CN111235105B - Method for differentiating human pluripotent stem cells into natural killer cells and application - Google Patents

Method for differentiating human pluripotent stem cells into natural killer cells and application Download PDF

Info

Publication number
CN111235105B
CN111235105B CN202010153358.3A CN202010153358A CN111235105B CN 111235105 B CN111235105 B CN 111235105B CN 202010153358 A CN202010153358 A CN 202010153358A CN 111235105 B CN111235105 B CN 111235105B
Authority
CN
China
Prior art keywords
cells
differentiation
cell
medium
culture medium
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
CN202010153358.3A
Other languages
Chinese (zh)
Other versions
CN111235105A (en
Inventor
不公告发明人
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.)
Nuwacell Ltd
Original Assignee
Nuwacell 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 Nuwacell Ltd filed Critical Nuwacell Ltd
Priority to CN202010153358.3A priority Critical patent/CN111235105B/en
Publication of CN111235105A publication Critical patent/CN111235105A/en
Application granted granted Critical
Publication of CN111235105B publication Critical patent/CN111235105B/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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
    • 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
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • 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
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-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
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/14Erythropoietin [EPO]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/145Thrombopoietin [TPO]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/10Growth factors
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2301Interleukin-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]
    • C12N2501/2303Interleukin-3 (IL-3)
    • 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]
    • C12N2501/2306Interleukin-6 (IL-6)
    • 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]
    • C12N2501/2307Interleukin-7 (IL-7)
    • 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]
    • C12N2501/2312Interleukin-12 (IL-12)
    • 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]
    • C12N2501/2318Interleukin-18 (IL-18)
    • 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]
    • C12N2501/2321Interleukin-21 (IL-21)
    • 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]
    • C12N2501/2327Interleukin-27 (IL-27)
    • 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
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • 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
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • 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
    • C12N2501/40Regulators of development
    • C12N2501/405Cell cycle regulated proteins, e.g. cyclins, cyclin-dependant kinases
    • 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
    • C12N2501/58Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
    • 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
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem 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
    • C12N2513/003D culture
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/52Fibronectin; Laminin

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Dentistry (AREA)
  • Environmental Sciences (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to the field of stem cell biology, in particular to a method for differentiating human pluripotent stem cells into natural killer cells and application thereof. The invention discloses a natural killer cell derived from pluripotent stem cells, which expresses CD56, Nkp30, Nkp44 and Nkp46 and also expresses markers of mature natural killer cells, namely CD16 and CD 94. The invention also discloses a method for preparing the natural killer cells, which comprises the following steps: s1: forming an embryoid body; s2: the embryoid body is differentiated into hematopoietic progenitor cells; s3: differentiating hematopoietic progenitor cells into NK cells; s4: maturation and expansion of NK cells. The differentiation method provided by the invention can rapidly, efficiently, simply and conveniently induce the pluripotent stem cells to differentiate into natural killer cells at lower cost based on the culture medium with definite components and the optimized combination of cytokines.

