CN115124619A - Preparation method and application of clinical blood immune cells - Google Patents

Preparation method and application of clinical blood immune cells Download PDF

Info

Publication number
CN115124619A
CN115124619A CN202210729772.3A CN202210729772A CN115124619A CN 115124619 A CN115124619 A CN 115124619A CN 202210729772 A CN202210729772 A CN 202210729772A CN 115124619 A CN115124619 A CN 115124619A
Authority
CN
China
Prior art keywords
cells
beta
catenin
monoclonal antibody
culture
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
CN202210729772.3A
Other languages
Chinese (zh)
Other versions
CN115124619B (en
Inventor
王正好
赵理
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.)
Guangdong Qimei Life Medicine Technology Research Institute
Guangdong Qimei Pharmaceutical Biotechnology Group Co ltd
Guangdong Stanfu International Stem Cell Medical Research Institute
Zhuhai Qimei Stem Cell Bank Co ltd
Original Assignee
Beijing Shunyuantian Biological Products 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 Shunyuantian Biological Products Co ltd filed Critical Beijing Shunyuantian Biological Products Co ltd
Priority to CN202210729772.3A priority Critical patent/CN115124619B/en
Publication of CN115124619A publication Critical patent/CN115124619A/en
Application granted granted Critical
Publication of CN115124619B publication Critical patent/CN115124619B/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • 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
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • 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]
    • C12N2501/2315Interleukin-15 (IL-15)
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/11Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system cells
    • C12N2506/115Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system cells from monocytes, from macrophages
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to a preparation method and application of clinical blood immune cells. The invention provides a method for preparing and amplifying NK cells and the NK cells obtained by the method, and verifies that the NK cells have better tumor killing capacity, and after the NK cells are combined with beta-catenin monoclonal antibodies, the growth of tumors in mice can be remarkably inhibited, so that the NK cells have an excellent tumor treatment effect.

