drawings
FIG. 1 is a diagram of the construction of recombinant vector FattbC31UGW-hGM-CSF by targeted cloning of hGM-CSF gene into a lentiviral ligation vector system via pacl restriction endonuclease sites.
FIG. 2 is the envelope plasmid VSVG.
FIG. 3 shows the packaging construct plasmid CMV Δ 8.9-D64N/D116N.
FIG. 4 is a bar graph comparing the ability of the multi-target complex antigen of the present invention to induce antigen-specific CTL to produce IFN-. Gamma..
FIG. 5 is a bar graph comparing the killing effect of the multi-target complex antigen-induced antigen-specific CTLs on specific tumor cells according to the present invention.
Detailed Description
The present invention is described in further detail below with reference to specific embodiments, and the examples are given only for illustrating the present invention and not for limiting the scope of the present invention. The examples provided below serve as a guide for further modifications by a person skilled in the art and do not constitute a limitation of the invention in any way.
The experimental procedures in the following examples are conventional unless otherwise specified. Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
Example 1
Synthesis of polypeptides
The polypeptides in the following examples were synthesized using standard Fmoc protocol by Shanghai intense Biotechnology Ltd, and subjected to purification and purity analysis by high performance liquid chromatography, identification by mass spectrometry, and molecular weight determination. The results show that the purity of the synthesized polypeptide is higher than 95%, and the molecular weight is consistent with the theoretical value.
This example provides a polypeptide designated MTCA consisting of EBV, HPV, HSV and CMV wherein:
the amino acid sequence of the EBV is SEQ ID NO: 1. SEQ ID NO: 7. SEQ ID NO: 8. SEQ ID NO: 9. SEQ ID NO: 10. SEQ ID NO: 11. SEQ ID NO:12 or SEQ ID NO:13;
the amino acid sequence of the HPV is SEQ ID NO: 2. SEQ ID NO: 14. the amino acid sequence of SEQ ID NO: 15. SEQ ID NO: 16. SEQ ID NO: 17. SEQ ID NO: 18. SEQ ID NO: 19. SEQ ID NO: 20. SEQ ID NO:21 or SEQ ID NO:22;
the HSV has an amino acid sequence shown as SEQ ID NO:3;
the amino acid sequence of CMV is SEQ ID NO:4 or SEQ ID NO:23.
wherein when the amino acid sequence of EBV is SEQ ID NO: 1. the amino acid sequence of the HPV is SEQ ID NO: 2. the HSV has an amino acid sequence shown as SEQ ID NO:3 and the amino acid sequence of CMV is SEQ ID NO:4, and connecting the N terminal to the C terminal in sequence to obtain the polypeptide MTCA. The amino acid sequence of MTCA is SEQ ID NO:5 and the sequence of the corresponding coding gene is the DNA molecule shown in the sequence 6.
Example 2 construction of LentihGM-CSF, a lentivirus
1. PCR primer F1 (F1' -G) was designed based on the CDS region sequence of human peripheral blood hGM-CSF gene (GeneID: NM-000758.3)TTAATTAACATGTGGCTGCTGCAGAGCCTGCTGCTCT-3 ') and R1 (R1: 5' -G)TTAATTAACTCACTCCTGGACTGGCTCCCAGCAG-3 '), primers F1 and R1 can be amplified by PCR to add enzyme recognition sites at both the 5' end and the 3' end of the above genes. In the above primers, the underlined part is an enzyme recognition site.
2. Using plasmid pORFhGM-CSF (purchased from Invivogen, cat # porf-hgmcsf) as a template, and amplifying by PCR method through primers F1 and R1 to obtain hGM-CSF gene fragment; then, the hGM-CSF gene is directionally cloned into a lentivirus ligation vector FattbC31UGW (Purpurin biotechnology (Beijing) Co., ltd., product number Biovector912008, the structure of which is shown in figure 1) through a PacI restriction endonuclease site to obtain a recombinant vector FattbC31UGW-hGM-CSF. The recombinant vector FattbC31UGW-hGM-CSF is obtained by inserting hGM-CSF gene into the PacI restriction endonuclease site of the lentivirus linking vector FattbC31UGW and keeping other sequences of the lentivirus linking vector FattbC31UGW unchanged.
3. Mixing a recombinant vector FattbC31UGW-hGM-CSF, an envelope plasmid VSVG (prohibitin biotechnology (Beijing) Co., ltd., product No. Biovector912007, the structure of which is shown in FIG. 2) and a packaging structure plasmid pCMVAR8.91-D64N/D116N (prohibitin biotechnology (Beijing) Co., ltd., product No. Biovector912006, the structure of which is shown in FIG. 3) to form a lentivirus vector system, transfecting 293T cells by using liposome LipofectamineTM, observing under a fluorescence microscope after 24-48h, collecting virus supernatant after 72h appears in a large amount of fluorescence, obtaining lentivirus particles Lenti-hGM-CSF encoding hGM-CSF, concentrating the collected virus supernatant, and subpackaging for later use or immediate use.
4. When the activity of the recombinant lentivirus is determined, the concentrated virus stock solution is diluted in different proportions, cells are infected for 72 hours, and then fluorescence counting is carried out under a fluorescence microscope to determine the titer. The lentivirus ligation vector FattbC31UGW was used as a control. The results showed that the site-specific integrating lentivirus titer was 7.1X 10 -7 TU/ml。
The method for detecting the virus titer comprises the following steps: taking logarithmic growth HT1080 cells, spreading in a 96-well plate in 6 × 103 cells/well, adding DMEM medium containing 10 FBS into each well, diluting the fluorescent expression virus with serum-free DMEM medium in a round bottom 96-well plate in 10-fold gradient in the next day, repeating three wells in each gradient, discarding 90ul of the medium in each well of the 96-well plate with HT1080 cells, adding 90ul of the diluted virus solution in the round bottom 96-well plate correspondingly, changing into 100ul of the DMEM medium containing 10 FBS after 24h, after 72h infection, counting the number of fluorescence in the wells with moderate fluorescence expression, wherein the volume ratio of the fluorescence to the virus solution in the wells is the titer of the fluorescent expression virus, the titer of the fluorescent expression virus is a standard curve, and the cq value of the fluorescent expression virus and the target virus obtained by combining the QPCR method is combined, so as to calculate the titer (TU/mL) of the target virus.
