WO2023228728A1 - 幹細胞の製造方法及びγδT細胞の製造方法 - Google Patents
幹細胞の製造方法及びγδT細胞の製造方法 Download PDFInfo
- Publication number
- WO2023228728A1 WO2023228728A1 PCT/JP2023/017460 JP2023017460W WO2023228728A1 WO 2023228728 A1 WO2023228728 A1 WO 2023228728A1 JP 2023017460 W JP2023017460 W JP 2023017460W WO 2023228728 A1 WO2023228728 A1 WO 2023228728A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- producing
- stem cells
- stem
- blood
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/30—Organic components
- C12N2500/42—Organic phosphate, e.g. beta glycerophosphate
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/125—Stem cell factor [SCF], c-kit ligand [KL]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/145—Thrombopoietin [TPO]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2302—Interleukin-2 (IL-2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2307—Interleukin-7 (IL-7)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/26—Flt-3 ligand (CD135L, flk-2 ligand)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/11—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/45—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
Definitions
- the present invention relates to cell technology, and relates to a method for producing stem cells and a method for producing ⁇ T cells.
- T cells derived from hematopoietic stem cells play an important role in immunity (for example, see Non-Patent Document 1).
- T cells express a wide variety of T cell receptors (TCRs) on their surface.
- TCR diversity is brought about by V(D)J gene rearrangement.
- VJ gene rearrangement is shown in Figure 1.
- DN1 double negative 1 cells, which are T cell precursor cells, proliferate in response to interleukin-7 (IL-7) and c-kit ligand (KL), which are strongly expressed in the subcortical region within the thymus. At the same time, they express CD25 (interleukin-2 receptor ⁇ chain) and become DN2 cells. DN2 cells gradually decrease CD44 expression and become DN3 cells.
- IL-7 interleukin-7
- KL c-kit ligand
- TCR includes a variable (V) region and a constant (C) region.
- V variable
- C constant
- the amino acid sequence of the V region is highly diverse and forms a binding site for antigens.
- the V region gene is divided into the V gene, D (diversity) gene, and J (joining) gene in germline DNA, and in the process of differentiation into T cells, these are divided into V-(D)-J and V-(D)-J. They are rearranged into a single link on the chromosome and become expressed genes.
- the J gene includes JP1 gene, JP gene, J1 gene, JP2 gene, and J2 gene.
- TCR ⁇ type T cells TCR consists of an ⁇ chain and a ⁇ chain.
- TCR ⁇ type T cells TCR consists of a ⁇ chain and a ⁇ chain. The decision to branch out into TCR ⁇ type T cells or TCR ⁇ type T cells is determined by control in the silencer regions present at the TCR ⁇ gene locus and the TCR ⁇ gene locus.
- TCR ⁇ type T cells are smaller in number than TCR ⁇ type T cells, they account for the majority of the interepithelial lymphocyte population in the intestinal mucosa. TCR ⁇ type T cells sense various stresses that cause damage to cells and induce immune responses. TCR ⁇ type T cells are said to sense not only stress from outside the body such as bacterial infection or viral infection, but also changes in properties associated with cell canceration.
- TCR ⁇ type T cells proliferate and become activated after recognizing intermediate products in the mevalonate metabolism of the cholesterol synthesis pathway of antigen presenting cells (APC) and isopentenyl pyrophosphate (IPP) as antigens. Therefore, cancer immunotherapy is being carried out in which TCR ⁇ type T cells of a patient are activated outside the body and returned to the body.
- APC antigen presenting cells
- IPP isopentenyl pyrophosphate
- One of the objects of the present invention is to provide a method for efficiently inducing stem cells having a rearranged ⁇ -TCR gene and a method for efficiently producing ⁇ T cells.
- a method for producing stem cells according to an aspect of the present invention includes applying tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate (PTA) to blood cells or immune cells; inducing stem cells from blood cells or immune cells.
- PTA tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate
- the above method for producing stem cells may further include applying interleukin to blood cells or immune cells.
- the interleukin may be at least one selected from the group consisting of IL-2, IL-4, IL-9, IL-18, and IL-33.
- the blood cells or immune cells may be mononuclear cells.
- the stem cells may be iPS cells.
- inducing factor RNA may be introduced into the blood cells or immune cells.
- an RNA virus vector may be used to induce stem cells from blood cells or immune cells.
- the RNA viral vector may be a single-stranded RNA viral vector.
- the RNA viral vector may be a single-stranded plus RNA viral vector.
- the RNA viral vector may be a single-stranded negative RNA viral vector.
- the RNA viral vector may be a chromosomally non-integrating RNA viral vector.
- the RNA viral vector may be a Mononegavirales viral vector.
- the RNA virus vector may be a Paramyxoviridae virus vector.
- the RNA virus vector may be a Respirovirus virus vector.
- the RNA virus vector may be a Sendai virus vector.
- a virus-derived genomic RNA carrying an inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA are used.
- a chimeric virus comprising the following may also be used.
- the stem cells may include a rearranged ⁇ -TCR gene.
- a method for producing blood cells or immune cells according to an aspect of the present invention includes providing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the blood cells or immune cells may be ⁇ type T cells.
- a cell mass of stem cells may be seeded onto feeder cells.
- the feeder cells may be stromal cells.
- a method for producing stem cells according to an aspect of the present invention includes adding a bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate (HMBPP) and an interleukin to blood cells or immune cells. and inducing stem cells from blood cells or immune cells, wherein the interleukin is at least one selected from the group consisting of IL-4, IL-9, IL-18, and IL-33. It is one.
- the interleukin may not include IL-2.
- the interleukin may include IL-18 and IL-33.
- the interleukin may include IL-4 and IL-18.
- the interleukin may include IL-9 and IL-18.
- the interleukins may include IL-4, IL-9, and IL-18.
- the interleukins may include IL-4, IL-18, and IL-33.
- IL-4 may be applied to blood cells or immune cells after IL-18 and IL-33.
- the interleukins may include IL-4, IL-9, IL-18, and IL-33.
- IL-4 may be applied to blood cells or immune cells after IL-9, IL-18 and IL-33.
- bisphosphonates include zoledronic acid, pamidronic acid, alendronic acid, risedronic acid, ibandronic acid, incadronic acid, etidronic acid, minodronic acid, tetrakis-pivaloyloxymethyl 2-(thiazole-2- ylamino)ethylidene-1,1-bisphosphonate (PTA), salts thereof and hydrates thereof.
- the bisphosphonate may be zoledronic acid.
- the blood cells or immune cells may be mononuclear cells.
- the stem cells may be iPS cells.
- inducing factor RNA may be introduced into the blood cells or immune cells.
- an RNA virus vector may be used to induce stem cells from blood cells or immune cells.
- the RNA viral vector may be a single-stranded RNA viral vector.
- the RNA viral vector may be a single-stranded plus RNA viral vector.
- the RNA viral vector may be a single-stranded negative RNA viral vector.
- the RNA viral vector may be a chromosomally non-integrating RNA viral vector.
- the RNA viral vector may be a Mononegavirales viral vector.
- the RNA virus vector may be a Paramyxoviridae virus vector.
- the RNA virus vector may be a Respirovirus virus vector.
- the RNA virus vector may be a Sendai virus vector.
- a virus-derived genomic RNA carrying an inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA are used.
- a chimeric virus comprising the following may also be used.
- the stem cells may include a rearranged ⁇ -TCR gene.
- a method for producing blood cells or immune cells according to an aspect of the present invention includes providing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the blood cells or immune cells may be ⁇ type T cells.
- a cell mass of stem cells may be seeded onto feeder cells.
- the feeder cells may be stromal cells.
- a method for producing stem cells according to an aspect of the present invention includes applying bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate (HMBPP) to blood cells or immune cells; inducing stem cells from cells or immune cells, without applying IL-2 to blood cells or immune cells.
- HMBPP bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate
- any interleukins, including IL-2 are not applied to blood cells or immune cells.
- bisphosphonates include zoledronic acid, pamidronic acid, alendronic acid, risedronic acid, ibandronic acid, incadronic acid, etidronic acid, minodronic acid, tetrakis-pivaloyloxymethyl 2-(thiazole-2- ylamino)ethylidene-1,1-bisphosphonate (PTA), salts thereof and hydrates thereof.
- the bisphosphonate may be zoledronic acid.
- the blood cells or immune cells may be mononuclear cells.
- the stem cells may be iPS cells.
- inducing factor RNA may be introduced into the blood cells or immune cells.
- an RNA virus vector may be used to induce stem cells from blood cells or immune cells.
- the RNA viral vector may be a single-stranded RNA viral vector.
- the RNA viral vector may be a single-stranded plus RNA viral vector.
- the RNA viral vector may be a single-stranded negative RNA viral vector.
- the RNA viral vector may be a chromosomally non-integrating RNA viral vector.
- the RNA viral vector may be a Mononegavirales viral vector.
- the RNA virus vector may be a Paramyxoviridae virus vector.
- the RNA virus vector may be a Respirovirus virus vector.
- the RNA virus vector may be a Sendai virus vector.
- a virus-derived genomic RNA carrying an inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA are used.
- a chimeric virus comprising the following may also be used.