Description

Preparation method and application of clinical blood immune cells
Technical Field
The invention relates to the field of biology, in particular to the field of immune cells, and relates to a preparation method and application of clinical blood immune cells.
Background
NK cells were discovered 40 years ago, mainly originated from bone marrow, mainly present in blood and lymphoid organs, do not require prior sensitization, do not depend on antibodies and complement, are not limited by Major Histocompatibility Complex (MHC), can directly kill tumor cells, and have important roles in early anti-tumor and immune surveillance of the body. In tumor immunity, NK cells belong to nonspecific immune cells, surface markers mainly comprise CD3, CD56, CD16 and the like, the surface markers account for about 5% -15% of the total number of lymphocytes in peripheral blood of healthy people, a first immune killing line of an organism is formed, the organism is usually in a dormant state, and after a signal factor is activated, the signal factor can permeate into tissues to attack tumor cells and virus infected cells.
The NK cell has 4 receptors including a killer cell activating receptor (KAR), a killer cell inhibiting receptor (KIR), a killer lectin-like receptor (KLR) and an Fc gamma receptor (CD16), the first 3 receptors can inhibit or activate the NK cell to enable the NK cell to recognize the capability of self-tissue cells and in-vivo abnormal tissue cells, the 4 th receptor mainly recognizes IgG1 and IgG3 markers on the surface of the NK cell and mediates the NK cell to recognize target cells coated by the antibody, so that antibody-dependent cell-mediated cytotoxicity (ADCC) is exerted, and further tumors specifically bound with the IgG antibody are killed. NK cells have a wide antitumor spectrum, can kill homologous, homologous or heterologous tumor cells, and can kill target cells by a mechanism that (i) perforin and granzyme are released to cause necrosis or apoptosis of the target cells. ② the target cell apoptosis is induced by death receptor regulation. ③ secreting a plurality of effector cytokines to resist the metastatic tumors.
And fourthly, stimulating the immune effect of the secondary tumor.
Abnormalities in the Wnt/β -eatenin signaling pathway are associated with human diseases, including tumors, osteoporosis, aging and degenerative disorders, among others. Therefore, the research on the Wnt/beta-eatenin signal transduction pathway not only helps to understand the occurrence mechanism of human diseases, but also provides a series of new targets for the treatment of diseases. At least 6 Wnt proteins (Wntl, Wnt2, Wnt3, Wnt3a, Wnt8, and Wnt8b) have been shown to activate the Wnt/β -eatenin signaling pathway. The activation of the Wnt/β -eatenin signalling pathway is determined by the levels of β -eatenin proteins in the cytoplasm. Normally, β -eatenin in cytoplasm is mediated by ubiquitin proteasome, which is a multi-protein complex composed of axin (axin), coliform protein, glycogen synthesis kinase 3 β, casein kinase let, etc., including ubiquitin activating enzyme (E1), ubiquitin conjugating enzyme (E2) and ubiquitin ligase (E3), and is in a degraded state and maintains low level expression. GSK-beta is used for marking protein by phosphorylating Ser3, Ser3 and Thr4, CK1 alpha is used for marking protein by phosphorylating Ser45 only, so that the protein is recognized by E3 subunit beta-transducin repeated protein, the degradation of beta-catenin molecules is mediated, and the proteasome of 26S is degraded. Wnt proteins initiate the aggregation of intracellular β -catenin by binding to the cell surface receptor complex transmembrane protein/co-receptor low density lipoprotein receptor-related proteins. However, the mechanism of cell signaling by binding of Wnt proteins to Fz/LRP receptors is not yet fully understood. Most scholars think that after 2 persons are combined, the downstream protein of the receptor complex, namely cytoplasmic dishevelled protein is phosphorylated, the activity of GSK-3 beta and CK1 alpha is inhibited, axin protein is fixed, the formation of a multi-protein complex is damaged, finally unphosphorylated beta-catenin is aggregated in cytoplasm, the recognition of beta-TrCP is escaped, the degradation process is avoided, and the translocation to nucleus is further carried out. In the nucleus, beta-catenin forms a complex with TCF. When no beta-catenin exists, TCF interacts with Groucho to form a complex to inhibit the transcription activity; when beta-catenin is present, it interferes with the interaction of TCF with Groucho, and co-initiates transcription of downstream target genes with TCF and other transcription cofactors such as CREB Binding Protein (CBP).
The expression level of beta-catenin of tumors with abnormal Wnt signals is up-regulated, so that the treatment targeting the beta-catenin attracts great attention. Currently, antisense oligonucleotide technology, RNA interference technology, and protein knock-out technology are under investigation. The antisense oligonucleotide aiming at the beta-catenin can reduce the expression of tumor beta-catenin protein and effectively inhibit the growth of tumors. The method for screening and obtaining the specific beta-catenin monoclonal antibody by taking the beta-catenin as a target is also an important direction of research. Especially, the combined application of immune cell therapy and beta-catenin monoclonal antibody is not studied sufficiently at present.
Disclosure of Invention
The invention overcomes the defects of the prior art, provides an improved preparation method of NK cells and a method for treating cancer by combining the NK cells and monoclonal antibodies, and has better effect.
In one aspect of the present invention, there are provided a highly active NK cell and a method for preparing the same.
Specifically, the preparation method of the NK cell comprises the following steps:
adding Ficoll-PaquePLUS mononuclear cell separation liquid into a centrifuge tube, slowly adding heparin anticoagulated peripheral blood along the tube wall, avoiding damaging the water phase of the Ficoll separation liquid, centrifuging at room temperature and 3500rpm for 15min, carefully sucking the upper plasma into another centrifuge tube, inactivating at 56 ℃ for 25min, for later use, sucking the yellowish mononuclear cell layer into the centrifuge tube, washing with sterile PBS for 3 times, discarding the supernatant, precipitating to obtain peripheral blood mononuclear cell PBMC, suspending the precipitated cells with serum-free PRMI-1640 medium into a plastic plate coated with goat anti-mouse IgG (25 μ g of goat anti-mouse IgG and 5ml of 0.05M-HCL buffer solution (PH PBS 9), incubating at room temperature for 2h, washing for 3 times, sealing with 1% FCS at room temperature for 15min, washing with PBS for 3 times), adding cells incubated with anti-CD3 into the Tris coated plate, incubating for 2h, slightly sucking the cell suspension to obtain NK cells at 4 ℃ for 2 hours; the mononuclear cells were resuspended in NK MACS GMP medium (Miltenyi), recombinant human IL-2 and IL-15 were added, mixed well and injected into small culture bags, and incubated at 37 ℃ in incubator. On day 3, NK MACS GMP culture medium and recombinant human IL-2 and IL-15 were supplemented, and the culture was continued after mixing. On day 4, the culture medium in the small culture bag was equally distributed to two large culture bags and the culture was continued, and NKMACS GMP culture medium and recombinant human IL-2 were supplied to each large culture bag every 3 days for a total of 14 days, i.e., the proliferated NK cells were collected.
The invention further provides application of the NK cells in preparing a pharmaceutical composition for treating cancer.
In addition, in another aspect, the invention also provides a monoclonal antibody specifically aiming at the beta-catenin.
Further, the antibody is a beta-catenin monoclonal antibody beta-catenin-2D 13, and the light chain variable region sequence of the beta-catenin-2D 13 monoclonal antibody is as follows:
DIVITQRPALMAASPGEKVTITCAEYKHHAELGHVWYQQKSGISPKPWIYRSFIYKGGVPARFSGSGSGTSYSLTITSMEAEDAATYYCNTWDWFTLPFGAGTKLELK
the heavy chain variable region sequence is:
EVQLEESATELARPGASVKLSCKASGYIFSFPGFNWIKQRPGQGLEWIGPGLQRLFHHCCFDMRWGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGPDDVCQHWGLGTTLAVSS。
furthermore, the invention also provides application of the beta-catenin monoclonal antibody in preparing a pharmaceutical composition for treating cancer.
Further, the specific kind of cancer in the use is lung cancer.
Further, the cancer is a cancer caused by PC9 human lung cancer cells.
Furthermore, the invention also provides the application of the beta-catenin monoclonal antibody and the NK cell in preparing a kit for treating cancer.
Furthermore, the beta-catenin monoclonal antibody in the medicine box is in the form of a pharmaceutical composition, and the beta-catenin monoclonal antibody also contains a pharmaceutically acceptable carrier.
Pharmaceutical carriers are known to those skilled in the art. These are most typically standard carriers for administering drugs to humans, including solutions at physiological pH, such as sterile water, saline, and buffered solutions. The composition can be administered intramuscularly or subcutaneously. Other compounds will be administered according to standard methods used by those skilled in the art.
In addition to the selected molecule, the pharmaceutical composition may include carriers, thickeners, diluents, buffers, preservatives, surfactants, and the like. The pharmaceutical compositions may also include one or more active ingredients, such as antimicrobial agents, anti-inflammatory agents, and the like.
Certain compositions can potentially be administered as pharmaceutically acceptable acid or base addition salts formed by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid,
further, the NK cells of the present invention are used at a concentration of 1X 10 in terms of cell density 7 /mL-1×10 9 mL, preferably, may be 2X 10 7 /mL。
Further, the NK cells and the mabs of the invention are administered simultaneously or substantially simultaneously, or half an hour apart.
Any route of introducing or delivering an agent to a subject is included. Administration can be by any suitable route, including orally, topically, intravenously, subcutaneously, transdermally, intramuscularly, intraarticularly, parenterally, by inhalation, via implanted reservoirs, parenterally (e.g., subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrastemal, intrathecal, intraperitoneal, intrahepatic, enteral and intracranial injection or infusion techniques), and the like, intraarterially, intradermally, intraventricularly, intracranial, intraperitoneal, intranasal, rectal, intravaginally. As used herein, "simultaneous administration," "co-administration," "simultaneous administration," or "substantially simultaneous administration" refers to administration at the same time point or substantially immediately following each other. In the latter case, the two compounds are administered close enough in time that no difference is observed in the results observed from the results obtained when administered at the same time point.
The frequency of administration (e.g., NK cells) of the compositions disclosed herein includes: but are not limited to, at least once every 12 months, once every 11 months, once every 10 months, once every 9 months, once every 8 months, once every 7 months, once every 6 months, once every 5 months, once every 4 months, once every 3 months, once every 2 months, once every month; or at least once every three weeks, once every two weeks, once per week, twice per week, three times per week, four times per week, five times per week, six times per week, or once per day. In some embodiments, the interval between each administration is less than about 4 months, less than about 3 months, less than about 2 months, less than about 1 month, less than about 3 weeks, less than about 2 weeks, or less than about 1 week, for example less than any of about 6, 5, 4, 3, 2, or 1 day. In some embodiments, the frequency of administration of the composition (e.g., expanded NK cells) includes, but is not limited to, at least once, twice, once or three times daily. In some embodiments, the interval between each administration is less than about 48 hours, 36 hours, 24 hours, 22 hours, 20 hours, 18 hours, 16 hours, 14 hours, 12 hours, 10 hours, 9 hours, 8 hours, or 7 hours. In some embodiments, the interval between each administration is less than about 24 hours, 22 hours, 20 hours, 18 hours, 16 hours, 14 hours, 12 hours, 10 hours, 9 hours, 8 hours, 7 hours, or 6 hoursThen (c) is performed. In some embodiments, the interval between each administration is constant. For example, administration may be daily, every second day, every third day, every fourth day, every fifth day, or weekly. Administration may also be continuous and adjusted to maintain the level of the compound within any desired and specified range. Each dose may comprise at least about 1 × 10 8 NK cells/kg (e.g., at least about 1 × 104, 1 × 10 5 ,1×10 6 ,1×10 7 ,1×10 8 ,1×10 9 ,1×10 10 ,1×10 11 ,1×10 12 NK cells/kg).
