CN116426475A - NK cell in-vitro activation and amplification method - Google Patents

NK cell in-vitro activation and amplification method Download PDF

Info

Publication number
CN116426475A
CN116426475A CN202310413829.3A CN202310413829A CN116426475A CN 116426475 A CN116426475 A CN 116426475A CN 202310413829 A CN202310413829 A CN 202310413829A CN 116426475 A CN116426475 A CN 116426475A
Authority
CN
China
Prior art keywords
cells
cell
biotin
labeled
antibody
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
CN202310413829.3A
Other languages
Chinese (zh)
Other versions
CN116426475B (en
Inventor
马士棋
林家会
程成
詹明高
陈旭
陈刚
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.)
Shanghai Geotech Biotech Co ltd
Suzhou Ecosai Biotechnology Co ltd
Original Assignee
Shanghai Geotech Biotech Co ltd
Suzhou Ecosai 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 Shanghai Geotech Biotech Co ltd, Suzhou Ecosai Biotechnology Co ltd filed Critical Shanghai Geotech Biotech Co ltd
Priority to CN202310413829.3A priority Critical patent/CN116426475B/en
Publication of CN116426475A publication Critical patent/CN116426475A/en
Application granted granted Critical
Publication of CN116426475B publication Critical patent/CN116426475B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • C12N2500/84Undefined extracts from animals from mammals
    • 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]
    • C12N2501/2301Interleukin-1 (IL-1)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to an NK cell in-vitro activation and amplification method. Specifically, mononuclear cells PBMC are separated and extracted from human peripheral blood, the extracted mononuclear cells are specifically activated by adopting nanoparticle suspension coupled with biotin-labeled 2B4 antibody and biotin-labeled IL-1 beta antibody on the day of NK cell activation culture, and the NK cells with high purity, high amplification efficiency and high killing power can be obtained after 14 days of amplification culture. The combined cell expansion factor IL-15 and IL-2 are used together, so that the purity of NK cells in PBMC and the expansion efficiency of the NK cells can be obviously improved, the amplified NK cells are in a better activation state, the purity of the NK cells can reach more than 85% when the NK cells are cultured on the 8 th day, the NK cells have stronger killing potential, the NK cells have better killing activity against tumor cells K562, and the killing rate of the NK cells can reach more than 80% under the condition that the effective target ratio is 5:1.

