WO2020152661A1 - Production method for cell population including nk cells - Google Patents

Production method for cell population including nk cells Download PDF

Info

Publication number
WO2020152661A1
WO2020152661A1 PCT/IB2020/052499 IB2020052499W WO2020152661A1 WO 2020152661 A1 WO2020152661 A1 WO 2020152661A1 IB 2020052499 W IB2020052499 W IB 2020052499W WO 2020152661 A1 WO2020152661 A1 WO 2020152661A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
population
cell
culture
donors
Prior art date
Application number
PCT/IB2020/052499
Other languages
French (fr)
Japanese (ja)
Inventor
米満 吉和
結 原田
Original Assignee
株式会社ガイアバイオメディシン
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
Priority claimed from JP2019007868A external-priority patent/JP6838750B2/en
Application filed by 株式会社ガイアバイオメディシン filed Critical 株式会社ガイアバイオメディシン
Priority to EP20745606.2A priority Critical patent/EP3915640A4/en
Priority to CN202080023218.2A priority patent/CN113613723A/en
Priority to AU2020211441A priority patent/AU2020211441B2/en
Priority to KR1020217026441A priority patent/KR20220138330A/en
Publication of WO2020152661A1 publication Critical patent/WO2020152661A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a method for producing a cell population containing natural killer cells (NK cells) and its use.
  • NK cells natural killer cells
  • Patent Document 1 discloses a step of preparing a cell population containing NK cells, a step of removing T cells from the cell population containing NK cells, and a medium containing the remaining remaining cells as a cytokine containing only IL-2.
  • a step of culturing without using feeder cells, and a method for expanding NK cells, and a pharmaceutical composition for cell therapy containing a cell population containing NK cells obtained by the expansion suggest.
  • Patent Document 2 a step of amplifying hematopoietic progenitor cells under a single culture condition containing IL-15, SCF, IL-7 and Flt3L, and a cell obtained in the step of amplifying IL-2,
  • a method for preparing NK cells which comprises the step of inducing differentiation into NK cells under culture conditions comprising 5, 6, 7, 8 or 9 days, and a cell therapy containing the prepared cell population containing the NK cells.
  • NK cells that are CD16-positive, CD56 high-expressing, CD57-negative, NKG2C-positive, NKG2A-negative-low-expressing, and CD94-positive.
  • a cell population containing NK cells thereof Patent Document 3
  • a CD3 negative cell expressing a chemokine receptor and a cell adhesion molecule Patent Document 4
  • a group of receptors that recognize HLA class I is expressed on the cell surface of NK cells.
  • the KIR (Killer cell Immunoglobulin-like Receptor) family is as diverse as HLA.
  • HLA is not matched as much as possible, and recently, an intentional KIR between a donor and a recipient, such as selecting a donor so that the recipient does not have a ligand for the KIR of the donor NK cell, has been proposed.
  • /HLA mismatch is being effectively used because it can bring about more desirable results from the viewpoint of recurrence, GVHD (graft versus host disease, graft-versus-host disease) and prognosis.
  • NK cells were cultured with feeder cells either matched or mismatched for inhibitory KIR ligands (lacking one or more ligands present in the NK cell donor). From a report (Non-patent Document 1) investigating to what extent the HLA class I-KIR interaction influences human NK cell proliferation in allogeneic environment (Non-patent Document 1), NK cell hyperactivation is indicated using an antitumor effect as an index. If desired, less signal from the KIR is suggested to be better.
  • Non-patent Document 2 a recent report (Non-patent Document 2) on allogeneic responsiveness of HLA heterozygous NK cells (in the case of unidirectional matching) to tissues derived from HLA haplotype homozygous (HLA homo) iPS cells is related to culture of NK cells. , Strongly suggests that mixed culture containing HLA/KIR mismatch is not established.
  • JP, 2013-27385, A (patent No. 5728863, patent No. 5989016).
  • JP, 2014-226079, A (patent No. 5511039, patent No. 6164650).
  • JP, 2018-193303, A Japanese Patent Application No. 2018-059624 Application specification (not disclosed on the priority date of the present application)
  • the present inventors are developing a cell population containing highly active NK cells.
  • the amount of peripheral blood as a raw material and the amount of blood obtained by the apheresis method are limited, it is not possible to convert the therapeutic NK cell population into an off-the-shelf in preparation for future treatment.
  • the in vitro amplification efficiency of NK cells may be extremely poor depending on the donor.
  • HLA class I immature NK cells that have undergone binding with a ligand
  • HLA class I immature NK cells that have undergone binding with a ligand
  • NK cells differentiated and matured from iPS cells or ES cells may not have sufficient signals required for license to obtain an antitumor effect.
  • the present invention provides the following.
  • Including NK cells comprising preparing a population of mononuclear cells derived from a plurality of donors and containing NK cells, and incubating the prepared population of mononuclear cells under conditions effective for treating NK cells Method for producing cell population.
  • the production method according to 1 or 2 wherein the step of preparing a population of mononuclear cells includes the step of removing CD34-positive cells.
  • step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from peripheral blood collected from a plurality of donors.
  • step of preparing a population of mononuclear cells includes a step of obtaining a population of mononuclear cells from apheresis blood collected from a plurality of donors.
  • any of the step of preparing a population of mononuclear cells selected from the group consisting of embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, and adult stem cells derived from a plurality of donors
  • the method for production according to any one of 1-3 which is for preparing a population of mononuclear cells derived from sea urchin.
  • a cell population containing NK cells having the following characteristics: (1) Derived from multiple donors.
  • cytotoxic activity is 50% or more when NK cells are used as effector cells (E) and K562 cells are used as target cells (T) at a mixing ratio (E:T) of 1:1.
  • a pharmaceutical composition for cell therapy comprising a cell population produced by the production method according to any one of 1-7.
  • a pharmaceutical composition for cell therapy comprising the cell population according to item 8.
  • blood or PBMC as a raw material can be mixed for multiple people, the amount of raw material can be increased and off-the-shelf conversion can be expected.
  • NK cells can be stably grown regardless of the donor, and the growth rate can be improved.
  • Culture test CD3 positive cells were removed from PBMCs of a plurality of persons, and the cells were cultured for 14 days using KBM-501 medium. Summary of experimental results (left) and statistical analysis (right): When mixed culture was performed, the growth rate was significantly improved as compared with the culture derived from a single donor.
  • Tumor cell cytotoxicity test The cytotoxic activity of NK cells prepared by mixed culture and culture derived from a single donor was evaluated using the damage to SKOV3 (human ovarian cancer cell line) as an index. Culture with 500 cm 2 culture bag. After 30 minutes from the start of culturing and on the 9th day of culturing, the bag was flipped over. Culture test.
  • Frozen apheresis blood (for 2 donors) was used as a material, and after removing CD3 positive cells and CD34 positive cells using an automatic closed cell processing device, using a T75 flask or a 500 cm 2 culture bag, using KBM-501 medium Cultured for a day.
  • Monocyte/NK cell swapping experiment The population of CD16 high increased under the condition that one or both of the signals from KIR3DL1 and KIR3DS1 entered. Higher ADCC activity can be expected for the CD16 high population.
  • the present invention relates to a cell population containing NK cells.
  • the NK cell-containing cell population of the present invention can be produced by a method including the following steps: Preparing a population of mononuclear cells from multiple donors, including NK cells, The prepared mononuclear cell population is incubated under conditions effective for treating NK cells.
  • the cell population of the present invention is obtained from a population of mononuclear cells derived from multiple donors.
  • the population of mononuclear cells referred to herein includes NK cells, and may include mononuclear cells other than NK cells, for example, monocytes.
  • the NK cells may be primary NK cells (NK cells collected from a living body and not passaged).
  • the population of mononuclear cells derived from a plurality of donors and containing NK cells includes a population of mononuclear cells (containing NK cells and may contain monocytes) obtained from one donor, and another population.
  • the population of mononuclear cells (including NK cells and may include monocytes) obtained from a donor may be a mixed one, and NK cells obtained from one donor and another donor may be mixed. It may be a mixture with monocytes obtained from.
  • plural means two or more, and is not particularly limited as long as the above conditions are satisfied.
  • mononuclear cells derived from three or more, four or more, seven or more, and nine or more donors were mixed.
  • the plurality of donors may include the patient himself/herself who is administered the cell population containing NK cells, or a relative of the patient.
  • KIRs include KIR2DL1, KIR2DL2, KIR2DL3, KIR3DL1, KIR3DL2, KIR2DL4, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DS1, KIR3DL3, KIR2DL5A and KIR2DL5B.
  • licensing refers to the process of acquiring the "missing-self response (detection of lack of self HLA)" characteristic of NK cells in the process of cell differentiation and maturation.
  • human and mice only immature NK cells that have undergone binding of HIR class I molecule with KIR or NKG2A in the process of differentiation of NK cells from hematopoietic stem cells in the bone marrow are licensed, and expression of self HLA class I after maturation is licensed. It is believed to acquire the ability to detect depressed cells.
  • KIRs known to be involved in licenses are KIR2DL1, KIR2DL2, KIR2DL3, KIR3DL1 and NKG2A/CD94.
  • HLA types that can be recognized by NK cells include HLA-C allotypes, about 1/3 of HLA-B allotypes, a part of HLA-A allotypes (those having a Bw4 motif), and non-classical HLA-E and HLA-G of HLA.
  • the KIR and HLA related to the license as described above are selected. Especially, it can be considered.
  • donors having different genotypes can be selected so as to increase the repertoire of molecules used for licensing NK cells.
  • the inhibitory KIR of NK cells may be selected not to be stimulated, or the inhibitory KIR may be selected to be stimulated.
  • ADCC antibody-dependent cellular cytotoxicity
  • Mixing is performed before administering NK cells to the subject.
  • the mixing can be carried out in various stages as long as the desired effects can be obtained.
  • blood taken from multiple donors may be mixed, and then the mixture may be used to obtain a population of mononuclear cells.
  • blood collected from each donor may be used to obtain a population of mononuclear cells, respectively.
  • the populations may be mixed.
  • the populations of mononuclear cells collected from a plurality of donors may be cultured and then the populations may be mixed. From the viewpoint of good growth, mixing is preferably performed before culturing.
  • the mixing ratio of the population of mononuclear cells derived from multiple donors can be set appropriately. For example, cells from multiple donors can be included in approximately equal proportions, and cells from a particular donor can be included in greater or lesser proportion.
  • the mixing ratio of cells is based on the number of cells, unless otherwise stated.
  • the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from peripheral blood collected from a plurality of donors.
  • the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from apheresis blood collected from a plurality of donors.
  • a population of mononuclear cells can be prepared using various procedures known to those of skill in the art.
  • red blood cells can be removed and a mononuclear cell fraction can be collected by performing density centrifugation at room temperature.
  • Peripheral Blood Mononuclear Cells (PBMC) isolated from peripheral blood include various lymphocytes such as T cells, B cells, NK cells, monocytes and dendritic cells.
  • lymphocytes such as T cells, B cells, NK cells, monocytes and dendritic cells.
  • mononuclear cells that are CD3 negative and CD56 positive can be called NK cells.
  • NK cells can be prepared using various procedures known to those of skill in the art.
  • a method for separating NK cells by removing unnecessary cells from whole blood or PBMC is known.
  • Monocytes can be prepared using various procedures known to those of skill in the art.
  • a method for separating monocytes by removing unnecessary cells from whole blood or PBMC is known.
  • the population of mononuclear cells may include NK cell precursors, T cells, NKT cells, hematopoietic progenitor cells, etc. in addition to monocytes and NK cells.
  • the desired NK cells may be selected after amplification using, for example, specific gravity centrifugation, immunomagnetic beads, FACS, flow cytometry, etc.
  • NK cells include anti-CD3 antibody, anti-CD16 antibody, anti-CD34 antibody, anti-CD56 antibody, anti-CD69 antibody, anti-CD94 antibody, anti-CD107a antibody, anti-KIR3DL1 antibody, anti-KIR3DL2 antibody, anti-KIR2DL3 antibody, anti-KIR2DL1 antibody, It may be selectively separated from the cell population using an anti-KIR2DS1 antibody, an anti-KIR2DL5 antibody, an anti-NKp46 antibody, an anti-NKp30 antibody, an anti-NKG2D antibody or the like.
  • the antibody may be a monoclonal antibody, a polyclonal antibody, or the like.
  • the selection of NK cells may be performed by selectively removing T cells, NKT cells, hematopoietic progenitor cells and other cells.
  • the step of preparing a population of mononuclear cells may include the step of removing T cells, which may be accomplished by removing CD3-positive cells.
  • the step of preparing a population of mononuclear cells may also include the step of removing hematopoietic progenitor cells, which may be accomplished by removing CD34-positive cells. That is, the step of preparing a population of mononuclear cells may include the step of removing CD3-positive cells or the step of removing CD34-positive cells.
  • the removal of CD3-positive cells and the removal of CD34-positive cells can be performed, for example, by using Dynabeads manufactured by Invitrogen, Dynabeads manufactured by Invitrogen, or immunomagnetic beads such as CliniMACS manufactured by Miltenyi Biotech, and the cell surface antigen CD3 and/or It can be carried out by separating and removing cells expressing CD34.
  • Such a step is preferably performed so as not to exhaust NK cells and prevent monocytes from exerting an extra phagocytic function.
  • using relatively small magnetic beads unreacted beads that are not bound to cells can be removed together with the supernatant by centrifugation), and the beads and cell population are incubated at low temperature. Examples of preferable operations include setting the time relatively short, removing unreacted beads, and then using a column to obtain cells not bound to the beads as a target cell population.
  • the cells may be removed using an automatic closed cell processing apparatus.
  • the population of mononuclear cells is prepared from hematopoietic stem cells derived from any one selected from the group consisting of embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, and adult stem cells. Good. Methods for inducing mononuclear cells including NK cells from these stem cells are known and can be applied to the present invention by those skilled in the art (Domogala A. et al., Natural killer cell immunotherapy: from bench to bedside, Frontiers in Immunology, 2015; 6, doi: 10.3389/fimmu.2015.0264, and Zeng J. et al., Generation of Shelf "Natural berrelloller Celloller Killer".
  • Stem cells may also be modified so that highly active NK cells can be obtained.
  • highly active NK cells For example, high-affinity CD16 (158V: 158th amino acid is valine) is known, and such findings can be applied. it can.
  • the prepared population of mononuclear cells is incubated in a state in which cells derived from a plurality of donors are mixed under conditions effective for NK cell treatment. Incubation may or may not be accompanied by cell proliferation. In one of the preferred embodiments of the invention, the incubating is accompanied by growth. Incubation with proliferation can be read as culturing or proliferation.
  • Effective conditions for NK cell treatment means conditions suitable for mixed cells derived from a plurality of donors to exert the desired mixing effect.
  • the medium or isotonic solution used for cell suspension, time, temperature, environment, etc. are not particularly limited as long as the desired effects are exhibited.
  • the condition effective for NK cell treatment includes the condition effective for activation of NK cell.
  • Such conditions include, for example, incubation in the presence of appropriate concentrations of activation factors such as IL-2 and IL-15 at 37° C., 5% CO 2 and saturated steam atmosphere for 4-18 hours. For example.
  • Effective conditions for NK cell treatment include effective conditions for NK cell proliferation.
  • Effective conditions for the growth of NK cells include using a medium suitable for the growth of NK cells. Examples of such a medium include KBM501 medium (Kojin Bio Co., Ltd.), CellGro SCGM medium (Celgenics, Iwai Chemical Co., Ltd.), X-VIVO15 medium (Lonza, Takara Bio Inc.), IMDM, MEM, DMEM. , RPMI-1640 and the like, but are not limited thereto.
  • Interleukin 2 may be added to the medium at a concentration that can achieve the object of the present invention.
  • the concentration of IL-2 can be greater than 2000 IU/mL and can be 2500-2815 IU/mL.
  • IL-2 preferably has a human amino acid sequence, and is preferably produced by recombinant DNA technology for safety.
  • the concentration of IL-2 may be indicated in national standard units (JRU) and international units (IU). Since 1 IU is about 0.622 JRU, 1750 JRU/mL is about 2813 IU/mL.
  • the subject's autologous serum, human AB type serum available from BioWhittaker or others, or blood donated human serum albumin available from the Japanese Red Cross Society may be added to the medium.
  • Autologous serum and human AB-type serum are preferably added at a concentration of 1-10%, and donated human serum albumin is preferably added at a concentration of 1-10%.
  • the test subject may be a healthy person or a patient suffering from a disease.
  • a composition formulated for the growth of immune cells may be added to the medium instead of or together with the serum.
  • Such compositions are commercially available. For example, UltraGro series (AventaCell), CTS Immune Cell SR (Thermo Fisher Scientific) can be used for the present invention.
  • the culture medium may contain appropriate proteins, cytokines, antibodies, compounds and other components, provided that the amplification effect of NK cells is not impaired.
  • Cytokines include interleukin 3 (IL-3), interleukin 7 (IL-7), interleukin 12 (IL-12), interleukin 15 (IL-15), interleukin 21 (IL-21), stem cell factor. (SCF) and/or FMS-like tyrosine kinase 3 ligand (Flt3L).
  • IL-3, IL-7, IL-12, IL-15, IL-21, SCF and Flt3L preferably have a human amino acid sequence, and are preferably produced by recombinant DNA technology for safety.
  • the medium may be replaced at any time after the start of culture, provided that the desired number of NK cells is obtained, but it is preferably every 3-5 days.
  • Culture vessels include, but are not limited to, commercially available dishes, flasks, plates, multiwell plates.
  • a culture container in which many cells can be cultured and which is easy to handle.
  • An example of such a culture container is a cell culture bag made of a material having high gas permeability.
  • both NK cells and monocytes adhere to the culture surface.
  • the cell density per unit volume as well as the cell density per unit volume greatly influences the culture efficiency including cell viability and growth rate.
  • the culture conditions are not particularly limited as long as the amplification effect of NK cells is not impaired, but the culture conditions are generally 37° C., 5% CO 2 and saturated steam atmosphere. Since one of the objects of the present invention is to prepare a large amount of NK cells, it is advantageous that the longer the culture period is, the more NK cells can be obtained.
  • the culture period is not particularly limited, provided that the NK cells are expanded to a desired cell number. For example, it may be performed for 7 to 28 days, may be 10 to 18 days, may be 12 to 16 days, for example, 14 days (Saito S. et al., Ex vivo generation of highly purified and activated natural killer cells from. human peripheral blood.Hum Gene Ther Methods.2013;24(4):241-252, and the above-mentioned Patent Document 1).
  • the prepared cell population containing mononuclear cells may be cryopreserved, thawed depending on the time of administration to a patient, and then provided for NK cell culture.
  • the population of mononuclear cells is frozen during or after amplification by the NK cell expansion method of the present invention, thawed according to the time of transplantation into a patient, and then thawed. May be offered. Freezing and thawing may use any method known to those skilled in the art. Any commercially available cell cryopreservation solution may be used for freezing the cells.
  • the predetermined NK cell population obtained by culture may contain NK cell precursors, T cells, NKT cells, hematopoietic progenitor cells, etc. in addition to the target NK cells.
  • the NK cells of interest or a population thereof may be selected using, for example, specific gravity centrifugation, immunomagnetic beads, FACS, flow cytometry, or the like.
  • the target NK cells or a population thereof may be selectively isolated using anti-KIR2DL5 antibody, anti-NKp46 antibody, anti-NKp30 antibody, anti-NKG2D antibody or the like.
  • the antibody may be a monoclonal antibody, a polyclonal antibody, or the like.
  • the NK cell of interest or a population thereof may be selected by selectively removing T cells, NKT cells, hematopoietic progenitor cells and other cells.
  • NK cell proliferation is improved by culturing a population of mononuclear cells derived from a plurality of donors and containing NK cells. It is considered that this is because more variations of the HLA/KIR combination resulted in more licensing signals entering each other. Good growth is compared with the case of culturing cells derived from any one of a plurality of donors in mixed culture, and the proliferation rate of cells (the number of cells after culture/the number of cells before culture) Is improving.
  • 70% or more preferably 80% or more, more preferably 90% or more may be NK cells.
  • the cytotoxic activity of NK cells obtained by mixed culture of a population of mononuclear cells containing NK cells derived from a plurality of donors is higher than that of NK cells derived from a single donor. It can be equal or better. Further, the proportion of NK cells that highly express CD16 may be equal to or higher than that of NK cells derived from a single donor.
  • the cytotoxic activity refers to the lytic ability of target cells (effector cells, E) to target cells (T), unless otherwise specified. The cytotoxic activity can be expressed as a percentage (%) of target cells that have been killed by effector cells, and is calculated by the following equation, for example.
  • the mixing ratio (E:T) of the effector cells to the target cells and the co-culture time of the effector cells to the target cells are Can be appropriately determined depending on the type of cells used and the strength of the activity.
  • the target cells may be, but are not limited to, K562 cells, SKOV3 cells (human ovarian cancer cell line), acute myelogenous leukemia cells, and chronic myelogenous leukemia cells. Effector cells and target cells, and living cells and dead cells can be distinguished and quantified by reagents such as radioactive substances and antibodies labeled with fluorescent dyes.
  • the population of NK cells of the present invention derived from a plurality of donors of the present invention is not suppressed by MDSC (Myeloid-delivered suppressor cells), or its suppression is extremely low.
  • MDSC Myeloid-delivered suppressor cells
  • the present inventors have confirmed that the population of NK cells derived from a single donor obtained by the above-mentioned culture method is not suppressed by MDSC.
  • TGF- ⁇ , IL-10 receptors for humoral factors
  • the cell population containing NK cells obtained by the present invention has the following characteristics: (1) Derived from multiple donors. (2) The cytotoxic activity is 50% or more when NK cells are used as effector cells (E) and K562 cells are used as target cells (T) at a mixing ratio (E:T) of 1:1. Instead of the above feature (2) or together with the feature (2), the following features may be provided: The cytotoxic activity is 50% or more when (2′) SKOV3 cells are co-cultured with target cells (T) at a mixing ratio (E:T) of 3:1.
  • Such a cell population may further have the following characteristics.
  • the proportion of NK cells is 70% or more, more specifically 80% or more, and further specifically 90% or more.
  • It may include both a population of NK cells having high CD16 expression and a population of NK cells having low CD16 expression.
  • the proportion of NK cells that highly express CD16 is 50% or more.
  • the cytotoxic activity of NK cells can be measured and calculated by a method well known to those skilled in the art.
  • the cytotoxic activity (%) is usually based on the viable cell number of the target cell (T) after the action of the effector cell (E), for example, by the formula: (1-viable cell number/negative control viable cell number) ⁇ 100. Can be calculated.
  • the cytotoxic activity of NK cells as effector cells (E) and K562 cells as target cells (T) at a mixing ratio (E:T) of 1:1 is preferably 60% or more, 70 % Or more, more preferably 80% or more, still more preferably 90% or more, still more preferably 95% or more.
  • the cytotoxic activity of NK cells as effector cells (E) and SKOV3 cells as target cells (T) at a mixing ratio (E:T) of 3:1 is preferably 60% or more, 70 % Or more, more preferably 80% or more, still more preferably 90% or more, still more preferably 95% or more.
  • a positive result may be represented by + and a negative result may be represented by-.
  • CD16 positive may be designated as CD16 + and CD16 negative may be designated as CD16 ⁇ .
  • Positive cases include cases of high expression and cases of low expression.
  • High expression may be expressed as high or bright.
  • high CD16 expression may be expressed as CD16 high or CD16 bright .
  • Low expression may be expressed as low and dim.
  • CD16 low expression may be represented as CD16 low , CD16 dim .
  • Positive, negative, high expression, low expression can be judged based on the chart obtained by the flow cytometry method.
  • the position appearing on the chart may vary depending on the voltage setting of the instrument, the sensitivity setting, the antibody clone used, the staining condition, the dye used, etc., but those skilled in the art can select the cell population recognized as a group in the obtained chart. It can be appropriately drawn so as not to be divided.
  • the isotype control antibody is an antibody that does not react with a specific antigen.
  • background may occur due to nonspecific binding to proteins other than the target or binding to Fc receptors on the cell surface.
  • control cells are NK cells obtained from peripheral blood that have not been substantially cultured, such as the primary NK cells described in the Examples section of the present specification.
  • the degree of CD16 expression in a certain NK cell population was determined by using flow cytometry, and the CD16 expression level in the NK cell population was determined from the peripheral blood-derived NK cell population that was not substantially cultured (control). In comparison with the CD16 expression level in the above, it can be judged that the expression is high if the expression is similar to that of the control, and that it is low when the expression is lower than that of the control cells.
  • the control NK cells are known to highly express CD16.
  • the present invention also provides a pharmaceutical composition for cell therapy, which comprises the above-mentioned cell population containing NK cells as an active ingredient.
  • Cell therapy refers to a method of treating a disease or condition in a subject by administering cells treated ex vivo to the subject, which includes immune cell therapy.
  • the pharmaceutical composition includes, in addition to the cell population, which is an active ingredient, a solution capable of suspending NK cells, such as physiological saline or phosphate buffered saline (PBS).
  • a solution capable of suspending NK cells such as physiological saline or phosphate buffered saline (PBS).
  • PBS physiological saline or phosphate buffered saline
  • the pharmaceutical composition or solution may also contain pharmaceutically acceptable additives.
  • the pharmaceutical composition can be administered, for example, into a vein, an artery, subcutaneously, intraperitoneally or the like. Cell therapy with a pharmaceutical composition can be performed alone or in combination with surgery, chemotherapy, radiation therapy and the like.
  • NK cells are expected to be applied to cancer treatment and infectious disease treatment (Dahlberg CM et al., Natural Killer Cell-Based Therapeutic Targeting Cancer: Possible Strategies Anesthetics). -Tormor Activity, Front. status and future perspectives, Oncotarget, 2018;9(29):20891-20907), and the pharmaceutical composition of the present invention can be used for treating such cancer or infectious disease. More specifically, it includes, but is not limited to, oral cancer, gallbladder cancer, bile duct cancer, lung cancer, liver cancer, colon cancer, kidney cancer, bladder cancer, leukemia; infectious diseases caused by viruses, bacteria and the like.
  • NK cells may be administered, for example, into veins, arteries, subcutaneously, intraperitoneally and the like.
  • Cell therapy may be performed alone or in combination with surgery, chemotherapy, radiation therapy, antibody drugs and the like.
  • NK cells and antibody may be mixed before administration to equip the NK cells with the antibody in advance. As a result, the effector is limited to NK cells, the amount of antibody to be administered can be reduced, and it is considered to be extremely effective in reducing side effects.
  • the pharmaceutical composition of the present invention may be prepared by mixing the NK cell population and the antibody, and then removing the antibody not bound to the NK cell. That is, one of the preferred embodiments of the pharmaceutical composition of the present invention comprises NK cells and an antibody, but the antibody is bound to NK cells and substantially free of antibodies not bound to NK cells. Is. (See the above-mentioned Patent Document 3).
  • the pharmaceutical composition of the present invention is manufactured under the conditions (good manufacturing practice, GMP) conforming to the manufacturing control and quality control rules for drugs and quasi-drugs, and the manufacturing control and quality control standards for products such as regenerative medicine (Good Gene). , Cellular, and Tissue-based Products, Manufacturing Structure, GCTP).
  • GMP good manufacturing practice
  • GCTP regenerative medicine
  • Example 1 Mixed culture 1 of NK cells obtained from fresh peripheral blood
  • Blood was collected from healthy volunteers and peripheral blood mononuclear cells were isolated by density gradient centrifugation using Ficoll (GE Healthcare, 17144002). Peripheral blood mononuclear cells from multiple isolated humans were mixed at approximately the same rate, CD3 beads *1 was added and suspended, incubated at 4°C for 15 minutes, and then well suspended by adding 1 mL *2 of separation buffer to 300 xg. Centrifugation was performed for 10 minutes.
  • the supernatant was removed, and the suspension was suspended in 0.5 mL of separation buffer, and 2 mL of the separation buffer was added to a previously moistened LD column (Miltenyi Biotech, 130-042-901) to elute from the LD column. The liquid was collected. Further, 1 mL of the separation buffer was added to the LD column, and the eluate was collected. Then, the column was washed with 1 mL of the separation buffer, the number of cells in the collected liquid was counted, and the total number of cells was calculated. After centrifugation at 500 ⁇ g for 5 minutes and removing the supernatant, the cells were suspended in KBM501 medium *3 at 5 ⁇ 10 5 cells / mL.
  • Some cells were collected for flow cytometer measurement, and the remaining cells were cultured. Culturing was performed in a CO 2 incubator using a 6-well plate (Thermo Fisher Scientific, 140675), a T-75 flask (Thermo Fisher Scientific, 156499), or a 500 cm 2 culture bag (Nipro) (37). °C, 5% CO 2). On the 5th and 9th day of culture, a part of the culture solution was taken and the number of cells was counted. On the 9th day, the final volume was 6 mL per well in the case of 6-well plate, and in the case of T-75 flask. KBM501 medium was added to 50 mL per flask. The cells were collected on the 14th day, the number of cells was counted, and a part of the cells was used to measure the cell surface antigen with a flow cytometer.
  • the results are shown in Fig. 1.
  • the cell growth was good in any of the 9-person mixture (A in FIG. 1), the 8-person mixture (B in FIG. 1), and the 7-person mixture (C in FIG. 1).
  • 90% or more of the CD3 - CD56 + NK cells were found in any of the mixed cultures.
  • the 8-person mixed culture the cells were cultured in a T-75 flask and a 500 cm 2 culture bag, and the cell growth was good in both cases.
  • the 9-person mixed culture, the 7-person culture, and the culture derived from a single donor (1 of 7 persons) were cultured using a 6-well plate. In the culture using the 500 cm 2 culture bag, the bag was turned inside out as described in Example 4 below.
  • the culture derived from a single donor grew from 6.8 ⁇ 10 6 cells to 1.9 ⁇ 10 7 cells (about 2.8 times).
  • the cells grew from 7.8 ⁇ 10 6 cells to 3.7 ⁇ 10 7 cells (about 4.7 times), and the mixed culture showed better cell growth (C in FIG. 1). )).
  • Example 2 Mixed culture 2 of NK cells obtained from fresh peripheral blood
  • Wilcoxon rank sum test was performed using JMP Pro13 as the analysis software.
  • Example 3 Tumor cytotoxicity test
  • NK cells obtained by the method described in Example 1 from healthy volunteers and cultured NK cells from a single donor (one of four) were collected and washed, and then washed with 10% FBS (Nichirei). Bioscience, 171012-500 ML), 100 units of penicillin, and 100 ⁇ g/mL streptomycin (Nacalai Tesque, 26253-84) in RPMI1640 medium (Wako Pure Chemical Industries, 189-02025) (hereinafter referred to as 10% FBS/RPMI1640). ), and was adjusted to a concentration of 1 ⁇ 10 6 cells/mL with the same medium.
  • 10% FBS RPMI1640 medium
  • SKOV3 cells human ovarian cancer cell line
  • RPMI1640 medium Wired Cell Linker Kit
  • FBS/RPMI1640 10% FBS/RPMI1640 at 2 ⁇ 10 6 cells/mL and 2 ⁇ 10 5 cells/mL. did.
  • MDSC Myeloid-derivatized suppressor cells
  • Peripheral blood mononuclear cells were isolated from healthy volunteers (separate from NK cell donors) using Ficoll, 10 ng/mL IL-6 (PEPROTECH, 200-06-5UG) and 10 ng/mL GM-CSF (CellGenix). , 1012-050), and MDSC was induced by culturing in 10% FBS/RPMI1640 for 7-10 days.
  • MDSCs were suspended in RPMI 1640 medium (Wako Pure Chemical Industries, 189-02025) containing no serum component, stained with PKH Green Fluorescent Cell Linker Kit (Sigma, MINI67-1KT), and finally 10% FBS/ It was adjusted to 8 ⁇ 10 5 cells/mL with RPMI1640.
  • ⁇ Cytotoxicity test> A group of mixed cultured NK cells and SKOV3 cells derived from a plurality of donors, a group of cultured NK cells derived from a single donor and SKOV3 cells, a group in which MDSC was added to each of these two groups, a total of 4 groups, and only SKOV3 cells as a negative control A group was prepared.
  • NK cells and SKOV3 cells were seeded in a 96-well plate (IWAKI, 4870-800SP) at a cell ratio of 3:1, mixed, and reacted at 37° C. under 5% CO 2 for 4 hours.
  • MDSC 96-well plate
  • NK cells and MDSC were first mixed in a 96-well plate so that the cell ratio of NK cells, SKOV3 and MDSC was 5:1:4, and the mixture was added at 37° C. and 5% CO 2 for 12 hours. The reaction was carried out for -18 hours. Then, after centrifugation (500 xg, 5 minutes) and removal of the supernatant, SKOV3 cells were added, mixed, and reacted at 37°C under 5% CO 2 for 4 hours.
  • the mixture was centrifuged (500 xg, 5 minutes), and after removing the supernatant, a Zombie solution (Biolegend, 423105) diluted with PBS was added and mixed, and the mixture was incubated at room temperature in the dark for 30 minutes. After centrifugation and removal of the supernatant, the cells were suspended in PBS, and AccuCheck Counting Beads (Thermo Fisher Scientific, PCB100) was added. The measurement was performed using a flow cytometer (BD LSR Fortessa, BD Bioscience) and analyzed by FlowJo software (FLOWJO, LLC) to calculate the cytotoxic activity rate (% Lysis) *4 .
  • Cytotoxic activity rate (1-live SKOV3 live cells/negative control SKOV3 live cells) ⁇ 100 Number of live cells: actual number of SKOV3 cells x number of beads in added bead solution/actual number of beads Actual number of SKOV3 cells: FSC/SSC gated (debris exclusion), PKH26 + gated after double exclusion.
  • Example 4 Mixed culture of NK cells prepared from frozen apheresis blood
  • Frozen apheresis blood of two donors HemaCare, PB001CLP
  • HBSS(-) solution Nacalai Tesque, 17461-05
  • FRESENIUS KABI automatically closed cell processing system Lovo Cell Processing System
  • the concentrated cell solution was used for CliniMACS Prodity TS 310 (Miltenyi Biotec, 130-097-183), CliniMACS CD3 MicroBeads (Miltenyi Biotec, 130-017-601), CliniMACS CD30Bi-oIcient Reactant (MINITENS). Were used to remove CD3-positive cells and CD34-positive cells, followed by washing and elution. The total number of cells was calculated by counting the number of cells in the eluate. After centrifugation at 500 xg for 5 minutes and removal of the supernatant, the cells were suspended in KBM501 medium at 1 x 10 6 cells/mL.
  • the culture was performed in a CO 2 incubator using a T-75 flask (Thermo Fisher Scientific, 156499) or a 500 cm 2 culture bag (Nipro) (37° C., 5% CO 2 ).
  • the bag was turned over 30 minutes after the start of the culture, and was turned over again on the 9th day of culture (see FIG. 4-1).
  • KBM 501 medium was added so that the final solution volume was 50 mL per flask for the T-75 flask and 500 mL per bag for the culture bag. did.
  • the cells were collected on the 14th day, the total number of cells was counted, and a part of the cells was used to measure the cell surface antigen with a flow cytometer.
  • the purity of NK cells in the obtained population of cultured NK cells was 90.5% for T-75 flask culture and 91.7% for 500 cm 2 culture bag culture.
  • the population of mixed culture NK cells was bimodal, as was the population of culture NK cells derived from a single donor (see JP-A-2018-193303). Specifically, the proportions of low CD16 expression and high CD16 expression were 50.9% and 49.1% in the case of T-75 flask culture, and 46 in the case of 500 cm 2 culture bag culture. It was 0.7% and 53.3% (FIG. 4-2, right).
  • Example 5 Monocyte/NK cell swapping experiment
  • the measurement of cell surface antigen was performed by staining the collected cells with an antibody and analyzing them as follows: Alexa Fluor (registered trademark) 700-labeled anti-human CD56 antibody (Biolegend, 318316), APC/Cy7-labeled anti-human CD3 antibody (Biolegend, 300426), FITC-labeled anti-human KIR2DL1 antibody (Miltenyi Biotec, 130-103-966), PerCP. /Cy5.5-labeled anti-human KIR3DL1 antibody (Biolegend, 312718) was stained at a concentration of 1 ⁇ g/mL at 4° C.
  • cytotoxic activity 4 groups were prepared by reacting cultured NK cells and K562 cells, each of which was formed by combining primary NK cells of two donors and monocytes, K562 cells alone as a negative control, and K562 as a positive control.
  • a group was prepared in which cells were fixed with 10% formalin.
  • K562 cells human chronic myelogenous leukemia cell line
  • RPMI1640 medium Wired Cell Linker Kit
  • PKH26GL-1KT PKH26GL-1KT
  • NK cells and K562 cells were added to a 96-well plate (IWAKI, 4870-800SP) at a cell ratio of 1:1 and mixed, and reacted at 37° C. under 5% CO 2 for 2 hours. After the reaction, the mixture was centrifuged (500 xg, 5 minutes), the supernatant was removed, 7-AAD solution diluted with PBS (Beckman Coulter, A07704) was added and suspended, and the mixture was incubated at room temperature for 20 minutes. The measurement was performed using a flow cytometer (BD LSR Fortessa, BD Bioscience) and analyzed by FlowJo software (FLOWJO, LLC) to calculate the cytotoxic activity rate (% Lysis) *5 .
  • Cytotoxic activity rate (K562 cell dead cell rate-negative control dead cell rate)/(positive control dead cell rate-negative control dead cell rate) x 100

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention addresses the problems of providing an effective method for producing a population of NK cells for cell therapy, improving NK cell expansion efficiency in vitro, and flexibly increasing the signalling necessary for NK cell licensing. Specifically provided is a production method for a cell population including NK cells by preparing a monocyte population that is derived from multiple donors and includes NK cells, incubating and culturing the prepared monocyte population under conditions effective for the processing and proliferation of NK cells, and inducing the proliferation of the NK cells.