Advantageous effects
The invention provides a method for preparing and amplifying NK cells and the NK cells obtained by the method, and verifies that the NK cells have better tumor killing capacity, and after the NK cells are combined with beta-catenin monoclonal antibodies, the growth of tumors in mice can be remarkably inhibited, so that the NK cells have an excellent tumor treatment effect.
Drawings
FIG. 1 is a diagram showing the identification result of the specific western blot of the beta-catenin monoclonal antibody
FIG. 2 is a graph showing the results of the inhibition rate of the beta-catenin monoclonal antibody on cancer cells
FIG. 3 is a graph showing the effect of monoclonal antibody on the activity of Wnt/beta-catenin pathway
FIG. 4 result graph of NK cell and/or monoclonal antibody effect on mouse tumor weight
Detailed Description
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
EXAMPLE 1 preparation of beta-catenin immunogen
Designing a primer according to a human beta-catenin gene sequence, wherein an upstream primer PF is 5'-AGGATCCAACTTGATTAACTATCAA-3'; downstream primer PR5'-ACTCGAGCAGGTCAGTATCAAACCA-3'. Amplifying beta-catenin fragment by using human blood DNA as a template. The PCR reaction conditions were 94 ℃ denaturation for 5min, 94 ℃ denaturation for 30s, 55 ℃ annealing for 30s, and 72 ℃ extension for 60s, and after 30 cycles, total extension at 72 ℃ for 10 min. The PCR amplification result was checked by 1.0% agarose gel electrophoresis to obtain a specific fragment of about 1932 bp. And connecting the fragment with a T vector, sequencing and identifying the fragment correctly, carrying out double enzyme digestion on the fragment and a pET-30a (+) vector, and connecting the fragment and the pET-30a (+) vector overnight at 4 ℃ by using T4 DNA ligase to construct a recombinant plasmid pET-30a (+) -beta-catenin. The recombinant plasmid is transformed into E.coli DH5 alpha competent cell, after kanamycin resistance screening, the identified plasmid is extracted, the recombinant plasmid is transformed into E.coli Rosetta (DE3) competent cell, LB solid medium is coated, and the culture is carried out overnight at 37 ℃. The single colony which is positive in identification is picked up and inoculated into 5ml LB liquid culture medium (containing 50ug/ml kanamycin), beta-catenin expression is induced (1mmol/L IPTG,25 ℃ induction for 10h), the supernatant is taken out, after 25 percent of saturated ammonium sulfate solution is used for precipitation, a proper amount of balanced solution (25mmol/L Tris, 0.5mol/L NaCl, 50mmol/L imidazole pH7.8) is used for dilution to prepare a crude extract. HisTrap TM The column was washed and equilibrated with 10 bed volumes of equilibration solution and loaded at a flow rate of 0.5 ml/min. Finally, gradient elution is carried out by eluent (25mmol/L Tris, 0.5mol/L NaCl, 0.5mol/L imidazole pH7.8) and the target protein is collected. After SDS-PAGE analysis of the purified β -catenin protein, the eluate containing the target protein was dialyzed against TBS (25mmol/L Tris, 0.15mol/L NaClpH8.0) and then quantified by BCA method at a concentration of 1.4 mg/ml.
EXAMPLE 2 preparation of beta-catenin monoclonal antibodies
Using the beta-catenin protein prepared in example 1 as immunogen, BALB/c female mice of 6 weeks old were selected for subcutaneous multi-point immunization, and the immunization dose was 50. mu.g/mouse. And finally screening 2 cell strains with the strongest positive by adopting a conventional cell fusion and screening method, wherein the cell strains are beta-catenin-1F 2 and beta-catenin-2D 13. The two hybridoma cells are respectively prepared into ascites type antibody by a mouse in vivo induction method, purified by a Protein G affinity chromatography column, and adjusted to the concentration of 1mg/ml for standby. Two monoclonal antibodies beta-catenin-1F 2 and beta-catenin-2D 13 and beta-catenin protein are respectively subjected to Westernblot detection. Specifically, PC9 human lung cancer cells (autumn living things-passing, cargo number: H086) are cracked, total proteins are extracted, the proteins are transferred to a PVDF membrane after SDS-PAGE electrophoresis, 2 diluted purified antibodies (1: 4000) are respectively added after 5% defatted milk powder seals the membrane, the membrane PBST is taken out after overnight reaction at 4 ℃ and washed for 3 times, HRP enzyme-labeled secondary antibody (1: 8000) is added for reaction at room temperature for 1.5H, washing is carried out for 6 times, chemiluminescent liquid is added for reaction, and imaging is carried out by a gel imager. The results are shown in FIG. 1.
As can be seen from the results in FIG. 1, 2 monoclonal antibodies prepared by the present invention all have good binding to the beta-catenin protein in tumor cells.
The SBACLonyping System-HRP kit is adopted to determine the class and subclass of the selected antibody Ig, and all the operations are strictly operated according to the kit instructions. The result shows that the beta-catenin-1F 2 and the beta-catenin-2D 13 are IgG 1.
Example 3 characterization and Performance analysis of the beta-catenin monoclonal antibody beta-catenin-2D 13