Example 3 preparation of DC vaccine
Monocytes were isolated from human peripheral blood, DC cells were obtained by induction with rhGM-CSF, rhIL-4 and TNF- α, and DC vaccines were obtained by infecting DC cells with the multi-target complex antigen peptide MTCA (SEQ ID NO: 5) synthesized in example 1 and the lentivirus Lenti-hGM-CSF prepared in example 2 simultaneously after identification by phase contrast microscopy and flow cytometry. The method specifically comprises the following steps:
1) Collecting peripheral blood of volunteer, separating autologous plasma (inactivated at 56 deg.C for 30min and preserved at 4 deg.C) and PBMC (peripheral blood mononuclear cell) by density gradient separation method using lymphocyte separation liquid (Ficoll-Hypaque, polysucrose-diatrizoate), and treating PBMC with serum-free RPMI-1640 (5-7) x 10 7 After 1h, suspension cells are collected and used for separating T lymphocytes, adherent cells are replaced by 1640 culture medium containing rhGM-CSF (1000U/ml), rhIL-4 (1000U/ml) and TNF-alpha (500U/ml) to stimulate and induce the monocyte to differentiate to the DC.
2) Immature DCs collected in 24-well plates in suspension were infected on day 5 with the polypeptide MTCA in example 1 and Lenti-hGM-CSF concentrate of lentiviral particles in example 2, and the volume required for lentivirus was calculated according to the pre-experimental optimal MOI values. After 24 hours of transfection, the medium was replaced with fresh DC induction medium and rhGM-CSF (1000U/ml), rhIL-4 (1000U/ml) and TNF- α (500U/ml) were added to promote differentiation and maturation of DC, to obtain DC vaccine.
EXAMPLE 4 preparation of antigen-specific CTL
Collecting peripheral blood of volunteers, subjecting to density gradient centrifugation with lymphocyte separation liquid (Ficoll-Hypaque), collecting interface cells rich in mononuclear cells, suspending in RPMI-1640 culture medium, placing at 37 deg.C, and 5% CO 2 Culturing in incubator for 2 hr, collecting non-adherent suspension cells, re-suspending with RPMI-1640 medium containing 3% serum, and regulating cell density to 1 × 10 6 And each ml, the suspension is rich in T lymphocytes. The DC vaccine prepared in example 3 was mixed with T lymphocytes in a ratio of 1 6 Per ml; and collecting the cells after culturing for 1 week to obtain epitope-induced specific CTL effector cells named CTL-MTCA.
EXAMPLE 5 preparation of control samples
The polypeptide MTCA in example 1 was replaced with the polypeptide EBV (amino acid sequence SEQ ID NO: 1), the polypeptide HPV (amino acid sequence SEQ ID NO: 2), the polypeptide HSV (amino acid sequence SEQ ID NO: 3) and the polypeptide CMV (amino acid sequence SEQ ID NO: 4), respectively, according to the methods of examples 1 to 4; specific CTL effector cells induced by the epitopes are prepared and named as CTL-EBV, CTL-HPV, CTL-HSV and CTL-CMV respectively.
EXAMPLE 6 measurement and comparative experiment of the ability of MTCA-Multi-target Complex antigen of the present invention to induce antigen-specific CTL to produce IFN-. Gamma.
The experimental groups are: 1. EBV antigen control group (CTL-EBV cell to be detected); 2. HPV antigen control group (CTL-HPV is the cell to be detected); 3. HSV antigen control group (tested cell is CTL-HSV); 4. a CMV antigen control group (the cell to be detected is CTL-CMV); 5. MTCA-multi-target complex antigen group (the cell to be detected is CTL-CMV); 6. blank control group (100 ul of RPMI-1640 medium only). The specific experimental method is as follows:
MTCA-multi-target complex antigen group: the capability of CTL-MTCA to secrete IFN-gamma is detected by an enzyme-linked immunospot method (ELISPOT): an ELISPOT assay kit was used, and a 96-well plate was added with PBS diluted at 1100ul of IFN-. Gamma.capture antibody was incubated at 4 ℃ overnight, washed with PBS, and then 100ul of a test cell (CTL-MTCA prepared in example 4) solution (the content of the test cell was adjusted by RPMI-1640 medium) was added thereto, and the amount of the antibody was 1X 10 6 Cell/well, at 37 ℃,5% CO 2 After 24h incubation under conditions, washed with PBS containing 0.1% Tween 20, the biotin-labeled anti-IFN-. Gamma.antibody diluted by 1 2 Incubate for 2h under conditions, incubate for 1h with streptavidin-labeled alkaline phosphatase 100ul diluted with 1% BSA in PBS at 1. The experiment was performed in triplicate.
1. EBV antigen control group: the same procedure was followed except that the test cells in the MTCA-multi-target complex antigen group were replaced with CTL-EBV prepared in example 5.
2. HPV antigen control group (the cell to be tested is CTL-HPV);
3. HSV antigen control group (tested cell is CTL-HSV);
4. a CMV antigen control group (the cell to be detected is CTL-CMV);
5. MTCA-multi-target complex antigen group (the cell to be detected is CTL-CMV);
6. blank control group: the procedure was the same except that 100ul of the cell sap to be tested was replaced with 100ul of RPMI-1640 medium.
The results are shown in FIG. 4 and show that: the capacity of the specific CTL (cytotoxic T lymphocyte) for secreting IFN-gamma in the MTCA-multi-target compound antigen group is obviously higher than that in the other 5 groups, the capacity of inducing and secreting IFN-gamma is strongest, and the capacity of secreting IFN-gamma is improved by more than 3 times compared with that of a control group in which a single antigen peptide loads the specific CTL.
Example 7 MTCA-Multi-target Complex antigen of the present invention induces killing Effect of antigen-specific CTL against specific tumor cells