Description

Method for differentiating human pluripotent stem cells into natural killer cells and application
Technical Field
The invention relates to the field of stem cell biology, in particular to lineage specific differentiation of pluripotent or multipotent stem cells, and specifically relates to a method for differentiating human pluripotent stem cells into natural killer cells and application thereof.
Background
The national latest cancer report in 2018 shows that the national malignancy estimates 380.4 ten thousand new cases in 2014, and more than 1 million people are diagnosed as cancer on average each day. The incidence of the tumor is 278.07/10 ten thousand, the cumulative incidence of the disease is 21.58% between 0 and 74 years old, and the cumulative mortality is 12.00%. In 2013, the journal of science lists tumor immunotherapy as the first ten scientific breakthroughs, and the immunotherapy becomes a new generation of tumor treatment means after surgery, chemotherapy, radiotherapy and tumor targeted therapy, thereby bringing new hopes for treating tumor patients.
Natural killer cells (NK) are the mainstay of the innate immune system, the first line of defense for the body against infection and tumors, and can recognize and kill diseased cells by either the "self-deletion" (missing-self) or "stress-induced" (stress-induced) pattern on the surface of malignant diseased cells. The activation of NK cells is dependent on the result of a balance of activated and inhibitory receptor signals on their cell surface. Activated NK cells can kill tumor cells by a variety of mechanisms: NK cells can release perforin and granzyme, and directly act on target cells to kill cancer cells; cancer cell apoptosis can be promoted by secretion of proinflammatory cytokines; killing can be accomplished by targeting cancer cells that recognize the monoclonal antibody drug marker.
The recognition and killing of NK cells have the characteristics of MHC-I non-restriction, pan-specificity and quick response, have no graft-versus-host reaction (GVHD), can adopt the feedback of allogeneic cells, have short survival period in vivo, have no side effects such as cytokine storm and the like, can be used together with tumor-targeted antibodies to enhance the anti-cancer effect, can be combined with a gene editing technology to develop targeted NK cell products aiming at different tumor targets, and have great application potential in tumor immunotherapy due to the characteristics.
Previous studies have demonstrated that NK cells in tumor patients are often reduced in number and impaired in function. Adoptive therapy of NK cells therefore has the efficacy of restoring immune function to patients against tumors. However, clinical studies after reinfusion of autologous NK cells show that their antitumor effects are very limited. The main reasons may include: (1) the KIR receptor of the autologous NK cells is matched with HLA of the tumor cells, and the activation of the NK cells is inhibited by the self-recognition signal; (2) the function of the patient's own NK cells is damaged, and the ideal killing effect is difficult to generate.
NK cells directly expanded from peripheral blood have certain difficulties, may fail to expand the cells of the patient, and usually the expanded cell mixture contains a certain proportion of T cells, which need to be cleared, otherwise it will cause Graft Versus Host Disease (GVHD). Meanwhile, the NK cytoplasm of different donors has great difference, directly influences the curative effect and is difficult to form a standardized product.
Human pluripotent stem cells (hpscs) including human embryonic stem cells (hescs) and human induced pluripotent stem cells (hipscs) have the ability to proliferate indefinitely and can differentiate into almost all functional cells in vitro, including NK cells. The in vitro large-scale and standardized preparation of the NK cells from the human pluripotent stem cells is an effective solution for the NK cell product patent medicine. However, the differentiation process reported at present has the problems of complex process, long period, feeder layer cell dependence, undefined culture medium components, functional defects of differentiated cells, low purity and activity and the like, so that the differentiation process is not beneficial to large-scale production.
The closest patents to the present invention are: patent CN102388130A discloses differentiation of a pluripotent cell, patent CN107429230A discloses a method and composition for inducing differentiation of hematopoietic cells, and patent CN102822332A discloses a method for generating natural killer cells and dendritic cells from hESC-derived angioblasts.
Patent CN102388130A has at least some of the following problems: only including differentiation of hpscs into hematopoietic precursor cells, but not including the stage of continued differentiation of hematopoietic precursor cells into NK cells. The factor combination used did not bypass the original hematopoietic phase.
Patent CN107429230A has at least some of the following problems: (1) EB 3D was very inefficient. (2) 2D differentiation conditions were used from hPSC to permanent pluripotent blood precursor cells. (3) The whole process uses expensive commercial culture medium (such as Stempro 34) with undefined components, bovine serum and the like, and is not beneficial to the production of clinical grade cell products. (4) The enriched CD34+/CD45+ cells are used for iNK (iPSC-derived NK, NK cells induced and differentiated by iPSC) differentiation, and the process is complicated.
Patent CN102822332A has at least some of the following problems: (1) expensive commercial media with undefined composition were used; (2) the obtained hemangioblast cells mostly belong to primitive hematopoietic cells; (3) the single cell obtained by EB digestion on the differentiation day4 is inoculated into a Methylellulose culture system for further differentiation to obtain vascular cells, and the system is not beneficial to the large-scale production of clinical NK cells; (4) the second and third steps of the method both use a medium containing human serum.
In conclusion, the NK cells have unique advantages and wide application prospects in immunotherapy of cancers, while NK cells of NK cells (PB-NK) from peripheral blood are limited in source and inconsistent in quality and yield, and are often polluted by T cells after being amplified. Therefore, the preparation of NK cells by using hPSC as a source can solve the problems and achieve stable large-scale production. However, the existing differentiation scheme has many problems that serum is an uncontrollable culture factor and further causes unstable factors such as batch difference and the like when a culture medium containing serum is used; the use of feeder layer cells of animal cell origin such as OP9 does not meet clinical production requirements; the commercial serum-free culture medium is expensive and is not suitable for large-scale production; the differentiation process is complex, the dosage and time of the cell factors are not fully optimized and simplified, and the cost is high; the long differentiation period also increases the production cost and improves the uncontrollable process control; the differentiated NK cells have low purity and lack of functions.
Therefore, the invention needs to invent a stable, efficient, low-cost, high-purity and functional NK cell differentiation system, and establishes a foundation for large-scale production and clinical application of NK cells.
Disclosure of Invention
Although the process of in vitro differentiation from pluripotent stem cells into natural killer cells is well-established in principle, and various methods for inducing differentiation are available at present, the existing differentiation methods have the problems of long differentiation period, low differentiation efficiency and purity, unclear differentiation components, feeder layer cell dependence, and complicated operation and high cost, so that it is difficult to expand the production and apply the methods to clinical application. The differentiation program provided by the invention can rapidly, efficiently, simply and conveniently induce the pluripotent stem cells to differentiate into natural killer cells at lower cost based on the culture medium with definite components and the optimized combination of cytokines.
Its advantages are as follows: firstly, the invention realizes the stable and high-efficiency differentiation of natural killer cells, and can obtain NK cells with purity of more than 90% and functionality in about 40 days; secondly, no culture system containing serum or feeder layer cells are used in the whole differentiation process, so that the method is suitable for expanded production and clinical application; third, we try to simplify and optimize the media and cytokines used in the differentiation process, avoiding the use of expensive commercial media such as Stemspan, etc., and thus the cost of the entire differentiation process is greatly reduced.
The key innovation points of the invention are as follows: 1) the EB 3D differentiation method has low cost, short period and high efficiency, can obtain NK cells which have large quantity, high purity and similar cytotoxicity with NK cells from peripheral blood sources only in about 25 days, and is suitable for large-scale production; 2) the components of the differentiation medium are clear, NK cells can be successfully differentiated under the condition of not using serum and trophoblast cells, and the method is suitable for clinical application; 3) the amplification conditions are optimized, and the NK cells obtained by differentiation can be massively amplified under the condition of no serum and trophoblast cells; 4) the cryopreservation condition is optimized, the NK cells can be stored for a long time, and higher cell activity can be maintained after recovery.
Specifically, the technical scheme of the invention is as follows:
the invention discloses a pluripotent stem cell-derived natural killer cell, which expresses CD56, Nkp30, Nkp44 and Nkp46 and also expresses markers of mature natural killer cells, namely CD16 and CD 94. In the present invention, NKp46, NKp30 and NKp44 all belong to the Natural Cytotoxic Receptor (NCR), and all three are immunoglobulin superfamily (IgSF) members, but have no homology with each other. NCR is expressed only on the surface of NK cells, is a unique marker of NK cells, and usually exerts a killing effect when KIR/KLR loses the ability to recognize "self". NKp30(NCR3) is an important member of the NCR family, expressed on the surface of all NK cells, and plays an important role in the process of NK cell activation and tumor killing. NKp44, CD336, is a member of IgSF, a natural cytotoxic receptor (NCR 2). It is expressed in activated NK cells with the ligand DAP12, involved in mediating NK cell killing activity. The NKp46 extracellular domain contains 2 Ig-like domains, and the NKp30 extracellular domain has only one V-type domain. NKp46 and NKp30 have short cytoplasmic domains, and both transmembrane regions contain positively charged arginine.
In a second aspect of the present invention, there is disclosed a method for preparing natural killer cells, comprising the steps of:
s1: forming an embryoid body;
s2: the embryoid body is differentiated into hematopoietic progenitor cells;
s3: differentiating hematopoietic progenitor cells into NK cells;
s4: maturation and expansion of NK cells.
Preferably, the S1 includes:
s11: placing the cell suspension of the human pluripotent stem cells on a shaking table to shake and culture overnight to form embryoid bodies.
Preferably, the S2 includes:
s21: removing the supernatant of the embryoid body, and adding a fresh first-step differentiation culture medium for cell culture; wherein, the first step differentiation culture medium is obtained by adding a small molecule GSK3 beta inhibitor and at least one of the following components in a basic differentiation culture medium: BMP signaling pathway activators, VEGF (vascular endothelial growth factor), bFGF (basic fibroblast growth factor), SCF (stem cell factor), Flt3L (FMS-like tyrosine kinase 3 ligand), IL3 (interleukin 3), IL6 (interleukin 6), insulin (insulin), IGF-1 (insulin-like growth factor 1), and TPO (human thrombopoietin);
s22: removing the first step differentiation culture medium, and adding the second step differentiation culture medium for cell culture; wherein, the second step differentiation culture medium is prepared by adding VEGF, bFGF and at least one of the following components in a basic differentiation culture medium: BMP signaling pathway activators, Noda inhibitors, SCF, Flt3L, IL15, IL3, IL6, insulin, IGF-1 and TPO.
S23: removing the differentiation culture medium of the second step, and adding the differentiation culture medium of the third step for cell culture to obtain hematopoietic progenitor cells; wherein, the third step differentiation culture medium is to add growth factors and colony stimulating factors in the basic differentiation culture medium.