Description

NK細胞を含む細胞集団の製造方法Method for producing cell population containing NK cells
 本発明は、ナチュラルキラー細胞(NK細胞)を含む細胞集団の製造方法とその利用に関する。 The present invention relates to a method for producing a cell population containing natural killer cells (NK cells) and its use.
 悪性腫瘍は国民の死亡原因の1位であり、その対策が急務とされている。特に、外科手術、放射線治療及び化学療法からなる既存の治療方法に抵抗性である進行期難治性悪性腫瘍に対する新しい治療方法の開発は、極めて重要で意義がある。近年、第4の治療方法として、免疫チェックポイント阻害剤やキメラ抗原受容体(chimeric antigen receptor,CAR)遺伝子改変T細胞を用いた療法(CAR−T療法)といった免疫療法が注目を集めている。しかし、その多くが抗原を認識することにより活性化されるT細胞をエフェクターとしているために特異抗原への拘束性という根源的な壁がある。  Malignant tumors are the number one cause of death among the people, and countermeasures against them are urgently needed. In particular, the development of new treatment methods for advanced-stage intractable malignant tumors that are resistant to existing treatment methods consisting of surgery, radiotherapy and chemotherapy is extremely important and significant. In recent years, as a fourth treatment method, immunotherapy such as an immune checkpoint inhibitor or a therapy using chimeric antigen receptor (CAR) gene-modified T cells (CAR-T therapy) has been attracting attention. However, since many of them use T cells that are activated by recognizing an antigen as effectors, there is a fundamental barrier that they are restricted to specific antigens.
 自然免疫の主要因子として働くNK細胞を用いる免疫療法として、NK細胞を体外で増殖させた後、患者に投与するNK細胞療法が、副作用の比較的少ない治療方法として注目されている。しかし、NK細胞は末梢血等から得られる数が比較的少ない上に体外での増殖性が低い。そのためNK細胞を培養し、増殖させるための技術が検討されてきた。例えば特許文献1は、NK細胞を含む細胞集団を調製するステップと、前記NK細胞を含む細胞集団からT細胞を除去するステップと、除去された残りの細胞をIL−2だけをサイトカインとして含む培地でフィーダー細胞を用いることなく培養するステップとを含むことを特徴とする、NK細胞の増幅方法、及び増幅により得られたNK細胞を含む細胞集団を含有する、細胞療法のための医薬組成物を提案する。また特許文献2は、造血前駆細胞を、IL−15、SCF、IL−7及びFlt3Lを含む単一の培養条件で増幅するステップと、前記増幅するステップで得られた細胞を、IL−2を含む培養条件下5、6、7、8又は9日間でNK細胞に分化誘導させるステップとを含む、NK細胞の調製方法、並びに調製されたNK細胞を含む細胞集団を含有する、細胞療法のための医薬組成物を提案する。 As immunotherapy using NK cells that act as a major factor of innate immunity, NK cell therapy in which NK cells are proliferated in vitro and then administered to patients is attracting attention as a therapeutic method with relatively few side effects. However, the number of NK cells obtained from peripheral blood and the like is relatively small, and in vitro proliferation is low. Therefore, techniques for culturing and expanding NK cells have been investigated. For example, Patent Document 1 discloses a step of preparing a cell population containing NK cells, a step of removing T cells from the cell population containing NK cells, and a medium containing the remaining remaining cells as a cytokine containing only IL-2. And a step of culturing without using feeder cells, and a method for expanding NK cells, and a pharmaceutical composition for cell therapy containing a cell population containing NK cells obtained by the expansion. suggest. Further, in Patent Document 2, a step of amplifying hematopoietic progenitor cells under a single culture condition containing IL-15, SCF, IL-7 and Flt3L, and a cell obtained in the step of amplifying IL-2, A method for preparing NK cells, which comprises the step of inducing differentiation into NK cells under culture conditions comprising 5, 6, 7, 8 or 9 days, and a cell therapy containing the prepared cell population containing the NK cells. The pharmaceutical composition of
 また本発明者らは、腫瘍細胞傷害活性を高めた培養細胞として、CD16陽性、CD56高発現性、かつCD57陰性であり、かつNKG2C陽性、NKG2A陰性‐低発現性、及びCD94陽性であるNK細胞、及びそのNK細胞を含む細胞集団(特許文献3)を報告し、またケモカインレセプターと細胞接着分子を発現するCD3陰性細胞について報告してきた(特許文献4)。 In addition, the inventors of the present invention have shown that, as cultured cells having enhanced tumor cytotoxicity, NK cells that are CD16-positive, CD56 high-expressing, CD57-negative, NKG2C-positive, NKG2A-negative-low-expressing, and CD94-positive. , And a cell population containing NK cells thereof (Patent Document 3), and a CD3 negative cell expressing a chemokine receptor and a cell adhesion molecule (Patent Document 4).
 一方、NK細胞の細胞表面上には、HLAクラスIを認識する受容体群が発現している。これら受容体のうち、KIR(Killer cell Immunoglobulin−like Receptor)ファミリーはHLAと同様に多様性を有する。造血幹細胞移植においては、HLAをできる限りマッチさせるのではなく、最近ではドナーNK細胞のKIRに対するリガンドをレシピエントがもたないようにドナーを選ぶこと等、ドナー・レシピエント間の意図的なKIR/HLAミスマッチが、再発、GVHD(graft versus host disease、移植片対宿主病)及び予後の観点からより望ましい結果をもたらしうるとして有効に利用されつつある。しかし、NK細胞の抗腫瘍活性に関しては、NK細胞を、抑制性KIRリガンドに関して一致又はミスマッチ(NK細胞ドナーに存在する1つ以上のリガンドが欠如している)のいずれかのフィーダー細胞と共に培養し、HLAクラスI−KIR相互作用が同種異系環境におけるヒトNK細胞増殖にどの程度影響するかを調べた報告(非特許文献1)からは、抗腫瘍効果を指標にNK細胞の高活性化を望む場合、KIRからのシグナルは少ないほうがよいことが示唆される。また、近年の、HLAハプロタイプホモ接合型(HLAホモ)iPS細胞由来の組織に対するHLAヘテロNK細胞(一方向適合の場合)の同種応答性に関する報告(非特許文献2)は、NK細胞の培養に関し、HLA/KIRミスマッチを含む混合培養が成立しないことを強く示唆するものである。 On the other hand, a group of receptors that recognize HLA class I is expressed on the cell surface of NK cells. Among these receptors, the KIR (Killer cell Immunoglobulin-like Receptor) family is as diverse as HLA. In hematopoietic stem cell transplantation, HLA is not matched as much as possible, and recently, an intentional KIR between a donor and a recipient, such as selecting a donor so that the recipient does not have a ligand for the KIR of the donor NK cell, has been proposed. /HLA mismatch is being effectively used because it can bring about more desirable results from the viewpoint of recurrence, GVHD (graft versus host disease, graft-versus-host disease) and prognosis. However, for the anti-tumor activity of NK cells, NK cells were cultured with feeder cells either matched or mismatched for inhibitory KIR ligands (lacking one or more ligands present in the NK cell donor). From a report (Non-patent Document 1) investigating to what extent the HLA class I-KIR interaction influences human NK cell proliferation in allogeneic environment (Non-patent Document 1), NK cell hyperactivation is indicated using an antitumor effect as an index. If desired, less signal from the KIR is suggested to be better. In addition, a recent report (Non-patent Document 2) on allogeneic responsiveness of HLA heterozygous NK cells (in the case of unidirectional matching) to tissues derived from HLA haplotype homozygous (HLA homo) iPS cells is related to culture of NK cells. , Strongly suggests that mixed culture containing HLA/KIR mismatch is not established.
特開2013−27385号公報(特許第5572863号、特許第5989016号)JP, 2013-27385, A (patent No. 5728863, patent No. 5989016). 特開2014−226079号公報(特許第5511039号、特許第6164650号)JP, 2014-226079, A (patent No. 5511039, patent No. 6164650). 特開2018−193303号公報JP, 2018-193303, A 特願2018−059624号出願明細書(本願優先日には未公開)Japanese Patent Application No. 2018-059624 Application specification (not disclosed on the priority date of the present application)
 本発明者らは、活性の高いNK細胞を含む細胞集団の開発を進めている。しかし、原料である末梢血やアフェレーシス法で得られる血液の量には限界があるため、治療用のNK細胞集団を将来の治療に備えてoff−the−shelf化しておくことができない。 The present inventors are developing a cell population containing highly active NK cells. However, since the amount of peripheral blood as a raw material and the amount of blood obtained by the apheresis method are limited, it is not possible to convert the therapeutic NK cell population into an off-the-shelf in preparation for future treatment.
 一方、本発明者らの検討によると、ドナーによってはNK細胞の体外での増幅効率が極めて悪い場合がある。 On the other hand, according to the studies by the present inventors, the in vitro amplification efficiency of NK cells may be extremely poor depending on the donor.
 さらに、体内であれば骨髄中の造血幹細胞から分化する過程において、リガンド(HLAクラスI)との結合を経験した未熟NK細胞が「ライセンス」され、成熟後に自己HLAクラスIの発現が低下した細胞を検知する「missing−self応答」の能力を備えると考えられているが、iPS細胞やES細胞から分化成熟させたNK細胞はライセンスに必要なシグナルが抗腫瘍効果を得るには十分でない可能性がある。 Furthermore, in the body, immature NK cells that have undergone binding with a ligand (HLA class I) are “licensed” in the process of differentiating from hematopoietic stem cells in the bone marrow, and the expression of autologous HLA class I is reduced after maturation. It is thought to have the ability of "missing-self response" to detect NK, but NK cells differentiated and matured from iPS cells or ES cells may not have sufficient signals required for license to obtain an antitumor effect. There is.
 本発明は、以下を提供する。
[1]複数のドナーに由来し、NK細胞を含む、単核細胞の集団を準備し、準備した単核細胞の集団をNK細胞処理上有効な条件でインキュベートすることを含む、NK細胞を含む細胞集団の製造方法。
[2]単核細胞の集団を準備する工程が、CD3陽性細胞を除去する工程を含む、1に記載の製造方法。
[3]単核細胞の集団を準備する工程が、CD34陽性細胞を除去する工程を含む、1又は2に記載の製造方法。
[4]単核細胞の集団を準備する工程が、複数のドナーから採取された末梢血から単核細胞の集団を得る工程を含む、1‐3のいずれか1項に記載の製造方法。
[5]単核細胞の集団を準備する工程が、複数のドナーから採取されたアフェレーシス血液から単核細胞の集団を得る工程を含む、1‐4のいずれか1項に記載の製造方法。
[6]単核細胞の集団を準備する工程が、複数のドナーに由来する、胚性幹(ES)細胞、人工多能性幹(iPS)細胞、及び成体幹細胞からなる群より選択されるいずれかに由来する単核細胞の集団を準備するものである、1‐3のいずれか1項に記載の製造方法。
[7]複数のドナーが、一のドナーと、それとはHLA及びKIRの少なくとも一方において遺伝子型が異なる他のドナーを含む、1‐6のいずれか1項に記載の製造方法。
[8]以下の特徴を備える、NK細胞を含む細胞集団:
(1)複数のドナーに由来する。
(2)NK細胞をエフェクター細胞(E)とし、K562細胞を標的細胞(T)として混合比(E:T)1:1で共培養した場合の細胞傷害活性が50%以上である。
[9]1‐7のいずれか1項に記載の製造方法によって製造される細胞集団を含む、細胞療法のための医薬組成物。
[10]8に記載の細胞集団を含む、細胞療法のための医薬組成物。
[11]感染症及び/又はがんを治療するための、9又は10に記載の医薬組成物。
The present invention provides the following.
[1] Including NK cells, comprising preparing a population of mononuclear cells derived from a plurality of donors and containing NK cells, and incubating the prepared population of mononuclear cells under conditions effective for treating NK cells Method for producing cell population.
[2] The production method according to 1, wherein the step of preparing a population of mononuclear cells includes the step of removing CD3-positive cells.
[3] The production method according to 1 or 2, wherein the step of preparing a population of mononuclear cells includes the step of removing CD34-positive cells.
[4] The production method according to any one of 1-3, wherein the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from peripheral blood collected from a plurality of donors.
[5] The production method according to any one of 1 to 4, wherein the step of preparing a population of mononuclear cells includes a step of obtaining a population of mononuclear cells from apheresis blood collected from a plurality of donors.
[6] Any of the step of preparing a population of mononuclear cells selected from the group consisting of embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, and adult stem cells derived from a plurality of donors The method for production according to any one of 1-3, which is for preparing a population of mononuclear cells derived from sea urchin.
[7] The production method according to any one of 1 to 6, wherein the plurality of donors include one donor and another donor having a genotype different from that of at least one of HLA and KIR.
[8] A cell population containing NK cells having the following characteristics:
(1) Derived from multiple donors.
(2) The cytotoxic activity is 50% or more when NK cells are used as effector cells (E) and K562 cells are used as target cells (T) at a mixing ratio (E:T) of 1:1.
[9] A pharmaceutical composition for cell therapy, comprising a cell population produced by the production method according to any one of 1-7.
[10] A pharmaceutical composition for cell therapy, comprising the cell population according to item 8.
[11] The pharmaceutical composition according to 9 or 10 for treating infectious disease and/or cancer.
 原料となる血液又はPBMCを複数人分混合することができるため、原材料量を増やすことができ、off−the shelf化が期待できる。 Since blood or PBMC as a raw material can be mixed for multiple people, the amount of raw material can be increased and off-the-shelf conversion can be expected.
 ドナーに拠らず安定してNK細胞を増殖でき、また増殖率を向上できる。 NK cells can be stably grown regardless of the donor, and the growth rate can be improved.
 HLA−KIRマッチングの多様化に対応可能となり、安定した活性化が期待できる。 ▽ Support for diversification of HLA-KIR matching and stable activation can be expected.
 NK細胞のライセンシングに必要なシグナルを柔軟に増やすことが可能となり、iPS細胞やES細胞由来のNK細胞集団の抗腫瘍効果の改善が期待できる。 It will be possible to flexibly increase the signals required for NK cell licensing, and it can be expected to improve the antitumor effect of the NK cell population derived from iPS cells or ES cells.
培養試験:複数人分のPBMCから、それぞれCD3陽性細胞を除去し、KBM−501培地を用いて14日間培養した。Culture test: CD3 positive cells were removed from PBMCs of a plurality of persons, and the cells were cultured for 14 days using KBM-501 medium. 実験結果のまとめ(左)と統計解析(右):混合培養を行うと、単独ドナー由来の培養と比較して優位に増殖率が向上した。Summary of experimental results (left) and statistical analysis (right): When mixed culture was performed, the growth rate was significantly improved as compared with the culture derived from a single donor. 腫瘍細胞傷害活性試験:混合培養と単独ドナー由来培養とで作成したNK細胞の傷害活性を、SKOV3(ヒト卵巣がん細胞株)に対する傷害を指標に評価した。Tumor cell cytotoxicity test: The cytotoxic activity of NK cells prepared by mixed culture and culture derived from a single donor was evaluated using the damage to SKOV3 (human ovarian cancer cell line) as an index. 500cm培養バッグによる培養。培養開始から30分後、及び培養9日目に、バッグを裏返す作業を行った。Culture with 500 cm 2 culture bag. After 30 minutes from the start of culturing and on the 9th day of culturing, the bag was flipped over. 培養試験。凍結アフェレーシス血液(2ドナー分)を材料とし、自動閉鎖系細胞処理装置を用いてCD3陽性細胞及びCD34陽性細胞を除去後、T75フラスコ又は500cm培養バッグを用い、KBM−501培地を用いて14日間培養した。Culture test. Frozen apheresis blood (for 2 donors) was used as a material, and after removing CD3 positive cells and CD34 positive cells using an automatic closed cell processing device, using a T75 flask or a 500 cm 2 culture bag, using KBM-501 medium Cultured for a day. 単球/NK細胞swapping実験:KIR3DL1、KIR3DS1からのシグナルが一方又は両方入る条件の場合にCD16highの集団が増えた。CD16highの集団は、より高いADCC活性が期待できる。Monocyte/NK cell swapping experiment: The population of CD16 high increased under the condition that one or both of the signals from KIR3DL1 and KIR3DS1 entered. Higher ADCC activity can be expected for the CD16 high population.
 本発明は、NK細胞を含む細胞集団に関する。 The present invention relates to a cell population containing NK cells.
[細胞集団の製造]
 本発明のNK細胞を含む細胞集団は、次の工程を含む方法で製造することができる:
 複数のドナーに由来し、NK細胞を含む、単核細胞の集団を準備し、
 準備した単核細胞の集団をNK細胞処理上有効な条件でインキュベートする。
[Production of cell population]
The NK cell-containing cell population of the present invention can be produced by a method including the following steps:
Preparing a population of mononuclear cells from multiple donors, including NK cells,
The prepared mononuclear cell population is incubated under conditions effective for treating NK cells.
(複数ドナーに由来する、単核細胞の集団の準備)
 本発明の細胞集団は、複数のドナーに由来する単核細胞の集団から得られる。ここでいう単核細胞の集団は、NK細胞を含み、NK細胞以外の単核細胞、例えば単球を含んでいてもよい。NK細胞は、初代NK細胞(生体から採取され、継代されていないNK細胞)であってもよい。
(Preparation of population of mononuclear cells derived from multiple donors)
The cell population of the present invention is obtained from a population of mononuclear cells derived from multiple donors. The population of mononuclear cells referred to herein includes NK cells, and may include mononuclear cells other than NK cells, for example, monocytes. The NK cells may be primary NK cells (NK cells collected from a living body and not passaged).
 複数のドナーに由来し、NK細胞を含む、単核細胞の集団は、一のドナーから得た単核細胞(NK細胞を含み、単球を含んでいてもよい。)の集団と、他のドナーから得た単核細胞(NK細胞を含み、単球を含んでいてもよい。)の集団が混合されたものである場合があり、また一のドナーから得たNK細胞と、他のドナーから得た単球とが混合されたものである場合がある。 The population of mononuclear cells derived from a plurality of donors and containing NK cells includes a population of mononuclear cells (containing NK cells and may contain monocytes) obtained from one donor, and another population. The population of mononuclear cells (including NK cells and may include monocytes) obtained from a donor may be a mixed one, and NK cells obtained from one donor and another donor may be mixed. It may be a mixture with monocytes obtained from.
 ドナーに関し、複数とは2以上であることをいい、上記の条件を満たせば特に限定されず、例えば、3以上、4以上、7以上、9以上のドナーに由来する単核細胞が混合されたものを用いることができる。複数のドナーには、NK細胞を含む細胞集団が投与される患者自身、該患者の近縁者が含まれる場合がある。 Regarding the donor, plural means two or more, and is not particularly limited as long as the above conditions are satisfied. For example, mononuclear cells derived from three or more, four or more, seven or more, and nine or more donors were mixed. One can be used. The plurality of donors may include the patient himself/herself who is administered the cell population containing NK cells, or a relative of the patient.