The non-competitive enzyme immunoassay determines the Affinity constant (Affinity constant), Ka value. According to the Method of a computer program, according to the formula Ka ═ 1/2 (n [ Ab']t-[Ab]t) calculating the value of the affinity constant Ka. The results show that the Ka value of beta-catenin-2D 13 is 8.71 multiplied by 10 9 L/mol, has better affinity property.
Amplification of V by PCR L And V H The gene PCR amplification kit is purchased from Takara company, the amplified product is sequenced and identified, the light chain variable region sequence of the beta-catenin-2D 13 monoclonal antibody is as follows:
the light chain variable region sequence is:
DIVITQRPALMAASPGEKVTITCAEYKHHAELGHVWYQQKSGISPKPWIYRSFIYKGGVPARFSGSGSGTSYSLTITSMEAEDAATYYCNTWDWFTLPFGAGTKLELK
the heavy chain variable region sequence is:
EVQLEESATELARPGASVKLSCKASGYIFSFPGFNWIKQRPGQGLEWIGPGLQRLFHHCCFDMRWGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGPDDVCQHWGLGTTLAVSS。
example 4 Activity analysis of the beta-catenin monoclonal antibody beta-catenin-2D 13
Placing PC9 human lung cancer cell in RPMI1640 culture solution containing 10% fetal calf serum, and adding 5% CO 2 And (5) incubating for 48h in an incubator, detecting the cell state before experiment, and selecting cells in logarithmic growth phase with good state for EDTA digestion for later use.
Take 5X 10 4 PC9 human lung cancer cells per mL are inoculated in a 96-well plate, and the culture medium is sucked out after adherent culture is carried out for 12 h. Adding 100.0 mu g/mL cetuximab into a DMEM medium (a positive control group), adding 1, 10, 50 and 100.0 mu g/mL beta-catenin-2D 13 monoclonal antibodies into the DMEM medium (corresponding to an experimental group 1-4), taking a pure DMEM medium as a blank group, after 48 hours of drug treatment, dripping 8 mu l of MTT solution into each hole, incubating for 8 hours at room temperature, sucking out the solution in the holes, dripping 80 mu l of dimethyl sulfoxide into each hole, oscillating for 30s, dissolving purple crystals, measuring the light absorption value by using a full-automatic enzyme standard analyzer, and calculating the cell proliferation inhibition rate, wherein the calculation formula is as follows: the cell inhibition ratio (%) × 1- (treatment group OD value/blank group OD value) × 100%. The results are shown in FIG. 2.
As can be seen from FIG. 2, after the monoclonal antibody is treated and cultured for 48h, the inhibition rate of the monoclonal antibody group for treating PC9 human lung cancer cell is significantly lower than that of the blank group (P < 0.05); under the same concentration, the beta-catenin-2D 13 monoclonal antibody has better inhibition effect than the PC9 human lung cancer cell proliferation of a positive control cetuximab group, and the cell inhibition rate reaches (95.9 +/-2.1)%, thus having better inhibition effect.
The activity detection of the Wnt/beta-catenin pathway adopts a dual-luciferase method, wherein a group of samples are placed in a 96-hole white fluorescence detection plate, 30 mu L of LAR II working solution is dripped into each hole, 8ul of each group of cell lysate is dripped into each hole, the solution is absorbed for 10min and uniformly mixed, the solution is immediately placed in a Modulus micropore plate multifunctional photometer for detection, the original value of firefly luciferase excited fluorescence is recorded, the detection plate is taken out, 30 mu LStop & Glo working solution is dripped into each hole, the solution is uniformly mixed and placed in the Modulus micropore plate multifunctional photometer for detection, the original value of renilla luciferase excited fluorescence is recorded, and data processing is carried out according to the following formula, wherein the relative luciferase activity is the luciferase activity of each hole sample/renilla luciferase activity of the same hole sample; activity of each Wnt/β -catenin pathway set TOPFlash relative luciferase activity/FOPFlash relative luciferase activity. The results are shown in FIG. 3.
As can be seen from FIG. 3, the active TOP/FOP values of the Wnt/beta-catenin pathway of each monoclonal antibody group are significantly lower than those of the blank group (P < 0.05) after the monoclonal antibody is cultured for 48 h. Under the same concentration, the beta-catenin-2D 13 monoclonal antibody has better effect of reducing the activity of the Wnt/beta-catenin pathway of the PC9 human lung cancer cells than that of positive control cetuximab, and has better inhibition effect by TOP/FOP value bit (4.21 +/-0.3)% under the concentration of 100.0 mu g/mL.
Example 5 isolation and culture of NK cells of immune cells
Adding 15 mLFicol-PaquePLUS mononuclear cell separating medium into a 50mL centrifuge tube, anticoagulating with heparin, slowly adding 30mL heparin anticoagulated peripheral blood along the tube wall, avoiding damaging the water phase of the Ficoll separating medium, centrifuging for 15min at room temperature and 3500rpm, carefully sucking the upper plasma into another 50mL centrifuge tube, inactivating for 25min at 56 ℃, for later use, sucking the yellowish mononuclear cell layer into the 15mL centrifuge tube, washing with sterile PBS for 3 times, discarding the supernatant, precipitating to obtain peripheral blood mononuclear cells, obtaining PBMC, adding the serum-free PRMI-1640 culture medium suspension precipitated cells into a plastic plate coated with goat anti-mouse IgG, incubating the plate with 25 μ g goat anti-mouse IgG and 5mL 0.05M Tris-HCL buffer solution (PH9), washing PBS for 3 times at room temperature for 2h, sealing with 1% FCS for 15min at room temperature, and washing with PBS for 3 times at room temperature, and adding the cells incubated with the anti-CD3 for 2h into a coated plate, and slightly sucking the cell suspension for 2h at 4 ℃ to obtain the required NK cells.
