CN112755051B - Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers - Google Patents

Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers Download PDF

Info

Publication number
CN112755051B
CN112755051B CN202110092687.6A CN202110092687A CN112755051B CN 112755051 B CN112755051 B CN 112755051B CN 202110092687 A CN202110092687 A CN 202110092687A CN 112755051 B CN112755051 B CN 112755051B
Authority
CN
China
Prior art keywords
monoclonal antibody
cells
chain variable
variable region
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110092687.6A
Other languages
Chinese (zh)
Other versions
CN112755051A (en
Inventor
陈小波
曹帆
张国军
亓爱杰
李少波
亓爱磊
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 Renda Biotechnology Co ltd
Original Assignee
Guangdong Renda Biotechnology 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 Guangdong Renda Biotechnology Co ltd filed Critical Guangdong Renda Biotechnology Co ltd
Priority to CN202110092687.6A priority Critical patent/CN112755051B/en
Publication of CN112755051A publication Critical patent/CN112755051A/en
Application granted granted Critical
Publication of CN112755051B publication Critical patent/CN112755051B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Abstract

The invention relates to preparation of NK cells and application of the NK cells in treating cancer. The invention provides a pharmaceutical composition prepared from NK cells and a monoclonal antibody of CD105, which can be used for treating colorectal cancer and has a good application prospect.