 複数のドナーは、一のドナーと、それとはHLA及びKIRの少なくとも一方において遺伝子型が異なる他のドナーを含むように選択することができる。KIRには、KIR2DL1、KIR2DL2、KIR2DL3、KIR3DL1、KIR3DL2、KIR2DL4、KIR2DS1、KIR2DS2、KIR2DS3、KIR2DS4、KIR2DS5、KIR3DS1、KIR3DL3、KIR2DL5A、KIR2DL5Bが含まれる。 Multiple donors can be selected to include one donor and other donors that differ in genotype in at least one of HLA and KIR. KIRs include KIR2DL1, KIR2DL2, KIR2DL3, KIR3DL1, KIR3DL2, KIR2DL4, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DS1, KIR3DL3, KIR2DL5A and KIR2DL5B.
 NK細胞に関し、ライセンス(ライセンシング)とは、NK細胞に特有の「missing−self応答(自己HLAの欠如の検出)」の能力を,細胞の分化成熟の過程で獲得するプロセスをいう。ヒトやマウスに関しては、骨髄中の造血幹細胞からNK細胞が分化する過程において、KIRやNKG2AとHLAクラスI分子の結合を経験した未熟NK細胞のみがライセンスされて成熟後に自己HLAクラスIの発現が低下した細胞を検知する能力を獲得すると考えられている。ライセンスに関わることが知られているKIRの例は、KIR2DL1、KIR2DL2、KIR2DL3,KIR3DL1及びNKG2A/CD94である。NK細胞が認識し得るHLAタイプは、古典的HLAのうち、HLA−C全アロタイプ、HLA−Bアロタイプの約1/3、HLA−Aアロタイプの一部(Bw4モティーフを有するもの)、非古典的HLAのHLA−E、HLA−Gである。本発明において、複数のドナーを、一のドナーと、それとはHLA及びKIRの少なくとも一方において遺伝子型が異なる他のドナーを含むように選択するに際しては、上記のようなライセンスに関わるKIR及びHLAを特に考慮することができる。 Regarding NK cells, licensing refers to the process of acquiring the "missing-self response (detection of lack of self HLA)" characteristic of NK cells in the process of cell differentiation and maturation. Regarding humans and mice, only immature NK cells that have undergone binding of HIR class I molecule with KIR or NKG2A in the process of differentiation of NK cells from hematopoietic stem cells in the bone marrow are licensed, and expression of self HLA class I after maturation is licensed. It is believed to acquire the ability to detect depressed cells. Examples of KIRs known to be involved in licenses are KIR2DL1, KIR2DL2, KIR2DL3, KIR3DL1 and NKG2A/CD94. Among classical HLA, HLA types that can be recognized by NK cells include HLA-C allotypes, about 1/3 of HLA-B allotypes, a part of HLA-A allotypes (those having a Bw4 motif), and non-classical HLA-E and HLA-G of HLA. In the present invention, when selecting a plurality of donors so as to include one donor and another donor having a genotype different from that of at least one of HLA and KIR, the KIR and HLA related to the license as described above are selected. Especially, it can be considered.
 本発明の好ましい態様の一つにおいては、NK細胞のライセンシングに供される分子のレパートリーを増やすように、遺伝子型の異なるドナーを選択することができる。また、NK細胞の抑制性KIRに刺激が入らないように、又は抑制性KIRに刺激が入るように選択してもよい。 In one of the preferred embodiments of the present invention, donors having different genotypes can be selected so as to increase the repertoire of molecules used for licensing NK cells. Alternatively, the inhibitory KIR of NK cells may be selected not to be stimulated, or the inhibitory KIR may be selected to be stimulated.
 ドナーの選択は、NK細胞による抗体依存性細胞傷害(Antibody dependent cellular cytotoxicity、ADCC)活性を期待する場合は、混合した単核細胞の集団を培養した際に、CD16highであるNK細胞が増えるように行ってもよい。NK細胞による細胞傷害の機構の一つとして、ADCCが知られているが、NK細胞はその細胞表面上のFc受容体であるCD16を介して、標的細胞に結合した抗体に結合し、標的細胞の傷害を行う。そのため、CD16highであるNK細胞が増えることにより、ADCC活性がより高くなることが期待できる。このようなドナーの組み合わせの条件の例は、NK細胞にKIR3DL1及びKIR3DS1の一方又は両方からのシグナルによるライセンシングが行われるようにドナーを選択して混合することである。 When anticipating antibody-dependent cellular cytotoxicity (ADCC) activity by NK cells, the selection of donors is such that NK cells that are CD16 high are increased when a mixed mononuclear cell population is cultured. You may go to ADCC is known as one of the mechanisms of cytotoxicity by NK cells. NK cells bind to an antibody bound to the target cell via CD16, which is an Fc receptor on the cell surface, Injuries. Therefore, it can be expected that the ADCC activity becomes higher by increasing the number of NK cells that are CD16 high . An example of such a donor combination condition is to select and mix donors so that NK cells are licensed by a signal from one or both of KIR3DL1 and KIR3DS1.
 混合は、NK細胞を対象へ投与する前に行われる。混合は、目的とする混合による効果が得られる限り、種々の段階で行うことができる。例えば、複数のドナーから採取された血液を混合し、次に混合したものから単核細胞の集団を得てもよく、また各ドナーから採取された血液から単核細胞の集団を各々得た後に、各集団を混合してもよい。また、複数のドナーから採取された単核細胞の集団を各々培養した後に、各集団を混合してもよい。増殖が良好になるとの観点からは、混合は培養前に行うことが好ましい。 Mixing is performed before administering NK cells to the subject. The mixing can be carried out in various stages as long as the desired effects can be obtained. For example, blood taken from multiple donors may be mixed, and then the mixture may be used to obtain a population of mononuclear cells. Alternatively, blood collected from each donor may be used to obtain a population of mononuclear cells, respectively. , The populations may be mixed. Alternatively, the populations of mononuclear cells collected from a plurality of donors may be cultured and then the populations may be mixed. From the viewpoint of good growth, mixing is preferably performed before culturing.
 複数ドナーに由来する、単核細胞の集団の混合割合は、適宜とすることができる。例えば、複数のドナー由来の細胞が、ほぼ等しい割合で含まれるようにすることができ、また特定のドナーに由来する細胞を、多く、又は少なく含まれるようにすることができる。一のドナー由来のNK細胞と他のドナー由来の単球を混合する場合も、混合割合は適宜とすることができる。例えば、NK細胞1に対し、その1−9倍の単球を混合することができ(NK細胞:単球=1:1‐9)、またNK細胞1に対し、その2‐4倍の単球を混合することができる(NK細胞:単球=1:2‐4)。本発明に関し、細胞の混合割合を表すときは、特に記載した場合を除き、細胞の数に基づいている。 The mixing ratio of the population of mononuclear cells derived from multiple donors can be set appropriately. For example, cells from multiple donors can be included in approximately equal proportions, and cells from a particular donor can be included in greater or lesser proportion. When mixing NK cells derived from one donor and monocytes derived from another donor, the mixing ratio can be set appropriately. For example, 1 to 9 times as many monocytes as NK cells 1 can be mixed (NK cells:monocytes=1:1-9), and 2 to 4 times as many monocytes as NK cells 1 can be mixed. The spheres can be mixed (NK cells:monocytes=1:2-4). In the context of the present invention, the mixing ratio of cells is based on the number of cells, unless otherwise stated.
 単核細胞の集団を得る原料としては、末梢血、臍帯血、骨髄及び/又はリンパ節から採取される血液細胞を用いることができる。好ましい原料の一つが末梢血であり、末梢血はアフェレーシス法により採取してもよい。すなわち、本発明の好ましい態様においては、単核細胞の集団を準備する工程は、複数のドナーから採取された末梢血から単核細胞の集団を得る工程を含む。また、別の好ましい態様においては、単核細胞の集団を準備する工程は、複数のドナーから採取されたアフェレーシス血液から単核細胞の集団を得る工程を含む。 As a raw material for obtaining a population of mononuclear cells, blood cells collected from peripheral blood, cord blood, bone marrow and/or lymph node can be used. One of the preferable raw materials is peripheral blood, which may be collected by the apheresis method. That is, in a preferred embodiment of the present invention, the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from peripheral blood collected from a plurality of donors. In another preferred embodiment, the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from apheresis blood collected from a plurality of donors.
 NK細胞と単球を含む、単核細胞の集団は、当業者に知られたさまざまな手順を用いて調製することができる。例えば、末梢血及び臍帯血のような血液からは、室温条件で密度遠心分離を行うことで赤血球の除去及び単核球細胞画分の回収が行える。末梢血から分離される単核細胞PBMC(Peripheral Blood Mononuclear Cells)は、T細胞、B細胞、NK細胞、単球及び樹状細胞などの多様なリンパ球を含む。本発明に関し、CD3陰性であり、かつCD56陽性である単核細胞をNK細胞ということができる。NK細胞は、当業者に知られたさまざまな手順を用いて調製することができる。例えば、全血又はPBMCから、不要な細胞を除いてNK細胞を分離するための方法が知られている。単球は、当業者に知られたさまざまな手順を用いて調製することができる。例えば全血又はPBMCから不要な細胞を除いて単球を分離するための方法が知られている。 A population of mononuclear cells, including NK cells and monocytes, can be prepared using various procedures known to those of skill in the art. For example, from blood such as peripheral blood and umbilical cord blood, red blood cells can be removed and a mononuclear cell fraction can be collected by performing density centrifugation at room temperature. Peripheral Blood Mononuclear Cells (PBMC) isolated from peripheral blood include various lymphocytes such as T cells, B cells, NK cells, monocytes and dendritic cells. In the context of the present invention, mononuclear cells that are CD3 negative and CD56 positive can be called NK cells. NK cells can be prepared using various procedures known to those of skill in the art. For example, a method for separating NK cells by removing unnecessary cells from whole blood or PBMC is known. Monocytes can be prepared using various procedures known to those of skill in the art. For example, a method for separating monocytes by removing unnecessary cells from whole blood or PBMC is known.
(CD3陽性細胞の除去、CD34陽性細胞の除去)
 単核細胞の集団は、単球、NK細胞以外に、NK細胞前駆体、T細胞、NKT細胞、造血前駆細胞等を含む場合がある。所望のNK細胞が、増幅後、例えば、比重遠心法、免疫磁気ビーズ、FACS、フロー・サイトメトリー等を用いて選択される場合がある。例えば、NK細胞は、抗CD3抗体、抗CD16抗体、抗CD34抗体、抗CD56抗体、抗CD69抗体、抗CD94抗体、抗CD107a抗体、抗KIR3DL1抗体、抗KIR3DL2抗体、抗KIR2DL3抗体、抗KIR2DL1抗体、抗KIR2DS1抗体、抗KIR2DL5抗体、抗NKp46抗体、抗NKp30抗体、抗NKG2D抗体等を用いて、細胞集団から選択的に分離される場合がある。抗体は、モノクローナル抗体、ポリクローナル抗体等の場合がある。NK細胞の選択は、T細胞、NKT細胞、造血前駆細胞その他の細胞を選択的に除去して行われる場合がある。
(Removal of CD3 positive cells, removal of CD34 positive cells)
The population of mononuclear cells may include NK cell precursors, T cells, NKT cells, hematopoietic progenitor cells, etc. in addition to monocytes and NK cells. The desired NK cells may be selected after amplification using, for example, specific gravity centrifugation, immunomagnetic beads, FACS, flow cytometry, etc. For example, NK cells include anti-CD3 antibody, anti-CD16 antibody, anti-CD34 antibody, anti-CD56 antibody, anti-CD69 antibody, anti-CD94 antibody, anti-CD107a antibody, anti-KIR3DL1 antibody, anti-KIR3DL2 antibody, anti-KIR2DL3 antibody, anti-KIR2DL1 antibody, It may be selectively separated from the cell population using an anti-KIR2DS1 antibody, an anti-KIR2DL5 antibody, an anti-NKp46 antibody, an anti-NKp30 antibody, an anti-NKG2D antibody or the like. The antibody may be a monoclonal antibody, a polyclonal antibody, or the like. The selection of NK cells may be performed by selectively removing T cells, NKT cells, hematopoietic progenitor cells and other cells.
 単核細胞の集団を準備する工程は、T細胞を除去する工程を含んでいてもよく、この工程はCD3陽性細胞を除去することにより達成される場合がある。また単核細胞の集団を準備する工程は、造血前駆細胞を除去するステップを含んでいてもよく、この工程は、CD34陽性細胞を除去することにより達成される場合がある。すなわち、単核細胞の集団を準備する工程は、CD3陽性細胞を除去する工程を含んでいてもよく、CD34陽性細胞を除去する工程を含んでいてもよい。 The step of preparing a population of mononuclear cells may include the step of removing T cells, which may be accomplished by removing CD3-positive cells. The step of preparing a population of mononuclear cells may also include the step of removing hematopoietic progenitor cells, which may be accomplished by removing CD34-positive cells. That is, the step of preparing a population of mononuclear cells may include the step of removing CD3-positive cells or the step of removing CD34-positive cells.
 CD3陽性細胞の除去、CD34陽性細胞の除去は、例えば、Invitrogen社から販売されるDynal Biotech社製Dynabeadsや、ミルテニーバイオテク社のCliniMACS等の免疫磁気ビーズを用いて、細胞表面抗原CD3及び/又はCD34を発現する細胞を分離除去することにより実施できる。このような工程は、NK細胞を疲弊させないように、また単球が余計な貪食機能を発揮しないように行うことが好ましい。具体的には、比較的小型の磁気ビーズを用いる(細胞と結合していない未反応のビーズを遠心操作により上清とともに除去することができる。)こと、ビーズと細胞集団との低温でのインキュベート時間を比較的短く設定すること、未反応のビーズを除いた上でカラムを用いてビーズと結合していない細胞を目的の細胞集団として得ること、等を好ましい操作として挙げることができる。細胞の除去は、自動閉鎖系細胞処理装置を用いて行ってもよい。 The removal of CD3-positive cells and the removal of CD34-positive cells can be performed, for example, by using Dynabeads manufactured by Invitrogen, Dynabeads manufactured by Invitrogen, or immunomagnetic beads such as CliniMACS manufactured by Miltenyi Biotech, and the cell surface antigen CD3 and/or It can be carried out by separating and removing cells expressing CD34. Such a step is preferably performed so as not to exhaust NK cells and prevent monocytes from exerting an extra phagocytic function. Specifically, using relatively small magnetic beads (unreacted beads that are not bound to cells can be removed together with the supernatant by centrifugation), and the beads and cell population are incubated at low temperature. Examples of preferable operations include setting the time relatively short, removing unreacted beads, and then using a column to obtain cells not bound to the beads as a target cell population. The cells may be removed using an automatic closed cell processing apparatus.
(iPS細胞等の利用)
 単核細胞の集団は、胚性幹(ES)細胞、人工多能性幹(iPS)細胞、及び成体幹細胞からなるグループから選択されるいずれかに由来する造血幹細胞から調製されたものであってもよい。これらの幹細胞からNK細胞を含む単核細胞の細胞を誘導する方法が知られており、当業者であれば、本発明に適用することができる(Domogala A.et al.,Natural killer cell immunotherapy:from bench to bedside,Frontiers in Immunology,2015;6,doi:10.3389/fimmu.2015.00264、及びZeng J.et al.,Generation of”Off−the−Shelf”Natural Killer Cells from Peripheral Blood Cell−Derived Induced Pluripotent Stem Cells,Stem Cell Reports,2017;9:1796−1812)。幹細胞は、また高活性のNK細胞が得られるように改変してもよく、例えば、高親和性CD16(158V:158番アミノ酸がバリン)が知られており、このような知見を適用することができる。
(Use of iPS cells, etc.)
The population of mononuclear cells is prepared from hematopoietic stem cells derived from any one selected from the group consisting of embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, and adult stem cells. Good. Methods for inducing mononuclear cells including NK cells from these stem cells are known and can be applied to the present invention by those skilled in the art (Domogala A. et al., Natural killer cell immunotherapy: from bench to bedside, Frontiers in Immunology, 2015; 6, doi: 10.3389/fimmu.2015.0264, and Zeng J. et al., Generation of Shelf "Natural berrelloller Celloller Killer". Derived Induced Pluripotent Stem Cells, Stem Cell Reports, 2017; 9: 1796-1812). Stem cells may also be modified so that highly active NK cells can be obtained. For example, high-affinity CD16 (158V: 158th amino acid is valine) is known, and such findings can be applied. it can.
(単核細胞の集団の培養)
 準備した単核細胞の集団は、複数のドナーに由来する細胞が混合された状態で、NK細胞処理上有効な条件でインキュベートされる。インキュベート(すること)は、細胞の増殖を伴わない場合と伴う場合がある。本発明の好ましい態様の一つにおいては、インキュベート(すること)は増殖を伴う。増殖を伴うインキュベート(すること)は、培養(すること)と、又は増殖(させること)と読み替えることができる。
(Culturing a population of mononuclear cells)
The prepared population of mononuclear cells is incubated in a state in which cells derived from a plurality of donors are mixed under conditions effective for NK cell treatment. Incubation may or may not be accompanied by cell proliferation. In one of the preferred embodiments of the invention, the incubating is accompanied by growth. Incubation with proliferation can be read as culturing or proliferation.
 NK細胞処理上有効な条件とは、複数のドナーに由来する混合された細胞が、目的とする混合による効果を奏するのに適した条件をいう。目的の効果が奏される限り、細胞懸濁のために使用する培地又は等張液、時間、温度、環境等に、特に限定はない。 “Effective conditions for NK cell treatment” means conditions suitable for mixed cells derived from a plurality of donors to exert the desired mixing effect. The medium or isotonic solution used for cell suspension, time, temperature, environment, etc. are not particularly limited as long as the desired effects are exhibited.
 NK細胞処理上有効な条件は、NK細胞の活性化上有効な条件を含む。このような条件は例えば、適切な濃度の活性化に必要な因子、例えばIL−2及びIL−15の存在下で、37℃、5%CO及び飽和水蒸気雰囲気下で、4−18時間インキュベートすることが含まれる。 The condition effective for NK cell treatment includes the condition effective for activation of NK cell. Such conditions include, for example, incubation in the presence of appropriate concentrations of activation factors such as IL-2 and IL-15 at 37° C., 5% CO 2 and saturated steam atmosphere for 4-18 hours. For example.
 NK細胞処理上有効な条件は、NK細胞の増殖上有効な条件を含む。NK細胞増殖上有効な条件は、NK細胞の増殖に適した培地を用いることを含む。そのような培地の例は、KBM501培地(コージンバイオ株式会社)、CellGro SCGM培地(セルジェニックス、岩井化学薬品株式会社)、X−VIVO15培地(ロンザ、タカラバイオ株式会社)、IMDM、MEM、DMEM、RPMI−1640等を含むが、これらに限定されない。 Effective conditions for NK cell treatment include effective conditions for NK cell proliferation. Effective conditions for the growth of NK cells include using a medium suitable for the growth of NK cells. Examples of such a medium include KBM501 medium (Kojin Bio Co., Ltd.), CellGro SCGM medium (Celgenics, Iwai Chemical Co., Ltd.), X-VIVO15 medium (Lonza, Takara Bio Inc.), IMDM, MEM, DMEM. , RPMI-1640 and the like, but are not limited thereto.