Description

NK cell in-vitro activation and amplification method
Technical Field
The invention relates to the technical field of culture media, in particular to the field of an NK cell in-vitro activation amplification method.
Background
Natural killer cells, namely NK cells (Natural Killer Cell), are lymphocytes which are identical to but different from T cells and B cells, and have certain regulation effect on T lymphocytes and B lymphocytes in human bodies. NK cells are a class of immune cells unrelated to specific immune responses and are characterized by the cell phenotype CD3 - CD56 + The mediated cell lysis is not influenced by a main tissue compatible complex (MHC), and NK cells are divided into two subgroups of NK cells with killing activity and NK cells secreting immune regulation factors according to the difference of the density of CD56 molecule expression of the NK cells, wherein the NK cells with killing activity account for more than 90% of the NK cells, mainly have cytotoxicity and have very strong killing activity; NK cells secreting immune modulating factors can produce a large number of cytokines, primarily playing an immune modulating role. As a first defense line of organisms, NK cells not only can remove parasitic bacteria, viruses and aging variant cells in cells, but also have extremely strong removal effect on cancer cells, can realize rapid activation under the condition of not generating pre-sensitization, and have strong anti-infection and anti-tumor effects. Therefore, the clinical use of NK cell adoptive immunotherapy is now becoming an important tool for tumor cell immunotherapy. Obtaining a sufficient number of NK cells of sufficient purity is thus a crucial step in developing NK cell drug development. However, NK cells obtained by culturing several current techniques have the following problems: (1) NK cells obtained by adding soluble recombinant growth factors, IL-2, IL-15 and other conventional NK cell culture methods have the defects of low purity, poor amplification efficiency and low killing activity of the NK cells. (2) NK cells obtained by using K562 as trophoblast cells in NK cell expansion culture have high purity, good expansion, but general killing activity and are not in compliance with GMP standards, including safety questioning. (3) Bottom of cell culture flask coated with certain stimulatory monoclonal antibodiesAnd the method is troublesome to operate, easy to cause pollution, and the obtained NK cells are limited in amplification factor and low in purity. (4) The magnetic beads are used for coupling various stimulatory factors, including interleukin cytokines IL-15, IL-21, IL-18 and the like, and various antibody molecules 4-1BB ligand, CD86, MICA and the like, so that the operation is troublesome, repeated addition is required in the whole cell culture process, the instability factors are more, and the obtained NK cells have different qualities.
Therefore, those skilled in the art have been devoted to research on an NK cell activation amplification method capable of rapidly and effectively activating NK cells in vitro, obtaining NK cells with high purity, high amplification efficiency, and high killing activity through long-term culture, and meeting the requirements of GMP standards and clinical use standards.
Disclosure of Invention
In view of the problems and disadvantages of the prior art, the present invention is directed to an in vitro activated expansion method of NK cells, which solves the problems set forth in the background art.
In order to solve the technical problems, the invention provides the following technical scheme:
an NK cell in vitro activation and amplification method,
separating and extracting mononuclear cell PBMC from human peripheral blood, adding nanoparticle suspension coupled with biotin-labeled 2B4 antibody and biotin-labeled IL-1 beta antibody into cell culture suspension for activation culture on the day of activation culture of NK cells in NK cell culture medium;
the number ratio of the nano particles to the NK cells is 1:2;
the concentration of the nanoparticle suspension is 1mg/mL;
the concentration of the biotin-labeled 2B4 antibody is 0.1-0.15mg/mL, and the concentration of the biotin-labeled IL-1 beta antibody is 0.1-0.15mg/mL.
More optimized scheme, the concentration of the biotin-marked 2B4 antibody is 0.1mg/mL, and the concentration of the biotin-marked IL-1 beta antibody is 0.1mg/mL.
In a more optimized scheme, the nano particles are biodegradable matrix material dextran.
In a more optimized scheme, the biodegradable matrix material dextran is streptavidin nano particles.
In an optimized scheme, the NK cell culture medium is any one of SCGM culture medium, corning KBM581 lymphocyte serum-free culture medium and Bao Ri medical NK cell serum-free culture medium.
In a more optimized scheme, IL-2 with the final concentration of 500IU/mL, IL-15 with the final concentration of 500IU/mL and 10% heat-inactivated autologous plasma are added into the NK cell culture medium.
More optimized scheme, the effective target ratio of NK cells to target cells is as follows: 1:1 to 10:1.
The more optimized scheme is characterized in that the effective target ratio of the NK cells to the target cells is as follows: 10:1.
Compared with the prior art, the invention has the following beneficial effects:
the invention provides a nanoparticle coupled with biotin-labeled 2B4 antibody and biotin-labeled IL-1 beta antibody, which is added into a cell culture suspension on the day of NK cell activation culture, is matched with a common NK cell culture medium on the market, and is used together with soluble cytokines IL-15 and IL-2, so that NK cells can be rapidly activated and greatly amplified while the high activity rate of the NK cells is ensured, NK cells with high purity and high killing activity are obtained, the purity of NK cells cultured for 8 days can reach more than 85%, and the conventional culture needs at least 14 days, thereby greatly shortening the CD3 of the NK cells - CD56 + The positive proportion rises in time, and simultaneously the killing capacity of NK cells is also obviously improved. The adopted nano-scale particles can be biodegraded without a magnetic separator, and the nano-scale particles can be completely removed from a culture system by a centrifugal method when the cells are harvested on the 14 th day, so that the method is simple to operate and is beneficial to controlling the quality of cell medicines.
The action mechanism of the composition of the invention is as follows:
2B4 (CD 244) is a member of the signaling lymphocyte activating molecule (SLAM/CD 150), expressed on all NK cells, and the use of 2B4 antibodies to bind to the NK cell surface 2B4 protein not only stimulates activating NK cells, but also mediates expression of CD48 expressing target cellsNatural cytotoxicity, and induces release of intracellular calcium, enhancing killing potential of NK cells. IL-1β is an important member of the IL-1 family of lymphocyte stimulators, and is involved in a variety of autoimmune inflammatory responses and a variety of cellular activities, including mediating NK cell activation, proliferation and differentiation, and in synergy with IL-2, can enhance NK cell activity, and IL-1β has potent pro-inflammatory activity, and activation of cells with IL-1β antibodies can induce a variety of pro-inflammatory mediators, such as cytokines and chemokines, and can induce NK cell lysis to virally infected cells. The streptavidin-Biotin (SA-Biotin) system has extremely high binding affinity and has wide application in the biological field. SA is covalently connected to the surface of the solid-phase carrier nano-particles, so that the SA can be efficiently combined with biotin-labeled antibody molecules to continuously and efficiently activate NK cells, and the activated NK cells kill target cells. Thus, one skilled in the art would be able to detect the activated NK cell marker molecule CD3 - CD56 + The purity of NK cells and the killing efficiency of NK cells on the target cell line K562 are evaluated to judge the killing potential of NK cells. From this, it can be demonstrated that the purity, amplification efficiency and killing potential of NK cells were significantly improved by adding a nanoparticle suspension coupled to the 2B4 and IL-1 β antibodies.
Drawings
The accompanying drawings are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate the invention and together with the embodiments of the invention, serve to explain the invention. In the drawings:
FIG. 1 is a graph showing the cell viability of example 1/2/3/4;
FIG. 2 is a graph of cell expansion for example 1/2/3/4;
FIG. 3 is a graph of NK cell killing activity of example 1/2/3/4;
FIG. 4 is a schematic representation of the proportion of CD56+ cells on day 8 of culture in example 1;
FIG. 5 is a schematic representation of the proportion of CD56+ cells on day 14 of culture in example 1;
FIG. 6 is a schematic representation of the proportion of CD56+ cells on day 8 of culture in example 2;
FIG. 7 is a schematic representation of the proportion of CD56+ cells on day 14 of culture in example 2;
FIG. 8 is a schematic representation of the proportion of CD56+ cells on day 8 of culture in example 3;
FIG. 9 is a schematic representation of the proportion of CD56+ cells on day 14 of culture in example 3;
FIG. 10 is a schematic representation of the proportion of CD56+ cells on day 8 of culture in example 4;
FIG. 11 is a schematic of the proportion of CD56+ cells on day 14 of culture in example 4.
Detailed Description
The following description of the embodiments of the present invention will be made clearly and completely with reference to the accompanying drawings, in which it is apparent that the embodiments described are only some embodiments of the present invention, but not all embodiments. All other embodiments, which can be made by those skilled in the art based on the embodiments of the invention without making any inventive effort, are intended to be within the scope of the invention.
And (3) a step of: experimental materials:
SCGM NK cell culture medium (manufacturer CellGenix (Sadoriis), cat# 130-090-0500), conning KBM581 lymphocyte serum-free medium (manufacturer Corning (Conning) cat# 88-581-CM), baozi NK cell serum-free medium (manufacturer Takara (Baozi), cat# GT-T561), IL-2 (manufacturer: prime, cat# 101-02), IL-15 (manufacturer: prime, cat# 101-15), streptavidin nanoparticle (manufacturer: meta. Mu., cat# 130-090-485), biotin-labeled 2B4 antibody (manufacturer: biolegend (hundred organism), cat# 302004), biotin-labeled IL-1 beta antibody (manufacturer: biolegend (hundred organism), cat# 511703), CD3 antibody (Biolegend (hundred organism), cat# 300439), CD56 antibody (Biolegend (hundred organism), cat# 362508), K cell line (ATCC (manufacturer: prime# 130-090-485), biotin-labeled IL-1 beta antibody (manufacturer: 62), cat# 40-carrier (HY-40), and stock solution (ChemMexico-40).
And II: the experimental method comprises the following steps:
(1) Preparation of nanoparticle suspensions coupling 2B4 with IL-1 beta antibodies