More preferably, the growth factor is selected from one or more of EGF (epidermal growth factor), VEGF (vascular endothelial growth factor), bFGF, insulin, IGF-1, PGF (platelet growth factor) and PDGF (platelet derived growth factor); the colony stimulating factor is selected from one or more of G-CSF (granulocyte colony stimulating factor), M-CSF (macrophage colony stimulating factor), GM-CSF (recombinant human granulocyte-macrophage colony stimulating factor), multi-CSF (multiple colony stimulating factor, also called IL-3), EPO (erythropoietin), TPO, SCF and Flt-3L.
Preferably, in S3, the third step differentiation medium is removed, cells are seeded in a cell culture vessel coated with matrix protein, and the fourth step differentiation medium is added for cell culture; the matrix protein at least comprises one of Notch pathway activator protein or integrin; wherein the fourth step of differentiation medium is a basal differentiation medium to which colony stimulating factors and interleukins are added.
The cell culture vessel coated with Notch pathway activator protein and integrin is used for culture, so that the growth rate of cells can be increased, and the differentiation efficiency of NK cells can be improved.
In some embodiments of the invention, the cell culture vessel is a cell culture flask.
More preferably, the colony stimulating factor is selected from one or more of G-CSF, M-CSF, GM-CSF, Multi-CSF (IL-3), EPO, TPO, SCF and Flt-3L; the interleukin is selected from one or more of IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21 and IL-27.
More preferably, the Notch pathway activating protein is selected from one or more of DLL1 recombinant protein, DLL4 recombinant protein, Jagged-1 recombinant protein, Jagged-2 recombinant protein and variants thereof, where "they" refers to DLL1 recombinant protein, DLL4 recombinant protein, Jagged-1 recombinant protein or Jagged-2 recombinant protein.
The integrin is selected from one or more of Fibronectin (Fibronectin), Laminin (Laminin), Vitronectin (Vitronectin), MAdCAM-1 (adhesion cell adhesion molecule-1), VCAM-1 (adhesion cell adhesion molecule-1), and ICAM (intercellular adhesion molecule), and variants of these integrins, where "they" refer to Fibronectin (Fibronectin), Laminin (Laminin), Vitronctin (Vitronectin), MAdCAM-1 (adhesion cell adhesion molecule-1), VCAM-1 (adhesion cell adhesion molecule-1), or ICAM (intercellular adhesion molecule).
Preferably, the S4 includes: removing the differentiation culture medium in the fourth step, and adding the differentiation culture medium in the fifth step for cell culture; wherein the fifth step differentiation medium is a basic differentiation medium to which interleukins and a substance promoting the maturation and expansion of NK cells are added.
More preferably, the interleukin is selected from one or more of IL-2, IL-12, IL-18, IL-21, and IL-27 and IL-15; the substance promoting NK cell maturation and expansion is selected from one or more of human AB plasma, human platelet lysate, Vitamin A, nicotinamide, Vitamin E and Heparin.
Preferably, the method further comprises step S5: freezing and storing the NK cells obtained by differentiation by using a freezing and storing solution; the cryopreservation liquid comprises: sodium chloride, sodium gluconate, sodium acetate, potassium chloride, magnesium chloride, human serum albumin and DMSO.
In some embodiments of the invention, the NK cell cryopreservation fluid composition is shown in table 1 below.
TABLE 1
Serial number Composition (I) Final concentration
1 Sodium chloride 1-10mg/ml
2 Sodium gluconate 1-10mg/ml
3 Sodium acetate 1-10mg/ml
4 Potassium chloride 0-1mg/ml
5 Magnesium chloride 0-1mg/ml
6 HSA (human serum albumin) 10-30%
7 DMSO (dimethyl sulfoxide) 5-20%
The steps of the method for preparing the natural killer cells are as follows:
first, Day-1 to Day 0: formation of Embryoid Bodies (EB)
The iPSC which is well undifferentiated and cultured until the polymerization degree is 70-90% is digested into single cell suspension, the single cell suspension is suspended in an iPSC maintenance culture medium according to a certain density, and the iPSC suspension is placed on a shaking table in an incubator at 37 ℃ for shaking culture overnight to form EB with uniform size and shape.
Operation details and optimization of Day-1-Day 0 experiment
1) Culture of human iPSC
The human ipscs used in the experiments were subjected to strict pluripotency validation (expressing various pluripotency markers and forming teratomas comprising three germ layers, inner, middle and outer, in immunodeficient mice). The ipscs are normally cultured in iPSC maintenance medium, and the medium used is E8 or TeSR or other similar medium.
2) Formation of EB
The formation experiment of the embryoid body is carried out when the iPSC is cultured to 70-90% of polymerization degree according to the method. The specific operation is as follows: use ofTrypLE or accutase digests ipscs into a complete single cell suspension, resuspended in iPSC maintenance medium, and Rock inhibitor added to the medium. Placing the cell suspension on a 3D shaking table in an incubator at 37 ℃ for shaking culture, wherein the rotation speed of the shaking table is 10-100 rpm; the shaking culture time in this step is 8-32 hours; at the end of the culture, EBs of more uniform size and morphology were obtained. For example, when T25 flask is used, the Rock inhibitor is Y-27632 at a concentration of 10. mu.M; the cell density was 0.1X 106-5×106Per mL; the rotating speed of the shaking table is 10-20 rpm; the culture time is 8-32 hours.
(II) Day 0 to Day 6-12: differentiation of pluripotent stem cells into hematopoietic progenitor cells
The EBs from D0 were changed to differentiation medium at different time points and EBs containing highly expressing CD34+ cells were harvested at D6-12.
Experimental details and optimization of Day 0-Day 6-12
1) Day 0: replacing with the first step differentiation culture medium
The flask containing the D0 EB was removed from the incubator, the flask was tilted to allow the EB to settle to the bottom, the supernatant was removed, and fresh first step differentiation medium was added. The first step differentiation medium is a basal differentiation medium supplemented with a small molecule GSK3 β inhibitor and one or more cytokines selected from BMP signaling pathway activators, VEGF, bFGF, SCF, Flt3L, IL3, IL6, insulin, IGF-1, and TPO. BMP signal pathway activators are BMP2, BMP4, SB4, ventromorphins (SJ000291942, SJ000063181, SJ000370178), Isoliquiritigenin, diosmetin (diosmin), apigenin (apigenin), biochanin (biochanin) and the like, wherein the isoliquirigenin is a flavonoid compound separated from glycyrrhiza glabra roots and has antitumor activity; the GSK3 beta inhibitor is NP031112, TWS119, SB216763, CHIR-98014, AZD2858, AZD1080, SB415286, LY2090314, CHIR99021, etc. For example, when the culture is carried out using T25 flask, in which the GSK3 beta inhibitor is CHIR99021, at a concentration of 0.5-20. mu.M; the BMP signal pathway activator is BMP4, and the concentration is 0-200 ng/mL; the cytokine comprises insulin, IGF-1, VEGF, bFGF, the concentration is 0-10 mug/mL, 0-100ng/mL, 5-100ng/mL and 0-100ng/mL respectively.
The time for culturing the EBs in the first-step differentiation medium was (2-4) days.
The composition of the basal differentiation medium is shown in Table 2 below.
TABLE 2
Figure BDA0002403196380000071
2) Day 2-4: replacing with the second step differentiation culture medium
Taking out the culture bottle filled with EB from the culture box, inclining the culture bottle to make EB sink to the bottom, removing the supernatant of the first step differentiation culture medium, and adding fresh second step differentiation culture medium. The second step differentiation medium is a basal differentiation medium supplemented with VEGF, bFGF, and one or more cytokines selected from the group consisting of BMP signaling pathway activators, Noda inhibitors, SCF, Flt3L, IL15, IL3, IL6, insulin, IGF-1, and TPO. BMP signaling pathway activators were selected similarly to D0; the Noda inhibitor is selected from the group consisting of Lefty-A, Lefty-B, Lefty-1, Lefty-2, SB431542, SB202190, SB505124, NPC30345, SD093, SD908, SD208, LY2109761, LY364947, LT580276, A83-01, and derivatives thereof. For example, when the culture is carried out using a T25 flask, wherein the BMP signaling pathway activator is BMP4 at a concentration of 0-200 ng/mL; the Noda inhibitor is SB431542 at a concentration of 0-20 μ M; the cytokine comprises insulin, IGF-1, VEGF, bFGF, IL3 and IL6, and the concentration is 0-10. mu.g/mL, 0-100ng/mL, 5-100ng/mL, 0-20ng/mL and 0-20ng/mL respectively.
The fluid change time point of the differentiation medium of the second step may be between Day2 and Day 4.
3) Day 3-6: changing into a third step differentiation culture medium
Taking out the culture bottle filled with EB from the culture box, inclining the culture bottle to make EB sink to the bottom, removing the supernatant of the differentiation culture medium of the second step, and adding fresh differentiation culture medium of the third step. The third step is to add growth factor and colony stimulating factor into the basic differentiation culture medium. The growth factor is one or more selected from EGF, VEGF, bFGF, insulin, IGF-1, PGF, PDGF and the like. The colony stimulating factor is one or more selected from the group consisting of G-CSF, M-CSF, GM-CSF, Multi-CSF (IL-3), EPO, TPO, SCF, Flt-3L, and the like. For example, when the culture is carried out using T25 flask, wherein the growth factors are insulin, VEGF, IGF-1 and bFGF, at concentrations of 0.5-10. mu.g/mL, 5-100ng/mL, 0-100ng/mL and 0.5-100ng/mL, respectively; colony stimulating factors are TPO, SCF and Flt-3L at concentrations of 0-100ng/mL, 0-100ng/mL and 0-100ng/mL, respectively;
the liquid changing time point of the differentiation medium of the third step may be between Day3 and Day 6.
4) Day 6-12: detecting the obtained hematopoietic progenitor cells
The diameters of the embryoid bodies were measured on days 0, 5 and 12 of the differentiation culture.
Selecting a proper time point during the period from Day6 to Day12, detecting the cell phenotype of the hematopoietic progenitor cells in the embryoid bodies by using a flow cytometer, and proving that the obtained cells contain the hematopoietic progenitor cells expressing CD34, and the percentage of the hematopoietic progenitor cells which are positive to CD34 in the total cells is between 20% and 80%.
Wherein, the information of the flow-type antibody is as follows:
APC Mouse Anti-Human CD34 Clone 581(RUO),BD,#555824;
FITC anti-human CD45,Biolegend,#304006。
(III) 1-2 weeks after EB inoculation (Day6-12 to Day 20-26): differentiation of hematopoietic progenitors into NK cells
The above-mentioned EB containing CD34+ hematopoietic progenitor cells of Day6-12 was resuspended in differentiation medium of the fourth step, inoculated in a cell matrix-coated flask and cultured for 2 weeks, and the medium was changed at the end of week 1 and 2 to maintain the following factors for different periods of action. NK cells containing high CD3-CD56+ were harvested at the end of week 2 (Day 20-26).
Details of experimental procedures at 1-2 weeks after EB inoculation (Day6-12 to Day 20-26):
1) day 6-12: replacing with the fourth step differentiation medium
The flasks were tilted to sink the EBs to the bottom, the third step differentiation medium supernatant was removed, and the EBs were resuspended in fresh fourth step differentiation medium. The fourth step is to add colony stimulating factors and interleukins into the basic differentiation medium. The colony stimulating factor is one or more selected from G-CSF, M-CSF, GM-CSF, multi-CSF (IL-3), EPO, TPO, SCF, Flt-3L, etc. The interleukin is one or more selected from IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21, IL-27, etc. For example, when cultured using T25 flasks, the colony stimulating factors are TPO, SCF, Flt-3L and IL-3 at concentrations of 0-100ng/mL, 5-100ng/mL and 5-100ng/mL, respectively, for a period of time of 1 week, 2 weeks or 1-2 weeks after inoculation. The interleukins are IL-2, L-7 and IL-15 at concentrations of 100-1000IU/mL, 5-100ng/mL and 5-100ng/mL, respectively, for periods of time 1 week, 2 weeks or 1-2 weeks after EB inoculation. The liquid changing time point of the differentiation medium of the fourth step may be between Day6 and Day 12.
The EBs resuspended in the fourth step differentiation medium were inoculated at the appropriate density into medium-protein-coated culture flasks. The matrix protein is at least one of the following components: notch pathway activator proteins and integrins. The Notch pathway activating protein is DLL1 recombinant protein, DLL4 recombinant protein, Jagged-1 recombinant protein, Jagged-2 recombinant protein, and variants of the Notch pathway activating proteins. The integrin is Fibronectin (Fibronectin), Laminin (Laminin), Vitronectin (Vitronectin), MAdCAM-1 (adhesion molecule for adhesion cell-1), VCAM-1 (adhesion molecule for vascular cell-1), ICAM (intercellular adhesion molecule), and variants thereof. For example, when cultured using T25 flasks, the Notch pathway activator protein is a DLL4-Fc recombinant protein. The integrin is VCAM-1.