- the stem cells may include a rearranged ⁇ -TCR gene.
- a method for producing blood cells or immune cells according to an aspect of the present invention includes providing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the blood cells or immune cells may be ⁇ type T cells.
- a cell mass of stem cells may be seeded onto feeder cells.
- the feeder cells may be stromal cells.
- a method for producing stem cells according to an aspect of the present invention includes applying tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate (PTA) to blood cells or immune cells; inducing stem cells from blood cells or immune cells, without applying IL-2 to blood cells or immune cells.
- PTA tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate
- the above method for producing stem cells may further include applying an interleukin other than IL-2 to blood cells or immune cells.
- the interleukin may be at least one selected from the group consisting of IL-4, IL-9, IL-18, and IL-33.
- the blood cells or immune cells may be mononuclear cells.
- the stem cells may be iPS cells.
- inducing factor RNA may be introduced into the blood cells or immune cells.
- an RNA virus vector may be used to induce stem cells from blood cells or immune cells.
- the RNA viral vector may be a single-stranded RNA viral vector.
- the RNA viral vector may be a single-stranded plus RNA viral vector.
- the RNA viral vector may be a single-stranded negative RNA viral vector.
- the RNA viral vector may be a chromosomally non-integrating RNA viral vector.
- the RNA viral vector may be a Mononegavirales viral vector.
- the RNA virus vector may be a Paramyxoviridae virus vector.
- the RNA virus vector may be a Respirovirus virus vector.
- the RNA virus vector may be a Sendai virus vector.
- a virus-derived genomic RNA carrying an inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA are used.
- a chimeric virus comprising the following may also be used.
- the stem cells may include a rearranged ⁇ -TCR gene.
- a method for producing blood cells or immune cells according to an aspect of the present invention includes providing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the blood cells or immune cells may be ⁇ type T cells.
- a cell mass of stem cells may be seeded onto feeder cells.
- the feeder cells may be stromal cells.
- a method for producing stem cells according to an aspect of the present invention includes applying (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate and interleukin to blood cells or immune cells; inducing stem cells from blood cells or immune cells, and the interleukin does not include IL-2.
- the interleukin may be at least one selected from the group consisting of IL-4, IL-9, IL-18, and IL-33.
- the interleukin may include IL-18 and IL-33.
- interleukins include IL-4, IL-9, IL-18, and IL-33, and IL-4, IL-9, IL-18, and IL-33 are added to blood cells or immune cells after IL-9, IL-18, and IL-33. -4 may be applied.
- the blood cells or immune cells may be mononuclear cells.
- the stem cells may be iPS cells.
- inducing factor RNA may be introduced into the blood cells or immune cells.
- an RNA virus vector may be used to induce stem cells from blood cells or immune cells.
- the RNA viral vector may be a single-stranded RNA viral vector.
- the RNA viral vector may be a single-stranded plus RNA viral vector.
- the RNA viral vector may be a single-stranded negative RNA viral vector.
- the RNA viral vector may be a chromosomally non-integrating RNA viral vector.
- the RNA viral vector may be a Mononegavirales viral vector.
- the RNA virus vector may be a Paramyxoviridae virus vector.
- the RNA virus vector may be a Respirovirus virus vector.
- the RNA virus vector may be a Sendai virus vector.
- a virus-derived genomic RNA carrying an inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA are used.
- a chimeric virus comprising the following may also be used.
- the stem cells may include a rearranged ⁇ -TCR gene.
- a method for producing blood cells or immune cells according to an aspect of the present invention includes providing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the blood cells or immune cells may be ⁇ type T cells.
- a cell mass of stem cells may be seeded onto feeder cells.
- the feeder cells may be stromal cells.
- the method for producing ⁇ T cells according to an aspect of the present invention includes inducing differentiation of ⁇ T cells from pluripotent stem cells derived from T cells.
- pluripotent stem cells may be derived from ⁇ T cells.
- the ⁇ T cells induced by pluripotent stem cells may express a ⁇ T cell receptor.
- the pluripotent stem cells may have a rearranged gene for the ⁇ T cell receptor.
- the differentiated ⁇ T cells may express a ⁇ T cell receptor.
- inducing differentiation of ⁇ T cells from pluripotent stem cells is the same as inducing hematopoietic stem progenitor cells from pluripotent stem cells and inducing ⁇ T cells from hematopoietic stem progenitor cells. , may also be included.
- cells may be cultured in a feeder-free manner while inducing hematopoietic stem progenitor cells from pluripotent stem cells.
- cells may be cultured in suspension while hematopoietic stem progenitor cells are induced from pluripotent stem cells.
- hematopoietic stem progenitor cells may be induced from pluripotent stem cells in a medium containing at least one of bone morphogenetic protein 4 (BMP-4), VEGF, and bFGF.
- BMP-4 bone morphogenetic protein 4
- VEGF vascular endothelial growth factor
- bFGF vascular endothelial growth factor
- hematopoietic stem progenitor cells may be induced from pluripotent stem cells using a medium containing an activin receptor-like kinase inhibitor.
- the cells are cultured in a medium containing at least one of bone morphogenetic protein 4 (BMP-4), VEGF, and bFGF,
- BMP-4 bone morphogenetic protein 4
- VEGF vascular endothelial growth factor
- bFGF vascular endothelial growth factor
- hematopoietic stem progenitor cells may be cultured on feeder cells.
- the feeder cells may be OP9 cells.
- ⁇ T cells are ⁇ T cells induced to differentiate from pluripotent stem cells derived from ⁇ T cells.
- a pharmaceutical composition for cancer treatment according to an aspect of the present invention contains ⁇ T cells induced to differentiate from pluripotent stem cells derived from ⁇ T cells.
- FIG. 2 is a schematic diagram showing VJ gene rearrangement.
- 1 is a photograph of induced stem cells according to Example 1.
- 1 is a photograph showing the results of PCR analysis of the genome of induced stem cells according to Example 1.
- 3 is a graph showing the percentage of ⁇ T cells according to Example 2.
- 2 is a graph showing the number of ⁇ T cells induced from 2 ⁇ 10 4 mononuclear cells according to Example 2.
- 3 is a photograph of induced stem cells according to Example 2.
- 3 is a photograph of induced stem cells according to Example 2.
- 3 is a graph showing the number of induced stem cell colonies according to Example 4.
- 3 is a flow cytometer dot plot showing the results of Example 5.
- 3 is a flow cytometer dot plot showing the results of Example 5.
- 3 is a flow cytometer dot plot showing the results of Example 5.
- 3 is a photograph showing the results of PCR analysis of the genome of induced stem cells according to Example 6.
- 7 is a graph showing the number of induced stem cell colonies according to Example 7.
- 3 is a photograph showing the results of PCR analysis of the genome of induced stem cells according to Example 7.
- 3 is a photograph showing the results of PCR analysis of the genome of induced stem cells according to Example 7.
- 3 is a flow cytometer dot plot showing the results of Example 8.
- 2 is a dot plot using a flow cytometer showing the results of Example 8, Comparative Example 1, and Comparative Example 2.
- 3 is a flow cytometer dot plot showing the results of Example 9.
- 3 is a photograph of cells showing the results of Example 9.
- a method for producing stem cells includes applying tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate (PTA) to blood cells or immune cells, and or inducing stem cells from immune cells.
- PTA tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate
- interleukin may be further applied to blood cells or immune cells.
- Examples of interleukins in combination with PTA include, but are not limited to, IL-2, IL-4, IL-9, IL-18, and IL-33. Interleukins may not include IL-2.
- the method for producing stem cells applies bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate (HMBPP) and interleukin to blood cells or immune cells. and inducing stem cells from blood cells or immune cells, wherein the interleukin in combination with the bisphosphonate or HMBPP is selected from the group consisting of IL-4, IL-9, IL-18, and IL-33. At least one is selected. Interleukins may not include IL-2.
- HMBPP bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate
- Interleukins in combination with bisphosphonates or HMBPP include, for example, IL-18 and IL-33.
- Interleukins in combination with bisphosphonates or HMBPP include, for example, IL-4 and IL-18.
- Interleukins in combination with bisphosphonates or HMBPP include, for example, IL-9 and IL-18.
- Interleukins in combination with bisphosphonates or HMBPP include, for example, IL-4, IL-9, and IL-18.
- Interleukins in combination with bisphosphonates or HMBP include, for example, IL-4, IL-18 and IL-33.
- IL-4 may be applied to blood cells or immune cells after IL-18 and IL-33.
- Interleukins in combination with bisphosphonates or HMBP include, for example, IL-4, IL-9, IL-18, and IL-33.
- IL-4 may be applied to blood cells or immune cells after IL-9, IL-18 and IL-33.
- Interleukins that are combined with bisphosphonates include, for example, IL-4.
- Interleukins in combination with bisphosphonates include, for example, IL-4 and IL-18.
- Interleukins in combination with bisphosphonates include, for example, IL-4, IL-9, and IL-18.
- Interleukins that are combined with bisphosphonates include, for example, IL-9.
- Interleukins in combination with bisphosphonates include, for example, IL-9 and IL-18.
- Interleukins that are combined with bisphosphonates include, for example, IL-18.