Adding 1mL of nano-particles with the diameter of 100nm and biodegradable matrix material dextran, specifically 1mg/mL of streptavidin nano-particle suspension into a centrifuge tube, wherein each milliliter of streptavidin nano-particle suspension contains 1X 10 8 Directly adding 0.1mg of biotin-labeled 2B4 antibody and 0.1mg of biotin-labeled IL-1 beta antibody into a centrifuge tube containing nanoparticle suspension, placing the centrifuge tube on a vortex oscillator for shaking for 20 seconds, placing the centrifuge tube on a rotary mixer, rotatably mixing for 30 minutes at room temperature, and placing the mixture into a refrigerator at 2-8 ℃ for standing and incubating for 30 minutes to obtain nanoparticle suspension of coupled biotin-labeled 2B4 and IL-1 beta antibody; wherein the concentration of nanoparticle suspension coupled with biotin-labeled 2B4 and IL-1β antibody is 1mg/mL.
(2) NK cell culture
PBMCs were freshly isolated from human whole blood using density gradient centrifugation for activated expansion of NK cells and autologous plasma was collected and inactivated at 56 ℃ for 30 min for cell culture. On day 0, the isolated PBMC cell suspension was transferred to a T75 flask and cell density was adjusted to 2X 10 using NK cell medium 6 Adding IL-2 with a final concentration of 500IU/mL, IL-15 with a final concentration of 500IU/mL and heat-inactivated autologous plasma accounting for 10% of the volume of NK cell culture medium, and simultaneously adding nanoparticle suspension coupled with 2B4 and IL-1 beta antibody, wherein the whole cell culture period only needs to add nanoparticle suspension coupled with 2B4 and IL-1 beta antibody once, and the ratio of microsphere particles to cell number is 1:2, namely the culture medium volume per milliliter contains 2X 10 6 The 10 mu L nanoparticle suspension of the coupled biotin-labeled 2B4 and IL-1 beta antibody is added into cells, and the cells are placed into a 37 ℃ and 5% carbon dioxide incubator for cell activation. On day 3 after activation, NK cell culture medium added with the IL-2, IL-15 and 10% heat inactivated autologous plasma is supplemented in the same volume as that on day 0, then cell viability and density are tested every 2 days, and cell density is regulated to 1-1.5X10 by fluid replacement 6 Culture was continued until day 14 at each mL to obtain cells to be treated.
(3) NK cell viability and proliferation efficiency detection
The cells were sampled and counted on days 0,5,7,9, 11 and 14 of the culture, respectively, to obtain data of cell proliferation curve and cell viability.
(4) NK cell identification
Cell immunophenotyping assays were performed on day 8 and day 14 of cell culture. About 1X 10 is taken respectively 6 The individual cells were centrifuged at 2000rpm for 5 minutes, the supernatant removed and resuspended in PBS, and the fluorescently labeled CD3 antibody and CD56 antibody were used to incubate the PBS-resuspended cells under the following conditions: at 4℃for 15 min, then washed 2 times with 1mL of PBS, the supernatant discarded, and resuspended in 0.5mL of PBS. And detecting and analyzing the treated cells by using a flow cytometer.
(5) NK cell killing Activity assay (CCK 8 method)
Taking K562 cell strain with vigorous growth as target cell, washing with RPMI-1640 culture solution for 2 times, and adjusting cell density to 1×10 6 And each mL. 50 μl of each well was plated in a 96-well plate. The NK cells cultured until day 14 were collected and the cell density was adjusted to 1X 10 6 Per mL, 2X 10 6 Per mL, 5X 10 6 Per mL, 1X 10 7 50 mu L of each/mL is taken and added into a 96-well plate, so that the effective target ratios are respectively as follows: 1:1, 2:1, 5:1, 10:1, into four groups, respectively experimental group (K562 + NK), control group (NK + RPMI-1640), maximum release group (K562 + RPMI-1640) and blank group (RPMI-1640), wherein the control group does not contain K562 cells, and the obtained absorbance is the value of K562 cell 0 killing; the maximum release group was subjected to K562 cell lysis, and the absorbance obtained was a value of 100% killing; the blank is medium, no cells are contained, and the absorbance obtained is the background value. Each group had 3 duplicate wells. After the inoculated cells are placed in an incubator for co-culture for 12 hours, 10 mu LCCK-8 reagent is added into each hole, and the cells are uniformly shaken for continuous culture for 3 hours. The enzyme label instrument detects the light absorption value at the wavelength of 450nm, and the killing rate is calculated according to the calculation mode: 100 are 1- [ experimental (OD value) -control (OD value) -blank (OD value) ]/[ maximum release (OD value) -blank (OD value) ].
Example 1:
culturing and identifying NK cells completely according to the experimental method, adding nanoparticle suspension coupled with 2B4 and IL-1 beta antibody into SCGM culture medium on the day of activation (D0), and continuing culturing until the 14 th day (D14) after NK activation;
the experimental results are shown in fig. 1: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody is added, and the NK cells keep a high-activity state in the whole cell culture period, and the activity rate of the NK cells is 97% when harvested on the 14 th day.
The experimental results are shown in fig. 2: with the addition of the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody, NK cells are rapidly amplified after the 5 th day, and the NK cells are amplified up to 1600 times when harvested on the 14 th day.
The experimental results are shown in fig. 3: the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody can obviously enhance the killing activity of the harvested NK cells, and the method comprises the following steps of: under the effective target ratio of 1, the NK cell killing rate on the 14 th day can reach 82%, and the NK cell killing rate is as high as 10:1, the NK cell killing rate on the 14 th day can reach 100%.