Half of the fresh fourth step medium was replaced every 3-7 days depending on the cell density.
2) The NK cells obtained were examined 2 weeks after EB inoculation (Day20-26)
At 2 weeks after EB inoculation, the suspension cells in the wells were collected. Detecting the expression of the cell surface related index protein by flow cytometry. The detection indexes comprise: CD56, NKp30, NKp44, NKp 46.
Wherein, the information of the flow-type antibody is as follows:
PE Mouse Anti-Human CD56 Clone B159(RUO),BD,#555516;
Alexa
Figure BDA0002403196380000091
647 Mouse anti-Human CD337(NKp30),BD,#558408;
Alexa
Figure BDA0002403196380000092
647 Mouse Anti-Human NKp44(CD336),BD,#558564;
APC Mouse Anti-Human CD335(NKp46),BD,#558051。
3) 3-4 weeks after EB inoculation (Day 27-Day 40): details of experimental procedures 3-4 weeks after NK cell maturation and expansion EB inoculation (Day 27-Day 40):
after two weeks of EB differentiation, the cell density reached (1-2). times.106Per mL, collecting cells, centrifuging to remove the fourth step differentiation medium at (0.5-1). times.106Cell density of one/mL was resuspended in fresh differentiation medium of the fifth step. The fifth step is to add interleukin and other substances for promoting NK cell maturation and expansion into the basic differentiation medium. The interleukin is one or more selected from IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21, IL-27, etc. Other substances promoting NK cell maturation and expansion are selected from human AB plasma, human platelet lysate, Vitamin A, Nicotinamide (NAM, Vitamin B3), Vitamin E and Heparin, etc. For example, when the culture is carried out using a T25 flask, the interleukins are IL-2, IL-12, IL-18, IL-21 and IL-27 and IL-15 at concentrations of 100-1000IU/mL, 0-100ng/mL and 5-100ng/mL, respectively; the additive substances for promoting NK cell maturation and amplification are human platelet lysate, Nicotinamide (NAM), Vitamin E and Heparin, and the concentrations are 1-10%, 1-10mmol/L, 0-10mg/mL and 0-100 μ g/mL respectively.
The NK cells obtained at 3-4 weeks (Day27-40) after EB inoculation were examined
Suspension cells in wells were collected at 3-4 weeks of EB differentiation (Day 27-40). Detecting the expression of the cell surface related index protein by flow cytometry. The detection indexes comprise: CD56, CD94, and CD 16.
Wherein, the information of the flow-type antibody is as follows:
PE Mouse Anti-Human CD56 Clone B159(RUO),BD,#555516;
APC Mouse Anti-Human CD94,BD,#559876;
APC Mouse Anti-Human CD16 Clone B73.1(RUO),BD,#561304。
in one embodiment of the present invention, the human pluripotent stem cells are prepared by the method disclosed in patent CN 108085299 a.
It is understood that one skilled in the art can select any commercial cell line or cell strain of human pluripotent stem cells as required to complete the present invention and all fall within the scope of the present invention.
In the invention:
1. the basic differentiation medium comprises the following components: IMDM, F-12, rHSA (recombinant Human serum albumin), MTG-Monothioglycerol (thioglycerol), Ascorbic Acid (Ascorbic Acid), Human Transferrin (Human Transferrin), Na Selenite (sodium Selenite), and Ethanolamine (Ethanolamine). Wherein, IMDM and F12 can be used as the basis for developing serum-free formula, and the culture medium is suitable for mammalian cell culture under the condition of low serum content; in the formula of the culture medium, IMDM and F-12 are mixed in a ratio of 1: 1. The combination and concentration of the other components is optimized, the supplement has the function of replacing serum, and the supplement is an additive for maintaining differentiation to blood progenitor cells and maintaining proliferation of differentiated cells.
GSK-3 inhibitors: substances which block the kinase activity of the GSK3 β protein, such as the GSK3 β inhibitor IX (6-bromoindirubin 3' -oxime), SB216763, the GSK3 β inhibitor VII (4-dibromoacetophenone), L803-mts, and CHIR99021 with high selectivity. CHIR99021 is preferred in the present invention.
The concentration of CHIR99021 in the medium is not particularly limited as long as it inhibits the kinase activity of GSK3 beta protein, but is not limited to, for example, 0.1. mu.M, 0.2. mu.M, 0.3. mu.M, 0.4. mu.M, 0.5. mu.M, 0.6. mu.M, 0.7. mu.M, 0.8. mu.M, 0.9. mu.M, 1. mu.M, 1.1. mu.M, 1.2. mu.M, 1.3. mu.M, 1.4. mu.M, 1.5. mu.M, 2. mu.M, 2.5. mu.M, 3. mu.M, 4. mu.M, 5. mu.M, 6. mu.M, 8. mu.M, or 10. mu.M. Preferably 1 to 8 μ M.
BMP signaling pathway activator: examples of the substance which activates the BMP signaling pathway include BMP2, BMP4, SB4, ventromorphins (SJ000291942, SJ000063181, SJ000370178), isooliquitinin, diosmetin, apigenin, biochaninA, etc. The BMP activator used in the present invention is BMP 4. The concentration of BMP4 in the medium is not particularly limited as long as it activates a BMP signaling pathway, and examples thereof include, but are not limited to, 5ng/ml, 10ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, 100ng/ml, 110ng/ml, 120dng/ml, 130ng/ml, 140ng/ml, 150ng/ml, 160ng/ml, 180ng/ml and 200 ng/ml. Preferably 5 to 50 ng/ml.
Nodal inhibitors: substances that inhibit Nodal signaling pathways. Lefty-A, Lefty-B, Lefty-1, Lefty-2, SB431542, SB202190, SB505124, NPC30345, SD093, SD908, SD208, LY2109761, LY364947, LT580276, A83-01, and derivatives thereof are selected. The Nodal inhibitor used in the present invention is SB 431542. The concentration of SB431542 in the medium is not particularly limited as long as it blocks the Nodal signaling pathway, and examples thereof include, but are not limited to, 0. mu.M, 1. mu.M, 2. mu.M, 3. mu.M, 4. mu.M, 5. mu.M, 6. mu.M, 7. mu.M, 8. mu.M, 9. mu.M, 10. mu.M, 11. mu.M, 12. mu.M, 13. mu.M, 14. mu.M, 15. mu.M, 20. mu.M, 25. mu.M, and 30. mu.M. Preferably 1-20. mu.M.
5. Colony stimulating factor: cytokines capable of stimulating hematopoietic stem cell proliferation and differentiation. G-CSF, M-CSF, GM-CSF, multi-CSF (IL-3), EPO, TPO, SCF, Flt-3L, and the like may be selected. Colony stimulating factors used in the present invention are G-CSF, GM-CSF, TPO, SCF and Flt-3L and multi-CSF. The concentrations of TPO, SCF and Flt-3L in the medium are not particularly limited as long as they stimulate the proliferation and differentiation of hematopoietic stem cells, and examples thereof include, but are not limited to, TPO at a concentration of 0ng/ml, 5ng/ml, 10ng/ml, 15ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml and 100 ng/ml. Preferably 0 to 100 ng/ml. SCF concentrations of 0ng/ml, 10ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 80ng/ml, 100ng/ml, 150ng/ml, 200ng/ml, but are not limited thereto. Preferably 0 to 100 ng/ml. The concentration of Flt-3L is 0ng/ml, 1ng/ml, 5ng/ml, 10ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 80ng/ml, 100ng/ml, 150ng/ml, 200ng/ml, but is not limited thereto. Preferably 0 to 100 ng/ml.
Notch signaling pathway activators: recombinant proteins that activate the Notch signaling pathway. The recombinant DLL1 protein, the recombinant DLL4 protein, the recombinant Jagged-1 protein, the recombinant Jagged-2 protein, and variants of these Notch pathway activating proteins, etc. can be selected. The Notch activator used in the present invention is DLL4-Fc recombinant protein. The density of the coating of the coated DLL4-Fc recombinant protein is not particularly limited as long as it can activate the Notch signaling pathway, and for example, the density of the coating of the DLL4-Fc recombinant protein is 0. mu.g/cm2、0.1μg/cm2、0.2μg/cm2、0.3μg/cm2、0.4μg/cm2、0.5μg/cm2、0.6μg/cm2、0.7μg/cm2、0.8μg/cm2、0.9μg/cm2、1μg/cm2、2μg/cm2、3μg/cm2、4μg/cm2、5μg/cm2But is not limited thereto. Preferably 0.5 to 2 mu g/cm2
7. Growth factor: a native protein that stimulates cell proliferation and cell differentiation. EGF, VEGF, bFGF, insulin, IGF-1, PGF, PDGF, and the like may be selected. The growth factors used in the present invention are insulin, VEGF, IGF-1 and bFGF. The concentration of the insulin, VEGF, and bFGF in the medium is not particularly limited as long as it can stimulate cell proliferation and cell differentiation, and examples thereof include concentrations of insulin of 0. mu.g/ml, 0.5. mu.g/ml, 1. mu.g/ml, 2. mu.g/ml, 3. mu.g/ml, 4. mu.g/ml, 5. mu.g/ml, 6. mu.g/ml, 7. mu.g/ml, 8. mu.g/ml, 9. mu.g/ml, 10. mu.g/ml, 20. mu.g/ml, and 30. mu.g/ml. Preferably 0 to 10. mu.g/ml. VEGF concentrations are 5ng/ml, 10ng/ml, 15ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, 100ng/ml, 150ng/ml, 200ng/ml, but are not so limited. Preferably 5 to 100 ng/ml. The concentration of IGF-1 is 5ng/ml, 10ng/ml, 15ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, 100ng/ml, 150ng/ml, 200 ng/ml. Preferably 0 to 100. mu.g/ml. The concentration of bEGF is 5ng/ml, 10ng/ml, 15ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, 100ng/ml, 150ng/ml, 200ng/ml, but is not limited thereto. Preferably 0 to 100 ng/ml.
8. Interleukin (IL): cytokines that mediate immune cell activation, proliferation and differentiation. One or more of IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21, IL-27, etc. may be selected at different differentiation stages.
The interleukins used in the present invention D6-12 to D20-26 (i.e., within two weeks after EB inoculation) were IL-2, IL-7 and IL-15. The concentration of IL-2, IL-7 and IL-15 in the medium is not particularly limited as long as it can mediate immune cell activation, proliferation and differentiation, and examples thereof include, but are not limited to, IL-2 concentrations of 0IU/ml, 50IU/ml, 100IU/ml, 200IU/ml, 300IU/ml, 400IU/ml, 500IU/ml, 700IU/ml, 800IU/ml, 1000IU/ml and 2000 IU/ml. Preferably 0 to 1000 IU/ml. IL-7 concentrations are, but not limited to, 10ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, 100ng/ml, 150ng/ml, 200 ng/ml. Preferably 5 to 100 ng/ml. IL-15 concentrations are, but not limited to, 1ng/ml, 5ng/ml, 10ng/ml, 15ng/ml, 20ng/ml, 25ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 80ng/ml, 100ng/ml, 150ng/ml, 200 ng/ml. Preferably 5 to 100 ng/ml.
ROCK inhibitor: a substance which inhibits Rho kinase (ROCK) function. For example: y-27632, HA100, HA1152, and Blebbistatin. Y-27632 is preferred in the present invention.
The concentration of Y-27632 in the medium is not particularly limited as long as it is a concentration that inhibits Rho kinase, and examples thereof include, but are not limited to, 1. mu.M, 1.5. mu.M, 2. mu.M, 2.5. mu.M, 3. mu.M, 3.5. mu.M, 4. mu.M, 4.5. mu.M, 5. mu.M, 5.5. mu.M, 6. mu.M, 6.5. mu.M, 7. mu.M, 7.5. mu.M, 8. mu.M, 9. mu.M, 10. mu.M, 15. mu.M, 20. mu.M, 30. mu.M, and 50. Preferably 10. mu.M.
In a third aspect, the invention discloses a natural killer cell prepared by the above method.
In a fourth aspect, the invention discloses a cell population enriched for the above natural killer cells.
In the fifth aspect of the invention, the invention discloses a medicament for preventing and/or treating tumors, which comprises the natural killer cells.
The key points and points to be protected of the invention are as follows:
1) based on the rapid, efficient and low-cost EB 3D differentiation method (including the types, combination modes, adding time, concentration and the like of the small molecular compounds and the cytokines) combined by the small molecular compounds and the cytokines, the method has the advantages of simpler operation, more stable result and lower cost, and is beneficial to large-scale iNK production.
2) NK cell differentiation and expansion media components. The key innovation points are as follows: the components of the differentiation and amplification culture medium are clear, NK cells can be successfully differentiated and amplified under the condition of not using serum and trophoblast cells, and the method is suitable for production and clinical application of large-scale cell preparations.
3) The cryopreservation condition is optimized, the NK cells can be stored for a long time, and higher cell activity can be maintained after recovery.
On the basis of the common general knowledge in the field, the above-mentioned preferred conditions can be combined arbitrarily without departing from the concept and the protection scope of the invention.
Compared with the prior art, the invention has the following remarkable advantages and effects:
first, the method uses 3D suspension culture during induced differentiation, which can be performed in a short period (about 40 days) of 1X 105The hiPSC gain of is greater than 1 x 107The NK cells of (1), are suitable for large-scale cell preparation production;
secondly, the differentiation process is finely regulated and controlled by the combined use of small molecular compounds and cytokines, so that stable and efficient differentiation is realized, and the proportion of CD56+ NK cells in a final culture system can reach more than 90%;