 培地には、インターロイキン2(IL−2)が、本発明の目的を達成できる濃度で添加される場合がある。IL−2の濃度は、2000IU/mLを超える場合があり、2500−2813IU/mLの場合がある。IL−2は、ヒトのアミノ酸配列を有することが好ましく、安全上、組換えDNA技術で生産されることが好ましい。IL−2の濃度は、国内標準単位(JRU)及び国際単位(IU)で示される場合がある。1IUが約0.622JRUであるから、1750JRU/mLは約2813IU/mLである。 Interleukin 2 (IL-2) may be added to the medium at a concentration that can achieve the object of the present invention. The concentration of IL-2 can be greater than 2000 IU/mL and can be 2500-2815 IU/mL. IL-2 preferably has a human amino acid sequence, and is preferably produced by recombinant DNA technology for safety. The concentration of IL-2 may be indicated in national standard units (JRU) and international units (IU). Since 1 IU is about 0.622 JRU, 1750 JRU/mL is about 2813 IU/mL.
 培地には、被験者の自家血清、BioWhittaker社その他から入手可能なヒトAB型血清や、日本赤十字社から入手可能な献血ヒト血清アルブミンが添加される場合がある。自家血清及びヒトAB型血清は1‐10%の濃度で添加されることが好ましく、献血ヒト血清アルブミンは1‐10%の濃度で添加されることが好ましい。被験者は、健常者と、疾患に罹患した患者との場合がある。また、培地には血清に替えて又は血清とともに、免疫細胞の増殖のために配合された組成物が添加される場合がある。このような組成物は市販されている。例えばUltraGroシリーズ(AventaCell社)、CTS Immune Cell SR(Thermo Fisher Scientific社)を本発明のためにも用いることができる。 The subject's autologous serum, human AB type serum available from BioWhittaker or others, or blood donated human serum albumin available from the Japanese Red Cross Society may be added to the medium. Autologous serum and human AB-type serum are preferably added at a concentration of 1-10%, and donated human serum albumin is preferably added at a concentration of 1-10%. The test subject may be a healthy person or a patient suffering from a disease. Further, a composition formulated for the growth of immune cells may be added to the medium instead of or together with the serum. Such compositions are commercially available. For example, UltraGro series (AventaCell), CTS Immune Cell SR (Thermo Fisher Scientific) can be used for the present invention.
 培地には、NK細胞の増幅効果を損なわないことを条件として、適切なタンパク質、サイトカイン、抗体、化合物その他の成分が含まれる場合がある。サイトカインは、インターロイキン3(IL−3)、インターロイキン7(IL−7)、インターロイキン12(IL−12)、インターロイキン15(IL−15)、インターロイキン21(IL−21)、幹細胞因子(SCF)、及び/又は、FMS様チロシンキナーゼ3リガンド(Flt3L)の場合がある。IL−3、IL−7、IL−12、IL−15、IL−21、SCF及びFlt3Lは、ヒトのアミノ酸配列を有することが好ましく、安全上、組換えDNA技術で生産されることが好ましい。 The culture medium may contain appropriate proteins, cytokines, antibodies, compounds and other components, provided that the amplification effect of NK cells is not impaired. Cytokines include interleukin 3 (IL-3), interleukin 7 (IL-7), interleukin 12 (IL-12), interleukin 15 (IL-15), interleukin 21 (IL-21), stem cell factor. (SCF) and/or FMS-like tyrosine kinase 3 ligand (Flt3L). IL-3, IL-7, IL-12, IL-15, IL-21, SCF and Flt3L preferably have a human amino acid sequence, and are preferably produced by recombinant DNA technology for safety.
 培地の交換は、所望のNK細胞の細胞数が得られることを条件として、培養開始後いつ行われてもかまわないが、3‐5日毎が好ましい。 The medium may be replaced at any time after the start of culture, provided that the desired number of NK cells is obtained, but it is preferably every 3-5 days.
 培養容器は、商業的に入手可能なディッシュ、フラスコ、プレート、マルチウェルプレートを含むが、これらに限定されない。治療用のNK細胞を含む細胞集団を得るためには、多くの細胞が培養でき、取り扱いが容易である培養容器を用いることが好ましい。このような培養容器の例として、高いガス透過性を有した素材からなる細胞培養用バッグが挙げられる。 Culture vessels include, but are not limited to, commercially available dishes, flasks, plates, multiwell plates. In order to obtain a cell population containing NK cells for treatment, it is preferable to use a culture container in which many cells can be cultured and which is easy to handle. An example of such a culture container is a cell culture bag made of a material having high gas permeability.
 バッグを用いる場合、培養期間中に上下を反転しながら用いることが好ましい。培養の際、NK細胞、単球とも培養面への接着がみられる。接着系の細胞の場合、単位容積当たり細胞密度とともに単位面積当たりの細胞密度が、細胞の生存率及び増殖速度を含む培養効率に大きく影響する。培養期間中にバッグを反転させることにより、それまで上側であったために細胞の接着が比較的少ない面へ一部のNK細胞と単球を移動させることができるため、培養をより効率的に行うことができる。 When using a bag, it is preferable to use it while turning it upside down during the culture period. During culturing, both NK cells and monocytes adhere to the culture surface. In the case of adherent cells, the cell density per unit volume as well as the cell density per unit volume greatly influences the culture efficiency including cell viability and growth rate. By inverting the bag during the culture period, it is possible to move some NK cells and monocytes to the surface where cell adhesion was relatively small because it was on the upper side until then, so that the culture is performed more efficiently. be able to.
 培養条件は、NK細胞の増幅効果を損なわないことを条件として特に限定されないが、37℃、5%CO及び飽和水蒸気雰囲気下の培養条件が一般的である。本発明の目的の一つはNK細胞を大量に調製することであるため、培地で培養する期間が長いほどより多くのNK細胞が得られるので有利である。培養期間は、NK細胞を所望の細胞数まで増幅することを条件として、特に限定されない。例えば、7‐28日間行うことができ、10‐18日間としてもよく、12‐16日間、例えば14日としてもよい(Saito S.et al.,Ex vivo generation of highly purified and activated natural killer cells from human peripheral blood.Hum Gene Ther Methods.2013;24(4):241−252、及び前掲特許文献1)。 The culture conditions are not particularly limited as long as the amplification effect of NK cells is not impaired, but the culture conditions are generally 37° C., 5% CO 2 and saturated steam atmosphere. Since one of the objects of the present invention is to prepare a large amount of NK cells, it is advantageous that the longer the culture period is, the more NK cells can be obtained. The culture period is not particularly limited, provided that the NK cells are expanded to a desired cell number. For example, it may be performed for 7 to 28 days, may be 10 to 18 days, may be 12 to 16 days, for example, 14 days (Saito S. et al., Ex vivo generation of highly purified and activated natural killer cells from. human peripheral blood.Hum Gene Ther Methods.2013;24(4):241-252, and the above-mentioned Patent Document 1).
 培養は、単核細胞の集団が準備された後に直ちに実施しなくてもよい。準備した単核細胞を含む細胞集団は凍結保存され、患者への投与時期に応じて解凍され、NK細胞の培養に供される場合がある。なお、単核細胞の集団は、本発明のNK細胞の増幅方法によって増幅される途中か、増幅が終わった後かに凍結され、患者への移植時期に応じて解凍され、患者への移植に供されてもよい。凍結及び解凍は当業者に周知のいかなる方法を用いてもかまわない。細胞の凍結には、いずれかの市販の細胞凍結保存液が用いられる場合がある。 Culturing need not be carried out immediately after the population of mononuclear cells is prepared. The prepared cell population containing mononuclear cells may be cryopreserved, thawed depending on the time of administration to a patient, and then provided for NK cell culture. The population of mononuclear cells is frozen during or after amplification by the NK cell expansion method of the present invention, thawed according to the time of transplantation into a patient, and then thawed. May be offered. Freezing and thawing may use any method known to those skilled in the art. Any commercially available cell cryopreservation solution may be used for freezing the cells.
 培養により得られた所定のNK細胞の集団は、目的のNK細胞に加えて、NK細胞前駆体、T細胞、NKT細胞、造血前駆細胞等を含む場合がある。培養後、目的のNK細胞、又はその集団が、例えば比重遠心法、免疫磁気ビーズ、FACS、フローサイトメトリー等を用いて選択される場合がある。例えば、抗CD3抗体、抗CD16抗体、抗CD34抗体、抗CD56抗体、抗CD69抗体、抗CD94抗体、抗CD107a抗体、抗KIR3DL1抗体、抗KIR3DL2抗体、抗KIR2DL3抗体、抗KIR2DL1抗体、抗KIR2DS1抗体、抗KIR2DL5抗体、抗NKp46抗体、抗NKp30抗体、抗NKG2D抗体等を用いて、目的のNK細胞、又はその集団が選択的に分離される場合がある。抗体は、モノクローナル抗体、ポリクローナル抗体等の場合がある。目的のNK細胞、又はその集団の選択は、T細胞、NKT細胞、造血前駆細胞その他の細胞を選択的に除去して行われる場合がある。 The predetermined NK cell population obtained by culture may contain NK cell precursors, T cells, NKT cells, hematopoietic progenitor cells, etc. in addition to the target NK cells. After culturing, the NK cells of interest or a population thereof may be selected using, for example, specific gravity centrifugation, immunomagnetic beads, FACS, flow cytometry, or the like. For example, anti-CD3 antibody, anti-CD16 antibody, anti-CD34 antibody, anti-CD56 antibody, anti-CD69 antibody, anti-CD94 antibody, anti-CD107a antibody, anti-KIR3DL1 antibody, anti-KIR3DL2 antibody, anti-KIR2DL3 antibody, anti-KIR2DL1 antibody, anti-KIR2DS1 antibody, The target NK cells or a population thereof may be selectively isolated using anti-KIR2DL5 antibody, anti-NKp46 antibody, anti-NKp30 antibody, anti-NKG2D antibody or the like. The antibody may be a monoclonal antibody, a polyclonal antibody, or the like. The NK cell of interest or a population thereof may be selected by selectively removing T cells, NKT cells, hematopoietic progenitor cells and other cells.
(混合培養の効果)
 本発明者らの検討によると、複数のドナーに由来し、NK細胞を含む、単核細胞の集団を培養することにより、NK細胞の増殖が良好となる。これは、HLA/KIRの組み合わせのバリエーションが増えることで、より多くのライセンシングシグナルが互いに入るようになったことに起因すると考えられる。増殖に関し良好とは、混合培養の複数のドナーのうちのいずれか一の単独ドナー由来の細胞を培養した場合に比較して、細胞の増殖率(培養後の細胞数/培養前の細胞数)が向上していることをいう。
(Effect of mixed culture)
According to the study by the present inventors, NK cell proliferation is improved by culturing a population of mononuclear cells derived from a plurality of donors and containing NK cells. It is considered that this is because more variations of the HLA/KIR combination resulted in more licensing signals entering each other. Good growth is compared with the case of culturing cells derived from any one of a plurality of donors in mixed culture, and the proliferation rate of cells (the number of cells after culture/the number of cells before culture) Is improving.
 また、混合培養により得られた細胞集団においては、70%以上、好ましくは80%以上、より好ましくは90%以上が、NK細胞でありうる。 Further, in the cell population obtained by the mixed culture, 70% or more, preferably 80% or more, more preferably 90% or more may be NK cells.
 また本発明者らの検討によると、複数ドナー由来のNK細胞を含む、単核細胞の集団の混合培養により、得られるNK細胞の細胞傷害活性は、単独ドナー由来のNK細胞と比較して、同等以上でありうる。さらにCD16高発現性であるNK細胞が含まれる割合も、単独ドナー由来のNK細胞と比較して、同等以上でありうる。細胞傷害活性とは、特に記載した場合を除き、対象細胞(エフェクター細胞、E)の標的細胞(T)に対する溶解能を指す。細胞傷害活性は、エフェクター細胞により死に至った標的細胞の百分率(%)で表すことができ、例えば次式により求められる。 Further, according to the study by the present inventors, the cytotoxic activity of NK cells obtained by mixed culture of a population of mononuclear cells containing NK cells derived from a plurality of donors is higher than that of NK cells derived from a single donor. It can be equal or better. Further, the proportion of NK cells that highly express CD16 may be equal to or higher than that of NK cells derived from a single donor. The cytotoxic activity refers to the lytic ability of target cells (effector cells, E) to target cells (T), unless otherwise specified. The cytotoxic activity can be expressed as a percentage (%) of target cells that have been killed by effector cells, and is calculated by the following equation, for example.
(エフェクター細胞と共培養した場合の細胞死−自然細胞死(陰性コントロール))/(最大細胞死(陽性コントロール)−自然細胞死(陰性コントロール))×100 (Cell death when co-cultured with effector cells-natural cell death (negative control))/(maximal cell death (positive control)-natural cell death (negative control)) x 100
 細胞傷害活性の測定に際しては、一般的には、エフェクター細胞の細胞傷害活性の程度等に応じ、エフェクター細胞と標的細胞の混合比(E:T)、エフェクター細胞と標的細胞の共培養の時間は、用いる細胞の種類や活性の強さに応じて適宜とすることができる。NK細胞をエフェクター細胞とするとき、標的細胞は、K562細胞、SKOV3細胞(ヒト卵巣がん細胞株)、急性骨髄性白血病細胞、慢性骨髄性白血病細胞の場合があるが、これらに限定されない。エフェクター細胞と標的細胞、生細胞と死細胞は、放射性物質、蛍光色素等で標識した抗体等の試薬により、区別し、また定量することができる。 When measuring the cytotoxic activity, generally, depending on the degree of the cytotoxic activity of the effector cells, the mixing ratio (E:T) of the effector cells to the target cells and the co-culture time of the effector cells to the target cells are Can be appropriately determined depending on the type of cells used and the strength of the activity. When NK cells are used as effector cells, the target cells may be, but are not limited to, K562 cells, SKOV3 cells (human ovarian cancer cell line), acute myelogenous leukemia cells, and chronic myelogenous leukemia cells. Effector cells and target cells, and living cells and dead cells can be distinguished and quantified by reagents such as radioactive substances and antibodies labeled with fluorescent dyes.
 また本発明による複数のドナーに由来する本発明のNK細胞の集団は、MDSC(Myeloid−derived suppressor cells)による抑制を受けないか、又はその抑制が著しく低い。本発明者らは、これまでの研究から、上述のような培養方法で得られた単独のドナーに由来するNK細胞の集団は、MDSCによる抑制を受けないことを確認してきた。この理由の一つとして、液性因子(TGF−β,IL−10)に対する受容体の発現が無いか、又は著しく低いことが挙げられる。本発明者らの今般の検討によると、また複数のドナーからの細胞を混合する場合であっても、上記の利点は損なわれないことが分かっている。 Further, the population of NK cells of the present invention derived from a plurality of donors of the present invention is not suppressed by MDSC (Myeloid-delivered suppressor cells), or its suppression is extremely low. From the previous studies, the present inventors have confirmed that the population of NK cells derived from a single donor obtained by the above-mentioned culture method is not suppressed by MDSC. One of the reasons for this is that there is no or extremely low expression of receptors for humoral factors (TGF-β, IL-10). According to the present studies by the present inventors, it has been found that the above advantages are not impaired even when cells from a plurality of donors are mixed.
[NK細胞を含む細胞集団]
 本発明により得られる、NK細胞を含む細胞集団は、以下の特徴を備える:
(1)複数のドナーに由来する。
(2)NK細胞をエフェクター細胞(E)とし、K562細胞を標的細胞(T)として混合比(E:T)1:1で共培養した場合の細胞傷害活性が50%以上である。
 上記の(2)の特徴の代わりに、または(2)の特徴ともに、下記の特徴を備えていてもよい:
(2’)SKOV3細胞を標的細胞(T)として混合比(E:T)3:1で共培養した場合の細胞傷害活性が50%以上である。
[Cell population including NK cells]
The cell population containing NK cells obtained by the present invention has the following characteristics:
(1) Derived from multiple donors.
(2) The cytotoxic activity is 50% or more when NK cells are used as effector cells (E) and K562 cells are used as target cells (T) at a mixing ratio (E:T) of 1:1.
Instead of the above feature (2) or together with the feature (2), the following features may be provided:
The cytotoxic activity is 50% or more when (2′) SKOV3 cells are co-cultured with target cells (T) at a mixing ratio (E:T) of 3:1.
 このような細胞集団はさらに、下記の特徴を備えていてもよい。
(3)NK細胞の割合が70%以上、より特定すると80%以上、さらに特定すると90%以上である。
(4)CD16高発現性であるNK細胞の集団、及びCD16低発現性であるNK細胞の集団の双方を含んでいてもよい。CD16高発現性であるNK細胞の割合が、50%以上である。
Such a cell population may further have the following characteristics.
(3) The proportion of NK cells is 70% or more, more specifically 80% or more, and further specifically 90% or more.
(4) It may include both a population of NK cells having high CD16 expression and a population of NK cells having low CD16 expression. The proportion of NK cells that highly express CD16 is 50% or more.
 NK細胞の細胞傷害活性は、当業者に周知の方法によって測定し、算出できる。細胞傷害活性(%)は、通常、エフェクター細胞(E)作用後の標的細胞(T)の生細胞数に基づき、例えば、式:(1−生細胞数/陰性コントロール生細胞数)×100により算出できる。
 NK細胞をエフェクター細胞(E)とし、K562細胞を標的細胞(T)として混合比(E:T)1:1で共培養した場合の細胞傷害活性は、60%以上であることが好ましく、70%以上であることがより好ましく、80%以上であることがさらに好ましく、90%以上であることがさらに好ましく、95%以上であることがさらに好ましい。
 NK細胞をエフェクター細胞(E)とし、SKOV3細胞を標的細胞(T)として混合比(E:T)3:1で共培養した場合の細胞傷害活性は、60%以上であることが好ましく、70%以上であることがより好ましく、80%以上であることがさらに好ましく、90%以上であることがさらに好ましく、95%以上であることがさらに好ましい。
The cytotoxic activity of NK cells can be measured and calculated by a method well known to those skilled in the art. The cytotoxic activity (%) is usually based on the viable cell number of the target cell (T) after the action of the effector cell (E), for example, by the formula: (1-viable cell number/negative control viable cell number)×100. Can be calculated.
The cytotoxic activity of NK cells as effector cells (E) and K562 cells as target cells (T) at a mixing ratio (E:T) of 1:1 is preferably 60% or more, 70 % Or more, more preferably 80% or more, still more preferably 90% or more, still more preferably 95% or more.
The cytotoxic activity of NK cells as effector cells (E) and SKOV3 cells as target cells (T) at a mixing ratio (E:T) of 3:1 is preferably 60% or more, 70 % Or more, more preferably 80% or more, still more preferably 90% or more, still more preferably 95% or more.
 CD16等のマーカーに関し、陽性であることは、+で表され、陰性であることは−で表されることがある。例えば、CD16陽性は、CD16と表され、CD16陰性は、CD16と表されることがある。陽性は、高発現性である場合と低発現性である場合を含む。高発現性であることは、high、brightと表されることがある。例えば、CD16高発現性は、CD16high、CD16brightと表されることがある。低発現性であることは、low、dimと表されることがある。例えば、CD16低発現性は、CD16low、CD16dimと表されることがある。 Regarding markers such as CD16, a positive result may be represented by + and a negative result may be represented by-. For example, CD16 positive may be designated as CD16 + and CD16 negative may be designated as CD16 . Positive cases include cases of high expression and cases of low expression. High expression may be expressed as high or bright. For example, high CD16 expression may be expressed as CD16 high or CD16 bright . Low expression may be expressed as low and dim. For example, CD16 low expression may be represented as CD16 low , CD16 dim .