The experimental groups are: MTCA-multi-target complex antigen group:
MTCA-multi-target complex antigen group: time resolved fluorescence cytotoxicity assays were used for the analysis. CTL-MTCA prepared in examples 4 and 5 were used as effector cells (E) and T2+ EBV, \8230; target cells \8230Specific killing activity of CTL was examined for target cells (T). At 37 ℃,5% CO 2 Under the condition, effector cells and target cells are mixed according to the quantitative ratio of 50:1 co-incubation for 4h, setting 3 multiple wells for each sample, taking an average value as a detection result, and calculating the cell killing activity of CTL-MTCA according to the following formula, wherein the specific lysis rate = (fluorescence intensity of the well to be detected-fluorescence intensity of the natural release well)/(fluorescence intensity of the maximum release well-fluorescence intensity of the natural release well) × 100%.
The experimental groups are: 1. EBV antigen control group (CTL-EBV); 2. HPV antigen control group (CTL-HPV); 3. HSV antigen control group (CTL-HSV); 4. a CMV antigen control group (CTL-CMV); 5. MTCA-Multi-target Complex antigen group (CTL-MTCA).
The results are shown in fig. 5, antigen-specific CTLs (CTL-MTCA) induced by MTCA-multi-target complex antigen can form specific lysis to target cells during the co-culture of effector target cells, while effector cells induced by each control polypeptide can only kill target cells loaded with the corresponding peptide, i.e., CTLs induced by MTCA-multi-target complex antigen can simultaneously kill target cells loaded with the above four control peptides, thus expanding the application range of experimental polypeptides. The MTCA-multi-target compound antigen group has higher specific cracking rate (more than 70%) for four groups of target cells T2+ EBV, T2+ HSV, T2+ CMV and T2+ HPV, among other four groups, the EBV antigen control group has the highest specific cracking rate for the target cells T2+ EBV, the HSV antigen control group has the highest specific cracking rate for the target cells T2+ HSV, the CMV antigen control group has the highest specific cracking rate for the target cells T2+ CMV, the HPV antigen control group has the highest specific cracking rate for the target cells T2+ HPV, but the highest value is smaller than the specific cracking rate corresponding to the MTCA-multi-target compound antigen group.
The present invention has been described in detail above. It will be apparent to those skilled in the art that the invention can be practiced in a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation. While the invention has been described with reference to specific embodiments, it will be appreciated that the invention can be further modified. In general, this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains. The use of some of the essential features is possible within the scope of the claims attached below.
Sequence listing
<110> Beijing kang Ericao cell technology Limited
<120> multi-target site composite antigen and application thereof
<160> 23
<170> SIPOSequenceListing 1.0
<210> 1
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Tyr Leu Leu Glu Met Leu Trp Arg Leu
1 5
<210> 2
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Thr Leu Gln Asp Ile Val Leu His Leu
1 5
<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Gly Leu Ala His Val Ala Ala Ala Val
1 5
<210> 4
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Val Leu Glu Glu Thr Ser Val Met Leu
1 5
<210> 5
<211> 36
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Tyr Leu Leu Glu Met Leu Trp Arg Leu Thr Leu Gln Asp Ile Val Leu
1 5 10 15
His Leu Gly Leu Ala His Val Ala Ala Ala Val Val Leu Glu Glu Thr
20 25 30
Ser Val Met Leu
35
<210> 6
<211> 108
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 6
tacctcctcg agatgctctg gcggctcacg ctccaagaca ttgtcctcca cctcggtctc 60
gcccacgtcg ccgccgccgt cgtcctcgag gagacgtccg tcatgctc 108
<210> 7
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Ala Leu Leu Val Leu Tyr Ser Phe Ala Leu
1 5 10
<210> 8
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Leu Leu Val Leu Tyr Ser Phe Ala Leu
1 5
<210> 9
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Val Leu Tyr Ser Phe Ala Leu Met Leu
1 5
<210> 10
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 10
Glu Ser Asp Ser Asn Ser Asn Glu Gly
1 5
<210> 11
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 11
Ile Ala Leu Tyr Leu Gln Gln Asn Trp
1 5
<210> 12
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 12
Ala Leu Tyr Leu Gln Gln Asn Trp Trp
1 5
<210> 13
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 13
Tyr Leu Gln Gln Asn Trp Trp Thr Leu
1 5
<210> 14
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 14
Lys Cys Ile Asp Phe Tyr Ser Arg Ile
1 5
<210> 15
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 15
Thr Ile His Asp Ile Ile Leu Glu Cys
1 5
<210> 16
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 16
Thr Leu Glu Asp Leu Leu Met Gly Thr
1 5
<210> 17
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 17
Gln Leu Leu Arg Arg Glu Val Tyr Asp Phe Ala Phe Arg Asp Leu
1 5 10 15
<210> 18
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 18
Ala Phe Arg Asp Leu Cys Ile Val Tyr Arg
1 5 10
<210> 19
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 19
Tyr Met Leu Asp Leu Gln Pro Glu Thr Thr
1 5 10
<210> 20
<211> 19
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 20
Gln Ala Glu Pro Asp Arg Ala His Tyr Asn Ile Val Thr Phe Cys Cys
1 5 10 15
Lys Cys Asp
<210> 21
<211> 14
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 21
Asp Leu Leu Met Gly Thr Leu Gly Ile Val Cys Pro Ile Cys
1 5 10
<210> 22
<211> 15
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 22
Ala Asp Asp Leu Arg Ala Phe Gln Gln Leu Phe Leu Asn Thr Leu
1 5 10 15
<210> 23
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 23
Asn Leu Val Pro Met Val Ala Thr Val
1 5