- the method for producing stem cells includes applying bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate (HMBPP) to blood cells or immune cells, and inducing stem cells from cells or immune cells, without applying IL-2 to blood cells or immune cells.
- HMBPP bisphosphonate or (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate
- any interleukins, including IL-2 are not applied to blood cells or immune cells.
- bisphosphonates include zoledronic acid, pamidronic acid, alendronic acid, risedronic acid, ibandronic acid, incadronic acid, etidronic acid, minodronic acid, PTA, salts thereof, and hydrates thereof.
- the stem cells are, for example, induced pluripotent stem cells (iPS cells).
- iPS cells induced pluripotent stem cells
- Blood cells or immune cells may be derived from humans or non-human animals. Blood cells are separated from the blood. Blood includes, but is not limited to, peripheral blood and umbilical cord blood. Blood may be collected from an adult or a minor. When blood is collected, anticoagulants such as ethylenediaminetetraacetic acid (EDTA), heparin, and Biological Products Standard Blood Preservation Solution A (ACD-A) may be used.
- EDTA ethylenediaminetetraacetic acid
- ACD-A Biological Products Standard Blood Preservation Solution A
- Immune cells are isolated from blood, bone marrow, thymus, lymph, cavity fluid, and tissue. Examples of cavity fluids include ascites, pleural effusions, and pericardial effusions. Examples of tissues include tumor tissue.
- Blood cells are, for example, nucleated cells such as mononuclear cells, neutrophils, eosinophils, basophils, and lymphocytes, and do not include red blood cells, granulocytes, and platelets.
- the blood cells may be, for example, vascular endothelial progenitor cells, blood stem/progenitor cells, T cells, or B cells.
- T cells may be, for example, ⁇ T cells or ⁇ T cells.
- the blood cells do not have to be ⁇ T cells.
- immune cells include lymphocytes. Examples of lymphocytes include T cells, NK cells, or B cells.
- PTA or bisphosphonate When applying PTA or bisphosphonate to blood cells or immune cells, PTA or bisphosphonate may be added to the medium in which the blood cells or immune cells are cultured.
- concentration of PTA or bisphosphonate in the medium is, for example, 0.5 ⁇ mol/L or more and 50 ⁇ mol/L or less, 1 ⁇ mol/L or more and 50 ⁇ mol/L or less, 3 ⁇ mol/L or more and 20 ⁇ mol/L or less, or 5 ⁇ mol/L or more and 12 ⁇ mol/L or less. It is.
- Blood cells or immune cells may be cultured in a medium supplemented with PTA or bisphosphonates for one or more days, two or more days, or three or more days.
- Blood cells or immune cells may be cultured in a medium supplemented with PTA or bisphosphonates for up to 14 days, up to 10 days, or up to 7 days.
- interleukin When applying interleukin to blood cells or immune cells, interleukin may be added to the medium in which the blood cells or immune cells are cultured.
- concentration of interleukin in the medium is, for example, 5 ng/mL or more and 100 ng/mL or less, 10 ng/mL or more and 90 ng/mL or less, or 15 ng/mL or more and 80 ng/mL or less.
- Blood cells or immune cells may be cultured in a medium supplemented with interleukin for one or more days, two or more days, or three or more days.
- Blood cells or immune cells may be cultured in a medium supplemented with interleukin for up to 14 days, up to 10 days, or up to 7 days.
- Interleukin may be added to the medium every day, once every two days, or once every three days.
- Interleukins may be applied to blood cells or immune cells after applying PTA or bisphosphonates to blood cells or immune cells.
- PTA or bisphosphonates and interleukins may be applied simultaneously to blood cells or immune cells.
- the PTA or bisphosphonate may be applied to the blood cells or immune cells after the interleukin is applied to the blood cells or immune cells.
- Examples of media for culturing blood cells or immune cells include, but are not limited to, RPMI 1640 medium, minimal essential medium ( ⁇ -MEM), Dulbecco's modified Eagle medium (DMEM), and F12 medium.
- the blood cells or immune cells may be expanded for 1, 2, or 3 days before introducing the inducer into the blood cells or immune cells.
- an inducing factor is introduced into the blood cells or immune cells to which at least PTA or bisphosphonate has been applied to induce stem cells from the blood cells or immune cells.
- inducing stem cells induction refers to reprogramming, reprogramming, transformation, cell fate reprogramming, and the like.
- the inducer introduced into blood cells or immune cells may be RNA.
- RNA may be mRNA.
- Inducers include, for example, OCT3/4, SOX2, KLF4, and c-MYC.
- M 3 O which is a modified version of OCT3/4, may be used as an inducing factor.
- the inducers are LIN28A, FOXH1, LIN28B, GLIS1, p53-dominant negative, p53-P275S, L-MYC, NANOG, DPPA2, DPPA4, DPPA5, ZIC3, BCL-2, E-RAS, TPT1, SALL 2.NAC1 , DAX1, TERT, ZNF206, FOXD3, REX1, UTF1, KLF2, KLF5, ESRRB, miR-291-3p, miR-294, miR-295, NR5A1, NR5A2, TBX3, MBD3sh, TH2A, TH2B, and P53DD group It may be at least one selected from. RNA for these inducers is available from TriLink.
- gene symbols are written here in humans, it is not intended to limit the species by uppercase or lowercase letters. For example, all capital letters do not exclude the inclusion of mouse or rat genes. However, in the examples, gene symbols are written according to the biological species actually used.
- vectors may be used to introduce inducers into blood cells or immune cells.
- the inducer may be introduced into blood cells or immune cells by lipofection.
- RNA virus can be used as a vector.
- the RNA virus may be a single-stranded RNA virus.
- the RNA virus may be a single-stranded positive RNA virus.
- the RNA virus may be a single-stranded negative RNA virus.
- the RNA virus may be a chromosomally non-integrating RNA virus.
- the RNA virus may be a Mononegavirales virus.
- the RNA virus may be a Paramyxoviridae virus.
- the RNA virus may be a virus belonging to the genus Respirovirus.
- the RNA virus may be Sendai virus (Sev).
- Sendai virus is a virus whose genome is RNA and belongs to the order Mononegavirales and the family Paramyxoviridae.
- Sendai virus has an RNA genome and an envelope made of a lipid bilayer membrane that encloses the RNA.
- CytoTune (registered trademark, Invitrogen) can be used as the Sendai virus loaded with inducer RNA.
- the Sendai virus vector may be a Sendai virus vector with improved persistence of infection.
- a stealth RNA vector may be used as the RNA virus vector.
- As the stealth RNA vector SRV iPSC-1 Vector, SRV iPSC-2 Vector, SRV iPSC-3 Vector, and SRV iPSC-4 Vector (registered trademark, Tokiwa Bio Co., Ltd.) can be used. Details of stealth RNA vectors are described in Japanese Patent No. 4478788, Japanese Patent No. 4936482, Japanese Patent No. 5633075, and Japanese Patent No. 5963309.
- Multiplicity of infection is an indicator of Sendai virus titer.
- the MOI of Sendai virus is, for example, 0.1 to 100.0, or 1.0 to 50.0.
- the inducer is introduced into blood cells or immune cells using a chimeric virus comprising a virus-derived genomic RNA carrying the inducer RNA and a virus-derived envelope that covers the genomic RNA and is different from the genomic RNA. Good too.
- the genomic RNA of the chimeric virus may be derived from a paramyxovirus.
- the chimeric virus genomic RNA may be a stealth RNA vector.
- the genomic RNA of the chimeric virus contains the nucleocapsid protein (NP) gene, phosphorylated protein (P) gene/C protein (C) gene, matrix protein (M) gene, membrane fusion protein (F) gene, hemagglutinin-neuraminidase (
- NP nucleocapsid protein
- P phosphorylated protein
- C C protein
- M matrix protein
- F membrane fusion protein
- HN hemagglutinin-neuraminidase
- the NP gene, P gene/C gene, and L gene are involved in transcription and replication of viral vectors.
- the F gene, M gene, and NH gene are involved in virus particle formation. Therefore, a viral vector in which the functions of the F gene, M gene, and NH gene are all deleted cannot form new virus particles after infecting cells.
- the F gene, M gene, and NH gene may be deleted in the genomic RNA.
- the L gene which has a mutation that enables sustained gene expression, encodes a large protein whose amino acid sequence is valine at position 1618.
- a virus vector having the mutant L gene has a reduced interferon-inducing ability, has no cytotoxicity, and has the ability to sustain infection. Therefore, the expression of the loaded inducer RNA is sustained within the cell.
- siRNA targeting at least one of the NP gene, P gene, and mutant L gene may be introduced into the cells in order to remove the viral vector from the cells.
- siRNA targeting a region containing the 527th or 1913rd nucleotide of the mutant L gene may be introduced into cells.
- an undifferentiated cell-specific microRNA target sequence may be added to the non-coding region of at least one of the NP gene, P gene, and mutant L gene.
- An example of an undifferentiated cell-specific microRNA is miR-302a.
- the expression of undifferentiated cell-specific miRNAs such as miR-302a is induced in the cells.
- the undifferentiated cell-specific miRNA binds to the target sequence, the expression of at least one of the NP gene, P gene, and mutant L gene is suppressed, and the viral vector is removed from the cell.