Description

Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers
Technical Field
The invention relates to the field of pharmacy, in particular to preparation of NK cells and application of the NK cells in treating cancer.
Background
NK cells are lymphoid cells with multiple immunological functions, and human NK cells are positive for CD56 and CD16, and negative for CD3 and CD 19. A small fraction of NK cells may be CD8 positive. NK cells kill tumor cells without MHC restriction, and do not need to be exposed to antigen or show any memory response. The killing of tumors by NK cells has the advantages of directly dissolving and secreting cytokines, and the tumors can be killed by perforin and Fas ligand. NK cells can produce TNF-a, IFN-gamma and IL-1, and these cytokines play a very important role in NK cell anti-cancer response. NK cells have the ability to rapidly reject allogeneic bone marrow, but do not mediate transplant rejection of solid tissues.
During the process of tumor development and development, NK cells can directly recognize malignant transformation cancer cells and be activated by means of "internal recognition" (such as NCRs, NKG2D, SLAMs, DNAMs and the like), and can also be activated by the action of helper cells (monocytes, macrophages, dendritic cells and the like). These helper cells respond to the change of internal and external environment through pattern recognition receptors (including cell surface receptors TLR2, TLR4, intracellular cytoplasmic receptors RIG-1, NALP3, NOD2, endosomal receptors TLR7, TLR9 and the like recognize various pathogens), and then transmit signals to NK cells through secreting various soluble factors or directly contacting, so that the NK cells can exert the killing function and the inflammatory cytokine secretion function. In humans, the presence of soluble factors has been demonstrated to be IL-12, IL-18, IFN, IL-2 and TNF; the molecules directly contacted with the antibody include GITRL/GITR, IL-12/IL-12R, CD48/2B4, MICAormICBorULBP1-ULBP3/NKG2D, AICL/NKp80, IL-15R-IL-15/IL-15. beta. gamma. and the like.
Using NK cells to treat tumors can use not only stimulating factors (such as IL-2, IL-12, IL-15, etc.), but also blocking agents against NK cells' KIR. It is also suggested that blocking the KIR function of NK cells can improve the anti-cancer immune response of the body as blocking the negative signal of CTLA4 to T cells. The data suggest that activated NK cells can either be directly used for adoptive infusion to treat tumors or block KIR of these NK cells to enhance their anti-cancer effect.
Recently, several approaches have improved the in vitro expansion of NK cells, persisting in vivo, promoting homing to the tumor microenvironment, increasing their anti-tumor effects. The culture solution used in the early NK cell expansion method contained only cytokines such as IL-2 and IL-15, and was expanded only 10-20 times. Subsequently, feeder cells in combination with cytokines were used to proliferate 80-10000 fold, especially in culture with CD3 depleted PBMC. NK cells with a purity of > 95% were recently obtained using CD3 depleted apheresis, using feeder cells free and MEM-alpha medium containing IL-2, IL-15 and vitamin B3. IL-15 promotes NK cell growth and proliferation without activating Treg cells. Heterodimeric IL-15(IL-15-sIL-15R alpha) is more effective in stimulating NK cell proliferation. NK cells express IL-12R beta 2, and high-dose IL-12 has an immunoregulation effect on the NK cells and promotes IFN-gamma production. The immunomodulatory drug lenalidomide indirectly improves proliferation and cytotoxic effects of NK cells through IL-2 and IFN-gamma. When PD1 and PDL 1-specific monoclonal antibodies were combined with lenalidomide, the NK cell anti-myeloma capacity was enhanced. The PD1 blocking drug can improve ADCC induced by NK cells and migration of the NK cells to tumors, and inhibit the function of Treg cells. The death ligand TRAIL of NK cell can induce TRAILR to express positive tumor cell to die, and histone acetylase and proteasome inhibitor can strengthen the function of the channel. In phase I clinical trials, bortezomib in combination with autologous NK cells resulted in tumor regression in refractory renal cell carcinoma and chronic leukemia.
Colorectal cancer is one of the most common malignant tumors of the digestive system, and the incidence rate is counted to be 3 rd of malignant tumors worldwide and has risen to 2 nd in western countries and economically developed regions of china. Colorectal cancer is also one of the leading causes of death from cancer worldwide. Although bevacizumab has been widely used in the field of colorectal cancer, the toxic and side effects of bevacizumab, including bleeding and thrombotic events, have attracted attention. Therefore, the development of a new monoclonal antibody capable of treating colorectal cancer (colorectal cancer) still has great application value. In particular, the use of NK cells together with monoclonal antibodies is the central direction of current combination therapy.
Disclosure of Invention
The present invention overcomes the disadvantages of the prior art and provides a novel method for effectively treating colorectal cancer, particularly colorectal cancer.