Claims (7)

1. A polypeptide consisting of EBV, HPV, HSV and CMV the amino acid sequence is SEQ ID NO:5, the CTL induced by the polypeptide composite antigen has a broad spectrum type for killing target cells loaded with at least one control peptide of EBV, HPV, HSV and CMV at the same time.
2. A nucleic acid molecule for encoding the polypeptide of claim 1, wherein the nucleic acid is a DNA molecule, and the DNA molecule is shown as a sequence 6 in a sequence table.
3. A recombinant expression vector, expression cassette, transgenic cell line, recombinant bacterium or recombinant viral vector comprising the nucleic acid molecule of claim 2.
4. The vaccine is characterized in that the active ingredients of the vaccine are any one of the following substances:
1) A polypeptide according to claim 1;
2) The nucleic acid molecule of claim 2;
3) The recombinant expression vector of claim 3;
4) The expression cassette of claim 3;
5) The transgenic cell line of claim 3;
6) The recombinant bacterium of claim 3;
7) The recombinant viral vector of claim 3.
5. The vaccine of claim 4, wherein the vaccine is a DC vaccine.
6. An epitope-induced specific CTL effector cell, wherein the epitope comprises the polypeptide of claim 1, and said CTL effector cell is capable of recognizing EBV, HPV, HSV and CMV simultaneously with the polypeptide of claim 1.
7. Use of any one of the following in the manufacture of a medicament for the treatment of cancer:
1) A polypeptide according to claim 1;
2) The nucleic acid molecule of claim 2;
3) The recombinant expression vector of claim 3;
4) The expression cassette of claim 3;
5) The transgenic cell line of claim 3;
6) The recombinant bacterium of claim 3;
7) The recombinant viral vector of claim 3;
8) The vaccine of claim 4 or 5;
9) An epitope-induced specific CTL effector cell of claim 6.
CN202010627451.3A 2020-07-01 2020-07-01 Multi-target-site composite antigen and application thereof Active CN111620955B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010627451.3A CN111620955B (en) 2020-07-01 2020-07-01 Multi-target-site composite antigen and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010627451.3A CN111620955B (en) 2020-07-01 2020-07-01 Multi-target-site composite antigen and application thereof