The mononuclear cells were resuspended in 90mL NK MACS GMP medium (Miltenyi), 5 ten thousand units of recombinant human IL-2 and IL-15 were added, mixed well and injected into a small culture bag, and placed in a 37 ℃ incubator for static culture. On day 3, 50mL of NK MACS GMP medium and 2.5 ten thousand units of recombinant human IL-2 and IL-15 were supplemented, and the mixture was mixed and cultured. On day 4, the culture medium in the small bag was equally distributed to two large bags and the culture was continued, and each large bag was supplemented with 100mL of NK MACS GMP culture medium and 15 ten thousand units of recombinant human IL-2 every 3 days for a total of 14 days, i.e., the proliferated NK cells were collected. Taking the amplified NK cell smear, drying, and adopting an Envision two-step method to carry out anti-CD56 labeling and anti-CD3 labeling, wherein the working concentration of the antibody is 1: 20. The results showed that CD56+ was 89.7% and CD3+ was 3.72%, indicating that the purity of the prepared NK cells was high.
Example 6 characterisation of immune cell NK cells
Placing the PC9 human lung cancer cell suspension at 37 ℃ and 5% CO 2 Culturing in incubator, collecting PC9 human lung cancer cell in logarithmic growth phase, adjusting cell number to 2.0 × 10 5 mL -1 Add 96 well U plate, 100. mu.L/well. Taking NK cells prepared in example 5, grouping the NK cells according to different effective target ratios, namely, an NK group, an NK + PC9 human lung cancer cell 10:1 and a 50: 1) group, a PC9 human lung cancer cell group and a blank group, culturing for 12 hours in a 96-hole U-shaped plate with the temperature of 37 ℃ and 5% CO2, adding 20ul MTT 5mg/ml into each hole, incubating for 4 hours at the temperature of 37 ℃, adding DMSO 100ul into each hole, fully blowing, uniformly mixing, detecting the OD value of the 570nm wavelength by using a microplate reader, and calculating according to the following formula: the killing rate (%) (1- (experimental well a value-effector cell well a value)/target cell well a value × 100%). The results are shown in Table 1.
TABLE 1 killing Activity of groups on cancer cells
Group of Killing rate (%)
10:1 group 83.46±2.02
50:1 group 90.31±3.47
As can be seen from the results in Table 1, NK has a killing effect on PC9 human lung cancer cells, and the killing rate was (90.31. + -. 3.47)%, at an effective target ratio of 50: 1.
Example 7 combination therapy experiment of NK cells and beta-catenin-2D 13 monoclonal antibody
Grouping nude mice: placing BALB/c nude mice in a sterile environment for feeding, and randomly grouping the mice into groups, wherein each group comprises 10 mice; a is a blank control group of a sterile nude mouse, B is a human lung cancer cell group transplanted with PC9 in vivo, C is a group injected with beta-catenin-2D 13 monoclonal antibody in vivo, D is a group injected with NK cells in vivo, E is a group injected with beta-catenin-2D 13 monoclonal antibody and NK cells in vivo, and F is a group injected with cetuximab in vivo.
Establishing a mouse model: digesting cultured PC9 human lung cancer cells with trypsin, centrifuging, preparing a glioblastoma cell suspension by using a serum-free DMEM/F12 culture medium, and injecting the cell suspension into the back subcutaneous of a BALB/c nude mouse by using a 0.2mL syringe through subcutaneous injection, wherein the injection amount is 2 multiplied by 10 6 (0.2 ml). In each experimental group, NK cell treatment was: the NK cell suspension prepared in example 5 was taken and injected into the body through the tail vein of mouse using 0.2mL syringe (cell density 2X 10) 7 mL), once more at intervals of 7 d. The monoclonal antibody treatment groups were: the beta-catenin-2D 13 monoclonal antibody or cetuximab is respectively injected into the mouse body by a 0.2mL syringe in the same way, the injection amount is 150.0 mu g/mouse, and the injection is administered 1 time at 3 days and 3 times in total. The above operations are all completed in the same time period and the injection of cancer cells is completed first. The NK cells and the monoclonal antibody are injected at an interval of 30min during the combination treatment. Nude mice were sacrificed at 14d after treatment, tumor tissues of the nude mice were dissected, and their weights were measured. The results are shown in FIG. 4.
As can be seen from FIG. 4, the tumor weights in the C-F treatment groups were significantly reduced (P < 0.05) relative to the PC9 human lung cancer cell group. After the beta-catenin-2D 13 monoclonal antibody and the NK cells are treated in a combined mode, the tumor weight is only (0.24 +/-0.05) g, while the tumor weight of the PC9 human lung cancer cell group is (2.70 +/-0.12) g, and the combined treatment group has a remarkable inhibition effect compared with a control group.
It is to be understood that the invention is not necessarily limited in its application to the details of construction and the arrangement of components set forth in the following description and/or illustrated in the drawings. The invention is capable of embodiments in addition to those described and of being practiced or carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein, as well as the abstract, are for the purpose of description and should not be regarded as limiting.
Sequence listing
<110> Beijing Shuyuansheng biological products Co., Ltd
<120> preparation method of clinical blood immune cells and application thereof
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 108
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Asp Ile Val Ile Thr Gln Arg Pro Ala Leu Met Ala Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ala Glu Tyr Lys His His Ala Glu Leu
20 25 30
Gly His Val Trp Tyr Gln Gln Lys Ser Gly Ile Ser Pro Lys Pro Trp
35 40 45
Ile Tyr Arg Ser Phe Ile Tyr Lys Gly Gly Val Pro Ala Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Thr Ser Met Glu
65 70 75 80
Ala Glu Asp Ala Ala Thr Tyr Tyr Cys Asn Thr Trp Asp Trp Phe Thr
85 90 95
Leu Pro Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210> 2
<211> 116
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Glu Val Gln Leu Glu Glu Ser Ala Thr Glu Leu Ala Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Ile Phe Ser Phe Pro
20 25 30
Gly Phe Asn Trp Ile Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Pro Gly Leu Gln Arg Leu Phe His His Cys Cys Phe Asp Met Arg
50 55 60
Trp Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Leu Ala Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Gly Pro Asp Asp Val Cys Gln His Trp Gly Leu Gly Thr Thr Leu
100 105 110
Ala Val Ser Ser
115