In one aspect of the invention, there is provided an isolated NK cell for use in the treatment of colorectal cancer, in particular colorectal cancer.
In another aspect, the invention provides a monoclonal antibody specific for CD 105.
Specifically, the light chain variable region sequence of the monoclonal antibody of CD105 provided by the invention is SEQ ID NO. 1, and the heavy chain variable region sequence thereof is SEQ ID NO. 2.
In another aspect of the present invention, a pharmaceutical composition is provided, wherein the pharmaceutical composition comprises the monoclonal antibody against CD105 provided by the present invention, and a pharmaceutically acceptable carrier.
In another aspect of the present invention, there is also provided a pharmaceutical composition comprising the NK cell provided by the present invention, and a pharmaceutically acceptable carrier.
In another aspect of the present invention, a pharmaceutical composition is provided, which comprises the NK cells provided by the present invention and the monoclonal antibody against CD105 provided by the present invention, and a pharmaceutically acceptable carrier.
In a further aspect of the invention, there is provided a use of NK cells in the manufacture of a medicament for the treatment of colorectal cancer.
In a further aspect of the invention, there is provided a use of a monoclonal antibody to CD105 in the manufacture of a medicament for the treatment of colorectal cancer.
In a further aspect of the invention, there is provided a use of a monoclonal antibody to NK cells and CD105 in the manufacture of a medicament for the treatment of colorectal cancer.
Further, the antibody provided by the invention is a full-length antibody, and the full-length protein of the antibody is conventional in the field and comprises a heavy chain variable region, a light chain variable region, a heavy chain constant region and a light chain constant region. The heavy chain variable region and the light chain variable region of the protein, the human heavy chain constant region and the human light chain constant region form a full-length protein of a fully human antibody. Preferably, the antibody full-length protein is IgG1, IgG2, IgG3 or IgG 4.
The invention also provides a nucleic acid encoding the antibody described above.
The reagents and starting materials used in the present invention are commercially available.
The positive progress effects of the invention are as follows: the invention provides a pharmaceutical composition prepared by NK cells and a monoclonal antibody of CD105, which can be used for treating colorectal cancer. Has better application prospect.
Drawings
FIG. 1 is a graph showing the result of subtype identification
FIG. 2 is a graph showing the inhibitory effect of monoclonal antibody on LOVO cells
FIG. 3 is a graph showing the effect of drugs on the inhibition of transplantable tumors
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
Example 1 preparation of NK cells
Using Ficoll-Hypaque densityCarrying out gradient centrifugation to separate cord blood mononuclear cells, washing with PBS for 3 times, and suspending with EX vivo-15 serum-free culture solution; the mononuclear cell concentration was adjusted to 3X106To each ml of culture flasks plated with CD3 antibody, 5ml of cell suspension was added, together with cytokines IL-2(1000u/ml), IL-15(50ng/ml), IL-21(30 ng/ml). Placing in 5% CO2And continuously culturing at 37 ℃ for 18 days. During the period, the medium was supplemented with half the amount of the medium and half the amount of the NK colony, and the cytokine was supplemented in total. The first addition of cytokines was performed by adding IL-21000u/ml, IL-1550ng/ml and IL-2130ng/ml, and from the second addition, cytokines IL-21000u/ml and IL-1550ng/ml were added. 18d post-flow cytometry detection of cell phenotype before and after expansion, CD56+ CD 3-cells reached score (81.53 + -2.53)%.
EXAMPLE 2 preparation of anti-CD 105 monoclonal antibody
1. Preparation of antigens
According to the amino acid sequence of human CD105, epitope peptide with high immunogenicity, also called antigen peptide FVLRSAYSSCGMQVSASMISNEAVVNILSSSSPQRK, is obtained by screening, and the polypeptide is sent to Shanghai for industrial synthesis for standby.
2. Preparation of hybridoma cells
BALB/c mice were immunized. Immunization 1. mu.L of a CD105 antigen peptide (0.5 mg/mL) was emulsified with CFA (100. mu.L) and 8-week-old mice were immunized with the emulsion. 2 nd immunization 3 weeks after completion of the 1 st immunization, 100. mu.L of IFA was further added to 100. mu.L of 0.5mg/mL of the CD105 antigen peptide to emulsify the peptide, and mice were immunized 2 nd time with the emulsion. 