- a miR-302a target sequence is added to the mutant L gene.
- the genomic RNA of the chimeric virus may have, from the 3' end, the viral NP gene, P gene, C gene, and a mutant L gene that enables continuous gene expression.
- the genomic RNA may have an inducer RNA between the C gene and the mutant L gene.
- the genomic RNA may have fluorescent protein RNA between the C gene and the mutant L gene.
- An example of a fluorescent protein is EGFP (Enhanced Green Fluorescent Protein).
- the genome may have the RNA of a drug resistance gene between the C gene and the mutant L gene.
- the envelope of the chimeric virus is, for example, derived from measles virus.
- the genome RNA has the NP gene, P gene, C gene, and mutant L gene of the virus, and has EGFP RNA and inducer RNA between the C gene and the mutant L gene, and the genome Tokiwa Bio was commissioned to produce a chimeric virus called MSRV-1, which has a measles virus-derived envelope that covers RNA. Tokiwa Bio was commissioned to produce a chimeric virus called MSRV-2 in which a target sequence for miR-302a was added to the mutated L gene of MSRV-1.
- the multiplicity of infection can be cited as an index of the titer of the chimeric virus.
- the MOI of the chimeric virus is, for example, 0.1 to 100.0, or 1.0 to 50.0.
- An inducing factor may be introduced into blood cells or immune cells that are adherently cultured, or may be introduced into blood cells or immunocytes that are cultured in suspension in a gel medium.
- the blood cells or immune cells into which the inducer is introduced are cultured feeder-free using a basement membrane matrix such as Matrigel (Corning), CELLstart (registered trademark, ThermoFisher), or Laminin 511 (iMatrix-511, Nippi). Good too.
- a basement membrane matrix such as Matrigel (Corning), CELLstart (registered trademark, ThermoFisher), or Laminin 511 (iMatrix-511, Nippi). Good too.
- a stem cell medium such as a human ES/iPS medium such as Stemfit (Ajinomoto) can be used.
- the stem cell culture medium is not limited to this, and various stem cell culture media can be used.
- Primate ES Cell Medium, mTeSR1, TeSR2 (STEMCELL Technologies), etc. may be used.
- the stem cell medium is placed, for example, in a dish, well, or tube.
- the gel medium does not need to be stirred. Moreover, the gel medium does not need to contain feeder cells.
- the gel medium may contain at least one substance selected from the group consisting of cadherin, laminin, fibronectin, and vitronectin.
- the cells After introducing an inducing factor into blood cells or immune cells, the cells may be initialized in a liquid medium other than a gel medium, or the cells may be initialized in a gel medium.
- the cells into which the inducer has been introduced are collected, and at least a portion of the collected and mixed cells is plated in a culture medium for at least one passage. It may be executed once. During passage, clones of cells into which the inducer has been introduced may be mixed. During passage, different clones of cells into which the inducer has been introduced may be mixed. Thereafter, the cells into which the inducer has been introduced may be collected, and at least a portion of the collected and mixed cells may be seeded in a medium and passaged multiple times.
- the cells into which the inducing factor has been introduced may be collected, and at least a portion of the collected mixed cells may be seeded in a culture medium and subcultured until stem cells are established. Note that all of the collected mixed cells may be seeded into the culture medium.
- collecting the cells into which the inducing factor has been introduced, and inoculating at least a portion of the collected and mixed cells into a culture medium means, for example, that the cells into which the inducing factor has been introduced are It means to passage without distinguishing between the two.
- cells into which an inducing factor has been introduced may be seeded in the same culture vessel without being differentiated by gene expression status.
- collecting the cells into which the inducing factor has been introduced, and inoculating at least a portion of the collected and mixed cells into a culture medium means, for example, that the cells into which the inducing factor has been introduced are It means to passage without distinguishing between the two.
- cells into which an induction factor has been introduced may be seeded in the same culture vessel without being differentiated by the degree of reprogramming.
- collecting the cells into which the inducing factor has been introduced, and inoculating at least a portion of the collected and mixed cells into a culture medium means, for example, distinguishing the cells into which the inducing factor has been introduced by morphology. It means to pass generations without any trouble.
- cells into which an inducing factor has been introduced may be seeded in the same culture vessel without being differentiated by morphology.
- collecting the cells into which the inducing factor has been introduced, and inoculating at least a portion of the collected and mixed cells into a culture medium means, for example, that the cells into which the inducing factor has been introduced are differentiated by size. It means to pass generations without any trouble.
- cells into which an inducing factor has been introduced may be seeded in the same culture vessel without distinguishing their sizes.
- collecting the cells into which the inducing factor has been introduced, and inoculating at least a portion of the collected and mixed cells into a medium for passage means that the cells into which the inducing factor has been introduced are passaged without cloning. It means to do something. For example, in the case of passage without cloning, it is not necessary to pick up colonies formed by cells into which an inducing factor has been introduced. For example, when subculturing without cloning, it is not necessary to separate multiple colonies formed by cells into which an inducing factor has been introduced. For example, during subculture, cells that have formed a plurality of different colonies may be mixed and seeded in the same culture vessel.
- the cells cultured adherently may be collected, and at least a portion of the collected and mixed cells may be seeded in a medium for subculture.
- cells may be detached from the culture vessel, and at least a portion of the detached and mixed cells may be seeded in the same culture vessel.
- the cells may be peeled from the culture vessel using a peeling solution, and the entire mixed cells may be subcultured.
- cells that do not form colonies may be passaged.
- the whole cells cultured in suspension may be passaged.
- the cells into which the inducing factor has been introduced may be cultured in a closed culture vessel and passaged.
- a closed incubator for example, does not exchange gases, viruses, microorganisms, impurities, etc. with the outside world.
- the cells into which the inducer has been introduced may be expanded by two-dimensional culture or three-dimensional culture.
- the entire adherent cultured cells may be cryopreserved as stem cells.
- whole cells detached from the culture vessel with a detachment solution may be cryopreserved as stem cells.
- the whole cells that are being cultured in suspension may be cryopreserved as stem cells.
- the induced stem cells can express undifferentiated cell markers such as Nanog, OCT4, and SOX2.
- the induced stem cells can express TERT.
- the induced stem cells may exhibit teostomerase activity.
- stem cells have been derived from blood cells or immune cells can be confirmed, for example, from the morphology of the cells.
- induced stem cells may form flat-shouldered colonies similar to ES cells and express alkaline phosphatase.
- whether or not stem cells are derived from blood cells or immune cells can be determined using a flow cytometer using cell surface markers that indicate undifferentiated cells such as TRA-1-60, TRA-1-81, SSEA-3, This may be carried out by analyzing whether at least one surface marker selected from SSEA-4 and SSEA5 is positive.
- TRA-1-60 is an antigen specific to iPS/ES cells and is not detected in somatic cells. Since iPS cells are generated only from the TRA-1-60 positive fraction, TRA-1-60 positive cells are considered to be iPS cells.
- the induced stem cells are equipped with, for example, a rearranged ⁇ -TCR gene.
- a rearranged ⁇ -TCR gene is a gene encoding TCR in which the TCR ⁇ region and TCR ⁇ region have been rearranged.
- the TCR ⁇ region includes V ⁇ -J ⁇ .
- the TCR ⁇ region includes V ⁇ -D ⁇ -J ⁇ .
- the induced stem cells are equipped with rearranged ⁇ -TCR genes with, for example, J1/J2 genes.
- a method for producing blood cells or immune cells includes preparing stem cells produced by the above-described method for producing stem cells, and inducing blood cells or immune cells from the stem cells.
- the method for inducing blood cells or immune cells from stem cells is not particularly limited, and includes, for example, glycogen synthase kinase-3 (GSK3) inhibitors such as CHIR99021, bone morphogenetic proteins such as bone morphogenetic protein-4 (BMP-4),
- GSK3 glycogen synthase kinase-3
- BMP-4 bone morphogenetic proteins
- the prepared cells are cultured for 4 days in a medium containing growth factors such as vascular endothelial growth factor (VEGF).
- VEGF vascular endothelial growth factor
- SB431542 growth factor
- growth factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF)
- SCF stem cell factor
- cells are cultured for 2 days in a medium containing growth factors such as VEGF, SCF, interleukins such as IL-3 and IL-6, cytokines such as Flt3L, and erythropoietin (EPO). Furthermore, cells are cultured in a medium containing SCF, interleukins such as IL-6, and EPO. This induces blood cells or immune cells.
- growth factors such as VEGF, SCF, interleukins such as IL-3 and IL-6, cytokines such as Flt3L, and erythropoietin (EPO).
- EPO erythropoietin
- stem cells may be seeded on stromal cells to induce blood cells or immune cells from the stem cells.
- the stromal cells may be derived from bone marrow.
- the stromal cells may be OP9 cells.
- OP9 cells do not produce macrophage stimulating factor (M-CSF) and have the function of supporting differentiation of stem cells into blood cells or immune cells.
- M-CSF macrophage stimulating factor
- a colony of stem cells is divided into a plurality of cell clusters, and the stem cell clusters are seeded onto OP9 cells as feeder cells.
- Blood cells or immune cells are thereby induced from the stem cells.
- the induced blood cells or immune cells are, for example, positive for CD34 and CD43.