thirdly, the NK cell prepared by the method has similar cell phenotype and functionality with PB-NK cell, and the preparation cost is low, so that the NK cell has great clinical and scientific research application potential;
fourth, the method uses serum-free media and no feeder cells during both induction and expansion, and is suitable for the production and use of subsequent clinical-grade cell preparations.
Drawings
FIG. 1 shows a flow chart for preparing natural killer cells disclosed in the present invention.
FIG. 2 shows the process of induced differentiation from pluripotent stem cells into mature NK cells in the method disclosed in the present invention (day-1, day 0, day 5, day12, after 2 weeks of EB inoculation and after 4 weeks of EB inoculation, respectively).
FIG. 3 shows that the specificity of D8 EB was measured by flow cytometry, which indicates that hematopoietic stem cells (CD34+) reached a high level.
FIG. 4 shows that the specificity indexes of iNK cells at 2 weeks and 4 weeks after EB inoculation are detected by a flow cytometer, and that iNK cells which are CD56 positive at 2 weeks after EB inoculation reach more than 80% and simultaneously highly express Nkp30, Nkp44 and Nkp 46. iNK cells simultaneously expressed specific indexes of mature NK cells, CD94 and CD16, at 4 weeks after EB inoculation, indicating that NK cells have matured.
FIG. 5 shows that iNK cells harvested at week 4 after EB inoculation produced a significant cytotoxic effect on target cell K562, with a cell killing capacity similar to that of PB-NK.
FIG. 6 shows that the concentration of CHIR99021 in the first step differentiation medium affects the differentiation efficiency of CD34+ hematologic progenitors.
FIG. 7 shows that the time for the second differentiation medium exchange affects the differentiation efficiency of CD34+ blood progenitor cells.
FIG. 8 shows that the time for the third differentiation medium exchange affects the differentiation efficiency of CD34+ blood progenitor cells.
FIG. 9 shows that EB seeding time points affect the differentiation efficiency of NK cells (CD3-CD56 +).
FIG. 10 shows that the effect concentration of BMP4 affects the differentiation efficiency of CD34+ blood progenitor cells.
FIG. 11 shows that the concentration of SB affects the differentiation efficiency of CD34+ blood progenitor cells.
FIG. 12 shows that the concentration of bFGF acting affects the differentiation efficiency of CD34+ blood progenitor cells.
Detailed Description
The technical solution of the present invention is described in detail below with reference to the drawings and the embodiments, but the present invention is not limited to the scope of the embodiments.
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. The reagents and starting materials used in the present invention are commercially available.
Example 1
This example discloses a method for preparing natural killer cells, comprising the steps of:
s1: forming an embryoid body;
s2: the embryoid body is differentiated into hematopoietic progenitor cells;
s3: differentiating hematopoietic progenitor cells into NK cells;
s4: maturation and expansion of NK cells.
Specifically, the flow chart is shown in fig. 1, and the steps are as follows:
first, Day-1 to Day 0: formation of Embryoid Bodies (EB)
1) Culture of human pluripotent stem cells (iPSCs)
The human ipscs used in the experiments were subjected to strict pluripotency validation (expressing various pluripotency markers and forming teratomas comprising three germ layers, inner, middle and outer, in immunodeficient mice). The ipscs are normally cultured in iPSC maintenance medium, and the medium used is E8 or TeSR or other similar medium.
The human iPSC was prepared by the method disclosed in patent CN 108085299 a.
2) Formation of EB
The formation experiment of the embryoid body is carried out when the iPSC is cultured to 70-90% of polymerization degree according to the method. The specific operation is as follows: ipscs were digested to a complete single cell suspension using TrypLE or accutase, resuspended in iPSC maintenance medium, and Rock inhibitors were added to the medium. Placing the cell suspension on a 3D shaking table in an incubator at 37 ℃ for shaking culture, wherein the rotation speed of the shaking table is 10-100 rpm; the shaking culture time in this step is 8-32 hours; at the end of the culture, EBs of more uniform size and morphology were obtained. When T25 flask is used, the Rock inhibitor is Y-27632 at a concentration of 10. mu.M; the cell density was 0.1X 106-5×106Per mL; the rotating speed of the shaking table is 10-20 rpm; the culture time is 8-32 hours.
(II) Day 0 to Day 6-12: differentiation of pluripotent stem cells into hematopoietic progenitor cells
1) Day 0: replacing with the first step differentiation culture medium
The flask containing the D0 EB was removed from the incubator, the flask was tilted to allow the EB to settle to the bottom, the supernatant was removed, and fresh first step differentiation medium was added. The first step differentiation culture medium is to add a small molecule GSK3 beta inhibitor and one or more components selected from the following components in a basic differentiation culture medium: BMP signaling pathway activator, VEGF, bFGF, SCF, Flt3L, IL3, IL6, insulin, IGF-1 and TPO. BMP signal pathway activators are BMP2, BMP4, SB4, ventromorphins (SJ000291942, SJ000063181, SJ000370178), isoliquiritigenin, diosmetin (diosmin), apigenin (apigenin), biochanin (biochanin), and the like; the GSK3 beta inhibitor is NP031112, TWS119, SB216763, CHIR-98014, AZD2858, AZD1080, SB415286, LY2090314, CHIR99021, etc. For example, when the culture is carried out using T25 flask, in which the GSK3 beta inhibitor is CHIR99021, at a concentration of 0.5-20. mu.M; the BMP signal pathway activator is BMP4, and the concentration is 0-200 ng/mL; the cytokine comprises insulin, IGF-1, VEGF, bFGF, the concentration is 0-10 mug/mL, 0-100ng/mL, 5-100ng/mL and 0-100ng/mL respectively.
Wherein Isooliquitinigenin is a flavonoid compound separated from Glycyrrhiza glabra root, and has antitumor activity.
The time for culturing the EBs in the first-step differentiation medium was (2-4) days.
The composition of the basal differentiation medium is shown in Table 2 below.
TABLE 2
Figure BDA0002403196380000151
2) Day 2-4: replacing with the second step differentiation culture medium
Taking out the culture bottle filled with EB from the culture box, inclining the culture bottle to make EB sink to the bottom, removing the supernatant of the first step differentiation culture medium, and adding fresh second step differentiation culture medium. The second step differentiation medium is a basal differentiation medium supplemented with VEGF, bFGF, and one or more cytokines selected from the group consisting of BMP signaling pathway activators, Noda inhibitors, SCF, Flt3L, IL15, IL3, IL6, insulin, IGF-1, and TPO. BMP signaling pathway activators were selected similarly to D0; the Noda inhibitor is selected from the group consisting of Lefty-A, Lefty-B, Lefty-1, Lefty-2, SB431542, SB202190, SB505124, NPC30345, SD093, SD908, SD208, LY2109761, LY364947, LT580276, A83-01, and derivatives thereof. When the culture is carried out by using a T25 culture flask, wherein the BMP signal pathway activator is BMP4, and the concentration is 0-200 ng/mL; the Noda inhibitor is SB431542 at a concentration of 0-20 μ M; the cytokine comprises insulin, IGF-1, VEGF, bFGF, IL3 and IL6, and the concentration is 0-10. mu.g/mL, 0-100ng/mL, 5-100ng/mL, 0-20ng/mL and 0-20ng/mL respectively.
The fluid change time point of the differentiation medium of the second step may be between Day2 and Day 4.
3) Day 3-6: changing into a third step differentiation culture medium
Taking out the culture bottle filled with EB from the culture box, inclining the culture bottle to make EB sink to the bottom, removing the supernatant of the differentiation culture medium of the second step, and adding fresh differentiation culture medium of the third step. The third step is to add growth factor and colony stimulating factor into the basic differentiation culture medium. The growth factor is one or more selected from EGF, VEGF, bFGF, insulin, IGF-1, PGF, PDGF and the like. The colony stimulating factor is one or more selected from the group consisting of G-CSF, M-CSF, GM-CSF, Multi-CSF (IL-3), EPO, TPO, SCF, Flt-3L, and the like. For example, when the culture is carried out using T25 flask, wherein the growth factors are insulin, VEGF, IGF-1 and bFGF, at concentrations of 0.5-10. mu.g/mL, 5-100ng/mL, 0-100ng/mL and 0.5-100ng/mL, respectively; colony stimulating factors are TPO, SCF and Flt-3L at concentrations of 0-100ng/mL, 0-100ng/mL and 0-100ng/mL, respectively;
the liquid changing time point of the differentiation medium of the third step may be between Day3 and Day 6.
4) Day 6-12: detecting the obtained hematopoietic progenitor cells
The diameters of the embryoid bodies at day 0, day 5 and day12 of the differentiation culture were 50-120. mu.M, 300-400. mu.M and 400-650. mu.M, respectively. The specific cell morphology is shown in FIG. 3.
Selecting a proper time point during the period from Day6 to Day12, detecting the cell phenotype of the hematopoietic progenitor cells in the embryoid bodies by using a flow cytometer, and proving that the obtained cells contain the hematopoietic progenitor cells expressing CD34, and the percentage of the hematopoietic progenitor cells which are positive to CD34 in the total cells is between 20% and 80%. See figure 5 for specific data.
Wherein, the information of the flow-type antibody is as follows:
APC Mouse Anti-Human CD34 Clone 581(RUO),BD,#555824;
FITC anti-human CD45,Biolegend,#304006。
(III) 1-2 weeks after EB inoculation (Day6-12 to Day 20-26): differentiation of hematopoietic progenitors into NK cells
The above-mentioned EB containing CD34+ hematopoietic progenitor cells of Day6-12 was resuspended in differentiation medium of the fourth step, inoculated in a cell matrix-coated flask and cultured for 2 weeks, and the medium was changed at the end of week 1 and 2 to maintain the following factors for different periods of action. NK cells containing high CD3-CD56+ were harvested at the end of week 2 (Day 20-26).
Details of experimental procedures at 1-2 weeks after EB inoculation (Day6-12 to Day 20-26):
1) day 6-12: replacing with the fourth step differentiation medium
The flasks were tilted to sink the EBs to the bottom, the third step differentiation medium supernatant was removed, and the EBs were resuspended in fresh fourth step differentiation medium. The fourth step is to add colony stimulating factors and interleukins into the basic differentiation medium. The colony stimulating factor is one or more selected from G-CSF, M-CSF, GM-CSF, multi-CSF (IL-3), EPO, TPO, SCF, Flt-3L, etc. The interleukin is one or more selected from IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21, IL-27, etc. When cultured using T25 flasks, the colony stimulating factors were TPO, SCF, Flt-3L and IL-3 at concentrations of 0-100ng/mL, 5-100ng/mL and 5-100ng/mL, respectively, for a period of time of 1 week, 2 weeks or 1-2 weeks after inoculation. The interleukins are IL-2, L-7 and IL-15 at concentrations of 100-1000IU/mL, 5-100ng/mL and 5-100ng/mL, respectively, for periods of time 1 week, 2 weeks or 1-2 weeks after EB inoculation. The liquid changing time point of the differentiation medium of the fourth step may be between Day6 and Day 12.
The EBs resuspended in the fourth step differentiation medium were inoculated at the appropriate density into medium-protein-coated culture flasks. The matrix protein is at least one of the following components: notch pathway activator proteins and integrins. The Notch pathway activating protein is DLL1 recombinant protein, DLL4 recombinant protein, Jagged-1 recombinant protein, Jagged-2 recombinant protein, and variants of the Notch pathway activating proteins. The integrin is Fibronectin (Fibronectin), Laminin (Laminin), Vitronectin (Vitronectin), MAdCAM-1 (adhesion molecule for adhesion cell-1), VCAM-1 (adhesion molecule for vascular cell-1), ICAM (intercellular adhesion molecule), and variants thereof. For example, when cultured using T25 flasks, the Notch pathway activator protein is a DLL4-Fc recombinant protein. The integrin is VCAM-1.
Half of the fresh fourth step medium was replaced every 3-7 days depending on the cell density.
2) The NK cells obtained were examined 2 weeks after EB inoculation (Day20-26)
At 2 weeks after EB inoculation, the suspension cells in the wells were collected. Detecting the expression of the cell surface related index protein by flow cytometry. The detection indexes comprise: CD56, NKp30, NKp44, NKp 46.
Wherein, the information of the flow-type antibody is as follows:
PE Mouse Anti-Human CD56 Clone B159(RUO),BD,#555516;
Alexa
Figure BDA0002403196380000171
647 Mouse anti-Human CD337(NKp30),BD,#558408;
Alexa
Figure BDA0002403196380000172
647 Mouse Anti-Human NKp44(CD336),BD,#558564;