3 rd immunization 3 weeks after completion of the 2 nd immunization, the 3 rd immunization was performed. The method is the same as the 2 nd immunization. Tail vein bleeds were performed on day 10 after completion of the 3 rd immunization and antibody titers were determined. Coating a 96-hole enzyme label plate with 1 mu g/mL of CD105 antigen peptide antigen solution; diluting the mouse serum with the ratio of 1: 100 in a 96-hole enzyme label plate in a multiple ratio, measuring the antibody titer in the mouse serum, and screening 3 mice with the serum antibody titer more than 1: 10000 for cell fusion. Spleens from 3 immunized mice were removed and 4mL of cell culture medium (2% RPMI1640, 0.2% NaHCO) was added to sterile tissue culture dishes 31% penicilin-streptomycin, 10% inactivated fetal bovine serum), spleen was placed into the above groupCrushing in a tissue culture dish; mixing NS-1 myeloma cells and mouse spleen cells in a 50mL centrifuge tube according to the ratio of 1: 10, centrifuging at 1500r/min for 5min, removing supernatant, and slowly adding 1mL 50% PEG 3000; washing off PEG3000 with RPMI1640 medium; screening hybridoma cells by adopting a semi-glial culture medium cloning method: hybridoma cells were cultured in 96-well cell culture plates (37 ℃, 5% CO)2) (ii) a After 3d of culture, the cell cloning wells with positive growth are screened by an indirect ELISA method, and the positive detection rate of the antibody of the hybridoma cells is 9.53 percent. The 4 monoclonal hybridoma cells with the most significant positive are cloned and subcloned by a limiting dilution method.
Finally, 2 hybridoma cell strains secreting the anti-CD 105 monoclonal antibody are obtained and named as 1A4 and 4C6 respectively.
Immunizing mice with the two hybridoma cells respectively, collecting ascites of the mice, conventionally purifying the monoclonal antibody in the ascites by using a protein A column, and then concentrating the protein for later use. The concentration of 1A4 antibody was 2.1mg/mL and 4C6 was 2.3mg/mL as determined by Lowry method.
Example 34 subtype identification and potency evaluation of the C6 monoclonal antibody
And (3) identifying by adopting a mouse monoclonal antibody typing kit, and strictly performing the operation steps according to the instruction. The results are shown in FIG. 1.
The 4C6 monoclonal antibody was detected to be an IgG1 class immunoglobulin (FIG. 1).
Measuring the titer of the monoclonal antibody: the concentrated antibody was mixed as follows: 1000. 1: 2000. 1: 4000. 1: 8000. 1: 16000. 1: 32000. 1: 64000. 1: 128000, 1: 256000, 1: 512000, adding the diluted solution into an ELISA plate coated with antigen peptide fragments, measuring the A450 nm value by an indirect ELISA method, drawing a line graph of the dilution to the A450 nm value, and taking the dilution when the A450 nm value is 0.1 as the titer. The 4C6 monoclonal antibody was tested to have a potency of about 1: 256000.
example 44 affinity identification and sequence identification of C6 monoclonal antibody
The binding ability of the 4C6 monoclonal antibody to the antigenic peptide was identified by the SPR method. The specific operation is that the obtained 4C6 monoclonal antibody is measured by a surface plasmon resonance (SRP) method through a BIAcoreX100 instrument aiming at the binding kinetics of the antigen peptide, and the antigen peptide is directly coated on a CMS biosensor chip. For kinetic measurements, the 4C6 monoclonal antibody was serially diluted three times with HBS-EP +1X buffer, injected at 25 ℃ for 120s with a dissociation time of 30min, and regenerated for 120s by addition of 1OmM glycine-HCl (pH 2.0). The equilibrium dissociation constant of the 4C6 monoclonal antibody and the antigenic peptide was calculated using a simple one-to-one Languir binding model. The calculation results are shown in table 2.
TABLE 2 dissociation constants of monoclonal antibodies
Monoclonal antibodies Equilibrium dissociation constant
4C6 monoclonal antibody 4.27E-11
As can be seen from Table 2, the equilibrium dissociation constant of the 4C6 monoclonal antibody is 4.27E-11, which indicates that the 4C6 monoclonal antibody of the present invention has a better effect of binding to antigenic peptides.
The sequences of the light chain variable region and the heavy chain variable region of the monoclonal antibody are identified by PCR as follows:
light chain variable region (SEQ ID NO: 1)
DIVITQSPALAAASPGEKVTITCCVSDDISASYLCWYQQKSGISPKPWIYSTAVLATGVPARFSGSGSGTSYSLTITSMEAEDAATYYCSQWYYSPLAFGAGTKLELK
Heavy chain variable region (SEQ ID NO: 2)
EVQLEESGTELRRPGASVKLSCKASGYIFSQYQLSWIKQRPGQGLEWIGSIYPGKLLTRSYQKFAGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGSNCASDSWGLGTTLAVSS
EXAMPLE 54C6 monoclonal antibody cytostatic experiments
Human colon carcinoma L0VO cell (Cat: CL-0144, Wuhan Punuo race Life technologies Co., Ltd.)
The MTT method detects the cytotoxic effect of the 4C6 monoclonal antibody on L0 VO. Collecting LOVO cells in logarithmic growth phase, digesting with 0.25% pancreatin to obtain single cell suspension, adding into 96-well culture plate with 100 μ l per well, and adjusting final cell density to 3 × 103One per ml. Dividing into experimental group and control group, each group has 4 multiple wells, placing at 37 deg.C and 5% CO2And culturing for 24 h. Respectively adding 200 mul of monoclonal antibody with final mass concentration of 0.1, 0.2, 0.3, 0.4 and 0.5mg/ml into the experimental group; the control group was incubated for 24h with addition of mouse IgG. 0.5mg/ml was added per well. Mu.l of MTT solution 2O, and continuously culturing for 4h in a dark place. The supernatant was discarded, 200. mu.l of DMSO was added to each well, and the absorbance at 490nm was measured by a microplate reader, and the measurement was repeated 3 times. Calculating the cell inhibition rate (%): inhibition rate (control value-experimental value)/control value × 100%.