The experimental results are shown in fig. 4 and 5: when the nanoparticle suspension of the invention, which is coupled with the 2B4 and the IL-1 beta antibody, is added, the positive rate of NK cells (CD 3-CD56+) rises fast, 86% can be reached on the 8 th day of culture, and the positive rate of NK cells (CD 3-CD56+) is 97.4% when the culture is harvested on the 14 th day.
Example 2:
unlike example 1, SCGM NK medium was replaced with corning KBM581 lymphocyte serum-free medium.
The test according to the above experimental method shows that:
the experimental results are shown in fig. 1: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody is added, and the NK cells keep a high-activity state in the whole cell culture period, and the activity rate of the NK cells is 92% when harvested on the 14 th day.
The experimental results are shown in fig. 2: with the addition of the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody, NK cells are rapidly amplified after the 5 th day, and the NK cells are amplified by 1250 times when harvested on the 14 th day.
The experimental results are shown in fig. 3: the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody can obviously enhance the killing activity of the harvested NK cells, and the method comprises the following steps of: 1, the NK cell killing rate on the 14 th day can reach 78%, and the NK cell killing rate is as follows: 1, the NK cell killing rate on the 14 th day can reach 100%.
The experimental results are shown in fig. 6 and 7: when the nanoparticle suspension of the invention, which is coupled with the 2B4 and the IL-1 beta antibody, is added, the positive proportion of NK cells (CD 3-CD56+) rises fast, 78.1% can be reached on 8 th day of culture, and the positive proportion of NK cells (CD 3-CD56+) is 92.2% when the culture is harvested on 14 th day.
Example 3:
unlike in examples 1 and 2, SCGM NK medium was replaced with baodial NK cell serum-free medium.
The test according to the above experimental method shows that:
the experimental results are shown in fig. 1: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody is added, and the NK cells keep a high-activity state in the whole cell culture period, and the activity rate of the NK cells is 94% when harvested on the 14 th day.
The experimental results are shown in fig. 2: with the addition of the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody, NK cells are rapidly amplified after the 5 th day, and the NK cells are amplified up to 1400 times when harvested on the 14 th day.
The experimental results are shown in fig. 3: the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody can obviously enhance the killing activity of the harvested NK cells, and the method comprises the following steps of: 1, the NK cell killing rate on the 14 th day can reach 81% under the effective target ratio of 10:1, the NK cell killing rate on the 14 th day can reach 100%.
The experimental results are shown in fig. 8 and 9: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody has high rising speed of NK cell positive proportion (CD 3-CD56+) which can reach 82.8% after 8 days of culture, and the NK cell positive proportion (CD 3-CD56+) is 91.6% when the culture is harvested on 14 days.
Example 4:
unlike example 1, a nanoparticle suspension of biotin-labeled 2B4 and IL-1 β antibody was used at a level of 0.15mg per ml of coupled 2B4 and IL-1 β antibody;
the test according to the above experimental method shows that:
the experimental results are shown in fig. 1: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody is added, and the NK cells keep a high-activity state in the whole cell culture period, and the activity rate of the NK cells is 90% when harvested on the 14 th day.
The experimental results are shown in fig. 2: with the addition of the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody, NK cells are rapidly amplified after the 5 th day, and the NK cells are amplified by 1100 times when harvested on the 14 th day.
The experimental results are shown in fig. 3: the nanoparticle suspension coupled with the 2B4 and IL-1 beta antibody can obviously enhance the killing activity of the harvested NK cells, and the method comprises the following steps of: 1, the NK cell killing rate on the 14 th day can reach 71%, and the NK cell killing rate is as high as 10:1, the NK cell killing rate on the 14 th day can reach 100%. Example 4 cultured NK cells were less killing against K562 cells than example 1.
The experimental results are shown in fig. 10 and 11: the nanoparticle suspension coupled with the 2B4 and the IL-1 beta antibody has the advantages that the positive rate of NK cells (CD 3-CD56+) rises fast, 74.4% can be achieved on 8 th day of culture, and 89.6% of NK cells are obtained on 14 th day of harvest. Example 4 cultured NK cells were not as pure as example 1.
It is noted that relational terms such as first and second, and the like are used solely to distinguish one entity or action from another entity or action without necessarily requiring or implying any actual such relationship or order between such entities or actions. Moreover, the terms "comprises," "comprising," or any other variation thereof, are intended to cover a non-exclusive inclusion, such that a process, method, article, or apparatus that comprises a list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, article, or apparatus.
Finally, it should be noted that: the foregoing description is only a preferred embodiment of the present invention, and the present invention is not limited thereto, but it is to be understood that modifications and equivalents of some of the technical features described in the foregoing embodiments may be made by those skilled in the art, although the present invention has been described in detail with reference to the foregoing embodiments. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (8)