 陽性、陰性、高発現性、低発現性は、フローサイトメトリー法により得られるチャートに基づき、判断することができる。チャートに現れる位置は、機器の電圧設定、感度設定、使用抗体クローン、染色条件、使用色素等により変動することがあるが、当業者であれば、得られたチャートにおいて一群と認められる細胞集団を切り分けないように適宜線引きすることができる。 Positive, negative, high expression, low expression can be judged based on the chart obtained by the flow cytometry method. The position appearing on the chart may vary depending on the voltage setting of the instrument, the sensitivity setting, the antibody clone used, the staining condition, the dye used, etc., but those skilled in the art can select the cell population recognized as a group in the obtained chart. It can be appropriately drawn so as not to be divided.
 目的のマーカーの発現が、陽性であるか陰性であるかの判断には、アイソタイプ・コントロール(Isotype control)抗体を用いた場合をネガティブ・コントロールとして用いて判断することができる。Isotype control抗体は特定の抗原とは反応しない抗体である。一般に、抗体を用いた実験においては、ターゲット以外のタンパク質との非特異的な結合や、細胞表面上のFcレセプターとの結合によって、バックグラウンドが発生する可能性がある。ネガティブ・コントロールとなる抗体を使用した系と比較することにより、目的の抗原に対する一次抗体の反応が特異的であることが明確になる。またバックグラウンドの影響が排除され、シグナルの強さを正確に解釈することができる。 To determine whether the expression of the target marker is positive or negative, it is possible to use the isotype control (Isotype control) antibody as a negative control. The Isotype control antibody is an antibody that does not react with a specific antigen. In general, in experiments using antibodies, background may occur due to nonspecific binding to proteins other than the target or binding to Fc receptors on the cell surface. By comparing with a system using an antibody as a negative control, it becomes clear that the reaction of the primary antibody against the antigen of interest is specific. In addition, the influence of background is eliminated, and the strength of the signal can be accurately interpreted.
 目的のマーカーの発現の程度(低発現性であるか、高発現性であるか)は、同一条件で測定した対照細胞の結果との比較により、判断することができる。対照細胞の例は、本願明細書の実施例の項に記載したプライマリーNK細胞のような、末梢血から得た、実質的な培養を行なっていないNK細胞である。 The degree of expression of the target marker (whether it is low expression or high expression) can be judged by comparison with the results of control cells measured under the same conditions. Examples of control cells are NK cells obtained from peripheral blood that have not been substantially cultured, such as the primary NK cells described in the Examples section of the present specification.
 例えば、あるNK細胞の集団におけるCD16の発現の程度は、フローサイトメトリーを用い、その細胞集団におけるCD16発現量を、末梢血から得た実質的な培養を行なっていないNK細胞の集団(対照)におけるCD16発現量と比較し、対照と同等の発現が見られる場合は高発現性であると判断し、対照細胞より発現が低い場合は低発現性であると判断することができる。なお、対照のNK細胞はCD16高発現性であることが知られている。 For example, the degree of CD16 expression in a certain NK cell population was determined by using flow cytometry, and the CD16 expression level in the NK cell population was determined from the peripheral blood-derived NK cell population that was not substantially cultured (control). In comparison with the CD16 expression level in the above, it can be judged that the expression is high if the expression is similar to that of the control, and that it is low when the expression is lower than that of the control cells. The control NK cells are known to highly express CD16.
[医薬用途]
 本発明はまた、上述のNK細胞を含む細胞集団を有効成分とする、細胞療法のための医薬品組成物を提供する。細胞療法とは、体外で処理した細胞を対象に投与することにより、対象における疾患又は状態を処置する方法をいい、これには免疫細胞療法が含まれる。
[Pharmaceutical use]
The present invention also provides a pharmaceutical composition for cell therapy, which comprises the above-mentioned cell population containing NK cells as an active ingredient. Cell therapy refers to a method of treating a disease or condition in a subject by administering cells treated ex vivo to the subject, which includes immune cell therapy.
 医薬組成物は、有効成分である細胞集団のほか、NK細胞を懸濁することができる溶液、例えば、生理食塩水、リン酸緩衝生理食塩水(PBS)等を含む。医薬組成物又は溶液はまた、薬学的に許容される添加剤を含んでいてもよい。医薬組成物は、例えば、静脈、動脈、皮下、腹腔内等へ投与することができる。医薬組成物による細胞療法は、単独か、あるいは外科療法、化学療法、放射線療法等と組み合わせて実施することができる。 The pharmaceutical composition includes, in addition to the cell population, which is an active ingredient, a solution capable of suspending NK cells, such as physiological saline or phosphate buffered saline (PBS). The pharmaceutical composition or solution may also contain pharmaceutically acceptable additives. The pharmaceutical composition can be administered, for example, into a vein, an artery, subcutaneously, intraperitoneally or the like. Cell therapy with a pharmaceutical composition can be performed alone or in combination with surgery, chemotherapy, radiation therapy and the like.
 NK細胞を含む、細胞集団はがん治療や感染症治療への適用が期待されているが(Dahlberg C.M.et al.,Natural Killer Cell−Based Therapies Targeting Cancer:Possible Strategies to Gain and Sustain Anti−Tumor Activity,Front.Immunol.,2015;30:https://doi.org/10.3389/fimmu.2015.00605、及びSchmidt S.et al,Natural killer cells as a therapeutic tool for infectious diseases−current status and future perspectives,Oncotarget,2018;9(29):20891−20907)、本発明の医薬組成物は、このようながん、又は感染症を治療するために用いることができる。より具体的には、口腔癌、胆嚢癌、胆管癌、肺癌、肝臓癌、大腸癌、腎臓癌、膀胱癌、白血病;ウイルス、細菌等による感染症を含むが、これらに限定されない。本発明の医薬組成物を用いた細胞療法において、NK細胞は、例えば、静脈、動脈、皮下、腹腔内等へ投与される場合がある。細胞療法は、単独か、あるいは外科療法、化学療法、放射線療法、抗体医薬品等と組み合わせて実施される場合がある。 Cell populations including NK cells are expected to be applied to cancer treatment and infectious disease treatment (Dahlberg CM et al., Natural Killer Cell-Based Therapeutic Targeting Cancer: Possible Strategies Anesthetics). -Tormor Activity, Front. status and future perspectives, Oncotarget, 2018;9(29):20891-20907), and the pharmaceutical composition of the present invention can be used for treating such cancer or infectious disease. More specifically, it includes, but is not limited to, oral cancer, gallbladder cancer, bile duct cancer, lung cancer, liver cancer, colon cancer, kidney cancer, bladder cancer, leukemia; infectious diseases caused by viruses, bacteria and the like. In the cell therapy using the pharmaceutical composition of the present invention, NK cells may be administered, for example, into veins, arteries, subcutaneously, intraperitoneally and the like. Cell therapy may be performed alone or in combination with surgery, chemotherapy, radiation therapy, antibody drugs and the like.
 抗体医薬品の多くは静脈内投与後、ADCC活性による抗腫瘍効果を示すとされる。その一方で、ADCC活性を発揮する際にはNK細胞の他に単球/マクロファージや好中球も動員される。そしてNK細胞以外のエフェクターは、正常細胞とがん細胞とを区別せずにADCC活性を示し、それは副作用の発現にもつながっていると考えられる。本発明においては、NK細胞と抗体とを、投与前に混合し、NK細胞に予め抗体を装備させてもよい。これによりエフェクターがNK細胞に限定され、投与する抗体の量を減じることができ、また副作用の低減にも極めて有効であると考えられる。すなわち、本発明の医薬組成物は、NK細胞集団と抗体とを混合した後、NK細胞と結合していない抗体を除去する工程を経て、調製されていてもよい。すなわち、本発明の医薬組成物の好ましい態様の一つは、NK細胞と抗体を含むが、抗体はNK細胞に結合しており、NK細胞に結合していない抗体が実質的に含まれないものである。(前掲特許文献3参照)。 Most antibody drugs are said to show an antitumor effect due to ADCC activity after intravenous administration. On the other hand, monocytes/macrophages and neutrophils are recruited in addition to NK cells when exerting ADCC activity. The effectors other than NK cells show ADCC activity without distinguishing between normal cells and cancer cells, which is considered to lead to the development of side effects. In the present invention, NK cells and antibody may be mixed before administration to equip the NK cells with the antibody in advance. As a result, the effector is limited to NK cells, the amount of antibody to be administered can be reduced, and it is considered to be extremely effective in reducing side effects. That is, the pharmaceutical composition of the present invention may be prepared by mixing the NK cell population and the antibody, and then removing the antibody not bound to the NK cell. That is, one of the preferred embodiments of the pharmaceutical composition of the present invention comprises NK cells and an antibody, but the antibody is bound to NK cells and substantially free of antibodies not bound to NK cells. Is. (See the above-mentioned Patent Document 3).
 本発明の医薬組成物の製造は、医薬品及び医薬部外品の製造管理及び品質管理規則に適合した条件(good manufacturing practice、GMP)及び再生医療等製品の製造管理及び品質管理の基準(Good Gene,Cellular,and Tissue−based Products Manufacturing Practice、GCTP)で実施されることが好ましい。 The pharmaceutical composition of the present invention is manufactured under the conditions (good manufacturing practice, GMP) conforming to the manufacturing control and quality control rules for drugs and quasi-drugs, and the manufacturing control and quality control standards for products such as regenerative medicine (Good Gene). , Cellular, and Tissue-based Products, Manufacturing Structure, GCTP).
 以下に説明する本発明の実施例は例示のみを目的とし、本発明の技術的範囲を限定するものではない。本発明の技術的範囲は特許請求の範囲の記載によってのみ限定される。本発明の趣旨を逸脱しないことを条件として、本発明の変更、例えば、本発明の構成要件の追加、削除及び置換を行うことができる。 The embodiments of the present invention described below are for illustrative purposes only and do not limit the technical scope of the present invention. The technical scope of the present invention is limited only by the description of the claims. Modifications of the present invention, for example, addition, deletion, and replacement of the constituent features of the present invention, can be made on the condition that the gist of the present invention is not deviated.
[実施例1:新鮮末梢血から得たNK細胞の混合培養1]
 健常人ボランティアから採血を実施し、フィコール(GEヘルスケア、17144002)を用いた密度勾配遠心により末梢血単核球を単離した。単離した複数人分の末梢血単核球をほぼ同率で混合し、CD3 beads※1を添加・懸濁し、4℃,15分間インキュベート後、分離バッファー1mL※2を加えてよく懸濁し、300xg、10分間、遠心分離を行った。上清を除去し、0.5mLの分離バッファーに懸濁し、分離バッファー2mLをあらかじめ添加して湿らせたLDカラム(ミルテニーバイオテク,130−042−901)に添加して、LDカラムからの溶出液を回収した。さらに分離バッファー1mLをLDカラムに添加し、溶出液を回収した。その後、分離バッファー1mLでカラムをwashし、回収された液中の細胞数をカウントし、総細胞数を算出した。500xg、5分間、遠心分離を行い、上清を除去後、5x10細胞mLとなるようにKBM501培地※3に懸濁した。
[Example 1: Mixed culture 1 of NK cells obtained from fresh peripheral blood]
Blood was collected from healthy volunteers and peripheral blood mononuclear cells were isolated by density gradient centrifugation using Ficoll (GE Healthcare, 17144002). Peripheral blood mononuclear cells from multiple isolated humans were mixed at approximately the same rate, CD3 beads *1 was added and suspended, incubated at 4°C for 15 minutes, and then well suspended by adding 1 mL *2 of separation buffer to 300 xg. Centrifugation was performed for 10 minutes. The supernatant was removed, and the suspension was suspended in 0.5 mL of separation buffer, and 2 mL of the separation buffer was added to a previously moistened LD column (Miltenyi Biotech, 130-042-901) to elute from the LD column. The liquid was collected. Further, 1 mL of the separation buffer was added to the LD column, and the eluate was collected. Then, the column was washed with 1 mL of the separation buffer, the number of cells in the collected liquid was counted, and the total number of cells was calculated. After centrifugation at 500×g for 5 minutes and removing the supernatant, the cells were suspended in KBM501 medium *3 at 5×10 5 cells / mL.
 一部の細胞をフローサイトメーター測定用に回収し、残りの細胞を培養した。培養は、6ウェルプレート(サーモフィッシャーサイエンティフィック,140675)、T−75フラスコ(サーモフィッシャーサイエンティフィック,156499)、又は500cm培養バッグ(ニプロ)を使用し、COインキュベーターで行った(37℃,5%CO)。培養5日目及び9日目に培養液の一部を取り、細胞数をカウントし、9日目には最終液量が、6ウェルプレートの場合は1ウェルあたり6mL、T−75フラスコの場合は1フラスコあたり50mLになるようにKBM501培地を添加した。14日目に細胞を回収し、細胞数をカウント、一部の細胞を用いてフローサイトメーターで細胞表面抗原の測定を行った。 Some cells were collected for flow cytometer measurement, and the remaining cells were cultured. Culturing was performed in a CO 2 incubator using a 6-well plate (Thermo Fisher Scientific, 140675), a T-75 flask (Thermo Fisher Scientific, 156499), or a 500 cm 2 culture bag (Nipro) (37). ℃, 5% CO 2). On the 5th and 9th day of culture, a part of the culture solution was taken and the number of cells was counted. On the 9th day, the final volume was 6 mL per well in the case of 6-well plate, and in the case of T-75 flask. KBM501 medium was added to 50 mL per flask. The cells were collected on the 14th day, the number of cells was counted, and a part of the cells was used to measure the cell surface antigen with a flow cytometer.
※1: CliniMACS CD3,ミルテニーバイオテク,130−017−601(1x10細胞あたり5μL)
※2: 0.5%ヒトAB型血清(コスモバイオ,12181301,56℃で30分の非働化処理したもの)、2mM EDTA(サーモフィッシャーサイエンティフィック,15575−020)を含むPBS((和光純薬工業,045−29795)
※3: 5%ヒトAB型血清(コスモバイオ,12181301,56℃で30分の非働化処理したもの)を添加したKBM 501(コージンバイオ,16025015)
*1: CliniMACS CD3, Miltenyi Biotech, 130-017-601 (5 μL per 1×10 7 cells)
*2: PBS containing 0.5% human type AB serum (Cosmobio, 12181301, 56°C inactivated for 30 minutes), 2mM EDTA (Thermo Fisher Scientific, 15575-020) ((Wako Jun Pharmaceutical industry, 045-29795)
*3: KBM 501 (Kojin Bio, 16025015) supplemented with 5% human AB type serum (Cosmo Bio, 12181301, inactivated at 56°C for 30 minutes)
 結果を図1に示した。9人混合(図1のA)、8人混合(図1のB)、7人混合(図1のC)のいずれにおいても、細胞の増殖は良好であった。純度を確認したところ、いずれの混合培養においても、90%以上がCD3CD56のNK細胞であった。8人混合培養では、T−75フラスコと500cm培養バッグとで培養を行ったが、双方とも細胞の増殖は良好であった。なお9人混合培養、7人培養、及び単独ドナー(7人のうちの1人)由来の培養は、6ウェルプレートを用いて培養を行った。500cm培養バッグを用いた培養においては、後述の実施例4に記載されているように、バッグを裏返す作業を行った。 The results are shown in Fig. 1. The cell growth was good in any of the 9-person mixture (A in FIG. 1), the 8-person mixture (B in FIG. 1), and the 7-person mixture (C in FIG. 1). When the purity was confirmed, 90% or more of the CD3 - CD56 + NK cells were found in any of the mixed cultures. In the 8-person mixed culture, the cells were cultured in a T-75 flask and a 500 cm 2 culture bag, and the cell growth was good in both cases. The 9-person mixed culture, the 7-person culture, and the culture derived from a single donor (1 of 7 persons) were cultured using a 6-well plate. In the culture using the 500 cm 2 culture bag, the bag was turned inside out as described in Example 4 below.
 また7人混合培養と単独ドナー由来の培養との細胞増殖を比較すると、単独ドナー由来の培養では6.8×10cellsから1.9×10cellsに増殖した(約2.8倍)のに対し、混合培養では7.8×10cellsから3.7×10cellsに増殖し(約4.7倍)、混合培養のほうが細胞の増殖が良好であった(図1のC))。 Further, comparing the cell growth between the mixed culture of 7 persons and the culture derived from a single donor, the culture derived from a single donor grew from 6.8×10 6 cells to 1.9×10 7 cells (about 2.8 times). On the other hand, in the mixed culture, the cells grew from 7.8×10 6 cells to 3.7×10 7 cells (about 4.7 times), and the mixed culture showed better cell growth (C in FIG. 1). )).
[実施例2:新鮮末梢血から得たNK細胞の混合培養2]
 実施例1に記載の方法で、単独ドナー由来の培養(n=17)、及び4人以上の混合培養(n=10)をT−75フラスコで行ったデータを合算し、得られたNK細胞数の細胞増殖率について統計解析を行った。解析ソフトにはJMP Pro13を用い、Wilcoxonの順位和検定を行った。
[Example 2: Mixed culture 2 of NK cells obtained from fresh peripheral blood]
By the method described in Example 1, NK cells obtained by adding together data obtained by culturing from a single donor (n=17) and mixed culture of 4 or more persons (n=10) in a T-75 flask Statistical analysis was performed on the number of cell proliferation rates. Wilcoxon rank sum test was performed using JMP Pro13 as the analysis software.
 結果を図2に示した。単独ドナー由来の細胞の培養の場合の増殖率(増殖後の全細胞数/増殖前の全細胞数)が3.10±0.56であるのに対し、混合培養の場合の増殖率は6.56±1.24であり、混合培養のほうが統計学的に有意に増殖率が高かった(P<0.01)。 The results are shown in Figure 2. The proliferation rate in the case of culturing cells derived from a single donor (total cell number after proliferation/total cell number before proliferation) is 3.10±0.56, whereas the proliferation rate in mixed culture is 6 0.56±1.24, and the growth rate was statistically significantly higher in the mixed culture (P<0.01).
[実施例3:腫瘍細胞傷害活性試験]
《NK細胞の調製》
 健常人ボランティアから実施例1に記載の方法で得られた4人の混合培養NK細胞と単独ドナー(4人のうちの1人)からの培養NK細胞を回収洗浄後、それぞれ10%FBS(ニチレイバイオサイエンス,171012−500ML)及び100ユニットのペニシリン、100μg/mLのストレプトマイシン(ナカライテスク,26253−84)を含むRPMI1640培地(和光純薬工業,189−02025)(以下、10%FBS/RPMI1640とする)にて懸濁し、同培地で1x10cells/mLの濃度に調製した。
[Example 3: Tumor cytotoxicity test]
<<Preparation of NK cells>>
Four mixed culture NK cells obtained by the method described in Example 1 from healthy volunteers and cultured NK cells from a single donor (one of four) were collected and washed, and then washed with 10% FBS (Nichirei). Bioscience, 171012-500 ML), 100 units of penicillin, and 100 μg/mL streptomycin (Nacalai Tesque, 26253-84) in RPMI1640 medium (Wako Pure Chemical Industries, 189-02025) (hereinafter referred to as 10% FBS/RPMI1640). ), and was adjusted to a concentration of 1×10 6 cells/mL with the same medium.