Publications (2)

Publication Number Publication Date
CN111620955A CN111620955A (en) 2020-09-04
CN111620955B true CN111620955B (en) 2022-11-08

Family

ID=72269608

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010627451.3A Active CN111620955B (en) 2020-07-01 2020-07-01 Multi-target-site composite antigen and application thereof

Country Status (1)

Country Link
CN (1) CN111620955B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1865065A1 (en) * 1998-12-09 2007-12-12 The Government of the United States of America, as represented by the Secretary, Department of Health and Human Services A recombinant vector expressing multiple costimulatory molecules and uses thereof
CN103328003A (en) * 2010-08-25 2013-09-25 亥姆霍兹慕尼黑中心德国研究健康与环境有限责任公司 An epstein-barr-virus vaccine
CN104926944A (en) * 2015-05-22 2015-09-23 北京康爱瑞浩生物科技股份有限公司空港分公司 Preparation method and application of multiple target complex antigen-loaded CD8<+> cytotoxic T lymphocyte
CN106103483A (en) * 2014-01-13 2016-11-09 贝勒研究院 The novel vaccine of the related disease of anti-HPV with HPV
CN109796536A (en) * 2019-02-22 2019-05-24 上海尚泰生物技术有限公司 A kind of preparation method for the CTL targeting a variety of epitopes of glioblastoma

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10350288B2 (en) * 2016-09-28 2019-07-16 Genocea Biosciences, Inc. Methods and compositions for treating herpes