- the induced blood cells or immune cells may be ⁇ type T cells.
- the method for producing ⁇ T cells includes inducing differentiation of ⁇ T cells from pluripotent stem cells derived from T cells.
- Pluripotent stem cells are, for example, iPS cells.
- Pluripotent stem cells are, for example, pluripotent stem cells derived from ⁇ T cells.
- Pluripotent stem cells are, for example, pluripotent stem cells derived from ⁇ T cells expressing the ⁇ T cell receptor.
- pluripotent stem cells may be induced from T cells by a method similar to the method for producing stem cells described above.
- pluripotent stem cells may be derived from T cells stimulated with IL-2 and zoledronic acid.
- the induced pluripotent stem cells may have a rearranged gene for the ⁇ T cell receptor.
- Inducing differentiation of ⁇ T cells from pluripotent stem cells may include inducing hematopoietic stem progenitor cells from pluripotent stem cells and inducing ⁇ T cells from hematopoietic stem progenitor cells.
- cells may be cultured feeder-free.
- cells may be cultured in suspension.
- Hematopoietic stem progenitor cells may be induced from pluripotent stem cells using a medium containing at least one of bone morphogenetic protein 4 (BMP-4), VEGF, and bFGF. Hematopoietic stem progenitor cells may be induced from pluripotent stem cells with a medium containing BMP-4, VEGF, and bFGF.
- BMP-4 bone morphogenetic protein 4
- VEGF vascular endothelial growth factor 4
- bFGF bone morphogenetic protein 4
- Hematopoietic stem progenitor cells may be induced from pluripotent stem cells using a medium containing an activin receptor-like kinase inhibitor.
- An activin receptor-like kinase inhibitor may be added to the medium containing BMP-4, VEGF, and bFGF.
- cells are cultured in a medium containing at least one of BMP-4, VEGF, and bFGF, and then, at least one of VEGF, bFGF, SCF, TPO, and FLT3L is cultured.
- Cells may be cultured in a medium containing
- cells While deriving hematopoietic stem progenitor cells from pluripotent stem cells, cells may be cultured in a medium containing BMP-4, VEGF, and bFGF, and then cells may be cultured in a medium containing VEGF, bFGF, and SCF. .
- cells are cultured in a medium containing BMP-4, VEGF, and bFGF; Cells may be cultured in a medium containing VEGF, bFGF, SCF, TPO, and FLT3L.
- Hematopoietic stem progenitor cells may be cultured on feeder cells.
- the feeder cells may be OP9 cells.
- ⁇ T cells induced to differentiate from pluripotent stem cells may express a ⁇ T cell receptor.
- ⁇ T cells can be used as pharmaceutical compositions for cancer treatment.
- Example 1 1 ⁇ mol/L to ⁇ mol/L tetrakis-pivaloyloxymethyl 2-(thiazol-2-ylamino)ethylidene-1,1-bisphosphonate (PTA, Techno Suzuta), 10% fetal bovine serum (Life Technologies), 1 RPMI1640 medium (Gibco) containing .0 ⁇ 10 ⁇ 5 mol/L 2-mercaptoethanol (Nacalai Tesque), 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin (Life Technologies) containing PTA according to Example 1. Prepared as a culture medium.
- Human peripheral blood mononuclear cells were placed in the PTA-containing medium according to Example 1, and the medium containing approximately 1 ⁇ 10 6 mononuclear cells was placed in a 24-well plate (day 1). The medium was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-2 daily. On the third day, the medium containing the cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, 2 mL of a new PTA-containing medium according to Example 1 was added to the cells, and 1 mL each was placed in two wells of a 24-well plate.
- the medium containing the cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, the PTA-containing medium according to Example 1 was added to the cells, and the medium containing 2 ⁇ 10 4 mononuclear cells was placed in a 96-well plate. KLF4, OCT3/4, SOX2, and c-MYC were introduced into the cells using MSRV (Tokiwa Bio). Multiplicity of infection (MOI) was adjusted to 5.
- Fresh PTA-containing medium according to Example 1 was added to the medium containing the cells collected from the 96-well plate on day 7 and placed in a 6-well plate coated with laminin (iMatrix-511, Nippi). On the 8th, 10th, and 12th days, a stem cell medium (Stem Fit, Ajinomoto) was added, and thereafter, the medium was replaced with the stem cell medium.
- laminin iMatrix-511, Nippi
- FIG. 3 A photograph of the established iPS cells is shown in Figure 2.
- genomic DNA was extracted from iPS cells and analyzed by PCR and electrophoresis to determine whether it contained the rearranged V ⁇ 9 gene.
- the genomic DNA of mononuclear cells (PBMC) was used as a positive control, and the genome of iPS cells that do not have the rearranged V ⁇ 9 gene was analyzed in the same manner as a negative control.
- PBMC mononuclear cells
- FIG. 3 it was confirmed that the iPS cells established in Example 1 had a rearranged V ⁇ 9 gene containing the JP gene and a rearranged V ⁇ 2 gene containing the J ⁇ 1 gene. It was done.
- Example 2 5 ⁇ mol/L zoledronic acid (Zol, Sigma-Aldrich), 10% fetal bovine serum (Life Technologies), 1.0 ⁇ 10 ⁇ 5 mol/L 2-mercaptoethanol (Nacalai Tesque), 100 U/mL penicillin. 1640 medium (Gibco) containing 100 ⁇ g/mL of streptomycin (Life Technologies) was prepared as the zoledronic acid-containing medium according to Example 2.
- Human peripheral blood mononuclear cells were placed in the zoledronic acid-containing medium according to Example 2, and the medium containing approximately 1 ⁇ 10 6 mononuclear cells was placed in a 24-well plate (day 1). 1 ⁇ L of 20 ⁇ g/mL IL-2 was added daily to the culture medium of the first mononuclear cell group. The culture medium of the second mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-9 daily. The medium of the third mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-33 daily.
- the culture medium of the fourth mononuclear cell group was supplemented with 1 ⁇ L each of 50 ⁇ g/mL IL-18 and 500 ⁇ g/mL IL-33 every day.
- the culture medium of the fifth mononuclear cell group was supplemented with 1 ⁇ L each of 5 ⁇ g/mL IL-4, 50 ⁇ g/mL IL-18, and 500 ⁇ g/mL IL-33 every day.
- the culture medium of the sixth mononuclear cell group was supplemented with 1 ⁇ L each of 5 ⁇ g/mL IL-4, 2 ⁇ g/mL IL-9, 50 ⁇ g/mL IL-18, and 500 ⁇ g/mL IL-33 every day. .
- the medium containing cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, 2 mL of a new zoledronic acid-containing medium according to Example 2 was added to the cells, and 1 mL each was placed in two wells of a 24-well plate.
- Figure 4 shows the percentage of ⁇ T cells examined on day 6 using an anti- ⁇ 9 antibody and an anti- ⁇ 2 antibody using a flow cytometer. The number of ⁇ T cells derived from 2 ⁇ 10 4 mononuclear cells is shown in FIG. 5.
- the medium containing the cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, the zoledronic acid-containing medium according to Example 2 was added to the cells, and the medium containing 2 ⁇ 10 4 mononuclear cells was placed in a 96-well plate. KLF4, OCT3/4, SOX2, and c-MYC were introduced into the cells using MSRV (Tokiwa Bio). Multiplicity of infection (MOI) was adjusted to 5.
- Fresh zoledronic acid-containing medium according to Example 2 was added to the medium containing the cells collected from the 96-well plate on the 7th day and placed in a 6-well plate. On the 8th, 10th, and 12th days, a stem cell medium (Stem Fit, Ajinomoto) was added, and thereafter, the medium was replaced with the stem cell medium.
- a stem cell medium Ste Fit, Ajinomoto
- Example 3 iPS cells were induced from mononuclear cells in the same manner as in Example 2, except that zoledronic acid in the zoledronic acid-containing medium according to Example 2 was changed to HMBPP. Note that only IL-2 was used as the interleukin.
- Example 4 Human peripheral blood mononuclear cells were placed in the zoledronic acid-containing medium according to Example 2, and the medium containing approximately 1 ⁇ 10 6 mononuclear cells was placed in a 24-well plate (day 1). No interleukin was added to the culture medium of the first mononuclear cell group.
- the culture medium of the second mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-2 daily.
- the medium of the third mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-9 daily.
- the culture medium of the fourth mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-18 daily.
- the culture medium of the fifth mononuclear cell group was supplemented with 1 ⁇ L of 20 ⁇ g/mL IL-4 every day.
- 1 ⁇ L each of 20 ⁇ g/mL IL-9 and 20 ⁇ g/mL IL-18 was added to the culture medium of the sixth mononuclear cell group every day.
- 1 ⁇ L each of 20 ⁇ g/mL IL-4 and 20 ⁇ g/mL IL-18 was added to the culture medium of the seventh mononuclear cell group every day.
- To the culture medium of the eighth mononuclear cell group, 1 ⁇ L each of 20 ⁇ g/mL IL-4, 20 ⁇ g/mL IL-9, and 20 ⁇ g/mL IL-18 were added daily.