《SKOV3の調製》
 SKOV3細胞(ヒト卵巣がん細胞株)を血清成分を含まないRPMI1640培地(和光純薬工業,189−02025)にて1x10cells/mLの濃度に調製した。調製したSKOV3細胞をPKH26 Red Fluorescent Cell Linker Kit(Sigma,PKH26GL−1KT)を用いて染色し、最終的に10%FBS/RPMI1640にて2x10cells/mL及び2x10cells/mLとなるように調製した。
<<Preparation of SKOV3>>
SKOV3 cells (human ovarian cancer cell line) were prepared at a concentration of 1×10 6 cells/mL in RPMI1640 medium (Wako Pure Chemical Industries, 189-02025) containing no serum component. The prepared SKOV3 cells were stained with PKH26 Red Fluorescent Cell Linker Kit (Sigma, PKH26GL-1KT), and finally prepared with 10% FBS/RPMI1640 at 2×10 6 cells/mL and 2×10 5 cells/mL. did.
《MDSC(Myeloid−derived suppressor cells)の調製》
 健常人ボランティア(NK細胞のドナーとは別)からフィコールを用いて末梢血単核球を単離し、10ng/mL IL−6(PEPROTECH,200−06−5UG)及び10ng/mL GM−CSF(CellGenix,1012−050)を含む10%FBS/RPMI1640で7‐10日間培養することによりMDSCを誘導した。MDSCを血清成分を含まないRPMI1640培地(和光純薬工業,189−02025)にて懸濁し、PKH Green Fluorescent Cell Linker Kit(Sigma,MINI67−1KT)を用いて染色し、最終的に10%FBS/RPMI1640にて8x10cells/mLとなるように調製した。
<<Preparation of MDSC (Myeloid-derivatized suppressor cells)>>
Peripheral blood mononuclear cells were isolated from healthy volunteers (separate from NK cell donors) using Ficoll, 10 ng/mL IL-6 (PEPROTECH, 200-06-5UG) and 10 ng/mL GM-CSF (CellGenix). , 1012-050), and MDSC was induced by culturing in 10% FBS/RPMI1640 for 7-10 days. MDSCs were suspended in RPMI 1640 medium (Wako Pure Chemical Industries, 189-02025) containing no serum component, stained with PKH Green Fluorescent Cell Linker Kit (Sigma, MINI67-1KT), and finally 10% FBS/ It was adjusted to 8×10 5 cells/mL with RPMI1640.
《細胞傷害活性試験》
 複数ドナー由来の混合培養NK細胞とSKOV3細胞の群、単独ドナー由来の培養NK細胞とSKOV3細胞の群、これら2群にそれぞれMDSCを加えた群の計4群と、陰性コントロールとしてSKOV3細胞のみの群を用意した。
<Cytotoxicity test>
A group of mixed cultured NK cells and SKOV3 cells derived from a plurality of donors, a group of cultured NK cells derived from a single donor and SKOV3 cells, a group in which MDSC was added to each of these two groups, a total of 4 groups, and only SKOV3 cells as a negative control A group was prepared.
 NK細胞とSKOV3細胞は、細胞比で3:1となるように96ウェルプレート(IWAKI,4870−800SP)に播種、混合し、37℃、5%CO下で4時間反応させた。MDSCを添加した群は、NK細胞とSKOV3とMDSCの細胞比が5:1:4となるように、まずNK細胞とMDSCを96ウェルプレートで混合し、37℃、5%CO下で12‐18時間反応させた。その後、遠心分離(500xg,5分間)し、上清を除去後、SKOV3細胞を添加、混合し、37℃、5%CO下で4時間反応させた。反応後、遠心分離(500xg,5分間)し、上清を除去後、PBSで希釈したZombie溶液(Biolegend,423105)を添加、混合し、室温、暗所で30分間インキュベートした。遠心、上清除去後、PBSで懸濁し、AccuCheck Counting Beads(サーモフィッシャーサイエンティフィック,PCB100)を添加した。フローサイトメーター(BD LSR Fortessa、BDバイオサイエンス社)を用いて測定を行い、FlowJoソフトウェア(FLOWJO,LLC)で解析し、細胞傷害活性率(%Lysis)を算出した※4NK cells and SKOV3 cells were seeded in a 96-well plate (IWAKI, 4870-800SP) at a cell ratio of 3:1, mixed, and reacted at 37° C. under 5% CO 2 for 4 hours. In the group to which MDSC was added, NK cells and MDSC were first mixed in a 96-well plate so that the cell ratio of NK cells, SKOV3 and MDSC was 5:1:4, and the mixture was added at 37° C. and 5% CO 2 for 12 hours. The reaction was carried out for -18 hours. Then, after centrifugation (500 xg, 5 minutes) and removal of the supernatant, SKOV3 cells were added, mixed, and reacted at 37°C under 5% CO 2 for 4 hours. After the reaction, the mixture was centrifuged (500 xg, 5 minutes), and after removing the supernatant, a Zombie solution (Biolegend, 423105) diluted with PBS was added and mixed, and the mixture was incubated at room temperature in the dark for 30 minutes. After centrifugation and removal of the supernatant, the cells were suspended in PBS, and AccuCheck Counting Beads (Thermo Fisher Scientific, PCB100) was added. The measurement was performed using a flow cytometer (BD LSR Fortessa, BD Bioscience) and analyzed by FlowJo software (FLOWJO, LLC) to calculate the cytotoxic activity rate (% Lysis) *4 .
※4: 細胞傷害活性率=(1−SKOV3生細胞数/陰性コントロールSKOV3生細胞数)×100
生細胞数:SKOV3細胞実測数×添加ビーズ液中のビーズ数/ビーズ実測数
SKOV3細胞実測数:FSC/SSC gated(debris exclusion)、doublet exclusionの後、PKH26 gated
*4: Cytotoxic activity rate=(1-live SKOV3 live cells/negative control SKOV3 live cells)×100
Number of live cells: actual number of SKOV3 cells x number of beads in added bead solution/actual number of beads Actual number of SKOV3 cells: FSC/SSC gated (debris exclusion), PKH26 + gated after double exclusion.
 結果を図3に示した。複数ドナー由来の混合培養NK細胞は、単独ドナー由来のNK細胞と比較して、同等以上の細胞傷害活性を示した。 The results are shown in Figure 3. Mixed culture NK cells derived from multiple donors showed equivalent or higher cytotoxic activity as compared to NK cells derived from a single donor.
[実施例4:凍結アフェレーシス血液から調製したNK細胞の混合培養]
 2ドナーの凍結アフェレーシス血液(HemaCare,PB001CLP)を解凍後、混合し、HBSS(−)溶液(ナカライテスク,17461−05)にて希釈し、自動閉鎖系細胞処理装置Lovo Cell Processing System(FRESENIUS KABI)を使用してPBS/EDTA溶液(Miltenyi Biotec,130−021−201)で洗浄、濃縮を行った。次に、濃縮した細胞液をCliniMACS Prodigy TS 310(Miltenyi Biotec,130−097−183)、CliniMACS CD3 MicroBeads(Miltenyi Biotec,130−017−601)、CliniMACS CD34 Reagent(Miltenyi Biotec,130−017−501)を使用してCD3陽性細胞、CD34陽性細胞を除去し、洗浄、溶出を行った。溶出液中の細胞数をカウントして総細胞数を算出した。500xg、5分間、遠心分離を行い、上清を除去後、1x10cells/mLとなるようにKBM501培地に懸濁した。培養は、T−75フラスコ(サーモフィッシャーサイエンティフィック,156499)又は500cm培養バッグ(ニプロ)を使用しCOインキュベーターで行った(37℃,5%CO)。バッグの培養は、培養開始から30分後にバッグを裏返し、培養9日目に再度裏返す作業を行った(図4−1参照)。また、培養9日目に培養液の一部を取り細胞数をカウントし、最終液量がT−75フラスコでは1フラスコあたり50mL、培養バッグでは1バッグあたり500mLになるようにKBM 501培地を添加した。14日目に細胞を回収し、総細胞数をカウント、一部の細胞を用いてフローサイトメーターで細胞表面抗原の測定を行った。
[Example 4: Mixed culture of NK cells prepared from frozen apheresis blood]
Frozen apheresis blood of two donors (HemaCare, PB001CLP) was thawed, mixed, diluted with HBSS(-) solution (Nacalai Tesque, 17461-05), and automatically closed cell processing system Lovo Cell Processing System (FRESENIUS KABI). Was washed with PBS/EDTA solution (Miltenyi Biotec, 130-021-201) and concentrated. Next, the concentrated cell solution was used for CliniMACS Prodity TS 310 (Miltenyi Biotec, 130-097-183), CliniMACS CD3 MicroBeads (Miltenyi Biotec, 130-017-601), CliniMACS CD30Bi-oIcient Reactant (MINITENS). Were used to remove CD3-positive cells and CD34-positive cells, followed by washing and elution. The total number of cells was calculated by counting the number of cells in the eluate. After centrifugation at 500 xg for 5 minutes and removal of the supernatant, the cells were suspended in KBM501 medium at 1 x 10 6 cells/mL. The culture was performed in a CO 2 incubator using a T-75 flask (Thermo Fisher Scientific, 156499) or a 500 cm 2 culture bag (Nipro) (37° C., 5% CO 2 ). Regarding the culture of the bag, the bag was turned over 30 minutes after the start of the culture, and was turned over again on the 9th day of culture (see FIG. 4-1). On the 9th day of culture, a part of the culture solution was taken and the number of cells was counted, and KBM 501 medium was added so that the final solution volume was 50 mL per flask for the T-75 flask and 500 mL per bag for the culture bag. did. The cells were collected on the 14th day, the total number of cells was counted, and a part of the cells was used to measure the cell surface antigen with a flow cytometer.
 回収した細胞を以下のように抗体で染色した:
 Alexa Fluor(登録商標)700標識抗ヒトCD56抗体(Biolegend,318316)、PerCP/Cy5.5標識抗ヒトCD3抗体(Biolegend,300430)、PE−Cy7標識抗ヒトCD16抗体(Biolegend,302016)を1μg/mLの濃度で4℃、30分間染色後、遠心分離(500xg,5分間,4℃)し、上清を除去し、PBS(−)(和光純薬工業)に懸濁後、フローサイトメーター(BD LSRFortessa,BDバイオサイエンス社)を用いて測定を行い、FlowJoソフトウェア(FLOWJO,LLC)で解析した。
Harvested cells were stained with antibody as follows:
Alexa Fluor (registered trademark) 700-labeled anti-human CD56 antibody (Biolegend, 318316), PerCP/Cy5.5-labeled anti-human CD3 antibody (Biolegend, 300430), PE-Cy7-labeled anti-human CD16 antibody (Biolegend, 302016) at 1 μg/ After staining at a concentration of mL for 30 minutes at 4°C, centrifugation (500 xg, 5 minutes, 4°C) was performed, the supernatant was removed, and the cells were suspended in PBS(-) (Wako Pure Chemical Industries, Ltd.) and then flow cytometer ( The measurement was carried out using BD LSR Fortessa (BD Bioscience) and analyzed by FlowJo software (FLOWJO, LLC).
 結果を図4−2に示した。凍結アフェレーシス血液から得たNK細胞を用いた場合も混合培養により、細胞は14日間で十分に増殖した。詳細には、T−75フラスコ培養の場合は1.50x10cellsから5.72x10cellsに増殖し(約3.81倍)、500cm培養バッグ培養の場合は1.50x10cellsから5.60×10cellsに増殖した(約3.73倍)(図4−2、左)。 The results are shown in Figure 4-2. Even when NK cells obtained from frozen apheresis blood were used, the cells were sufficiently grown in 14 days by the mixed culture. Specifically, in the case of T-75 flask culture, it grew from 1.50×10 8 cells to 5.72×10 8 cells (about 3.81 times), and in the case of 500 cm 2 culture bag culture, 1.50×10 8 cells to 5. It proliferated in 60×10 8 cells (about 3.73 times) (FIG. 4-2, left).
 得られた培養NK細胞の集団におけるNK細胞の純度は、T−75フラスコ培養の場合は90.5%、500cm培養バッグ培養の場合は91.7%であった。またCD16の発現に関しては、混合培養NK細胞の集団は、単独ドナー由来の培養NK細胞の集団(特開2018−193303参照)と同様、二峰性であった。詳細には、CD16低発現性、及びCD16高発現性の割合は、T−75フラスコ培養の場合は順に50.9%、及び49.1%であり、500cm培養バッグ培養の場合は順に46.7%、及び53.3%であった(図4−2、右)。 The purity of NK cells in the obtained population of cultured NK cells was 90.5% for T-75 flask culture and 91.7% for 500 cm 2 culture bag culture. Regarding the expression of CD16, the population of mixed culture NK cells was bimodal, as was the population of culture NK cells derived from a single donor (see JP-A-2018-193303). Specifically, the proportions of low CD16 expression and high CD16 expression were 50.9% and 49.1% in the case of T-75 flask culture, and 46 in the case of 500 cm 2 culture bag culture. It was 0.7% and 53.3% (FIG. 4-2, right).
[実施例5:単球/NK細胞 swapping実験]
《プライマリーNK細胞と単球の調製》
 健常人ボランティア2名(ドナー1、2とする)から採血を実施、フィコールを用いた密度勾配遠心により末梢血単核球を単離した。単離した末梢血単核球からEasySepTM Human NK Cell Enrichment Kit(STEMCELL,19055)を使用しプライマリーNK細胞を、EasySepTM Human Monocyte Enrichment Kit without CD16 Depletion(STEMCELL,19058)を使用し、単球を分離し、それぞれ細胞数をカウントした。プライマリーNK細胞は1x10cells/mL、単球は3x10cells/mLとなるようにKBM 501培地で懸濁した。
[Example 5: Monocyte/NK cell swapping experiment]
<<Preparation of primary NK cells and monocytes>>
Blood was collected from two healthy volunteers (donors 1 and 2), and peripheral blood mononuclear cells were isolated by density gradient centrifugation using Ficoll. From the isolated peripheral blood mononuclear cells, primary NK cells were used using the EasySep Human NK Cell Enrichment Kit (STEMCELL, 19055), and EasySep Human Monocyte Enrichment Kit with ELEMCT 58 (CD16DST) was used. The cells were separated and the number of cells was counted. The primary NK cells were suspended in KBM 501 medium at 1×10 5 cells/mL and monocytes at 3×10 5 cells/mL.
《swapping培養》
 ドナー1のプライマリーNK細胞とドナー1の単球、ドナー1のプライマリーNK細胞とドナー2の単球、ドナー2のプライマリーNK細胞とドナー2の単球、ドナー2のプライマリーNK細胞とドナー1の単球をそれぞれ組み合わせた、計4群を、プライマリーNK細胞と単球の細胞比が1:3となるように混合して6ウェルプレート(サーモフィッシャーサイエンティフィック,140675)で培養を開始した(37℃,5%CO)。実施例1に記載の方法と同様に培養9日目にKBM501培地を追加し、14日目に細胞を回収した。得られた細胞を用いて細胞表面抗原とK562(ヒト慢性骨髄性白血病細胞株)に対する細胞傷害活性率の測定を行った。
<<swapping culture>>
Donor 1 primary NK cells and donor 1 monocytes, donor 1 primary NK cells and donor 2 monocytes, donor 2 primary NK cells and donor 2 monocytes, donor 2 primary NK cells and donor 1 monocytes A total of 4 groups in which spheres were combined were mixed so that the cell ratio of primary NK cells and monocytes was 1:3, and the culture was started in a 6-well plate (Thermo Fisher Scientific, 140675). ℃, 5% CO 2). Similar to the method described in Example 1, KBM501 medium was added on the 9th day of culture, and the cells were collected on the 14th day. The obtained cells were used to measure the cell surface antigen and the cytotoxic activity rate against K562 (human chronic myelogenous leukemia cell line).
 細胞表面抗原の測定は回収した細胞を以下のように抗体で染色し解析した:
 Alexa Fluor(登録商標)700標識抗ヒトCD56抗体(Biolegend,318316)、APC/Cy7標識抗ヒトCD3抗体(Biolegend,300426)、FITC標識抗ヒトKIR2DL1抗体(Miltenyi Biotec,130−103−966)、PerCP/Cy5.5標識抗ヒトKIR3DL1抗体(Biolegend,312718)を1μg/mLの濃度で4℃、30分間染色後、遠心分離(500xg,5分間,4℃)し、上清を除去し、PBS(−)(和光純薬工業)に懸濁後、フローサイトメーター(BD LSRFortessa,BDバイオサイエンス社)を用いて測定を行い、FlowJoソフトウェア(FLOWJO,LLC)で解析を行った。
The measurement of cell surface antigen was performed by staining the collected cells with an antibody and analyzing them as follows:
Alexa Fluor (registered trademark) 700-labeled anti-human CD56 antibody (Biolegend, 318316), APC/Cy7-labeled anti-human CD3 antibody (Biolegend, 300426), FITC-labeled anti-human KIR2DL1 antibody (Miltenyi Biotec, 130-103-966), PerCP. /Cy5.5-labeled anti-human KIR3DL1 antibody (Biolegend, 312718) was stained at a concentration of 1 μg/mL at 4° C. for 30 minutes, and then centrifuged (500×g, 5 minutes, 4° C.) to remove the supernatant, and PBS ( -) (Wako Pure Chemical Industries, Ltd.), followed by measurement using a flow cytometer (BD LSR Fortessa, BD Bioscience) and analysis with FlowJo software (FLOWJO, LLC).
 細胞傷害活性の測定には、2ドナーのプライマリーNK細胞と単球を組み合わせてできたそれぞれの培養NK細胞とK562細胞を反応させた4群、陰性コントロールとしてK562細胞のみの群、陽性コントロールとしてK562細胞を10%ホルマリンで固定した群を用意した。 To measure the cytotoxic activity, 4 groups were prepared by reacting cultured NK cells and K562 cells, each of which was formed by combining primary NK cells of two donors and monocytes, K562 cells alone as a negative control, and K562 as a positive control. A group was prepared in which cells were fixed with 10% formalin.
《K562の調製》
 K562細胞(ヒト慢性骨髄性白血病細胞株)を血清成分を含まないRPMI1640培地(和光純薬工業,189−02025)にて1x10cells/mLの濃度に調製した。調製したK562細胞をPKH26 Red Fluorescent Cell Linker Kit(Sigma,PKH26GL−1KT)を用いて染色し、最終的に10%FBS/RPMI1640にて2x10cells/mLとなるように調製した。
<<Preparation of K562>>
K562 cells (human chronic myelogenous leukemia cell line) were prepared at a concentration of 1×10 6 cells/mL in RPMI1640 medium (Wako Pure Chemical Industries, 189-02025) containing no serum component. The prepared K562 cells were stained with PKH26 Red Fluorescent Cell Linker Kit (Sigma, PKH26GL-1KT), and finally adjusted to 2×10 6 cells/mL with 10% FBS/RPMI1640.
《培養NK細胞の調製》
 上記の方法で得られた2ドナーのプライマリーNK細胞と単球を組み合わせて培養して得た4群の培養NK細胞を回収洗浄後、それぞれ10%FBS/RPMI1640にて懸濁し、同培地で1x10cells/mLの濃度に調製した。
<<Preparation of cultured NK cells>>
After collecting and washing the 4 groups of cultured NK cells obtained by culturing by combining the primary NK cells of the two donors obtained by the above method and monocytes, they were suspended in 10% FBS/RPMI1640 and 1×10 6 in the same medium. The concentration was adjusted to 6 cells/mL.
《細胞傷害活性試験》
 NK細胞とK562細胞は、細胞比で1:1となるように96ウェルプレート(IWAKI,4870−800SP)に添加、混合し、37℃、5%CO下で2時間反応させた。反応後、遠心分離(500xg,5分間)し、上清を除去後、PBSで希釈した7−AAD溶液(Beckman Coulter,A07704)を添加、懸濁し、室温で20分間インキュベートした。フローサイトメーター(BD LSR Fortessa、BDバイオサイエンス社)を用いて測定を行い、FlowJoソフトウェア(FLOWJO,LLC)で解析し、細胞傷害活性率(%Lysis)を算出した※5
<Cytotoxicity test>
NK cells and K562 cells were added to a 96-well plate (IWAKI, 4870-800SP) at a cell ratio of 1:1 and mixed, and reacted at 37° C. under 5% CO 2 for 2 hours. After the reaction, the mixture was centrifuged (500 xg, 5 minutes), the supernatant was removed, 7-AAD solution diluted with PBS (Beckman Coulter, A07704) was added and suspended, and the mixture was incubated at room temperature for 20 minutes. The measurement was performed using a flow cytometer (BD LSR Fortessa, BD Bioscience) and analyzed by FlowJo software (FLOWJO, LLC) to calculate the cytotoxic activity rate (% Lysis) *5 .
※5: 細胞傷害活性率=(K562細胞死細胞率−陰性コントロール死細胞率)/(陽性コントロール死細胞率−陰性コントロール死細胞率)×100 *5: Cytotoxic activity rate = (K562 cell dead cell rate-negative control dead cell rate)/(positive control dead cell rate-negative control dead cell rate) x 100
 ドナータイピング情報を下表に、結果を図5に示した。 Donor typing information is shown in the table below and the results are shown in Figure 5.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
 KIR3DL1、KIR3DS1からのシグナルが一方あるいは両方入る条件の場合に、CD16highの集団が増えることから、より高いADCC活性が期待できる。 When the signal from KIR3DL1 or KIR3DS1 enters one or both of them, the population of CD16 high increases, and thus higher ADCC activity can be expected.