APC Mouse Anti-Human CD335(NKp46),BD,#558051。
3) 3-4 weeks after EB inoculation (Day 27-Day 40): maturation and expansion of NK cells
Details of experimental procedures 3-4 weeks after EB inoculation (Day 27-Day 40):
after two weeks of EB differentiation, the cell density reached (1-2). times.106Per mL, collecting cells, centrifuging to remove the fourth step differentiation medium at (0.5-1). times.106Cell density of one/mL was resuspended in fresh differentiation medium of the fifth step. The fifth step is to add interleukin and other substances for promoting NK cell maturation and expansion into the basic differentiation medium. The interleukin is one or more selected from IL-1, IL-2, IL-6, IL-7, IL-12, IL-15, IL-18, IL-21, IL-27, etc. Other substances promoting NK cell maturation and expansion are selected from human AB plasma, human platelet lysate, Vitamin A, Nicotinamide (NAM, Vitamin B3), Vitamin E and Heparin, etc. For example, when the culture is carried out using a T25 flask, the interleukins are IL-2, IL-12, IL-18, IL-21 and IL-27 and IL-15 at concentrations of 100-1000IU/mL, 0-100ng/mL and 5-100ng/mL, respectively; the additive substances for promoting NK cell maturation and amplification are human platelet lysate, Nicotinamide (NAM), Vitamin E and Heparin, and the concentrations are 1-10%, 1-10mmol/L, 0-10mg/mL and 0-100 μ g/mL respectively.
The NK cells obtained were examined at 4 weeks (Day27-40) after EB inoculation
At the 4 th week of EB differentiation (Day27-40), the suspension cells in the wells were collected. Detecting the expression of the cell surface related index protein by flow cytometry. The detection indexes comprise: CD56, CD94, and CD 16.
Wherein, the information of the flow-type antibody is as follows:
PE Mouse Anti-Human CD56 Clone B159(RUO),BD,#555516;
APC Mouse Anti-Human CD94,BD,#559876;
APC Mouse Anti-Human CD16 Clone B73.1(RUO),BD,#561304。
specific cell morphology patterns at day-1, day 0, day 5, day12, week 2 after EB inoculation, and week 4 after EB inoculation in differentiation culture are shown in fig. 2.
Example 2
This example investigates the effect of different EB culture methods on EB differentiation rates. In the experimental group, a culture method of forming an embryoid body through 3D suspension culture is adopted in the whole induced differentiation process, namely, iPSC is directly formed into EB with a smaller volume on a 3D shaking table, and then the EB is changed into different special differentiation culture media at different differentiation stages (the specific steps are shown in example 1). During the culture process, the volume of EB increases gradually and the cells proliferate continuously. Compared with a 2D differentiation scheme (see patent CN107429230A), the 3D differentiation condition greatly saves culture space and culture volume, and the number of cells obtained in the same culture system is remarkably increased, thereby being beneficial to the large-scale production of the hPSC to the multipotential blood precursor cells. Compared with the differentiation scheme of Spin EB (US20180072992A1) and a method for further differentiation after sorting and purifying hematopoietic progenitor cells in EB cells in the literature, the 3D differentiation scheme of the invention is simpler and easier, has higher efficiency, and is more suitable for clinical-level large-scale production. Meanwhile, the present protocol directly uses EBs from day6 to day12 for differentiation, and obtains higher differentiation efficiency (the proportion of NK cells positive to CD56 reaches more than 90% at the fourth cycle of EB differentiation), and the results are shown in FIG. 4.
Example 3
The invention discovers that in the 3D induction culture, the small molecular reagent, the cell factor and the cell matrix can replace serum and trophoblast cells, accelerate the differentiation process, improve the differentiation efficiency and be beneficial to the production of the subsequent clinical cell preparation.
In our differentiation method, in addition to the use of 3D to increase differentiation efficiency, the combined use of small molecule agents, cytokines and cell matrices not only avoids the use of serum and trophoblast cells, but also further increases the proportion of blood precursor cells. The GSK3 β inhibitor selected CHIR99021, BMP signaling pathway activator used BMP4, Nodal inhibitor used SB431542, and Notch signaling pathway activator in the cell matrix used DLL4-Fc recombinant protein. On days 6-12, the proportion of blood progenitor cells (CD34+) obtained can be as high as 20-80%, which is higher than the proportion of positive cells obtained by differentiation method using serum-containing medium and trophoblast cells reported in the literature (for example, in patent CN102822332A, the proportion of CD56+ cells only reaches 20-30%).
Example 4
The invention finds that the cell density and the culture time of the initial human iPSC can influence the size of EB formation, thereby influencing the efficiency of 3D differentiation. On day 0 of differentiation, we digested human ipscs into single cells using accutase (cell digest), initial cell density was controlled at 0.1-5 million cells/T25, hpscs were maintained in medium for 8-32 hours.
Example 5
The invention discovers that the efficiency of differentiating pluripotent stem cells into blood progenitor cells can be obviously improved by optimizing the concentration of CHIR99021 in the differentiation medium of the first step.
When EB differentiation was performed using T25 flask, other conditions were as described in example 1, and concentration of CHIR99021 in the first differentiation step was gradient-optimized by treating cells with 0. mu.M, 1. mu.M, 2. mu.M, 4. mu.M, 6. mu.M, 8. mu.M or 10. mu.M of CHIR99021, respectively, and measuring CD34, which is an index of blood progenitor cells, using a flow cytometer at D12, and the results are shown in FIG. 6.
It can be seen that the addition of optimized CHIR99021 increases the efficiency of differentiation of blood progenitor cells; at lower concentrations, the differentiation rate of blood progenitor cells is low, and at higher concentrations, the differentiation of blood progenitor cells is inhibited; optimizing the concentration of CHIR99021 can improve the purity of the blood progenitor cells at D6-12, namely, the proportion of CD34+ cells is improved; the concentration of CHIR99021 is preferably 1-8 μ M.
Example 6
The invention discovers that the efficiency of differentiating pluripotent stem cells into blood progenitor cells can be obviously improved by optimizing the liquid changing time point of the differentiation culture medium of the second step.
When EB differentiation was performed using T25 flask, other condition methods were as described in example 1, the point of time for changing to the second-step differentiation medium was optimized, i.e., differentiation was continued by changing to the second-step differentiation medium at D1, D2, D2.5, D3, D3.5, D4 or D5, and the index of blood progenitor cells, CD34, was measured using flow cytometry at D12, and the results are shown in FIG. 7. Therefore, the proportion of the obtained blood progenitor cells can be obviously improved by optimizing the liquid changing time point of the differentiation culture medium of the second step; the proportion of blood progenitor cells that differentiate is low when fluid is changed at an earlier or later time point; the liquid change time point of the differentiation medium in the second step is preferably D2 to D4.
Example 7
The invention discovers that the efficiency of differentiating pluripotent stem cells into blood progenitor cells can be obviously improved by optimizing the liquid changing time point of the differentiation culture medium in the third step.
When EB differentiation was performed using T25 flask, other condition method was as described in example 1, the time point for changing to the differentiation medium of the third step was optimized, i.e., differentiation was continued by changing D3, D4, D5, D6, D7 or D8 to the differentiation medium of the fourth step, and the index CD34 of blood progenitor cells was measured using flow cytometry at D12, and the result was shown in FIG. 8. It can be seen that the proportion of blood progenitor cells obtained can be significantly increased by optimizing the time point for changing to the differentiation medium of the third step; at later time points, the proportion of blood progenitor cells differentiated is low; the time point for changing the differentiation medium in the third step is preferably D3-D6.
Example 8
The invention discovers that the optimized EB inoculation time point can obviously improve the differentiation efficiency of NK cells.
In the case of EB differentiation using T25 flasks, other conditions were optimized as described in example 1, i.e., EBs were formed according to the above differentiation method, and EBs were removed on days 4, 6, 8, 10, 12 and 14 of differentiation, inoculated into matrix protein-coated plates, and subsequently differentiated. The proportion of differentiated NK cells (CD56+) was measured two weeks after inoculation and the results are shown in FIG. 9.
Therefore, optimizing the EB inoculation time point can improve the differentiation efficiency of NK cells; NK cell differentiation efficiency was low for EBs at early stage (day 4) and EBs at late stage (day 14) at time point; the preferred EB inoculation time points should be D6-D12.
Example 9
The invention finds that optimizing the action concentration of BMP4 can obviously improve the proportion of blood progenitor cells.
In EB differentiation using T25 flasks, other conditions were performed as described in example 1, and the concentration of BMP4 in the first and second differentiation media was gradient optimized by treating the cells with BMP4 at 0ng/ml, 10ng/ml, 20ng/ml, 30ng/ml, 40ng/ml, 50ng/ml, 60ng/ml, 70ng/ml, 80ng/ml, 90ng/ml, or 100ng/ml, respectively, and measuring the CD34, which is an index of blood progenitor cells, using a flow cytometer at D12, as shown in FIG. 10. It can be seen that the addition of optimized BMP4 increases the efficiency of differentiation of blood progenitor cells; at lower concentrations, the differentiation rate of blood progenitor cells is low, and at higher concentrations, the differentiation of blood progenitor cells is inhibited; optimizing the concentration of BMP4 can improve the purity of blood progenitor cells at D12, namely, the proportion of CD34+ cells is improved; the concentration of BMP4 is preferably 10-50 ng/ml.
Example 10
The invention discovers that the proportion of the obtained blood progenitor cells can be obviously improved by optimizing the acting concentration of SB 431542.
In EB differentiation using T25 flask, other conditions were as described in example 1, and the concentration of SB431542 in the differentiation medium of the second step was gradient optimized by treating cells with 0. mu.M, 2. mu.M, 4. mu.M, 6. mu.M, 8. mu.M, 10. mu.M, 20. mu.M, 30. mu.M, 40. mu.M or 50. mu.M of SB431542, respectively, and measuring the CD34 index of blood progenitor cells using flow cytometry at D12, and the results are shown in FIG. 11.
It can be seen that the addition of optimized SB431542 increases the efficiency of differentiation of blood progenitor cells, but at higher concentrations, differentiation of blood progenitor cells is inhibited; optimizing SB431542 concentration increases the purity of blood progenitor cells at D6-12, i.e., increases the proportion of CD34+ cells; the preferable concentration of SB431542 is 0-20 μ M.
Example 11
The invention discovers that the proportion of the obtained blood progenitor cells can be obviously improved by optimizing the action concentration of the bFGF.
In EB differentiation using T25 flasks, other conditions were optimized by gradient as described in example 1 for bFGF concentration in the first, second and third differentiation media, i.e., cells were treated with bFGF at 0ng/ml, 0.1ng/ml, 0.5ng/ml, 1ng/ml, 2.5ng/ml, 5ng/ml, 10ng/ml or 20ng/ml, respectively, and the CD34 index of blood progenitor cells was determined by flow cytometry at D6-12, as shown in FIG. 12.
It can be seen that the addition of optimized bFGF increases the efficiency of differentiation of blood progenitor cells, but at higher concentrations, differentiation of blood progenitor cells is inhibited; the optimized bFGF concentration can improve the purity of the blood progenitor cells at D6-12, namely, the proportion of CD34+ cells is improved; the preferable concentration of bFGF is 0.1-5 ng/ml.
The above embodiments are preferred embodiments of the present invention, but the present invention is not limited to the above embodiments, and any other changes, modifications, substitutions, combinations, and simplifications which do not depart from the spirit and principle of the present invention should be construed as equivalents thereof, and all such changes, modifications, substitutions, combinations, and simplifications are intended to be included in the scope of the present invention.