- a zoledronic acid-free medium which is the same as the zoledronic acid-containing medium according to Example 2 except that it does not contain zoledronic acid, was prepared, and the zoledronic acid-free medium for the ninth mononuclear cell group contained 20 ⁇ g/mL daily. 1 ⁇ L of IL-2 was added.
- the medium containing cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, 2 mL of a new zoledronic acid-containing medium according to Example 2 was added to the first to eighth cell groups, and 1 mL each was placed in two wells of a 24-well plate. In addition, 2 mL of fresh zoledronic acid-free medium was added to the ninth cell group, and 1 mL each was added to two wells of a 24-well plate.
- the medium containing the cells was collected from the plate, the medium was centrifuged, and the supernatant was removed. Thereafter, the zoledronic acid-containing medium according to Example 2 was added to the first to eighth cell groups, and the zoledronic acid-free medium was added to the ninth cell group, containing 2 ⁇ 10 4 mononuclear cells.
- the medium was placed in a 96-well plate. KLF4, OCT3/4, SOX2, and c-MYC were introduced into the cells using MSRV (Tokiwa Bio). Multiplicity of infection (MOI) was adjusted to 5.
- Fresh zoledronic acid-containing medium according to Example 2 was added to the medium containing the first to eighth cell groups collected from the 96-well plate on the seventh day, and fresh zoledronic acid was added to the medium containing the ninth cell group. Acid-free medium was added and placed in a 6-well plate. On the 8th, 10th, and 12th days, a stem cell medium (Stem Fit, Ajinomoto) was added, and thereafter, the medium was replaced with the stem cell medium.
- Ste cell medium Ste Fit, Ajinomoto
- Example 5 ⁇ MEM medium supplemented with FBS at a final concentration of 20% and penicillin/streptomycin at a final concentration of 1% was prepared as a medium for OP9 cells.
- Vitamin C at a final concentration of 100 mmol/L
- interleukin-7 IL-7
- FLT3 ligand at a final concentration of 10 ⁇ g/mL
- SCF stem cell factor
- Each colony of iPS cells induced using only IL-2 as the interleukin in Examples 2 and 3 was detached from the dish using a cell scraper and divided into multiple cell clusters by pipetting.
- a dish in which OP9 cells were cultured as feeder cells was prepared.
- OP9 cells were cultured in OP9 cell medium.
- a plurality of cell clusters consisting of iPS cells were seeded on the feeder cells, and cultured for 2 days using a stem cell medium supplemented with Y27632 at a final concentration of 10 ⁇ mol/L.
- the medium was replaced with a medium for OP9 cells supplemented with vitamin C, and the cells were cultured for an additional 12 days.
- half of the medium was removed from the dish and replaced with a fresh OP9 medium supplemented with vitamin C.
- Figure 10 shows the results of flow cytometry analysis of cells cultured on OP9/DLL1 for 28 days.
- the cells were positive for both lymphoid progenitor cell markers CD5-PE and CD7-FITC.
- a cell population positive for ⁇ TCR-APC, a ⁇ T cell marker was confirmed among the CD3-PE positive population, a Pan-T cell marker.
- a CD3-PE monopositive ⁇ T cell population was also confirmed.
- T progenitor cells In order to induce cells that had been induced to become T progenitor cells to become ⁇ T cells, strong stimulation was applied to the cells via ⁇ T cell receptor (TCR).
- HMBPP (0.01 ⁇ g/mL to 1 ⁇ g/mL) or zoledronic acid (5 ⁇ mol/L) was added to the medium as a stimulant, and the cells were cultured until day 35.
- the feeder cells were changed from OP9/DLL1 cells that express Notch ligands to OP9 cells that do not express Notch ligands to inhibit induction into ⁇ T cells.
- Figure 11 shows the results of flow cytometry analysis of cells on day 35. It was confirmed that CD3 and ⁇ TCR (V ⁇ 9) were expressed on the cell surface. The marker expression was higher when the feeder cells were OP9-DLL1 cells than when the feeder cells were OP9 cells. In the negative control, in which no stimulant was added, there was no marker expression.
- Genomic DNA was extracted from the iPS cells established in Example 2, and analyzed by PCR and electrophoresis to determine whether they contained the rearranged V ⁇ 9 gene.
- the genomic DNA of mononuclear cells (PBMC) was used as a positive control, and the genome of iPS cells that do not have the rearranged V ⁇ 9 gene was analyzed in the same manner as a negative control.
- PBMC mononuclear cells
- FIG. 12 it was confirmed that the iPS cells established in Example 1 had a rearranged V ⁇ 9 gene containing the JP gene and a rearranged V ⁇ 2 gene containing the J ⁇ 1 gene. It was done.
- Example 7 In the same manner as in Example 4, iPS cells were established by adding only IL-2 to a zoledronic acid-containing medium. In addition, by the same method as in Example 4, iPS cells were established by adding IL-4, IL-9, and IL-18 to the zoledronic acid-containing medium. As shown in Figure 13, the number of iPS cell colonies established was higher when IL-4, IL-9, and IL-18 were added to the medium than when only IL-2 was added to the medium. There were many.
- iPS cells induced from ⁇ T cells were prepared in a medium supplemented with IL-2 and zoledronic acid, and the iPS cells were adherently cultured. iPS cells were collected using TrypLe Select and suspended in iPS cell medium (Puel, I Peace, Inc.). iPS cells were placed in a low adhesion well plate, 10 ⁇ mol/L ROCK inhibitor and 10 ⁇ mol/L GSK-3 inhibitor were added to the medium, and the cells were cultured at 37° C. in a 5% CO 2 environment. Thereafter, the cell suspension was collected, and the cell suspension was centrifuged to remove the supernatant.
- L-glutamine replacement (Gibco GlutaMAX supplement), 50 ⁇ g/mL ascorbic acid 2-phosphate, 40 mmol/L monothioglycerol (MTG), insulin-transferrin-selenium solution (ITS), and penicillin/streptomycin.
- the added serum-free medium (Gibco StemPro-34 SFM) was prepared as a differentiation medium.
- BMP-4 bone morphogenetic protein 4
- VEGF vascular endothelial growth factor
- bFGF vascular endothelial growth factor
- the cell suspension was collected, centrifuged, and the supernatant was removed.
- the cells were suspended in a differentiation medium supplemented with 50 ng/mL VEGF, 50 ng/mL bFGF, and 50 ng/mL SCF, and cultured at 37° C. in a 5% CO 2 environment.
- the cell suspension was collected, centrifuged, and the supernatant was removed.
- Cells were suspended in differentiation medium supplemented with 50 ng/mL VEGF, 50 ng/mL bFGF, 50 ng/mL SCF, 30 ng/mL TPO, and 10 ng/mL FLT3L, and incubated at 37°C in a 5% CO 2 environment. Cells were cultured under Thereafter, cells were collected every 2 to 3 days and cultured in a differentiation medium containing the same additives until the 14th day to form embryoid bodies.
- the cell suspension was collected and the embryoid bodies were disrupted by pipetting. Furthermore, the cells obtained by disrupting the embryoid bodies were passed through a 40 ⁇ m filter to collect only hematopoietic stem progenitor cells.
- OP9/DLL1 cells were prepared as feeder cells. Additionally, ⁇ -MEM supplemented with FBS, GlutaMAX supplement, and penicillin/streptomycin was prepared as OP9 medium. OP9/DLL1 cells were seeded in a 6 cm dish and cultured overnight in OP9 medium.
- Hematopoietic stem progenitor cells were suspended in OP9 medium supplemented with SCF, IL-7, FLT3L, and ascorbic acid 2-phosphate, and seeded onto OP9/DLL1 cells. Thereafter, the medium was replaced with fresh OP9 medium every 2 to 3 days, and the cells were passaged onto new OP9/DLL1 cells every week to culture the cells for 3 weeks.
- stem cells become positive for CD45, a blood cell marker, and then become positive for CD3, a T cell marker, and positive for TCR ⁇ 9 and TCR ⁇ 2, which are ⁇ T cell markers. It is thought that it will become. Therefore, cells exhibiting a combination of CD3-positive and CD8-positive, a combination of CD7-positive and CD5-negative, a combination of CD7-positive and CD5-positive, a combination of CD45-positive, TCR ⁇ 9-positive, and TCR ⁇ 2-negative are in the process of being induced into ⁇ T cells. It can be a cell.
- IL-2 and a serum-free lymphocyte medium (X-VIVO10, registered trademark) supplemented with beads (Dynabeads, registered trademark, Human T-Activator CD3/CD28, ThermoFisher) with anti-CD3 and anti-CD28 monoclonal antibodies bound to the surface.
- iPS cells induced from ⁇ T cells ( ⁇ T-iPS cells) using KLF4, OCT3/4, SOX2, and c-MYC were prepared in the same manner as in Example 8.
- Hematopoietic stem progenitor cells were induced.
- hematopoietic stem progenitor cells were cultured for 3 weeks in the same manner as in Example 8. When the cells cultured for 3 weeks were analyzed using a flow cytometer, they were positive for CD45, but as shown in FIG. 17, they were negative for TCR ⁇ 9 and TCR ⁇ 2, and ⁇ T cells were not induced.