Claims (11)

  1.  複数のドナーに由来し、NK細胞を含む、単核細胞の集団を準備し、
     準備した単核細胞の集団をNK細胞処理上有効な条件でインキュベートする
    ことを含む、NK細胞を含む細胞集団の製造方法。
    Preparing a population of mononuclear cells from multiple donors, including NK cells,
    A method for producing a cell population containing NK cells, which comprises incubating the prepared population of mononuclear cells under conditions effective for treating NK cells.
  2.  単核細胞の集団を準備する工程が、CD3陽性細胞を除去する工程を含む、請求項1に記載の製造方法。 The method according to claim 1, wherein the step of preparing a population of mononuclear cells includes the step of removing CD3-positive cells.
  3.  単核細胞の集団を準備する工程が、CD34陽性細胞を除去する工程を含む、請求項1又は2に記載の製造方法。 The method according to claim 1 or 2, wherein the step of preparing a population of mononuclear cells includes the step of removing CD34-positive cells.
  4.  単核細胞の集団を準備する工程が、複数のドナーから採取された末梢血から単核細胞の集団を得る工程を含む、請求項1‐3のいずれか1項に記載の製造方法。 The method according to any one of claims 1 to 3, wherein the step of preparing a population of mononuclear cells includes the step of obtaining a population of mononuclear cells from peripheral blood collected from a plurality of donors.
  5.  単核細胞の集団を準備する工程が、複数のドナーから採取されたアフェレーシス血液から単核細胞の集団を得る工程を含む、請求項1‐4のいずれか1項に記載の製造方法。 The production method according to any one of claims 1 to 4, wherein the step of preparing a population of mononuclear cells includes a step of obtaining a population of mononuclear cells from apheresis blood collected from a plurality of donors.
  6.  単核細胞の集団を準備する工程が、複数のドナーに由来する、胚性幹(ES)細胞、人工多能性幹(iPS)細胞、及び成体幹細胞からなる群より選択されるいずれかに由来する単核細胞の集団を準備するものである、請求項1‐3のいずれか1項に記載の製造方法。 The step of preparing a population of mononuclear cells is derived from any one selected from the group consisting of embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, and adult stem cells derived from multiple donors. The method according to any one of claims 1 to 3, wherein a population of mononuclear cells to be prepared is prepared.
  7.  複数のドナーが、一のドナーと、それとはHLA及びKIRの少なくとも一方において遺伝子型が異なる他のドナーを含む、請求項1‐6のいずれか1項に記載の製造方法。 The production method according to any one of claims 1 to 6, wherein the plurality of donors include one donor and another donor having a genotype different from that of at least one of HLA and KIR.
  8.  以下の特徴を備える、NK細胞を含む細胞集団:
    (1)複数のドナーに由来する。
    (2)NK細胞をエフェクター細胞(E)とし、K562細胞を標的細胞(T)として混合比(E:T)1:1で共培養した場合の細胞傷害活性が50%以上である。
    A cell population including NK cells, which has the following characteristics:
    (1) Derived from multiple donors.
    (2) The cytotoxic activity is 50% or more when NK cells are used as effector cells (E) and K562 cells are used as target cells (T) at a mixing ratio (E:T) of 1:1.
  9.  請求項1‐7のいずれか1項に記載の製造方法によって製造される細胞集団を含む、細胞療法のための医薬組成物。 A pharmaceutical composition for cell therapy, comprising a cell population produced by the production method according to any one of claims 1 to 7.
  10.  請求項8に記載の細胞集団を含む、細胞療法のための医薬組成物。 A pharmaceutical composition for cell therapy, comprising the cell population according to claim 8.
  11.  感染症及び/又はがんを治療するための、請求項9又は10に記載の医薬組成物。 The pharmaceutical composition according to claim 9 or 10 for treating infectious disease and/or cancer.
PCT/IB2020/052499 2019-01-21 2020-03-19 Production method for cell population including nk cells WO2020152661A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP20745606.2A EP3915640A4 (en) 2019-01-21 2020-03-19 Production method for cell population including nk cells
CN202080023218.2A CN113613723A (en) 2019-01-21 2020-03-19 Method for producing cell population containing NK cells
AU2020211441A AU2020211441B2 (en) 2019-01-21 2020-03-19 A method for producing a cell population including nk cells
KR1020217026441A KR20220138330A (en) 2019-01-21 2020-03-19 Methods of Making a Cell Population Comprising NK Cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2019007868A JP6838750B2 (en) 2019-01-21 2019-01-21 Method for producing a cell population containing NK cells
JP2019-007868 2019-01-21
US16/808,989 US20210000871A1 (en) 2019-01-21 2020-03-04 Method for producing a cell population including nk cells
US16/808,989 2020-03-04