Claims (7)

1. The beta-catenin monoclonal antibody is characterized in that the antibody is beta-catenin-2D 13, and the variable region sequence of the light chain of the monoclonal antibody is as follows:
DIVITQRPALMAASPGEKVTITCAEYKHHAELGHVWYQQKSGISPKPWIYRSFIYKGGVPARFSGSGSGTSYSLTITSMEAEDAATYYCNTWDWFTLPFGAGTKLELK
the heavy chain variable region sequence is:
EVQLEESATELARPGASVKLSCKASGYIFSFPGFNWIKQRPGQGLEWIGPGLQRLFHHCCFDMRWGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGPDDVCQHWGLGTTLAVSS。
2. use of the beta-catenin monoclonal antibody of claim 1 in the preparation of a pharmaceutical composition for treating lung cancer.
3. The use according to claim 2, wherein said composition further comprises a pharmaceutically acceptable carrier.
4. Use according to claim 3, characterized in that the carrier also contains a preservative.
Use of NK cells in combination with beta-catenin monoclonal antibodies for the preparation of a kit for the treatment of cancer lung cancer; wherein, the beta-catenin monoclonal antibody is beta-catenin-2D 13, and the light chain variable region sequence of the monoclonal antibody is as follows:
DIVITQRPALMAASPGEKVTITCAEYKHHAELGHVWYQQKSGISPKPWIYRSFIYKGGVPARFSGSGSGTSYSLTITSMEAEDAATYYCNTWDWFTLPFGAGTKLELK
the heavy chain variable region sequence is:
EVQLEESATELARPGASVKLSCKASGYIFSFPGFNWIKQRPGQGLEWIGPGLQRLFHHCCFDMRWGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGPDDVCQHWGLGTTLAVSS;
the preparation method of the NK cells comprises the following steps: separating peripheral blood mononuclear cells, adding suspension precipitated cells in a serum-free PRMI-1640 culture medium into a plastic plate coated with goat anti-mouse IgG for 15 minutes at room temperature, sealing the plastic plate with PBS containing 1% FCS (FCS), washing the plastic plate with PBS for 3 times, adding cells incubated with anti-CD3 for 2 hours into the coated plate, slightly sucking cell suspension at 4 ℃ for 2 hours, suspending the mononuclear cells by using NK MACS GMP culture medium, adding recombinant human IL-2 and IL-15, uniformly mixing, injecting into a small culture bag, and placing into an incubator at 37 ℃ for standing culture; on the 3 rd day, half amount of NK MACS GMP culture medium and half amount of recombinant human IL-2 and IL-15 are supplemented, and the culture is continued after the mixture is evenly mixed; on day 4, the culture medium in the small culture bag was equally distributed to two large culture bags and the culture was continued, and NK MACS GMP culture medium and recombinant human IL-2 were supplied to each large culture bag every 3 days for a total of 14 days, i.e., the proliferated NK cells were collected.
6. Use according to claim 5, characterized in that the β -catenin monoclonal antibody is itself in the form of a pharmaceutical composition containing a pharmaceutically acceptable carrier.
7. Use according to claim 6, characterized in that the carrier also contains a preservative.
CN202210729772.3A 2022-06-24 2022-06-24 Preparation method and application of clinical blood immune cells Active CN115124619B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210729772.3A CN115124619B (en) 2022-06-24 2022-06-24 Preparation method and application of clinical blood immune cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210729772.3A CN115124619B (en) 2022-06-24 2022-06-24 Preparation method and application of clinical blood immune cells