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1865065A1 (en) * 1998-12-09 2007-12-12 The Government of the United States of America, as represented by the Secretary, Department of Health and Human Services A recombinant vector expressing multiple costimulatory molecules and uses thereof
CN103328003A (en) * 2010-08-25 2013-09-25 亥姆霍兹慕尼黑中心德国研究健康与环境有限责任公司 An epstein-barr-virus vaccine
CN106103483A (en) * 2014-01-13 2016-11-09 贝勒研究院 The novel vaccine of the related disease of anti-HPV with HPV
CN104926944A (en) * 2015-05-22 2015-09-23 北京康爱瑞浩生物科技股份有限公司空港分公司 Preparation method and application of multiple target complex antigen-loaded CD8<+> cytotoxic T lymphocyte
CN109796536A (en) * 2019-02-22 2019-05-24 上海尚泰生物技术有限公司 A kind of preparation method for the CTL targeting a variety of epitopes of glioblastoma

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
A CTL epitope from human cytomegalovirus IE1 defined by combining prediction of HLA binding and proteasomal processing is the target of dominant immune responses in patients after allogeneic stem cell transplantation;Holger Hebart等;《Experimental Hematology》;20031031;966–973 *
EBV, HSV, CMV and HPV in Laryngeal and Oropharyngeal Carcinoma in Polish Patients;DOROTA POLZ-GRUSZKA等;《ANTICANCER RESEARCH》;20150301;第35卷(第1期);1657-1662 *
Generating CTLs against the subdominant Epstein-Barr virus LMP1 antigen for the adoptive immunotherapy of EBV-associated malignancies;Stephen Gottschalk等;《IMMUNOBIOLOGY》;20020301;第101卷(第5期);1905-1912 *
Human T-Cell Responses to HLA-A-restricted High Binding Affinity Peptides of Human Papillomavirus Type 18 Proteins E6 and E7;Michael P. Rudolf等;《Clinical Cancer Research》;20010331;第7卷;788-795 *

Also Published As

Publication number Publication date
CN111620955A (en) 2020-09-04

Similar Documents

Publication Publication Date Title
CN105602992B (en) It is a kind of based on the CAR-T transgene carrier and its construction method of replication defective recombinant slow virus and application
WO2021189571A1 (en) Vaccine vector capable of efficiently inducing body humoral immune response as well as preparation method therefor and application thereof
CN107286247B (en) The Dendritic Cells and application thereof of Chimeric antigen receptor modification containing anti-mesothelin single-chain antibody
CN111286493A (en) Oncolytic virus vaccine and medicine for treating tumor by combining oncolytic virus vaccine with immune cells
CN105949316B (en) Anti- EGFRvIII Chimeric antigen receptor, encoding gene, recombinant expression carrier and its construction method and application
CN104926944A (en) Preparation method and application of multiple target complex antigen-loaded CD8&lt;+&gt; cytotoxic T lymphocyte
CN110144326A (en) A kind of antitumor T cell of targeting and its preparation method and application
CN105950664B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD123 and its construction method and application
CN111606999B (en) Replicative oncolytic adenovirus with functions of activating immune co-stimulatory signaling pathway and blocking immune checkpoint and application thereof
CN113755448B (en) Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof
CN105950663B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD30 and its construction method and application
CN110358735B (en) Preparation method and application of CTL cell
CN111378624B (en) Targeting anti-tumor T cell and preparation method and application thereof
CN114592010A (en) NK-CAR-MbIL-15 cell and preparation method and application thereof
CN105950662B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD22 and its construction method and application
CN111704674B (en) Chimeric antigen receptor targeting c-Met and autocrine PD-L1 scFv and application thereof
CN111620955B (en) Multi-target-site composite antigen and application thereof
CN111849905A (en) Immunotherapy of mesenchymal stem cell targeted transport of chemokines and cytokines
CN110157674A (en) A kind of targeting T lymphocyte and its preparation method and application
CN105969805B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting Mesothelin and its construction method and application
CN109796536B (en) Preparation method of CTL (cytotoxic T lymphocyte) targeting multiple epitopes of glioblastoma
US11718827B2 (en) LRFFT2 cell
WO2021163874A1 (en) Recombinant viral vector, immunogenic composition comprising same, and uses
Kung et al. Lentiviral vector-transduced dendritic cells induce specific T cell response in a nonhuman primate model
CN109790224A (en) Tumor-antigen peptide and its application derived from CACNA1H

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