1. An NK cell in vitro activation and amplification method is characterized in that,
separating and extracting mononuclear cell PBMC from human peripheral blood, adding nanoparticle suspension coupled with biotin-labeled 2B4 antibody and biotin-labeled IL-1 beta antibody into cell culture suspension for activation culture on the day of activation culture of NK cells in NK cell culture medium;
the number ratio of the nano particles to the NK cells is 1:2;
the concentration of the nanoparticle suspension is 1mg/mL;
the concentration of the biotin-labeled 2B4 antibody is 0.1-0.15mg/mL, and the concentration of the biotin-labeled IL-1 beta antibody is 0.1-0.15mg/mL.
2. The method for in vitro activation and expansion of NK cells of claim 1 wherein,
the concentration of the biotin-labeled 2B4 antibody is 0.1mg/mL, and the concentration of the biotin-labeled IL-1 beta antibody is 0.1mg/mL.
3. The method for in vitro activated expansion of NK cells according to claim 1, wherein said nanoparticle is a biodegradable matrix material dextran.
4. The method for in vitro activated expansion of NK cells according to claim 3, wherein said biodegradable matrix material dextran is streptavidin nanoparticle.
5. The method for in vitro activation and amplification of NK cells according to claim 1, wherein the NK cell culture medium is any one of SCGM culture medium, corning KBM581 lymphocyte serum-free medium and Bao Ri Yi NK cell serum-free medium.
6. The method for in vitro activation and expansion of NK cells according to claim 1, wherein IL-2 with a final concentration of 500IU/mL, IL-15 with a final concentration of 500IU/mL, 10% heat-inactivated autologous plasma are added to the NK cell culture medium.
7. The method for in vitro activation and expansion of NK cells according to claim 1, wherein the NK cells to target cells have an effective target ratio of: 1:1 to 10:1.
8. The method for in vitro activation and expansion of NK cells according to claim 7, wherein the NK cells to target cells have an effective target ratio of: 10:1.
CN202310413829.3A 2023-04-18 2023-04-18 NK cell in-vitro activation and amplification method Active CN116426475B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310413829.3A CN116426475B (en) 2023-04-18 2023-04-18 NK cell in-vitro activation and amplification method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310413829.3A CN116426475B (en) 2023-04-18 2023-04-18 NK cell in-vitro activation and amplification method