Publications (2)

Publication Number Publication Date
CN115124619A true CN115124619A (en) 2022-09-30
CN115124619B CN115124619B (en) 2023-05-05

Family

ID=83379203

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210729772.3A Active CN115124619B (en) 2022-06-24 2022-06-24 Preparation method and application of clinical blood immune cells

Country Status (1)

Country Link
CN (1) CN115124619B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1323732A1 (en) * 2000-09-22 2003-07-02 Kyowa Hakko Kogyo Co., Ltd. Beta-catenin nuclear localizing protein
CN101532008A (en) * 2009-04-29 2009-09-16 朱晓林 Preparation of anti beta-catenin monoclonal antibody and application thereof in pathologic examination of colon cancer
CN103173413A (en) * 2012-12-07 2013-06-26 天津三箭生物技术有限公司 Mouse anti-human beta-catenin monoclonal antibody and hybridoma cell strain for secreting monoclonal antibody
CN112457400A (en) * 2020-12-09 2021-03-09 福州迈新生物技术开发有限公司 Anti-beta-catenin protein monoclonal antibody, cell line, preparation method and application thereof
CN113073079A (en) * 2021-04-08 2021-07-06 北京广未生物科技有限公司 Use of NK immune cells with enhanced activity for the preparation of a medicament for the treatment of cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1323732A1 (en) * 2000-09-22 2003-07-02 Kyowa Hakko Kogyo Co., Ltd. Beta-catenin nuclear localizing protein
CN101532008A (en) * 2009-04-29 2009-09-16 朱晓林 Preparation of anti beta-catenin monoclonal antibody and application thereof in pathologic examination of colon cancer
CN103173413A (en) * 2012-12-07 2013-06-26 天津三箭生物技术有限公司 Mouse anti-human beta-catenin monoclonal antibody and hybridoma cell strain for secreting monoclonal antibody
CN112457400A (en) * 2020-12-09 2021-03-09 福州迈新生物技术开发有限公司 Anti-beta-catenin protein monoclonal antibody, cell line, preparation method and application thereof
CN113073079A (en) * 2021-04-08 2021-07-06 北京广未生物科技有限公司 Use of NK immune cells with enhanced activity for the preparation of a medicament for the treatment of cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
JESSICA C. KLING ET AL.: "Temporal regulation of natural Killer T cell interferon gamma responses by β-catenin-Dependent and -independent Wnt signaling" *
XIN LI ET AL.: "WNT/b-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment" *
牛夏忆等: "大鼠抗人β-catenin 多克隆抗体的制备与鉴定" *
陈晶,单保恩,贺占国,董稚明,马洪: "抗β-连环蛋白抗体的制备及其在乳腺癌诊断中的应用" *
陈晶等: "抗β-catenin抗体制备及在结肠癌病理检测中的应用" *

Also Published As

Publication number Publication date
CN115124619B (en) 2023-05-05

Similar Documents

Publication Publication Date Title
JP7241207B2 (en) TIGIT and PD-1/TIGIT binding molecules
WO2020227159A2 (en) Methods of modulating immune activity
JP7090204B2 (en) Bispecific antibodies targeting immune checkpoints
JP2022058740A (en) Combined cancer therapy of immune checkpoint modulator and fermented product by symbiotic microbial flora
CN110981958A (en) PD-L1 antibody
TW202115113A (en) Anti-bcma antibody and application in the field of car-t thereof
Chen et al. Programmed cell death protein-1/programmed death-ligand 1 blockade enhances the antitumor efficacy of adoptive cell therapy against non-small cell lung cancer
CN113876946B (en) Combined pharmaceutical application of PD-1 antibody and pseudomonas aeruginosa and pharmaceutical composition
TW202017943A (en) Cd226 agonist antibodies
WO2024056098A1 (en) Nkg2d-nkp46 cell adapter molecule and use thereof
BR112020014446A2 (en) METHODS FOR ADMINISTERING CHEMICAL ANTIGEN RECEPTOR IMMUNOTHERAPY IN COMBINATION WITH A 4-1BB AGONIST
CN111956795A (en) Application of chimeric antigen receptor combined anti-tumor drug taking CD99 as target
CN116120465B (en) Chimeric antigen receptor targeting BCMA and/or FCRH5 and application thereof
EP3885357A1 (en) Interleukin 21 protein (il21) mutant and use thereof
CN116143943B (en) Targeting BAFFR chimeric antigen receptor, CAR-T cell and application
CN115124618B (en) Application of immune cells separated from autoblood in treating diseases
CN115124619B (en) Preparation method and application of clinical blood immune cells
JP6961090B2 (en) Anti-PD-1 / CD47 bispecific antibody and its application
CN113980133A (en) Antibody and application thereof in anti-tumor
CN114456267B (en) anti-CD 73 humanized monoclonal antibody and application thereof
CN117143254B (en) Chimeric Antigen Receptor (CAR) and application thereof in anticancer
CN116063569B (en) EPHA2 chimeric antigen receptor and uses thereof
RU2811918C2 (en) Methods of treating cancer diseases and tumors using pde1 inhibitors
CN112755051B (en) Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers
AU2019275453B2 (en) Organic compounds

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
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20230419

Address after: Room 2304, Building 10, No. 739 Qinzheng Road, Hengqin New District, Zhuhai City, Guangdong Province, 519000

Applicant after: Guangdong Qimei Pharmaceutical Biotechnology Group Co.,Ltd.

Address before: Room 2152, Building 2, East Ring Road, Yanqing Garden, Zhongguancun, Yanqing District, Beijing 100000

Applicant before: Beijing Shunyuantian Biological Products Co.,Ltd.

GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right
TR01 Transfer of patent right

Effective date of registration: 20230717

Address after: Room 2108, Building 3, No. 78, Dongcheng Section, Guanlong Road, Dongcheng Street, Dongguan City, Guangdong Province, 523000

Patentee after: Guangdong Qimei Life Medicine Technology Research Institute

Patentee after: Guangdong Qimei Pharmaceutical Biotechnology Group Co.,Ltd.

Patentee after: Zhuhai Qimei Stem Cell Bank Co.,Ltd.

Patentee after: Guangdong Stanfu International Stem Cell Medical Research Institute

Address before: Room 2304, Building 10, No. 739 Qinzheng Road, Hengqin New District, Zhuhai City, Guangdong Province, 519000

Patentee before: Guangdong Qimei Pharmaceutical Biotechnology Group Co.,Ltd.