As can be seen from FIG. 2, the 4C6 monoclonal antibody has growth inhibitory effect on LOVO cells, and the growth inhibitory rate of LOVO cells increases with the increase of the antibody concentration, which is obviously dose-dependent, and the inhibitory rate reaches 82% at the concentration of 0.5 mg/ml.
Example 6 Weathern blotting assay Panel Effect on expression of apoptotic proteins
Taking LOVO cells in logarithmic growth phase, digesting with 0.25% pancreatin to obtain single cell suspension, adding into 6-well culture plate, and making final cell density 3X105Per ml, put at 37 ℃ and 5% CO2And culturing for 24 h. Pouring out the culture solution, adding monoclonal antibody with final concentration of 0.5mg/ml in experimental group 1, adding monoclonal antibody with final concentration of 0.5mg/ml and 3X10 in experimental group 25NK cells at individual/ml, Experimental group 3 plus final concentration 3X105NK cells of each/ml and DMEM culture solution of a control group were used for 24 hours. And (3) cracking and separating LOVO cells by using RAPI lysate on ice to extract total protein, separating protein by 12% SDS-PAGE, placing the gel in a membrane transferring liquid for 90V, and transferring the protein to a PVDF membrane in 45min. Adding primary antibody, reacting for 1h, and washing with a washing solution for 3 times; adding a secondary antibody labeled by human HRP, reacting for 1h, washing and developing ECI. Caspase-3 and clear caspase-3 expression was detected by Western blotting. The results were shown in Table 3, in which the expression level of the control group was used as the relative expression level。
TABLE 3 relative expression of apoptotic proteins as a result
Group of Relative expression level of caspase-3 Relative expression level of clearedcaspase-3
Experimental group 1 0.77±0.06 1.31±0.10
Experimental group 2 0.60±0.05 1.57±0.08
Experimental group 3 0.82±0.04 1.20±0.06
As seen from the results in Table 3, the results of the experimental groups show that the three groups all can lead to the enhancement of clear caspase-3 band signal and the reduction of caspase-3 band signal, wherein the combination of the monoclonal antibody and the NK cell can induce the apoptosis of tumor cell LOVO most effectively, and the relative expression amount of the clear caspase-3 also reaches 1.57 +/-0.08.
Example 7 in vivo antitumor experiment to observe the influence of the application of NK cells and monoclonal antibody of the invention alone or in combination on the growth of LOVO cell transplanted tumor
L0VO cells in logarithmic growth phase were digested, washed 2 times with PBS, and cell density was adjusted to 1X 107Per BAL in/mlB/nu male nude mice were inoculated subcutaneously with 0.2 ml. The tumor mass is 100mm long3The nude mice were randomly divided into a normal saline control group, an NK combined monoclonal antibody group, an NK group and a monoclonal antibody treatment group, 6 mice in each group were injected with drugs intravenously, 100. mu.l of NK cells were administered at a concentration of 11X 1075mg/kg of monoclonal antibody per ml, 4d once, and 4 weeks in total. The maximum longitudinal diameter (a) and the maximum transverse diameter (b) of the tumor body of the nude mice were measured with a vernier caliper every 4d after the start of the treatment, and the growth of the tumor was observed, and the tumor volume was 0.5 × a × b2. On day 28, the nude mice were sacrificed, tumor tissues were stripped off, tumor weights were weighed, and tumor inhibition rates were calculated. The tumor inhibition ratio (%) × 100% (1-mean tumor mass in experimental group/mean tumor mass in control group).
From fig. 3, it can be seen that the inhibitory effect of NK-conjugated mab-administered group on transplanted tumors is significantly stronger than the effect of both single drugs (fig. 3). The most remarkable tumor weight increase inhibition of the NK combined monoclonal antibody group is only 144mm in volume3And the inhibitor has better inhibiting effect.
Sequence listing
<110> Beijing Daxi Biotechnology Ltd
<120> preparation of NK cell and application in treating cancer
<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 Ser Pro Ala Leu Ala Ala Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Cys Val Ser Asp Asp Ile Ser Ala Ser
20 25 30
Tyr Leu Cys Trp Tyr Gln Gln Lys Ser Gly Ile Ser Pro Lys Pro Trp
35 40 45
Ile Tyr Ser Thr Ala Val Leu Ala Thr 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 Ser Gln Trp Tyr Tyr Ser Pro
85 90 95
Leu Ala 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 Gly Thr Glu Leu Arg Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Ile Phe Ser Gln Tyr
20 25 30
Gln Leu Ser Trp Ile Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ser Ile Tyr Pro Gly Lys Leu Leu Thr Arg Ser Tyr Gln Lys Phe
50 55 60
Ala 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 Ser Asn Cys Ala Ser Asp Ser Trp Gly Leu Gly Thr Thr Leu
100 105 110
Ala Val Ser Ser
115