Publications (2)

Publication Number Publication Date
CN116426475A true CN116426475A (en) 2023-07-14
CN116426475B CN116426475B (en) 2023-10-31

Family

ID=87079411

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310413829.3A Active CN116426475B (en) 2023-04-18 2023-04-18 NK cell in-vitro activation and amplification method

Country Status (1)

Country Link
CN (1) CN116426475B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117384839A (en) * 2023-08-30 2024-01-12 广州达博生物制品有限公司 NK cell in-vitro amplification method

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180155690A1 (en) * 2016-09-28 2018-06-07 Korea Institute Of Radiological & Medical Sciences Method for preparing natural killer cells using irradiated pbmcs, and anti-cancer cell therapeutic agent comprising the nk cells
WO2020175960A1 (en) * 2019-02-28 2020-09-03 재단법인 아산사회복지재단 Microparticles for detecting natural killer cell activity and detection method using same
KR20200143578A (en) * 2019-06-13 2020-12-24 한국원자력의학원 Method for Isolating NK cells Directly from whole blood and Method for Mass Proliferation of the NK cells
US20220347216A1 (en) * 2019-09-13 2022-11-03 Ohio State Innovation Foundation Nk cell immunotherapy compositions, methods of making and methods of using same

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180155690A1 (en) * 2016-09-28 2018-06-07 Korea Institute Of Radiological & Medical Sciences Method for preparing natural killer cells using irradiated pbmcs, and anti-cancer cell therapeutic agent comprising the nk cells
WO2020175960A1 (en) * 2019-02-28 2020-09-03 재단법인 아산사회복지재단 Microparticles for detecting natural killer cell activity and detection method using same
KR20200143578A (en) * 2019-06-13 2020-12-24 한국원자력의학원 Method for Isolating NK cells Directly from whole blood and Method for Mass Proliferation of the NK cells
US20220347216A1 (en) * 2019-09-13 2022-11-03 Ohio State Innovation Foundation Nk cell immunotherapy compositions, methods of making and methods of using same

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CASEY W BULLER等: "Roles of NK Cell Receptors 2B4 (CD244), CS1 (CD319), and LLT1 (CLEC2D) in Cancer", 《CANCERS》, vol. 12, no. 7, pages 1 - 13 *
MINA M SANDUSKY等: "Regulation of 2B4 (CD244)-mediated NK cellactivation by ligand-induced receptor modulation", 《EUR J IMMUNOL》, vol. 36, no. 12, pages 3268 - 3276 *
PAOLO AMBROSINI等: "IL-1β inhibits ILC3 while favoring NK-cell maturation of umbilical cord bloodCD34 precursors", 《EUROPEAN JOURNAL OF IMMUNOLOGY》, vol. 45, no. 8, pages 2061 - 2071, XP055222638, DOI: 10.1002/eji.201445326 *
郭振兴等: "利用饲养细胞体外扩增高纯度的效应NK细胞", 《同济大学学报(医学版)》, no. 2, pages 48 - 53 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117384839A (en) * 2023-08-30 2024-01-12 广州达博生物制品有限公司 NK cell in-vitro amplification method

Also Published As

Publication number Publication date
CN116426475B (en) 2023-10-31

Similar Documents

Publication Publication Date Title
US20210115401A1 (en) Methods and Materials for the Generation of Regulatory T Cells
CN109666639B (en) NK (Natural killer) cell with enhanced killing activity and preparation method thereof
CN108588022B (en) Method for enriching human CD4+ and CD8+ TCM cells through in vitro culture
CN116426475B (en) NK cell in-vitro activation and amplification method
CN115651903B (en) High-lethality immune cell population, and culture method, reagent composition and application thereof
CN111235209A (en) Method for evaluating immune regulation function of stem cells
CN111117958B (en) In-vitro amplification kit and amplification method for human peripheral blood regulatory T cells
CN115558641A (en) High-purity effector immune cell population, and culture method, reagent composition and application thereof
CN113249321A (en) Peripheral blood NK cell culture method
CN112852731A (en) Method for inducing hematopoietic stem cells to differentiate into regulatory T cells in vitro
CN112251405A (en) Method for efficiently inducing and amplifying NK cells in vitro
CN108486055B (en) Culture medium and application thereof in central memory type T lymphocyte culture
CN113005081B (en) Culture method for amplifying stem cell-like memory T cells
CN115521914A (en) Human primary natural killer cell in-vitro amplification system and method
CN115678845A (en) Method for culturing tumor-specific CTL cells and cell therapy product
CN105861484B (en) Cell culture medium composition containing resveratrol and silk sericin
US8313945B2 (en) Methods for inducing the differentiation of blood monocytes into functional dendritic cells
CN113195706A (en) CIML NK cells and methods thereof
CN114958740B (en) Method for in vitro culture and enrichment of human NK cells
CN112961827B (en) Application of forskolin in T cell culture
TWI768356B (en) A culture medium for in vitro expansion and activation of gamma delta t cells
CN114990061B (en) Culture method for inducing expansion of central memory T cells
CN110628714B (en) Serum-free cell culture solution for in vitro amplification of natural killer cells and natural killer T cells
CN117603911A (en) Natural killer cell culture system, culture kit and culture method
CN116063449A (en) Method for preparing tumor antigen specific T cells

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