Publications (1)

Publication Number Publication Date
WO2020152661A1 true WO2020152661A1 (en) 2020-07-30

Family

ID=71736538

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2020/052499 WO2020152661A1 (en) 2019-01-21 2020-03-19 Production method for cell population including nk cells

Country Status (1)

Country Link
WO (1) WO2020152661A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5511039B2 (en) 1978-05-11 1980-03-21
JP2013027385A (en) 2011-06-24 2013-02-07 Kyushu Univ Method for amplifying nk cells
JP2014080431A (en) * 2011-06-24 2014-05-08 Kyushu Univ Amplification method of nk cells
WO2014112491A1 (en) * 2013-01-15 2014-07-24 Abe Hiroyuki Method for manufacturing immunocyte-containing composition, and cancer-treating composition
JP2014226079A (en) 2013-05-22 2014-12-08 国立大学法人九州大学 Method for preparing nk cells
JP2016187341A (en) * 2009-12-29 2016-11-04 ガミダ セル リミテッド Population of natural killer cells cultured ex vivo
JP2017508479A (en) * 2014-03-07 2017-03-30 エメルセル エスエーエス Pooled NK cells derived from cord blood and their use for the treatment of cancer and chronic infections
JP6164650B2 (en) 2014-01-20 2017-07-19 国立大学法人九州大学 Method for preparing NK cells
JP2018059624A (en) 2016-08-22 2018-04-12 ジョンソン エレクトリック ソシエテ アノニム Ring gear and gear device
JP2018193303A (en) 2017-05-12 2018-12-06 米満 吉和 Highly active nk cells and application thereof

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5511039B2 (en) 1978-05-11 1980-03-21
JP2016187341A (en) * 2009-12-29 2016-11-04 ガミダ セル リミテッド Population of natural killer cells cultured ex vivo
JP2013027385A (en) 2011-06-24 2013-02-07 Kyushu Univ Method for amplifying nk cells
JP2014080431A (en) * 2011-06-24 2014-05-08 Kyushu Univ Amplification method of nk cells
JP5572863B2 (en) 2011-06-24 2014-08-20 国立大学法人九州大学 Method for amplifying NK cells
JP5989016B2 (en) 2011-06-24 2016-09-07 国立大学法人九州大学 Method for amplifying NK cells
WO2014112491A1 (en) * 2013-01-15 2014-07-24 Abe Hiroyuki Method for manufacturing immunocyte-containing composition, and cancer-treating composition
JP2014226079A (en) 2013-05-22 2014-12-08 国立大学法人九州大学 Method for preparing nk cells
JP6164650B2 (en) 2014-01-20 2017-07-19 国立大学法人九州大学 Method for preparing NK cells
JP2017508479A (en) * 2014-03-07 2017-03-30 エメルセル エスエーエス Pooled NK cells derived from cord blood and their use for the treatment of cancer and chronic infections
JP2018059624A (en) 2016-08-22 2018-04-12 ジョンソン エレクトリック ソシエテ アノニム Ring gear and gear device
JP2018193303A (en) 2017-05-12 2018-12-06 米満 吉和 Highly active nk cells and application thereof

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
DAHLBERG C.M. ET AL.: "Natural Killer Cell-Based Therapies Targeting Cancer: Possible Strategies to Gain and Sustain Anti-Tumor Activity", FRONT. IMMUNOL., 2015, pages 30, Retrieved from the Internet <URL:https://doi.org/10.3389/fimmu.2015.00605>
DOMOGALA A. ET AL.: "Natural killer cell immunotherapy: from bench to bedside", FRONTIERS IN IMMUNOLOGY, 2015, pages 6
HIROSHI ICHISE ET AL.: "NK Cell Alloreactivity against KIR-Ligand-Mismatched HLA-Haploidentical Tissue Derived from HLA Haplotype-Homozygous iPSCs", STEM CELL REPORTS, vol. 9, 2017, pages 853 - 867
MASUDA, AKIKO: "II. Basic Knowledge of Lymphoid Malignancies. 4. Cell Surface Markers.", NIHON NAIKA GAKKAI ZASSHI , vol. 100, no. 7, 1 July 2011 (2011-07-01), JP, pages 1807 - 1816, XP009529457, ISSN: 0021-5384, DOI: 10.2169/naika.100.1807 *
MINGUS J.J. ROSE ET AL.: "Killer Ig-Like Receptor Ligand Mismatch Directs NK Cell Expansion In Vitro", THE JOURNAL OF IMMUNOLOGY, vol. 183, 2009, pages 4502 - 4508
SAITO S. ET AL.: "Ex vivo generation of highly purified and activated natural killer cells from human peripheral blood", HUM. GENE THER. METHODS, vol. 24, no. 4, 2013, pages 241 - 252, XP055154762, DOI: 10.1089/hgtb.2012.183
SCHMIDT S. ET AL.: "Natural killer cells as a therapeutic tool for infectious diseases - current status and future perspectives", ONCOTARGET, vol. 9, no. 29, 2018, pages 20891 - 20907
SUGITA, SUNAO ET AL.: "Natural Killer Cell Inhibition by HLA-E Molecules on Induced Pluripotent Stem Cell -Derived Retinal Pigment Epithelial Cells", INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 59, no. 5, April 2018 (2018-04-01), pages 1719 - 1731, XP055832695 *
ZENG J. ET AL.: "Generation of ''Off-the-Shelf' Natural Killer Cells from Peripheral Blood Cell-Derived Induced Pluripotent Stem Cells", STEM CELL REPORTS, vol. 9, 2017, pages 1796 - 1812, XP055534970, DOI: 10.1016/j.stemcr.2017.10.020

Similar Documents

Publication Publication Date Title
Mehta et al. Cord blood as a source of natural killer cells
US20100310533A1 (en) Methods of using il-21 for adoptive immunotherapy and identification of tumor antigens
JP6647240B2 (en) Highly active NK cells and their use
Shin et al. Ex vivo expansion of canine cytotoxic large granular lymphocytes exhibiting characteristics of natural killer cells
JP6543375B1 (en) Population of CD3 negative cells expressing chemokine receptor and cell adhesion molecule and use thereof
Schmid et al. Culture-expanded human invariant natural killer T cells suppress T-cell alloreactivity and eradicate leukemia
JP5989016B2 (en) Method for amplifying NK cells
Boor et al. Characterization of antigen-presenting cell subsets in human liver-draining lymph nodes
TWI757709B (en) A method for producing a cell population including nk cells
KR20030032961A (en) Method of Amplifying Natural Killer T Cells
Plantinga et al. Clinical grade production of wilms’ tumor-1 loaded cord blood-derived dendritic cells to prevent relapse in pediatric AML after cord blood transplantation
Zahorchak et al. In vivo mobilization and functional characterization of nonhuman primate monocytic myeloid-derived suppressor cells
US20220249567A1 (en) Low density cell culture
WO2020152661A1 (en) Production method for cell population including nk cells
JP6697611B2 (en) Highly active NK cells and use thereof
JP2020108405A (en) Highly active nk cells and application thereof
JP2021035400A (en) Method for producing cell population comprising nk cells
Galaverna Outcome and immune reconstitution of children and young adults with acute leukemia after alfa/beta T-cell and B-cell depleted HLA-Haploidentical transplantation
SUPPORTIVE Cord blood oral abstracts

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20745606

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020211441

Country of ref document: AU

Date of ref document: 20200319

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020745606

Country of ref document: EP

Effective date: 20210823