- Example 9 The ⁇ T cells produced in Example 8 were collected, centrifuged, and the supernatant was removed. Next, IL-7, IL-2, dexamethasone, and anti-CD3 (OKT3) antibody were added to the differentiation medium, the cells were suspended in the differentiation medium, and the cells were cultured at 37°C in a 5% CO 2 environment. did. After 3 days, the cells were harvested and washed with PBS, the cells were centrifuged, and the supernatant was removed. Next, IL-7, IL-2, and dexamethasone were added to the differentiation medium, the cells were suspended in the differentiation medium, and the cells were cultured at 37° C. in a 5% CO 2 environment. After 4 days, mature ⁇ T cells were obtained. When mature ⁇ T cells were analyzed using a flow cytometer, as shown in FIG. 18, more than 98% of the cells were positive for the combination of CD3 and CD45 and the combination of V ⁇ 9 and CD3.
- Mature ⁇ T cells were collected, centrifuged, and the supernatant was removed.
- IL-2, IL-4, IL-9, and IL-18 were added to the differentiation medium, the cells were suspended in the differentiation medium, and the cells were placed in wells coated with anti-CD3 (OKT3) antibody and retronectin. were seeded, and the cells were cultured at 37° C. in a 5% CO 2 environment.
- the cells were harvested and washed with PBS, the cells were centrifuged, and the supernatant was removed.
- the cells were suspended in a differentiation medium supplemented with IL-2, IL-4, IL-9, and IL-18, and cultured at 37° C. in a 5% CO 2 environment.
- the cells were repeatedly subcultured to a wider well, thereby expanding mature ⁇ T cells.
- the cell morphology indicates that proliferating ⁇ T cells are activated by IL-2, IL-4, IL-9, and IL-18, and anti-CD3 (OKT3) antibody. confirmed.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- Organic Chemistry (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Hematology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Virology (AREA)
- Developmental Biology & Embryology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
1μmol/Lからμmol/Lのテトラキス-ピバロイルオキシメチル2-(チアゾール-2-イルアミノ)エチリデン-1,1-ビスホスホネート(PTA、テクノ鈴田)、10%のウシ胎児血清(Life Technologies)、1.0×10-5mol/Lの2-メルカプトエタノール(ナカライテスク)、100U/mLのペニシリン、及び100μg/mLのストレプトマイシン(Life Technologies)を含むRPMI1640培地(Gibco)を実施例1に係るPTA含有培地として用意した。
5μmol/Lのゾレドロン酸(Zol、Sigma-Aldrich)、10%のウシ胎児血清(Life Technologies)、1.0×10-5mol/Lの2-メルカプトエタノール(ナカライテスク)、100U/mLのペニシリン、及び100μg/mLのストレプトマイシン(Life Technologies)を含む)1640培地(Gibco)を実施例2に係るゾレドロン酸含有培地として用意した。
実施例2に係るゾレドロン酸含有培地のゾレドロン酸をHMBPPに変えた以外は、実施例2と同様に、単核球からiPS細胞を誘導した。なお、インターロイキンはIL-2のみを用いた。
実施例2に係るゾレドロン酸含有培地にヒト末梢血単核球を入れ、約1×106個の単核球を含む培地を24ウェルプレートに入れた(1日目)。第1の単核球群の培地には、インターロイキンを添加しなかった。第2の単核球群の培地には、毎日20μg/mLのIL-2を1μL添加した。第3の単核球群の培地には、毎日20μg/mLのIL-9を1μL添加した。第4の単核球群の培地には、毎日20μg/mLのIL-18を1μL添加した。第5の単核球群の培地には、毎日20μg/mLのIL-4を1μL添加した。第6の単核球群の培地には、毎日20μg/mLのIL-9と20μg/mLのIL-18をそれぞれ1μL添加した。第7の単核球群の培地には、毎日20μg/mLのIL-4と20μg/mLのIL-18をそれぞれ1μL添加した。第8の単核球群の培地には、毎日20μg/mLのIL-4と、毎日20μg/mLのIL-9と、20μg/mLのIL-18をそれぞれ1μL添加した。また、実施例2に係るゾレドロン酸含有培地とはゾレドロン酸を含有しない以外は同じゾレドロン酸非含有培地を用意し、第9の単核球群のゾレドロン酸非含有培地には、毎日20μg/mLのIL-2を1μL添加した。
終濃度が20%のFBS、及び終濃度が1%のペニシリン/ストレプトマイシンを添加されたαMEM培地をOP9細胞用培地として用意した。
実施例2で樹立したiPS細胞からゲノムDNAを抽出し、再構成されたVγ9遺伝子を有するかを、PCRと電気泳動により解析した。陽性コントロールとして単核球(PBMC)のゲノムDNA、陰性コントロールとして再構成されたVγ9遺伝子を有しないiPS細胞のゲノムを同様に解析した。その結果、図12に示すように、実施例1で樹立されたiPS細胞は、JP遺伝子を有する再構成されたVγ9遺伝子と、Jδ1遺伝子を有する再構成されたVδ2遺伝子をと、有することが確認された。
実施例4と同様の方法により、ゾレドロン酸含有培地にIL-2のみを添加してiPS細胞を樹立した。また、実施例4と同様の方法により、ゾレドロン酸含有培地にIL-4、IL-9、及びIL-18を添加してiPS細胞を樹立した。図13に示すように、培地にIL-2のみを添加した場合と比較して、培地にIL-4、IL-9、及びIL-18を添加したほうが、樹立されたiPS細胞のコロニー数は多かった。
IL-2及びゾレドロン酸を添加した培地中でγδT細胞から誘導されたiPS細胞(γδT-iPS細胞)を用意し、iPS細胞を接着培養した。TrypLe Selectを用いてiPS細胞を回収し、iPS細胞をiPS細胞用培地(Puel、I Peace,Inc.)に懸濁した。iPS細胞を低接着ウェルプレートに入れ、培地に10μmol/LのROCK阻害剤と10μmol/LのGSK-3阻害剤を添加し、37℃、5%CO2環境下で細胞を培養した。その後、細胞浮遊液を回収し、細胞浮遊液を遠心して上清を除いた。
IL-2と、表面に抗CD3及び抗CD28モノクローナル抗体を結合したビーズ(Dynabeads、登録商標、Human T-Activator CD3/CD28、ThermoFisher)を添加した無血清リンパ球培地(X-VIVO10、登録商標、Lonza)中で、KLF4、OCT3/4、SOX2、c-MYCを用いてαβT細胞から誘導されたiPS細胞(αβT-iPS細胞)を用意し、実施例8と同じ方法で、αβT-iPS細胞から造血幹前駆細胞を誘導した。さらに、実施例8と同じ方法で、造血幹前駆細胞を3週間培養した。3週間培養された細胞をフローサイトメーターで分析したところ、CD45陽性であったものの、図17に示すように、TCRγ9及びTCRδ2が陰性であり、γδT細胞が誘導されなかった。
IL-6、SCF、TPO、FLT3リガンド、及びIL-3を添加した血球培地(StemSpan H3000、STEMCELLTechnologies)中で、KLF4、OCT3/4、SOX2、c-MYCを用いて、再構成されたTCR遺伝子を有しないnonT細胞から誘導されたiPS細胞(nonT-iPS細胞)を用意し、実施例8と同じ方法で、αβT-iPS細胞から造血幹前駆細胞を誘導した。さらに、実施例8と同じ方法で、造血幹前駆細胞を3週間培養した。3週間培養された細胞をフローサイトメーターで分析したところ、CD45陽性であったものの、図17に示すように、TCRγ9及びTCRδ2が陰性であり、γδT細胞が誘導されなかった。
実施例8で作製したγδT細胞を回収し、遠心して上清を除去した。次に、分化用培地にIL-7、IL-2、デキサメタゾン、及び抗CD3(OKT3)抗体を添加し、分化用培地に細胞を懸濁し、37℃、5%CO2環境下で細胞を培養した。3日後、細胞を回収してPBSで洗浄し、細胞を遠心して、上清を除去した。次に、分化用培地にIL-7、IL-2、及びデキサメタゾンを添加し、分化用培地に細胞を懸濁し、37℃、5%CO2環境下で細胞を培養した。4日後、成熟したγδT細胞が得られた。成熟したγδT細胞フローサイトメーターで分析したところ、図18に示すように、98%以上の細胞において、CD3及びCD45の組み合わせ、Vγ9及びCD3の組み合わせが陽性であった。
Claims (28)
- 血液細胞又は免疫細胞にテトラキス-ピバロイルオキシメチル2-(チアゾール-2-イルアミノ)エチリデン-1,1-ビスホスホネートを適用することと、
前記血液細胞又は前記免疫細胞から幹細胞を誘導することと、
を含む、幹細胞の製造方法。 - 前記血液細胞又は前記免疫細胞にインターロイキンを適用することをさらに含む、請求項1に記載の幹細胞の製造方法。
- 前記インターロイキンが、IL-2、IL-4、IL-9、IL-18、及びIL-33からなる群から選択される少なくとも一つである、請求項2に記載の幹細胞の製造方法。
- 前記幹細胞が、iPS細胞である、請求項1に記載の幹細胞の製造方法。
- 請求項1に記載の幹細胞の製造方法で製造された幹細胞を用意することと、
前記幹細胞から血液細胞又は免疫細胞を誘導することと、
を含む、血液細胞又は免疫細胞の製造方法。 - 血液細胞又は免疫細胞に、ビスホスホネート又は(E)-4-ヒドロキシ-3-メチル-ブト-2-エニルピロリン酸と、インターロイキンと、を適用することと、
前記血液細胞又は前記免疫細胞から幹細胞を誘導することと、
を含み、
前記インターロイキンが、IL-4、IL-9、IL-18、及びIL-33からなる群から選択される少なくとも一つである、
幹細胞の製造方法。 - 前記幹細胞が、iPS細胞である、請求項6に記載の幹細胞の製造方法。
- 請求項6に記載の幹細胞の製造方法で製造された幹細胞を用意することと、
前記幹細胞から血液細胞又は免疫細胞を誘導することと、
を含む、血液細胞又は免疫細胞の製造方法。 - 血液細胞又は免疫細胞に、ビスホスホネート又は(E)-4-ヒドロキシ-3-メチル-ブト-2-エニルピロリン酸を適用することと、
前記血液細胞又は免疫細胞から幹細胞を誘導することと、
を含み、
前記血液細胞又は免疫細胞にIL-2を適用しない、
幹細胞の製造方法。 - 前記血液細胞又は前記免疫細胞にあらゆるインターロイキンを適用しない、請求項9に記載の幹細胞の製造方法。
- 前記幹細胞が、iPS細胞である、請求項9に記載の幹細胞の製造方法。
- 前記幹細胞が、再構成されたγδ-TCR遺伝子を備える、請求項9に記載の幹細胞の製造方法。
- 請求項9に記載の幹細胞の製造方法で製造された幹細胞を用意することと、
前記幹細胞から血液細胞又は免疫細胞を誘導することと、
を含む、血液細胞又は免疫細胞の製造方法。 - 血液細胞又は免疫細胞にテトラキス-ピバロイルオキシメチル2-(チアゾール-2-イルアミノ)エチリデン-1,1-ビスホスホネートを適用することと、
前記血液細胞又は前記免疫細胞から幹細胞を誘導することと、
を含み、
前記血液細胞又は前記免疫細胞にIL-2を適用しない、
幹細胞の製造方法。 - 前記血液細胞又は前記免疫細胞にIL-2以外のインターロイキンを適用することをさらに含む、請求項14に記載の幹細胞の製造方法。
- 前記幹細胞が、再構成されたγδ-TCR遺伝子を備える、請求項14に記載の幹細胞の製造方法。
- 請求項14に記載の幹細胞の製造方法で製造された幹細胞を用意することと、
前記幹細胞から血液細胞又は免疫細胞を誘導することと、
を含む、血液細胞又は免疫細胞の製造方法。 - 血液細胞又は免疫細胞に、(E)-4-ヒドロキシ-3-メチル-ブト-2-エニルピロリン酸と、インターロイキンと、を適用することと、
前記血液細胞又は前記免疫細胞から幹細胞を誘導することと、
を含み、
前記インターロイキンがIL-2を含まない、
幹細胞の製造方法。 - 前記インターロイキンが、IL-4、IL-9、IL-18、及びIL-33からなる群から選択される少なくとも一つである、請求項18に記載の幹細胞の製造方法。
- 請求項18に記載の幹細胞の製造方法で製造された幹細胞を用意することと、
前記幹細胞から血液細胞又は免疫細胞を誘導することと、
を含む、血液細胞又は免疫細胞の製造方法。 - T細胞から誘導された多能性幹細胞からγδT細胞を分化誘導することを含む、γδT細胞の製造方法。
- 前記多能性幹細胞がγδT細胞から誘導されている、請求項21に記載のδT細胞の製造方法。
- 前記多能性幹細胞に誘導されたγδT細胞が、γδT細胞受容体を発現している、請求項21に記載のδT細胞の製造方法。
- 前記多能性幹細胞が、γδT細胞受容体の再構成遺伝子を有する、請求項21に記載のδT細胞の製造方法。
- 前記分化誘導されたγδT細胞が、γδT細胞受容体を発現している、請求項21に記載のδT細胞の製造方法。
- 前記多能性幹細胞から前記γδT細胞を分化誘導することが、
前記多能性幹細胞から造血幹前駆細胞を誘導することと、
前記造血幹前駆細胞から前記γδT細胞を誘導することと、
を含む、請求項21に記載のδT細胞の製造方法。 - γδT細胞から誘導された多能性幹細胞から分化誘導されたγδT細胞。
- γδT細胞から誘導された多能性幹細胞から分化誘導されたγδT細胞を含む、がん治療のための医薬品組成物。
Priority Applications (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2024523014A JPWO2023228728A1 (ja) | 2022-05-23 | 2023-05-09 | |
| US18/868,650 US20250354115A1 (en) | 2022-05-23 | 2023-05-09 | Method for producing stem cells and method for producing gamma delta t cells |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263344860P | 2022-05-23 | 2022-05-23 | |
| US63/344,860 | 2022-05-23 | ||
| US202363492196P | 2023-03-24 | 2023-03-24 | |
| US63/492,196 | 2023-03-24 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2023228728A1 true WO2023228728A1 (ja) | 2023-11-30 |
Family
ID=88919052
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2023/017460 Ceased WO2023228728A1 (ja) | 2022-05-23 | 2023-05-09 | 幹細胞の製造方法及びγδT細胞の製造方法 |
Country Status (3)
| Country | Link |
|---|---|
| US (1) | US20250354115A1 (ja) |
| JP (1) | JPWO2023228728A1 (ja) |
| WO (1) | WO2023228728A1 (ja) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2018143243A1 (ja) * | 2017-02-03 | 2018-08-09 | 国立大学法人神戸大学 | 人工多能性幹細胞の作製方法 |
Family Cites Families (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2020027193A1 (ja) * | 2018-07-31 | 2020-02-06 | 国立大学法人三重大学 | γδ型T細胞への遺伝子導入方法 |
-
2023
- 2023-05-09 JP JP2024523014A patent/JPWO2023228728A1/ja active Pending
- 2023-05-09 US US18/868,650 patent/US20250354115A1/en active Pending
- 2023-05-09 WO PCT/JP2023/017460 patent/WO2023228728A1/ja not_active Ceased
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2018143243A1 (ja) * | 2017-02-03 | 2018-08-09 | 国立大学法人神戸大学 | 人工多能性幹細胞の作製方法 |
Non-Patent Citations (1)
| Title |
|---|
| OKUNO DAISUKE, SUGIURA YUKI, SAKAMOTO NORIHO, TAGOD MOHAMMED S. O., IWASAKI MASASHI, NODA SHUTO, TAMURA AKIHIRO, SENJU HIROAKI, UM: "Comparison of a Novel Bisphosphonate Prodrug and Zoledronic Acid in the Induction of Cytotoxicity in Human Vγ2Vδ2 T Cells", FRONTIERS IN IMMUNOLOGY, vol. 11, 21 July 2020 (2020-07-21), pages 1405, XP093092610, DOI: 10.3389/fimmu.2020.01405 * |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2023228728A1 (ja) | 2023-11-30 |
| US20250354115A1 (en) | 2025-11-20 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP5984217B2 (ja) | 少量の末梢血からの人工多能性幹細胞の作製 | |
| JP6736003B2 (ja) | 多能性幹細胞からt細胞への誘導方法 | |
| JP6948072B2 (ja) | Cd8陽性t細胞を誘導する方法 | |
| JP7224021B2 (ja) | 人工多能性幹細胞の作製方法 | |
| JP7610817B2 (ja) | 造血前駆細胞マーカー | |
| US20230220344A1 (en) | Induced pluripotent cell comprising a controllable transgene for conditional immortalisation | |
| WO2015099134A1 (ja) | 再構成されたt細胞レセプター遺伝子を有する多能性幹細胞由来のt前駆細胞を用いる免疫細胞療法 | |
| CN115087732B (zh) | 来自人T细胞来源的iPS细胞的细胞毒性T细胞 | |
| JPWO2019070021A1 (ja) | iPS細胞由来の遺伝的多様性を有するT細胞集団の製造方法 | |
| US20250354115A1 (en) | Method for producing stem cells and method for producing gamma delta t cells | |
| JP2023086679A (ja) | 幹細胞の製造方法及び体細胞の製造方法 | |
| JP7758307B2 (ja) | 幹細胞の製造方法 | |
| EP3564385A1 (en) | Evaluation method and selection method for induced pluripotent stem cells, and production method for induced pluripotent stem cells | |
| Kawai et al. | Redifferentiation of Adaptive Naïve-Like CTL from T-Cell-Derived iPSC | |
| CN120829933A (zh) | 嵌合抗原受体多能干细胞分化为免疫细胞的方法和应用 | |
| Shanmugam | Generation of induced pluripotent stem cells and mesenchymal stromal cells. | |
| HK40013499A (en) | Evaluation method and selection method for induced pluripotent stem cells, and production method for induced pluripotent stem cells |
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: 23811603 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024523014 Country of ref document: JP |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 18868650 Country of ref document: US |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 23811603 Country of ref document: EP Kind code of ref document: A1 |
|
| WWP | Wipo information: published in national office |
Ref document number: 18868650 Country of ref document: US |