Claims (7)

1. A method of producing natural killer cells comprising the steps of:
s1: forming an embryoid body;
s2: the embryoid body is differentiated into hematopoietic progenitor cells;
wherein the S2 includes:
s21: removing the supernatant of the embryoid body, and adding a fresh first-step differentiation culture medium for cell culture;
wherein, the first step differentiation culture medium is obtained by adding a small molecule GSK3 beta inhibitor and at least one of the following components in a basic differentiation culture medium: BMP signal pathway activator, insulin, IGF-1, VEGF, bFGF; the GSK3 beta inhibitor is CHIR99021, and the concentration is 0.5-20 μ M; the BMP signal pathway activator is BMP4, and the concentration is 0-200 ng/mL; the concentrations of insulin, IGF-1, VEGF and bFGF are respectively 0-10 mug/mL, 0-100ng/mL, 5-100ng/mL and 0-100 ng/mL;
s22: removing the first step differentiation culture medium, and adding the second step differentiation culture medium for cell culture; wherein, the second step differentiation culture medium is prepared by adding VEGF, bFGF and at least one of the following components in a basic differentiation culture medium: BMP signaling pathway activators, Nodal inhibitors, insulin, IGF-1, IL3 and IL 6; wherein the concentration of VEGF and bFGF is respectively 5-100ng/mL, 0-100ng/mL and is not 0; the BMP signal pathway activator is BMP4, and the concentration is 0-200 ng/mL; the Nodal inhibitor is SB431542 at a concentration of 0-20 μ M; the concentrations of insulin, IGF-1, IL3 and IL6 are 0-10. mu.g/mL, 0-100ng/mL, 0-20ng/mL and 0-20ng/mL, respectively;
s23: removing the differentiation culture medium of the second step, and adding the differentiation culture medium of the third step for cell culture to obtain hematopoietic progenitor cells; wherein, the third step differentiation culture medium is to add growth factors and colony stimulating factors in a basic differentiation culture medium;
the growth factor is selected from one or more of Insulin, VEGF, IGF-1 and bFGF, wherein the concentrations of the Insulin, the VEGF, the IGF-1 and the bFGF are respectively 0.5-10 mug/mL, 5-100ng/mL, 0-100ng/mL and 0.5-100 ng/mL; the colony stimulating factor is selected from one or more of TPO, SCF and Flt-3L; wherein the concentrations of TPO, SCF and Flt-3L are 0-100ng/mL, 0-100ng/mL and 0-100ng/mL respectively;
s3: differentiating hematopoietic progenitor cells into NK cells;
wherein, in S3, the differentiation medium of the third step is removed, cells are inoculated into a cell culture vessel coated with matrix protein, and the differentiation medium of the fourth step is added for cell culture;
the matrix protein at least comprises one of Notch pathway activator protein or integrin; wherein, the differentiation culture medium of the fourth step is to add colony stimulating factors and interleukins in a basic differentiation culture medium; colony stimulating factors are TPO, SCF, Flt-3L and IL-3 at concentrations of 0-100ng/mL, 5-100ng/mL and 5-100ng/mL, respectively; the interleukin is IL-2, IL-7 and IL-15, and the concentration is respectively 100-1000IU/mL, 5-100ng/mL and 5-100 ng/mL;
s4: maturation and expansion of NK cells;
wherein the S4 includes: removing the differentiation culture medium in the fourth step, and adding the differentiation culture medium in the fifth step for cell culture; wherein the fifth step differentiation medium is a basic differentiation medium to which interleukins and a substance promoting the maturation and expansion of NK cells are added;
the interleukin is selected from one or more of IL-2, IL-12, IL-18, IL-21, IL-27 and IL-15; the substance promoting NK cell maturation and expansion is selected from one or more of human AB plasma, human platelet lysate, Vitamin A, nicotinamide, Vitamin E and Heparin; wherein the concentrations of IL-2, IL-12, IL-18, IL-21, IL-27 and IL-15 are respectively 100-1000IU/mL, 0-100ng/mL and 5-100 ng/mL; human platelet lysate, Nicotinamide (NAM), Vitamin E and Heparin at concentrations of 1-10%, 1-10mmol/L, 0-10mg/mL and 0-100. mu.g/mL, respectively;
the basal differentiation medium of S2-S4 comprises IMDM 50% at final concentration, F-12 50% at final concentration, rHSA 0.2-20mg/ml, thioglycerol 0-1000. mu.M, ascorbic acid 0-200. mu.g/ml, transferrin 1-50. mu.g/ml, sodium selenite 1-50 ng/ml, and ethanolamine 5-100. mu.M.
2. The method according to claim 1, wherein the S1 includes:
s11: placing the cell suspension of the human pluripotent stem cells on a shaking table to shake and culture overnight to form embryoid bodies.
3. The method of claim 1, wherein in S3, the Notch pathway activating protein is selected from one or more of DLL1 recombinant protein, DLL4 recombinant protein, Jagged-1 recombinant protein, Jagged-2 recombinant protein, and variants thereof;
the integrin is selected from one or more of Fibronectin (Fibronectin), Laminin (Laminin), Vitronectin (Vitronectin), MAdCAM-1 (adhesion cell adhesion molecule-1), VCAM-1 (vascular cell adhesion molecule-1), ICAM (intercellular adhesion molecule), and their variants.
4. The method according to claim 1, further comprising step S5: freezing and storing the NK cells obtained by differentiation by using a freezing and storing solution; the cryopreservation liquid comprises: sodium chloride, sodium gluconate, sodium acetate, potassium chloride, magnesium chloride, human serum albumin and DMSO.
5. A natural killer cell produced by the method according to any one of claims 1 to 4.
6. A cell population enriched for the natural killer cells of claim 5.
7. A pharmaceutical agent for preventing and/or treating tumor, which comprises the natural killer cell according to claim 5.
CN202010153358.3A 2020-03-06 2020-03-06 Method for differentiating human pluripotent stem cells into natural killer cells and application Active CN111235105B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010153358.3A CN111235105B (en) 2020-03-06 2020-03-06 Method for differentiating human pluripotent stem cells into natural killer cells and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010153358.3A CN111235105B (en) 2020-03-06 2020-03-06 Method for differentiating human pluripotent stem cells into natural killer cells and application

Publications (2)

Publication Number Publication Date
CN111235105A CN111235105A (en) 2020-06-05
CN111235105B true CN111235105B (en) 2021-08-10

Family

ID=70876842

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010153358.3A Active CN111235105B (en) 2020-03-06 2020-03-06 Method for differentiating human pluripotent stem cells into natural killer cells and application

Country Status (1)

Country Link
CN (1) CN111235105B (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2021211414A1 (en) * 2020-01-21 2022-08-04 Takeda Pharmaceutical Company Limited Compositions and methods of cryopreserving cells
CN111849913B (en) * 2020-07-31 2021-05-25 南京北恒生物科技有限公司 Engineered immune cells and uses thereof
US11591381B2 (en) 2020-11-30 2023-02-28 Crispr Therapeutics Ag Gene-edited natural killer cells
CN112608895B (en) * 2020-12-18 2024-02-27 深圳市济因生物科技有限公司 Natural killer cell differentiated from human pluripotent stem cells, preparation method and application thereof
WO2022144632A1 (en) * 2020-12-30 2022-07-07 Crispr Therapeutics Ag Compositions and methods for differentiating stem cells into nk cells
CN112662627B (en) * 2021-01-19 2022-09-02 上海爱萨尔生物科技有限公司 Culture solution for differentiating pluripotent stem cells into natural killer cells and differentiation method
CN112980788B (en) * 2021-03-08 2021-11-05 河北森朗生物科技有限公司 Preparation method of NK (natural killer) cells with low expression of CD7
CN113373114A (en) * 2021-06-11 2021-09-10 杭州原生生物科技有限公司 Culture medium and method for improving efficiency of differentiation of pluripotent stem cells into hematopoietic stem cells
CN113388579A (en) * 2021-07-07 2021-09-14 杭州原生生物科技有限公司 A culture medium containing cytokine for differentiating hematopoietic stem cell into natural killer cell and its differentiation method
CN115261318B (en) * 2021-09-29 2024-05-28 苏州艾凯利元生物科技有限公司 Method for producing natural killer cells
CN114517176B (en) * 2022-01-27 2022-12-02 广东普罗凯融生物医药科技有限公司 Kit for inducing IPS (in-plane switching) cells into NK (natural killer) cells and application method of kit
CN114621920B (en) * 2022-02-22 2023-09-05 中国医学科学院血液病医院(中国医学科学院血液学研究所) Method for differentiating hematopoietic stem progenitor cells into NK cells and culture medium
CN114507641B (en) * 2022-04-20 2022-07-12 山东兴瑞生物科技有限公司 Method for inducing and differentiating human embryonic stem cells into NK cells
WO2023233339A1 (en) * 2022-06-01 2023-12-07 Crispr Therapeutics Ag Compositions and methods for differentiating stem cells into nk cells
CN114958771B (en) * 2022-06-27 2023-12-05 广东康盾创新产业集团股份公司 Preparation method of IPSC-CAR-NK cells
WO2024010053A1 (en) * 2022-07-06 2024-01-11 キリンホールディングス株式会社 Embryoid body cell composition, differentiated cell composition and method for producing differentiated cell composition
CN115216443A (en) * 2022-07-21 2022-10-21 中国科学院广州生物医药与健康研究院 Method for obtaining NK (natural killer) cells from human pluripotent stem cells through in-vitro rapid and efficient differentiation and application of NK cells
CN116334082A (en) * 2023-02-15 2023-06-27 深圳市济因生物科技有限公司 NK (natural killer) cell with CD16a gene knocked out as well as preparation method and application thereof
CN115896019B (en) * 2023-02-23 2023-05-26 山东兴瑞生物科技有限公司 Method for inducing and differentiating induced pluripotent stem cells into NK cells
CN117050940B (en) * 2023-10-11 2024-01-26 苏州艾凯利元生物科技有限公司 Method for preparing natural killer cells
CN117050941B (en) * 2023-10-11 2024-01-26 苏州艾凯利元生物科技有限公司 Method for preparing natural killer cells
CN117305241B (en) * 2023-11-28 2024-03-19 上海兴瑞一达生物科技有限公司 Method for inducing and differentiating hiPSCs into NK cells

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2841563B1 (en) * 2012-04-24 2019-06-12 Dan S. Kaufman Method for developing natural killer cells from stem cells
WO2018147801A1 (en) * 2017-02-07 2018-08-16 Agency For Science, Technology And Research Methods and kits for generating mimetic innate immune cells from pluripotent stem cells

Also Published As

Publication number Publication date
CN111235105A (en) 2020-06-05

Similar Documents

Publication Publication Date Title
CN111235105B (en) Method for differentiating human pluripotent stem cells into natural killer cells and application
CN112608895B (en) Natural killer cell differentiated from human pluripotent stem cells, preparation method and application thereof
WO2017221975A1 (en) Method for producing cd4/cd8 double-positive t cells
JP6339998B2 (en) How to generate natural killer cells from stem cells
KR102292843B1 (en) Induced pluripotent stem cell(iPSC) derived natural killer cell and its use
JP2021510527A (en) Methods for Differentiation of Human Pluripotent Stem Cell Lines in Suspension Culture
KR20150126943A (en) Methods and materials for hematoendothelial differentiation of human pluripotent stem cells under defined conditions
CN111607566B (en) Method for differentiating human pluripotent stem cells into hematopoietic progenitor cells and application thereof
CN113631699A (en) Methods and systems for producing cells of hematopoietic lineage
WO2023240763A1 (en) Method for inducing differentiation of ipscs to obtain cd34+ cells and nk cells and use thereof
JP2013194049A (en) Composition and method for amplifying human hematopoietic stem cell
WO2013086029A1 (en) Compositions and methods for differentiating pluripotent stem cells into primitive blood cells and uses thereof
Lyadova et al. Macrophages derived from human induced pluripotent stem cells: the diversity of protocols, future prospects, and outstanding questions
CN115261318B (en) Method for producing natural killer cells
WO2020056158A1 (en) Organoid compositions for the production of hematopoietic stem cells and derivatives thereof
Sivalingam et al. Superior red blood cell generation from human pluripotent stem cells through a novel microcarrier-based embryoid body platform
US20200345789A1 (en) Production method for ips cell-derived population of genetically diverse t cells
JP4706208B2 (en) Method for producing hematopoietic stem cells
CN117050941B (en) Method for preparing natural killer cells
CN117050940B (en) Method for preparing natural killer cells
WO2024113358A1 (en) Methods and compositions for differentiation of pluripotent stem cells and derived natural killer cells
WO2024113352A1 (en) Methods and compositions for differentiation of pluripotent stem cells and derived hematopoietic lineage cells
JPWO2005056778A1 (en) Method of inhibiting or proliferating hematopoietic stem cells
WO2023243627A1 (en) Thymic epithelial cell production method
JP2012165660A (en) Composition and method for amplifying human hematopoietic stem cell

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