Claims (5)

1. A pharmaceutical composition comprising a monoclonal antibody to CD105, and a pharmaceutically acceptable carrier; the monoclonal antibody is 4C6 CD105 monoclonal antibody, and has the light chain variable region sequence of SEQ ID No. 1 and the heavy chain variable region sequence of SEQ ID No. 2.
2. A pharmaceutical composition comprising NK cells and a monoclonal antibody to 4C6 CD105, and a pharmaceutically acceptable carrier; wherein, the monoclonal antibody is a monoclonal antibody of 4C6 CD105, the light chain variable region sequence of the monoclonal antibody is SEQ ID NO. 1, and the heavy chain variable region sequence of the monoclonal antibody is SEQ ID NO. 2; the preparation method of the NK cells comprises the following steps: separating umbilical blood mononuclear cells by Ficoll-Hypaque density gradient centrifugation, washing with PBS for 3 times, and suspending with culture solution; the mononuclear cell concentration was adjusted to 3X106(ii) to a culture flask that has been plated with CD3 antibody, 5ml of cell suspension, and 1000u/ml of IL-2, 50ng/ml of IL-15, and 30ng/ml of IL-21; placing in 5% CO2Continuously culturing at 37 ℃ for 18 d; during the period, the culture solution is supplemented in half amount according to the color of the culture solution and the size of NK colony, and the cell factor is supplemented in full amount; the first cytokine addition was performed with 1000u/ml IL-2, 50ng/ml IL-15 and 30ng/ml IL-21, and from the second time onwards, 1000u/ml IL-2 and 50ng/ml IL-15 were added.
3. Use of a monoclonal antibody to CD105 in the manufacture of a medicament for the treatment of colorectal cancer; the monoclonal antibody is 4C6 CD105 monoclonal antibody, and has the light chain variable region sequence of SEQ ID No. 1 and the heavy chain variable region sequence of SEQ ID No. 2.
4. Use of a monoclonal antibody to NK cells and CD105 in the manufacture of a medicament for the treatment of colorectal cancer; the monoclonal antibody is 4C6 CD105 monoclonal antibody, and has the light chain variable region sequence of SEQ ID No. 1 and the heavy chain variable region sequence of SEQ ID No. 2.
5. The monoclonal antibody of 4C6 CD105 is characterized in that the light chain variable region sequence is SEQ ID NO. 1, and the heavy chain variable region sequence is SEQ ID NO. 2.
CN202110092687.6A 2021-01-25 2021-01-25 Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers Active CN112755051B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110092687.6A CN112755051B (en) 2021-01-25 2021-01-25 Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110092687.6A CN112755051B (en) 2021-01-25 2021-01-25 Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers

Publications (2)

Publication Number Publication Date
CN112755051A CN112755051A (en) 2021-05-07
CN112755051B true CN112755051B (en) 2021-08-31

Family

ID=75706957

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110092687.6A Active CN112755051B (en) 2021-01-25 2021-01-25 Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers

Country Status (1)

Country Link
CN (1) CN112755051B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103333247A (en) * 2013-05-30 2013-10-02 北京东方百泰生物科技有限公司 Novel monoclonal antibody of VEGFR2 and preparation and application thereof
CN109384845A (en) * 2017-08-14 2019-02-26 中国科学院上海生命科学研究院 A kind of CD40 monoclonal antibody, preparation method and its application

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103333247A (en) * 2013-05-30 2013-10-02 北京东方百泰生物科技有限公司 Novel monoclonal antibody of VEGFR2 and preparation and application thereof
CN109384845A (en) * 2017-08-14 2019-02-26 中国科学院上海生命科学研究院 A kind of CD40 monoclonal antibody, preparation method and its application

Also Published As

Publication number Publication date
CN112755051A (en) 2021-05-07

Similar Documents

Publication Publication Date Title
JP6207783B2 (en) Method for the proliferation of antigen-specific T cells
CN112426526B (en) Preparation method of NK (natural killer) cells and application of NK cells in treatment of cancers
CN109400713B (en) Use of novel chimeric antigen receptor modified T cells for the treatment of cancer
CN109385403B (en) GPC 3-targeting CAR NK cells
CN108752482B (en) Carry the Chimeric antigen receptor and its application of truncation or not truncated myeloid cell triggering property receptor signal structure
CN111944054B (en) anti-BCMA CAR and expression vector and application thereof
WO2023046110A1 (en) Engineered immune cell co-expressing ccr2b, preparation therefor and application thereof
US11925662B2 (en) Compositions and methods of enhancing anti-tumor response using hybrid neutrophils
CN113402619B (en) Targeting B7H3 co-expression IL-21 fully human chimeric antigen receptor, iNKT cell and application thereof
JP2021523090A (en) Methods and compositions for treating tumors
KR20230129979A (en) Dendritic cell activation chimeric antigen receptor and uses thereof
CN110317822B (en) TROP2 chimeric antigen receptor, T cell thereof, and preparation method and application thereof
KR20230018376A (en) Treatment and prevention of cancer using virus-specific immune cells expressing chimeric antigen receptors
WO2023083192A1 (en) Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof
CN110055269B (en) Human mesothelin chimeric antigen receptor, T cell thereof, preparation method and application thereof
CN113214408B (en) Chimeric antigen receptor macrophage and preparation method and application thereof
CN110713977B (en) Culture amplification method of CD8T cells
CN108822216B (en) Carry the Chimeric antigen receptor and its application of truncation or not truncated nature cell toxin receptor signal structure
CN111944053B (en) anti-BCMA CAR and expression vector and application thereof
CN113621077B (en) TIM-3/CD28 fusion protein and CAR-T cell modified by fusion protein
WO2018205472A1 (en) Non-antibody binding protein binding with pd-1 receptor, and application thereof
CN109232740B (en) anti-PD-L1 antibody and application thereof in anti-tumor treatment
CN113117073A (en) Use of immune cell-associated antibodies in the treatment of cancer
CN113481167A (en) Immune cell exosome and application thereof
CN112755051B (en) Preparation of NK (natural killer) cells and application of NK cells in treatment of cancers

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
CB03 Change of inventor or designer information

Inventor after: Chen Xiaobo

Inventor after: Cao Fan

Inventor after: Zhang Guojun

Inventor after: Qi AI Jie

Inventor after: Li Shaobo

Inventor after: Qi AI Lei

Inventor before: Qi Aijie

Inventor before: Li Shaobo

Inventor before: Qi Ailei

CB03 Change of inventor or designer information
TA01 Transfer of patent application right

Effective date of registration: 20210813

Address after: 510320 units 401 and 407, 4th floor, No. 5, helix 4th Road, International Biological Island, Guangzhou, Guangdong

Applicant after: Guangdong Renda Biotechnology Co.,Ltd.

Address before: 1408, 14th floor, building 1, yard 5, Ronghua Middle Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing

Applicant before: Beijing Daxi Biotechnology Co., Ltd

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant