WO2024171957A1 - Transplantation of pituitary hormone-producing cells - Google Patents
Transplantation of pituitary hormone-producing cells Download PDFInfo
- Publication number
- WO2024171957A1 WO2024171957A1 PCT/JP2024/004478 JP2024004478W WO2024171957A1 WO 2024171957 A1 WO2024171957 A1 WO 2024171957A1 JP 2024004478 W JP2024004478 W JP 2024004478W WO 2024171957 A1 WO2024171957 A1 WO 2024171957A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- pituitary
- cell
- tissue
- producing cells
- 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
Images
Classifications
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K67/00—Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
-
- 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
-
- 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
- A61K35/48—Reproductive organs
- A61K35/54—Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
- A61K35/545—Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/06—Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/10—Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/38—Drugs for disorders of the endocrine system of the suprarenal hormones
-
- 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
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
Definitions
- the present invention relates to a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized in that the pituitary hormone-producing cells are used to be transplanted into the subcutaneous tissue and/or muscle tissue of a subject.
- the pituitary gland is an endocrine organ located in the head that produces various pituitary hormones, such as adrenocorticotropic hormone (ACTH) and growth hormone, which are important for the maintenance and growth of the body.
- ACTH adrenocorticotropic hormone
- growth hormone which are important for the maintenance and growth of the body.
- ACTH adrenocorticotropic hormone
- pituitary gland malfunctions due to diseases such as pituitary aplasia, hypopituitarism, or pituitary adenoma
- serious symptoms such as growth disorders, reproductive organ abnormalities, and abnormalities of the adrenal gland or thyroid gland occur.
- it is rare for damaged pituitary tissue to regenerate naturally and regain function.
- the objective of the present invention is therefore to provide a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used for transplantation into a transplantation site (subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue) that is less invasive and can be easily surgically removed even if the transplanted cells become tumorous and require removal, in order to realize clinical application of the pituitary hormone-producing cells.
- a transplantation site subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue
- the present inventors have found that pituitary hormone-producing cells induced to differentiate in vitro from pluripotent stem cells can be engrafted when transplanted into subcutaneous tissue, despite the existing belief that subcutaneous tissue is unsuitable for transplantation of cells because of the lack of blood vessels in the subcutaneous tissue itself.
- the inventors have found that transplantation into these regions, particularly into the groin, armpits, abdomen, back, groin, thighs, buttocks, upper arms, etc., which are rich in white fat, is more effective, and that angiogenesis is involved in cell engraftment.
- the inventors have also found that muscle tissue present under the subcutaneous tissue is suitable for transplantation of the pituitary hormone-producing cells, just like subcutaneous tissue.
- the inventors have completed the present invention, which is characterized in that the pituitary hormone-producing cells are used for transplantation into subcutaneous tissue and/or muscle tissue.
- the present invention is as follows.
- a therapeutic agent for a disease caused by a disorder of the pituitary gland comprising pituitary hormone-producing cells, characterized in that the pituitary hormone-producing cells are used to be transplanted into the subcutaneous tissue and/or muscle tissue of a subject.
- the subcutaneous tissue is subcutaneous tissue present in at least one area selected from the groin, armpit, back, abdomen, thigh, buttocks, upper arm, and scalp.
- the therapeutic agent described in [1] or [2], wherein the subcutaneous tissue is subcutaneous adipose tissue.
- the therapeutic agent described in [3], wherein the adipose tissue is white adipose tissue.
- the therapeutic agent described in [1], wherein the muscle tissue is muscle tissue present in at least one area selected from the back, abdomen, thighs, buttocks, and upper arms.
- the disease caused by a pituitary disorder is at least one disease selected from hypopituitarism, pituitary dwarfism, adrenal insufficiency, partial hypopituitarism, isolated anterior pituitary hormone deficiency, and craniopharyngioma.
- the pituitary hormone-producing cells are cell aggregates containing pituitary hormone-producing cells.
- the pituitary hormone-producing cells are pituitary hormone-producing cells that express an angiogenic factor.
- the angiogenic factor is one or more selected from the group consisting of VEGFA, VEGFB, VEGFC and ANGPT2.
- the pituitary hormone-producing cells are pituitary hormone-producing cells produced by a method comprising the steps of: (1) a first step of culturing pluripotent stem cells in the presence of a c-jun N-terminal kinase (JNK) signaling pathway inhibitor and a Wnt signaling pathway inhibitor to obtain a cell population; (2) A second step of culturing the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary hormone-producing cells.
- JNK c-jun N-terminal kinase
- [15] The therapeutic agent according to any one of [1] to [14], wherein angiogenesis occurs around the transplanted pituitary hormone-producing cells in a human subject after transplantation.
- the therapeutic agent according to any one of [1] to [15] which is used in combination with an angiogenesis promoter.
- the therapeutic agent according to any one of [1] to [16], wherein the pituitary hormone-producing cells are engrafted in subcutaneous tissue and/or muscle tissue of a human subject after transplantation.
- the therapeutic agent according to any one of [1] to [17] characterized in that a pituitary hormone is secreted into the blood in a human subject after transplantation.
- a method for treating a disease caused by a pituitary gland disorder comprising administering pituitary hormone-producing cells to the subcutaneous tissue and/or muscle tissue of a human subject.
- a method for producing a non-human animal model bearing human pituitary tissue which comprises administering human pituitary hormone-producing cells into the subcutaneous tissue and/or muscle tissue of the non-human animal.
- a non-human animal model bearing human pituitary gland tissue which comprises human pituitary hormone-producing cells in subcutaneous tissue and/or muscle tissue, and which is capable of secreting pituitary hormones produced by the human pituitary hormone-producing cells into blood.
- a method for evaluating the efficacy and safety of a test substance comprising administering the test substance to the non-human animal model bearing human pituitary tissue according to [21].
- the present invention makes it possible to provide a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used for transplantation into subcutaneous tissue and/or muscle tissue that is less invasive and can be easily surgically removed even if the transplanted cells become tumorous and require removal. Furthermore, the therapeutic agent of the present invention makes it possible to realize clinical applications of pituitary hormone-producing cells. Furthermore, the method of the present invention makes it possible to create model animals having human pituitary tissue that maintains the ability to secrete human hormones for a long period of time, which is useful for evaluating the efficacy and safety of compounds.
- FIG. 1 shows immunofluorescence micrographs of cultured pituitary organoids (PO) 100 days after the start of differentiation induction.
- A Adrenocorticotropic hormone (ACTH),
- B E-cadherin (E-cad),
- C LIM homeobox protein 3 (LHX3),
- D E
- Figure 2 Inguinal subcutaneous white adipose tissue (ISWAT) implantation of hESC-derived PO.
- ISWAT Inguinal subcutaneous white adipose tissue
- FIG. 1 A Schematic diagram, mouse processing protocol (hypophysectomy, confirmation of hypopituitarism, PO implantation, and corticotropin-releasing hormone (CRH) challenge).
- B Shaved left groin, mouse in supine position restrained under inhalation anesthesia.
- C Membranous adipose tissue vessels (arrowheads) overlying the femoral vein and artery (arrows) as viewed through a 4 mm vertical skin incision.
- D Pocket created in ISWAT.
- E Placement of PO into pocket via syringe fitted with wide-bore tip.
- F Placed PO.
- G Nylon suture closure of adipose tissue over PO. Scale interval, 1 mm.
- a gelatin hydrogel sustained release device (Gel) containing basic fibroblast growth factor with heparin was placed under the skin. After one week, the device was replaced with a PO. The placement site was vascularized and prone to bleeding.
- C Subcutaneous device-less (DL) replacement. The PO was placed in the cavity where the nylon catheter was embedded.
- D, E Inguinal subcutaneous white adipose tissue (ISWAT) implantation method.
- F The PO was placed untreated (NT) in a poorly vascularized area of the back subcutaneous layer of the mouse.
- FIG. 1 Comparison of basal and corticotropin-releasing hormone (CRH)-stimulated plasma adrenocorticotropic hormone (ACTH) levels for each method.
- Figure 4 relates to the evaluation of transplanted mice. All data are presented as mean ⁇ SEM. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
- B Running wheel activity test.
- FIG. 5 Macroscopic and histological findings after ISWAT transplantation.
- A Macroscopic appearance, transplantation site, 4 weeks after transplantation.
- Transplanted PO (arrow), blood vessel associated with the graft (arrowhead).
- B Macroscopic appearance, 21 weeks after transplantation.
- Transplanted PO (arrow).
- C I) Skin and underlying tissue containing graft. Multiple POs are included. Hematoxylin/eosin (HE).
- D-H, J-N Immunofluorescence micrographs. Counterstained with 4,6-diamidine-2-phenylindole dihydrochloride (DAPI).
- D, J Pituitary progenitor cell markers adrenocorticotropic hormone (ACTH) and LIM homeobox protein 3 (LHX3).
- E, K Pituitary progenitor cell marker adrenocorticotropic hormone (ACTH).
- F, L Human nuclei (hunuclei) and E-cadherin (E-cad), an oral ectoderm marker.
- G, M Human nuclei (hunuclei).
- H Smooth muscle actin
- SMA Smooth muscle actin
- Scale bars are 100 ⁇ m.
- Figure 6 shows histological findings of AR mice.
- A Hematoxylin/eosin (HE) staining. Black boxes B-F. Grey boxes G-K.
- B-K Immunofluorescence micrographs.
- B, G Adrenocorticotropic hormone (ACTH) and LIM homeobox protein 3 (LHX3). Staining in the dermis is a nonspecific reaction.
- C, H Adrenocorticotropic hormone (ACTH).
- Inguinal (inguinal subcutaneous white adipose tissue transplantation), Axilla (axillary white adipose tissue transplantation), Muscle (intramuscular transplantation).
- Figure 9 relates to the evaluation of transplanted mice. All data are shown as mean ⁇ SEM.
- FIG. 10 Plasma ACTH levels, basal and corticotropin releasing hormone (CRH) stimulated, in mice subjected to pituitary hormone-producing cell transplantation up to 3 months after transplantation.
- ISWAT inuinal white adipose tissue transplantation
- n 2.
- Figure 11 relates to the evaluation of transplanted mice. All data are shown as mean ⁇ SEM.
- AR group back avascular field transplantation group
- bFGF group bFGF + sodium hyaluronate mixture was locally injected subcutaneously into the back
- Duragen group bFGF + sodium hyaluronate mixture was soaked in a Duragen sheet and left subcutaneously on the back for one week, then removed
- Surgicel group bFGF + sodium hyaluronate mixture was soaked in a Surgicel sheet and left subcutaneously on the back for one week).
- the present invention relates to a therapeutic agent for diseases due to disorders of the pituitary gland, comprising pituitary hormone-producing cells, characterized in that the pituitary hormone-producing cells are used to be transplanted into subcutaneous tissue, preferably subcutaneous adipose tissue, more preferably subcutaneous white adipose tissue and/or muscle tissue.
- pituitary hormone-producing cells refers to cells that produce growth hormone (GH), prolactin (PRL), adrenocorticotropic hormone (ACTH), thyroid stimulating hormone (TSH), follicle stimulating hormone (FSH), luteinizing hormone (LH), melanocyte stimulating hormone (MSH), or the like, and is a general term for cells that produce at least one of these pituitary hormones.
- GH growth hormone
- PRL prolactin
- ACTH adrenocorticotropic hormone
- TSH thyroid stimulating hormone
- FSH follicle stimulating hormone
- LH luteinizing hormone
- MSH melanocyte stimulating hormone
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention are not particularly limited in terms of their origin, production method, etc., as long as they are capable of producing the pituitary hormones described above, but are preferably pituitary hormone-producing cells induced to differentiate from pluripotent stem cells.
- the pituitary hormone-producing cells in the present specification include at least one type selected from the group consisting of adrenocorticotropic hormone (ACTH)-producing cells, growth hormone (GH)-producing cells, and prolactin (PRL)-producing cells.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention include at least one type (one, two or three types) selected from the group consisting of adrenocorticotropic hormone (ACTH)-producing cells, growth hormone (GH)-producing cells and prolactin (PRL)-producing cells.
- the cell population containing the pituitary hormone-producing cells in the present specification has the following characteristics.
- the cell population containing the pituitary hormone-producing cells preferably contains cells expressing an angiogenic factor.
- the angiogenic factor is not particularly limited as long as it is a factor involved in angiogenesis, preferably a factor involved in promoting angiogenesis, and examples thereof include angiogenin, angiopoietin-1, Del-1, fibroblast growth factors: acidic (aFGF) and basic (bFGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), follistatin, granulocyte macrophage-colony stimulating factor (GM-CSF), hepatocyte growth factor (HGF), monocyte chemotactic protein-1 (MCP-1), sphingosine-1-phosphate (S1P), platelet-derived growth factor (PDGF), transforming growth factor ⁇ (TGF- ⁇ ), transforming growth factor ⁇ (TGF- ⁇ ), tumor necrosis factor ⁇ (TNF- ⁇ ), and the like.
- aFGF acidic
- the angiogenic factor is one or more selected from the group consisting of vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor B (VEGFB), vascular endothelial growth factor C (VEGFC), and angiopoietin-2 (ANGPT2).
- VAGFA vascular endothelial growth factor A
- VEGFB vascular endothelial growth factor B
- VGFC vascular endothelial growth factor C
- ANGPT2 angiopoietin-2
- the cells have a characteristic that, by expressing an angiogenic factor, angiogenesis can occur around the pituitary hormone-producing cells contained in the therapeutic agent of the present invention after transplantation. Furthermore, angiogenesis can occur around the pituitary hormone-producing cells, allowing the pituitary hormone-producing cells to be engrafted in subcutaneous tissue or muscle tissue.
- the cell population containing the pituitary hormone-producing cells may contain cells that can differentiate into pituitary hormone-producing cells.
- examples of such cells include pituitary stem cells or pituitary progenitor cells (precursor cells of pituitary hormone-producing cells). That is, the cell population containing the pituitary hormone-producing cells of the present invention includes cell populations containing pituitary hormone-producing cells, and pituitary stem cells and/or pituitary progenitor cells. That is, the cell population may contain cells expressing a pituitary stem cell marker.
- the pituitary stem cell marker examples include at least one of Sox2, Sox9, E-Cadherin, Nestin, S100 ⁇ , GFR ⁇ 2, Prop1, CD133, ⁇ -Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE.
- the pituitary stem cell marker may be SOX2 and/or CXADR.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be contained as at least one type of pituitary hormone-producing cells, or may be a cell population containing a single type or multiple types of pituitary hormone-producing cells.
- the cell population may be a dispersed cell population, a cell aggregate containing pituitary hormone-producing cells, or a two-dimensional cell sheet.
- the cell population includes a cell aggregate containing pituitary tissue containing pituitary hormone-producing cells and tissue other than pituitary tissue (for example, hypothalamic tissue, etc.).
- the cell population includes pituitary tissue containing pituitary hormone-producing cells, and pituitary tissue recovered from a cell population containing tissue other than pituitary tissue (e.g., hypothalamic tissue, etc.).
- pituitary tissue containing pituitary hormone-producing cells
- pituitary tissue recovered from a cell population containing tissue other than pituitary tissue (e.g., hypothalamic tissue, etc.).
- cell aggregates containing pituitary hormone-producing cells are also referred to as pituitary organoids (PO).
- PO pituitary organoids
- the cell population is a cell aggregate.
- the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention is preferably a cell population containing pituitary hormone-producing cells at least 5% or more, preferably 10% or more of the total cell number.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be identified using the pituitary hormone-producing markers described below as indicators.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention contain at least ACTH-producing cells.
- the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention is preferably a cell population containing ACTH-positive cells at 3% or more, preferably 5% or more of the total cell number.
- the major axis (or equivalent circle diameter) of the cell aggregate containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be, for example, 0.1 to 7 mm, and preferably 0.4 to 5 mm.
- the method for measuring the major axis, minor axis and height of the cell aggregate is not particularly limited, and may be measured from an image captured under a microscope.
- the cell aggregate during culture in a 96-well culture plate can be imaged with a 10x lens of a Keyence inverted microscope, and the height can be measured from the image.
- the major axis refers to the longest line segment and its length among the line segments connecting the two end points of the cell aggregate in the captured image.
- the circle equivalent diameter refers to the diameter of a perfect circle, which corresponds to the area of a figure (circular or elliptical) obtained when projected onto a two-dimensional surface.
- the aggregate is a spherical aggregate, and the major axis and the circle equivalent diameter are approximate, and the difference between the major axis and the circle equivalent diameter becomes smaller as the aggregate approaches a spherical aggregate close to a perfect sphere.
- the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may contain cells of hypothalamic tissue.
- Such cells include Rx-positive, Pax6-positive and Nkx2.1-negative cells (dorsal hypothalamus) and Rx-positive, Pax6-negative and Nkx2.1-positive cells (ventral hypothalamus).
- the cell population containing pituitary hormone-producing cells included in the therapeutic agent of the present invention may contain pituitary stem cells.
- the cell population contains at least 1% or more, preferably 2% or more, of the total number of cells that are pituitary stem cells, i.e., cells that are positive for any of the above-mentioned pituitary stem cell markers.
- Examples of the pituitary stem cell marker include SOX2 and CXADR.
- the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be a cell population from which non-target cells other than pituitary hormone-producing cells generated in the differentiation induction step have been removed.
- a cell population that does not contain non-target cells or has a reduced proportion of non-target cells can be obtained by selecting pituitary hormone-producing cells using as an index the expression of a marker protein expressed on the cell surface that is expressed in pituitary hormone-producing cells or their intermediates (precursor cells of pituitary hormone-producing cells) and not expressed in non-target cells.
- the marker protein include EpCAM (see International Publication No. WO 2021/201175).
- the cell population (hereinafter sometimes referred to as "purified body") that does not contain the above-mentioned undesired cells or in which the proportion of undesired cells is reduced can be obtained, for example, by the method described below in “7. Method for producing purified body".
- the cell population (purified body) is characterized in that the proportion of EpCAM and/or E-cadherin positive cells in the cell population (cell aggregate) in which the proportion of undesired cells is reduced may be 80% or more, and preferably 85% or more, 90% or more, or 95% or more.
- the proportion of neural cells in a cell population in which the proportion of non-target cells is reduced may be 20% or less, and preferably 15% or less, 10% or less, or 5% or less.
- the proportion of ACTH-positive cells in a cell population in which the proportion of non-target cells is reduced may be 5% or more, and preferably 10% or more.
- ACTH-positive cells simultaneously express NeuroD1 and Tbx19. Therefore, the proportion of NeuroD1-positive and/or Tbx19-positive cells to the total number of cells present in the cell population may be the same as the proportion of ACTH-positive cells.
- the ACTH secretion ability in a cell population in which the proportion of non-target cells is reduced may be 200 to 1,000,000 pg/ml, 1,000 to 500,000 pg/ml, 5,000 to 300,000 pg/ml, or 10,000 to 100,000 pg/ml.
- ACTH secretion ability is affected by the number of ACTH cells and culture conditions.
- ACTH secretion ability can be expressed as the concentration of ACTH secreted into the medium from one cell population (cell aggregate) under specific culture conditions.
- the specific culture conditions refer to the following conditions: (i) a medium that can be used for culturing pituitary hormone-producing cells, preferably a serum-free medium containing KSR (e.g., IMDM, DMEM, EMEM, ⁇ MEM, GMEM, F-12 medium, DMEM/F12, IMDM/F12, epithelial cell medium (e.g., CnT-Prime epithelial culture medium), or a mixed medium thereof, is used; (ii) no substance that affects ACTH production from the outside is added (e.g., Notch signal inhibitors that increase ACTH production); (iii) the medium volume is set to 0.18 to 20 ml; and (iv) the culture is performed for 3 to 4 days. After cul
- the ratio of the number of pituitary stem cells to the total number of cells present in a cell population in which the proportion of non-target cells is reduced may be 1% or more, preferably 3% or more or 5% or more.
- pituitary stem cells simultaneously express pituitary stem cell markers such as Sox2, Sox9, E-Cadherin, Nestin, S100 ⁇ , GFR ⁇ 2, Prop1, CD133, ⁇ -Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE. Therefore, the proportion of pituitary stem cells present can be determined by measuring the proportion of cells positive for the above pituitary stem cell markers (e.g., Sox2, CXADR) to the total number of cells present in the cell population.
- the proportion of pituitary stem cell markers e.g., Sox2, CXADR
- the cell density of the cell population in which the proportion of non-target cells is reduced may be 3,000 to 20,000 cells/mm 2 , preferably 5,000 to 10,000 cells/mm 2 , and more preferably 6,000 to 9,000 cells/mm 2 .
- the cell density of a cell population in which the proportion of non-target cells has been reduced can be expressed as the number of cells per unit area on the cut surface of the cell population, for example, at a position within 200 ⁇ m from the center.
- a person skilled in the art can measure the cell density of a cell population using known methods. Specifically, a slice is prepared by cutting the cell population, and the number of cells is measured by a method such as immunostaining using DAPI or anti-E-cadherin antibody, and then divided by the area of the slice.
- the diameter of the cell population in which the proportion of non-target cells is reduced may be 200 to 3,000 ⁇ m, preferably 300 to 2,000 ⁇ m, and more preferably 400 to 1,500 ⁇ m.
- the cell population in which the proportion of non-target cells is reduced may be a spherical cell aggregate.
- spherical cell aggregate refers to a cell aggregate having a three-dimensional shape close to a sphere.
- a three-dimensional shape close to a sphere is a shape having a three-dimensional structure, and examples of such shapes include a spherical shape that is circular or elliptical when projected onto a two-dimensional surface.
- the cell aggregate is preferably a spherical cell aggregate close to a perfect sphere.
- a spherical cell aggregate close to a perfect sphere means a cell aggregate having a three-dimensional shape close to a perfect sphere.
- a three-dimensional shape close to a perfect sphere means a spherical shape in which the multiple circles obtained when projected onto a two-dimensional surface from multiple directions all have the same length from the center to the circumference of the circles, or the difference between the longest and shortest line segments is 10% or less, 5% or less, 3% or less, 2% or less, or 1% or less.
- a cell population in which the proportion of non-target cells has been reduced may have, on its surface, EpCAM-positive cells and cells positive for at least one of E-cadherin that adhere to each other to form an epithelial structure.
- surface adhesion means that adjacent cells adhere to each other at their surfaces. More specifically, surface adhesion means that the proportion of the surface area of a cell that is adhered to the surface of another cell is, for example, 1% or more, preferably 3% or more, more preferably 5% or more, and even more preferably 10% or more.
- the cell surface means the cell membrane surface. The cell surface can be observed by immunostaining for cell adhesion factors (e.g., E-cadherin, EpCAM, etc.). Point adhesion is an example of a state that is not surface adhesion. Point adhesion means that cells adhere to each other at points.
- epithelial structure refers to a structure that epithelial tissue has characteristically, such as the apical surface or basement membrane. This can be confirmed by immunostaining using markers for the apical surface and basement membrane, which will be described later.
- Epithelial tissue is polarized to form an "apical surface” and a “basement membrane.”
- basement membrane refers to a basement membrane that contains a 50-100 nm basal layer (basement membrane) produced by epithelial cells that is rich in laminin and type IV collagen, which are markers of the basement membrane.
- apical surface refers to the surface (superficial surface) that is formed on the opposite side of the “basement membrane.” Such apical surfaces can be identified by immunostaining methods well known to those skilled in the art using antibodies against apical surface markers (e.g., atypical-PKC (hereinafter sometimes abbreviated as "aPKC”), E-cadherin, and N-cadherin).
- aPKC atypical-PKC
- E-cadherin E-cadherin
- N-cadherin N-cadherin
- Epithelial tissue is tissue formed when cells tightly cover the surface of the body, lumen (such as the digestive tract), and body cavity (such as the pericardial cavity). The cells that form epithelial tissue are called epithelial cells. Epithelial cells have apical-basal polarity. Epithelial cells form strong bonds with each other through adherens junctions and/or tight junctions, and can form cell layers. Epithelial tissue is a tissue formed when one to a dozen or so cell layers are stacked on top of each other.
- the cell population in which the proportion of non-target cells is reduced may have a plurality of island-like structures formed by surface adhesion of EpCAM-positive cells and/or E-cadherin-positive cells to each other.
- the cell population may have a sponge-like structure formed by a collection of a plurality of island-like structures.
- islet-like structure refers to a structure formed by surface adhesion of about 2 to 20 (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 15, 20) EpCAM-positive cells and/or E-cadherin-positive cells to each other.
- the number of cells contained in each island-like structure may differ for each island-like structure.
- Surface adhesion means that adjacent cells are adhered to each other by their surfaces. More specifically, surface adhesion means that the rate at which cells are adhered to each other by their surfaces is, for example, 1% or more, preferably 3% or more, more preferably 5% or more, and even more preferably 10% or more.
- the major axis of the cells present in the island-like structure may be, for example, 2 to 50 ⁇ m, preferably 4 to 30 ⁇ m, and the minor axis may be, for example, 0.5 to 20 ⁇ m, preferably 1 to 10 ⁇ m.
- the island structure may contain pituitary hormone-producing cells.
- Specific examples include ACTH-positive cells, PRL-positive cells, FSH-positive cells, LH-positive cells, GH-positive cells, TSH-positive cells, and MSH-positive cells.
- a "sponge-like structure” is a structure formed by a group of multiple island structures with voids between the island structures. It is not necessary that there are voids between all of the island structures. The presence of voids allows nutrients and oxygen from the culture medium to reach the inside of the cell mass, so the proportion of necrotic cells is low even in the center of the cell mass.
- Island-like structures and “sponge-like structures” are typically not observed before sorting with EpCAM, i.e., in cell aggregates obtained by culturing pluripotent stem cells in suspension.
- Island-like structures and “sponge-like structures” are typically structures that are first identified upon re-aggregation after sorting with EpCAM.
- the GH secretion ability of the cell population in which the proportion of non-target cells is reduced may be 0.05 to 100 ng/ml, and in another embodiment, may be 0.1 to 50 ng/ml, 0.3 to 30 ng/ml, or 0.5 to 10 ng/ml.
- GH secretion ability is affected by the number of GH cells and culture conditions. GH secretion ability can be expressed as the concentration of GH secreted into the medium from one cell group (cell aggregate) under specific culture conditions.
- the specific culture conditions refer to the following conditions: (i) a medium that can be used for culturing pituitary hormone-producing cells, preferably a serum-free medium containing KSR (e.g., DMEM, MEM, GMEM, epithelial cell medium (e.g., CnT-Prime epithelial culture medium) is used, (ii) no substances that affect GH production from the outside are added (e.g., adrenal cortical hormones that increase GH production (e.g., dexamethasone)), (iii) the medium volume is set to 0.18 to 20 ml, and (iv) the culture is performed for 3 to 4 days. The medium after culture under the above conditions is collected, and the GH concentration in the medium is measured.
- KSR
- a cell population with a reduced proportion of non-target cells may be cultured in the presence of a specific substance depending on the intended use of the cell population to increase the amount of secretion of a specific pituitary hormone.
- the GH secretion ability is improved by changing the condition (ii) to a condition of culturing in a medium containing adrenal cortical hormones.
- adrenal cortical hormones include artificially synthesized glucocorticoids such as dexamethasone, betamethasone, prednisolone, methylprednisolone, and triamcinolone; and natural glucocorticoids such as hydrocortisone, cortisone acetate, and fludrocortisone acetate.
- the concentration of the adrenal cortical hormones can appropriately set the concentration of the adrenal cortical hormones, but in one embodiment, the concentration of dexamethasone may be 4 ng/ml to 40 ⁇ g/ml, preferably 40 ng/ml to 4 ⁇ g/ml.
- the GH secretion ability of the cell population containing pituitary hormone-producing cells after culturing in the presence of adrenal cortical hormones may be 0.1 to 10,000 ng/ml, 0.5 to 5,000 ng/ml, 5 to 1,000 ng/ml, or 20 to 500 ng/ml.
- the ACTH secretion ability is 10 to 500,000 pg/ml, preferably 100 to 100,000 pg/ml, more preferably 500 to 30,000 pg/ml, and even more preferably 1,000 to 10,000 pg/ml.
- the cell population containing pituitary hormone-producing cells after culturing in the presence of corticosteroids contains more pituitary hormone-producing cells than before the culturing.
- the ratio of the number of ACTH-positive (producing) cells to the total number of cells present in the cell population may decrease.
- the proportion of ACTH-positive cells present is 2% or more, preferably 3% or more, and more preferably 5% or more.
- the cell population (purified product) in which the proportion of undesired cells is reduced may, in one embodiment, have at least one of the following characteristics (i) to (vi).
- the proportion of cells positive for at least one of EpCAM and E-cadherin in the cell population is 80% or more.
- the proportion of neural cells in the cell population is 20% or less.
- the proportion of ACTH-positive cells in the cell population is 5% or more.
- the cell density of the cell population is 3,000 to 20,000 cells/ mm2 .
- the major axis of the cell population is 200 to 3,000 ⁇ m.
- On the surface of the cell population cells positive for at least one of EpCAM and E-cadherin adhere to each other to form an epithelial structure.
- the cell population forms a sponge-like structure.
- the cell population (purified product) having at least one of the characteristics (i) to (vi) may also have at least one of the characteristics (viii) to (x) below.
- the proportion of pituitary stem cells in the cell population is 3% or more
- the ACTH secretion ability of the cell population is 200 to 1,000,000 pg/ml.
- the cell population has a GH secretion ability of 0.05 to 100 ng/ml.
- the cell population (purified body) in which the proportion of non-target cells is reduced may be characterized in that the proportion of EpCAM-positive cells and E-cadherin-positive cells is 80% or more, the proportion of nervous system cells is 20% or less, and the ACTH secretion ability of the cell population is 200 to 1,000,000 pg/ml.
- the therapeutic agent of the present invention is used to be implanted into subcutaneous tissue and/or muscle tissue.
- subcutaneous tissue refers to the tissue located at the bottom of the three-layer structure of the skin (i.e., on the visceral side) and supporting the epidermis and dermis.
- Subcutaneous tissue can also be said to be tissue sandwiched between the dermis and muscle (including fascia).
- muscle tissue refers to tissue located below the subcutaneous tissue and including muscle and fascia. Muscle is broadly divided into skeletal muscle, which is striated muscle, cardiac muscle, and smooth muscle.
- the site "subcutaneous tissue and/or muscle tissue” can be used interchangeably with "site located deeper than the dermis” or "site including from the subcutaneous tissue to the muscle.”
- the therapeutic agent of the present invention is not particularly limited to the type of subcutaneous tissue or muscle tissue or the site of transplantation, as long as it is used to be transplanted into subcutaneous tissue and/or muscle tissue.
- the therapeutic agent of the present invention is used to be transplanted into subcutaneous tissue, it is preferably used in subcutaneous adipose tissue, specifically brown adipose tissue or white adipose tissue, more preferably white adipose tissue.
- the subcutaneous adipose tissue is not particularly limited as long as it is a subcutaneous adipose tissue at a site where cells can be surgically transplanted, but is preferably subcutaneous adipose tissue present at at least one site selected from the groin, armpit, back, and abdomen.
- the therapeutic agent of the present invention is used to be transplanted into white adipose tissue in the groin. In another embodiment, the therapeutic agent of the present invention is used to be transplanted into white adipose tissue in the armpit.
- angiogenesis may occur around the pituitary hormone-producing cells after transplantation of the therapeutic agent of the present invention.
- angiogenesis around the pituitary hormone-producing cells may promote the survival of the pituitary hormone-producing cells in the subcutaneous tissue. This survival allows the pituitary hormones to be secreted into the blood vessels.
- the therapeutic agent of the present invention is used such that the pituitary hormone-producing cells are transplanted near the blood vessels in the subcutaneous tissue.
- the therapeutic agent of the present invention when used to be transplanted into muscle tissue, it may be used to be transplanted into either striated muscle or smooth muscle, and is not particularly limited, but one embodiment is striated muscle, particularly skeletal muscle.
- Skeletal muscle is typically composed of muscles (superficial muscles) located in the superficial layer, which have relatively long tendon tissue at both ends and a relatively small attachment area to bone, and muscles (deep muscles) located in the deep layer, which directly attach to bone over a wide area and have little tendon tissue.
- the therapeutic agent of the present invention when used to be transplanted into skeletal muscle, it may be transplanted into either superficial muscle or deep muscle, or between deep muscle and superficial muscle.
- the transplant site examples include muscle tissue present in areas such as the back (including the lower back), abdomen, thighs, buttocks, upper arms, and chest. Since muscle tissue has a vascular network formed in the same manner as the subcutaneous tissue described above, from the viewpoint of the transplantation site, angiogenesis may occur around the pituitary hormone-producing cells after transplantation of the therapeutic agent of the present invention. Furthermore, angiogenesis around the pituitary hormone-producing cells may promote the engraftment of the pituitary hormone-producing cells into the muscle tissue. This engraftment may cause the pituitary hormones to be secreted into the blood vessels. From the viewpoint of engraftment, in one embodiment, the therapeutic agent of the present invention is used such that the pituitary hormone-producing cells are transplanted near the blood vessels in the muscle tissue.
- the pituitary hormone-producing cells are used to be transplanted into an artificially created space (pocket) in the tissue.
- the size of the space may be changed appropriately depending on the size and number of cell aggregates (cell populations) contained in the therapeutic agent of the present invention.
- the size of the artificially created space is typically 0.001 to 50 cm 3. More specifically, for example, when transplanting five pituitary organoids (e.g., 2 to 5 mm diameter), the size of the space may be 1 to 15 cm 3.
- the artificially created space is preferably a space that can accommodate only the desired number of purified bodies, and is also preferably directly under a (small) blood vessel in the subcutaneous tissue or muscle tissue.
- angiogenesis promoter e.g., bFGF, etc.
- the term "diseases caused by pituitary gland disorders” may refer to animal diseases caused by pituitary gland disorders, or non-human animal diseases caused by pituitary gland disorders.
- Specific examples of “diseases caused by pituitary gland disorders” include panhypopituitarism, pituitary dwarfism, adult growth hormone deficiency, adrenal insufficiency, partial hypopituitarism, isolated anterior pituitary hormone deficiency, pituitary hypogonadism, autoimmune hypothalamic hypophysitis, and pituitary function and hormone secretion deficiency after surgery for pituitary tumors and other tumors.
- the animal is preferably a mammal, and includes rodents, ungulates, felines, lagomorphs, primates, etc.
- Rodents include mice, rats, hamsters, guinea pigs, etc.
- Ungulates include pigs, cows, goats, horses, sheep, etc.
- Felidae include dogs, cats, etc.
- Lagomorphs include rabbits, etc.
- Primates refer to mammals belonging to the order Primates, and include prosimians such as lemurs, lorises, and tree shrews, and anthropoids such as monkeys, apes, and humans.
- the pituitary hormone-producing cells, cell aggregates containing pituitary hormone-producing cells, or two-dimensional cell sheets contained in the therapeutic agent of the present invention are not particularly limited in terms of their origin or production method, etc., as long as they are capable of producing the hormones described above.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be produced by the method described in PCT/JP2022/36019, as described below.
- stem cells refer to undifferentiated cells that have differentiation and proliferation capabilities (particularly self-renewal capabilities). Stem cells include pluripotent stem cells, multipotent stem cells, unipotent stem cells, etc., depending on their differentiation capabilities.
- pluripotent stem cells refer to stem cells that can be cultured in vitro and have the ability to differentiate into all cells that constitute a living body (pluripotency). All cells are cells derived from the three germ layers of ectoderm, mesoderm, and endoderm.
- Multipotent stem cells refer to stem cells that have the ability to differentiate into multiple types of tissues and cells, although not all types.
- Unipotent stem cells refer to stem cells that have the ability to differentiate into specific tissues and cells.
- Pluripotent stem cells can be derived from fertilized eggs, cloned embryos, germline stem cells, tissue stem cells, somatic cells, etc.
- pluripotent stem cells include embryonic stem cells (ES cells), EG cells (embryonic germ cells), and induced pluripotent stem cells (iPS cells).
- Muse cells multi-lineage differentiating stress ending cells obtained from mesenchymal stem cells (MSCs), and GS cells produced from germ cells (e.g. testes) are also included in pluripotent stem cells.
- Human embryonic stem cells are established from human embryos within 14 days of fertilization.
- Embryonic stem cells were first established in 1981, and have been used to create knockout mice since 1989. Human embryonic stem cells were established in 1998, and are also being used in regenerative medicine. ES cells can be produced by culturing the inner cell population on feeder cells or in a medium containing leukemia inhibitory factor (LIF). Methods for producing ES cells are described in, for example, International Publication No. 96/22362, International Publication No. 02/101057, U.S. Patent No. 5,843,780, U.S. Patent No. 6,200,806, and U.S. Patent No. 6,280,718, etc. Embryonic stem cells are available from designated institutions, and can also be purchased commercially.
- LIF leukemia inhibitory factor
- ntES cells nuclear transfer ES cells
- EG cells can be produced by culturing primordial germ cells in a medium containing mouse stem cell factor (mSCF), LIF, and basic fibroblast growth factor (bFGF) (Cell, 70:841-847, 1992).
- mSCF mouse stem cell factor
- LIF basic fibroblast growth factor
- bFGF basic fibroblast growth factor
- “Induced pluripotent stem cells” are cells in which pluripotency has been induced by reprogramming somatic cells using known methods.
- Specific examples of induced pluripotent stem cells include cells in which pluripotency has been induced by reprogramming somatic cells differentiated into fibroblasts, peripheral blood mononuclear cells, etc., through the expression of multiple genes selected from a group of reprogramming genes including Oct3/4, Sox2, Klf4, Myc (c-Myc, N-Myc, L-Myc), Glis1, Nanog, Sall4, lin28, Esrrb, etc.
- Oct3/4, Sox2, Klf4, Myc (c-Myc, N-Myc, L-Myc), Glis1, Nanog, Sall4, lin28, Esrrb, etc. Yamanaka et al.
- artificial pluripotent stem cells were established from human fibroblasts, and like embryonic stem cells, they have pluripotency and the ability to self-replicate (Cell, 2007, 131(5) pp.861-872; Science, 2007, 318(5858) pp.1917-1920; Nat. Biotechnol., 2008, 26(1) pp.101-106).
- artificial pluripotent stem cells can also be induced from somatic cells by adding compounds (Science, 2013, 341, pp.651-654).
- Somatic cells used in producing induced pluripotent stem cells are not particularly limited, but include tissue-derived fibroblasts, blood cells (e.g., peripheral blood mononuclear cells, T cells, etc.), liver cells, pancreatic cells, intestinal epithelial cells, smooth muscle cells, etc.
- tissue-derived fibroblasts e.g., peripheral blood mononuclear cells, T cells, etc.
- blood cells e.g., peripheral blood mononuclear cells, T cells, etc.
- liver cells e.g., pancreatic cells, intestinal epithelial cells, smooth muscle cells, etc.
- the means for expressing the genes is not particularly limited.
- means for expressing genes include infection methods using viral vectors (e.g., retroviral vectors, lentiviral vectors, Sendai virus vectors, adenoviral vectors, and adeno-associated virus vectors), gene transfer methods using plasmid vectors (e.g., plasmid vectors, episomal vectors) (e.g., calcium phosphate method, lipofection method, retronectin method, and electroporation method), gene transfer methods using RNA vectors (e.g., calcium phosphate method, lipofection method, and electroporation method), and direct protein injection methods.
- viral vectors e.g., retroviral vectors, lentiviral vectors, Sendai virus vectors, adenoviral vectors, and adeno-associated virus vectors
- gene transfer methods using plasmid vectors e.g., plasmid vectors, episomal vectors
- RNA vectors e
- induced pluripotent stem cell lines for example, human induced pluripotent cell lines such as 201B7 cells, 201B7-Ff cells, 253G1 cells, 253G4 cells, 1201C1 cells, 1205D1 cells, 1210B2 cells, and 1231A3 cells established at Kyoto University are available from Kyoto University and iPS Academia Japan Inc.
- established induced pluripotent stem cell lines for example, Ff-I01 cells, Ff-I14 cells, and QHJI01s04 cells established at Kyoto University are available from Kyoto University.
- Pluripotent stem cells may be genetically modified.
- Genetically modified pluripotent stem cells can be produced, for example, by using homologous recombination techniques.
- Genes on chromosomes that can be modified include, for example, cell marker genes, histocompatibility antigen genes, and disease-related genes based on disorders of nervous system cells. Modification of target genes on chromosomes can be carried out using methods described in Manipulating the Mouse Embryo, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1994), Gene Targeting, A Practical Approach, IRL Press at Oxford University Press (1993), Biomanual Series 8, Gene Targeting, Creation of Mutant Mice Using ES Cells, Yodosha (1995), etc.
- the genomic gene of the target gene to be modified (e.g., a cell marker gene, a gene for a histocompatibility antigen, or a disease-related gene) is isolated, and a target vector is prepared using the isolated genomic gene for homologous recombination of the target gene.
- the target vector thus prepared is introduced into stem cells, and cells in which homologous recombination has occurred between the target gene and the target vector are selected, thereby producing stem cells in which genes on chromosomes have been modified.
- Methods for isolating the genomic gene of the target gene include known methods described in Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989) and Current Protocols in Molecular Biology, John Wiley & Sons (1987-1997).
- a genomic DNA library screening system manufactured by Genome Systems
- Universal GenomeWalker Kits manufactured by Clontech
- the preparation of a target vector for homologous recombination of a target gene and efficient selection of a homologous recombinant can be performed according to the methods described in Gene Targeting, A Practical Approach, IRL Press at Oxford University Press (1993), Biomanual Series 8, Gene Targeting, Generation of Mutant Mice Using ES Cells, Yodosha (1995), etc. Either a replacement type or an insertion type target vector can be used. Selection methods that can be used include positive selection, promoter selection, negative selection, and poly A selection. Methods for selecting the desired homologous recombinant from the selected cell lines include Southern hybridization and PCR for genomic DNA.
- Pluripotent stem cells that have undergone genome editing can also be used as pluripotent stem cells.
- Gene editing is a technique for intentionally modifying target genes or genomic regions using principles such as site-specific cleavage of genomic DNA strands using nucleases or chemical conversion of bases. Examples of site-specific nucleases include zinc finger nucleases (ZFNs), TALENs, and CRISPR/Cas9. Using genome editing technology, it is possible to create knockout cell lines in which specific genes have been deleted, knock-in cell lines in which a different sequence has been artificially inserted into a specific gene locus, etc.
- the pluripotent stem cells used in the present invention are mammalian pluripotent stem cells, and when producing pituitary hormone-producing cells for use in the therapeutic agent of the present invention, it is preferable that the pluripotent stem cells are the same mammalian pluripotent stem cells as the subject to be transplanted.
- human pluripotent stem cells are used.
- the pluripotent stem cells used in the present invention are human pluripotent stem cells.
- Cell adhesion includes adhesion between cells (cell-cell adhesion) and adhesion between cells and extracellular matrix (substrate) (cell-substrate adhesion).
- Cell adhesion also includes adhesion of cells to culture equipment and the like that occurs in an in vitro artificial culture environment.
- the bond formed in cell-cell adhesion is a cell-cell junction
- the bond formed in cell-substrate adhesion is a cell-substrate junction.
- Types of cell adhesion include, for example, anchoring junction, communicating junction, and occluding junction.
- Tight junctions are relatively strong cell-cell junctions that can occur in epithelial cells. The presence of tight junctions between cells can be detected by techniques such as immunohistochemistry using antibodies against components of tight junctions (anti-claudin antibodies, anti-ZO-1 antibodies, etc.).
- suspension culture refers to culturing cells while maintaining a state in which they exist suspended in the culture solution.
- suspension culture is carried out under conditions that do not allow cells to adhere to the cultureware and feeder cells, etc. on the cultureware (hereinafter referred to as "cultureware, etc.”), and is distinguished from culture carried out under conditions that allow cells to adhere to the cultureware, etc. (adherent culture).
- suspension culture refers to culture under conditions that do not allow strong cell-substrate bonds to be formed between the cells and the cultureware, etc. Those skilled in the art can easily distinguish whether the cultured cells are in a suspension culture state or an adherent culture state, for example, by rocking the cultureware during microscopic observation.
- “Adherent culture” refers to culturing cells while maintaining a state in which the cells are attached to culture equipment, etc.
- cells adhering to culture equipment, etc. refers to, for example, the formation of strong cell-substrate bonds, which is a type of cell adhesion, between the cells and the culture equipment, etc.
- Plant attachment between cells refers to cells adhering to each other through a plane. More specifically, “plane attachment between cells” refers to the proportion of the surface area of a cell that is attached to the surface of another cell, for example, 1% or more, preferably 3% or more, and more preferably 5% or more.
- the cell surface can be observed by staining with a membrane-staining reagent (e.g., DiI) or immunostaining with cell adhesion factors (e.g., E-cadherin, N-cadherin, etc.).
- a membrane-staining reagent e.g., DiI
- cell adhesion factors e.g., E-cadherin, N-cadherin, etc.
- the cultureware used for adhesion culture is not particularly limited as long as it is capable of adhesion culture, and a person skilled in the art can appropriately determine the cultureware.
- Examples of such cultureware include flasks, flasks for tissue culture, dishes, dishes for tissue culture, multi-dishes, microplates, microwell plates, micropores, multi-plates, multi-well plates, chamber slides, petri dishes, tubes, trays, culture bags, and biofunctional chips such as organ-on-chips.
- Examples of cell-adhesive cultureware that can be used include cultureware whose surface has been artificially treated for the purpose of improving adhesion to cells.
- Examples of artificial treatments include coating treatments with extracellular matrices, polymers, etc., and surface treatments such as gas plasma treatment and positive charge treatment.
- Examples of extracellular matrices to which cells are attached include basement membrane preparations, laminin, entactin, collagen, gelatin, etc.
- Examples of polymers include polylysine, polyornithine, etc.
- the culture surface of the cultureware may be flat or uneven.
- Laminin is a heterotrimeric molecule consisting of ⁇ , ⁇ , and ⁇ chains, and is an extracellular matrix protein with isoforms that differ in the composition of the subunit chains. Specifically, laminin has approximately 15 isoforms, which are heterotrimeric combinations of five types of ⁇ chains, four types of ⁇ chains, and three types of ⁇ chains. The names of laminins are determined by combining the numbers of the ⁇ chains ( ⁇ 1- ⁇ 5), ⁇ chains ( ⁇ 1- ⁇ 4), and ⁇ chains ( ⁇ 1- ⁇ 3). For example, a laminin made up of a combination of ⁇ 5, ⁇ 1, and ⁇ 1 chains is called laminin 511.
- the cultureware used for suspension culture is not particularly limited as long as it is capable of suspension culture, and a person skilled in the art can appropriately determine the cultureware.
- Examples of such cultureware include flasks, flasks for tissue culture, dishes, Petri dishes, dishes for tissue culture, multi-dishes, microplates, microwell plates, micropores, multi-plates, multi-well plates, chamber slides, petri dishes, tubes, trays, culture bags, spinner flasks, and roller bottles.
- These cultureware are preferably non-adhesive to cells in order to enable suspension culture.
- non-adhesive cultureware those whose surfaces have not been artificially treated to improve adhesion to cells can be used.
- non-adhesive cultureware those whose surfaces have been artificially treated to reduce adhesion to cells can also be used.
- the culture surface of the cultureware may be flat, U- or V-bottom, or may be uneven.
- treatments that reduce adhesion to cells include superhydrophilic treatments using coatings such as 2-methacryloyloxyethyl phosphorylcholine (MPC) polymer, Poly(2-hydroxyethyl methacrylate) (Poly-HEMA), and polyethylene glycol (PEG), as well as low protein adsorption treatments.
- MPC 2-methacryloyloxyethyl phosphorylcholine
- Poly-HEMA Poly(2-hydroxyethyl methacrylate)
- PEG polyethylene glycol
- Shaking culture is a culture method in which the culture medium is stirred by shaking the culture equipment, promoting the supply of oxygen to the culture medium and the exchange of materials with the surrounding cells. Agitation culture, flow path culture, etc. can also be performed.
- the cell aggregates can be embedded in a gel or encapsulated in a substance-permeable capsule before suspension culture (Nature, 2013, 501.7467:373).
- the encapsulated cell aggregates may be cultured with shaking.
- the gel or capsule used for embedding may be made of either a biological or synthetic polymer.
- gels or capsules used for such purposes examples include Matrigel (manufactured by Corning), PuraMatrix (manufactured by 3D Matrix), VitroGel 3D (manufactured by TheWell Bioscience), collagen gel (manufactured by Nitta Gelatin), alginate gel (manufactured by PG Research), and Cell-in-a-Box (manufactured by Austrianova).
- the medium used for cell culture can be prepared as a basal medium using a medium commonly used for culturing animal cells.
- basal media include Basal Medium Eagle (BME), BGJb medium, CMRL 1066 medium, Glasgow Minimum Essential Medium (Glasgow MEM), Improved MEM Zinc Option, Iscove's Modified Dulbecco's Medium (IMDM), Medium 199, Eagle Minimu
- Examples of such medium include Eagle MEM, Alpha Modified Eagle Minimum Essential Medium ( ⁇ MEM), Dulbecco's Modified Eagle Medium (DMEM), F-12 medium, DMEM/F12, IMDM/F12, Ham's medium, RPMI 1640, Fischer's medium, and mixtures of these.
- a medium for pluripotent stem cell culture based on the above-mentioned basal medium a medium for preferably known embryonic stem cells or induced pluripotent stem cells, a medium for culturing pluripotent stem cells under feeder-free conditions (feeder-free medium), etc.
- feeder-free medium a medium for culturing pluripotent stem cells under feeder-free conditions
- Many synthetic media have been developed and are commercially available as feeder-free media, for example, Essential 8 medium.
- Essential 8 medium contains, in DMEM/F12 medium, the following additives: L-ascorbic acid-2-phosphate magnesium (64 mg/l), sodium selenium (14 ⁇ g/l), insulin (19.4 mg/l), NaHCO 3 (543 mg/l), transferrin (10.7 mg/l), bFGF (100 ng/mL), and a TGF ⁇ family signaling pathway active substance (TGF ⁇ 1 (2 ng/mL) or Nodal (100 ng/mL)) (Nature Methods, 8, 424-429 (2011)).
- feeder-free media include, for example, Essential 8 (manufactured by Thermo Fisher Scientific), S-medium (manufactured by DS Pharma Biomedical), StemPro (manufactured by Thermo Fisher Scientific), hESF9, mTeSR1 (manufactured by STEMCELL Technologies), mTeSR2 (manufactured by STEMCELL Technologies), TeSR-E8 (manufactured by STEMCELL Technologies), mTeSR Plus (manufactured by STEMCELL Technologies), StemFit (manufactured by Ajinomoto Co., Inc.), and ReproMed iPSC.
- Essential 8 manufactured by Thermo Fisher Scientific
- S-medium manufactured by DS Pharma Biomedical
- StemPro manufactured by Thermo Fisher Scientific
- hESF9 hESF9
- mTeSR1 manufactured by STEMCELL Technologies
- mTeSR2 manufactured by STEMCELL Technologies
- TeSR-E8
- Examples of such medium include Cellartis DEF-CS Xeno-Free Culture Medium (manufactured by ReproCELL), NutriStem XF (manufactured by Biological Industries), NutriStem V9 (manufactured by Biological Industries), Cellartis DEF-CS Xeno-Free Culture Medium (manufactured by Takara Bio Inc.), Stem-Partner SF (manufactured by Kyokuto Pharmaceutical Co., Ltd.), PluriSTEM Human ES / iPS Cell Medium (manufactured by Merck), and StemSure hPSC Medium ⁇ (manufactured by Fujifilm Wako Pure Chemical Industries).
- Cellartis DEF-CS Xeno-Free Culture Medium manufactured by ReproCELL
- NutriStem XF manufactured by Biological Industries
- NutriStem V9 manufactured by Biological Industries
- Cellartis DEF-CS Xeno-Free Culture Medium manufactured by Takara Bio Inc.
- Stem-Partner SF
- the serum-free medium may contain a serum substitute.
- serum substitutes include those that contain albumin, transferrin, fatty acids, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, or equivalents thereof, as appropriate.
- serum substitutes can be prepared, for example, by the method described in WO 98/30679. Commercially available products may be used as serum substitutes.
- KSR Knockout Serum Replacement
- Glutamax manufactured by Thermo Fisher Scientific
- B27 Supplement manufactured by Thermo Fisher Scientific
- N2 Supplement manufactured by Thermo Fisher Scientific
- the serum-free medium used in suspension culture and adherent culture may contain fatty acids or lipids, amino acids (e.g., non-essential amino acids), vitamins, growth factors, cytokines, antioxidants, 2-mercaptoethanol, pyruvic acid, buffers, inorganic salts, etc., as appropriate.
- a serum-free medium containing an appropriate amount e.g., about 0.5% to about 30%, preferably about 1% to about 20%
- an appropriate amount e.g., about 0.5% to about 30%, preferably about 1% to about 20%
- KSR commercially available KSR
- An example of a KSR equivalent is the medium disclosed in JP2001-508302A.
- “Serum medium” refers to a medium containing unconditioned or unpurified serum.
- the medium may contain fatty acids or lipids, amino acids (e.g., non-essential amino acids), vitamins, growth factors, cytokines, antioxidants, 2-mercaptoethanol, 1-monothioglycerol, pyruvic acid, buffers, inorganic salts, etc.
- the culture in the present invention is preferably carried out under xeno-free conditions.
- Xeno-free refers to conditions in which components derived from organisms other than the organism of the cells to be cultured are excluded.
- the medium used in the present invention is preferably a medium whose components are chemically defined (CDM) in order to avoid contamination with chemically undefined components.
- Basis membrane refers to a thin membrane-like structure composed of extracellular matrix.
- basement membranes are formed on the basal side of epithelial cells.
- Components of basement membranes include type IV collagen, laminin, heparan sulfate proteoglycan (perlecan), entactin/nidogen, cytokines, growth factors, etc.
- tissue staining such as PAM staining
- immunohistochemistry using antibodies against components of basement membranes (anti-laminin antibodies, anti-type IV collagen antibodies, etc.).
- base membrane preparation refers to a preparation containing basement membrane components that have the function of controlling epithelial cell-like cell morphology, differentiation, proliferation, movement, functional expression, etc., when desired cells having basement membrane formation ability are seeded and cultured thereon. In this specification, when cells are cultured for adhesion, they can be cultured in the presence of a basement membrane preparation.
- basement membrane components refers to thin membrane-like extracellular matrix molecules that exist between epithelial cell layers and interstitial cell layers, etc., in animal tissues.
- a basement membrane preparation can be prepared, for example, by removing cells having basement membrane formation ability that are adhered to a support via a basement membrane from the support using a solution capable of dissolving the lipids of the cells, an alkaline solution, or the like.
- basement membrane preparations include products commercially available as basement membrane preparations, such as Matrigel (manufactured by Corning) and Geltrex (manufactured by Thermo Fisher Scientific), and those that contain extracellular matrix molecules known as basement membrane components (e.g., laminin, type IV collagen, heparan sulfate proteoglycan, entactin, etc.).
- basement membrane preparations such as Matrigel (Corning) extracted and solubilized from tissues or cells such as Engelbreth-Holm-Swarm (EHS) mouse sarcoma can be used.
- EHS Engelbreth-Holm-Swarm
- basement membrane components for cell culture human solubilized amniotic membrane (BioResource Application Research Institute), human recombinant laminin produced in HEK293 cells (BioLamina), human recombinant laminin fragments (Nippi), human recombinant vitronectin (Thermo Fisher Scientific), etc. can also be used. From the viewpoint of avoiding contamination with components from different biological species and from the viewpoint of avoiding the risk of infection, it is preferable to use recombinant proteins with known components as basement membrane preparations.
- medium containing substance X and "in the presence of substance X” respectively mean a medium to which exogenous substance X has been added or a medium containing exogenous substance X, and in the presence of exogenous substance X.
- Exogenous substance X is distinguished from endogenous substance X, for example, in which cells or tissues present in the medium endogenously express, secrete, or produce the substance X.
- Substance X in the medium may have undergone slight changes in concentration due to decomposition of substance X or evaporation of the medium.
- the start of culture in a medium in which the concentration of substance X is Y preferably refers to the point in time when the concentration of substance X in the medium becomes uniform at Y, but if the culture vessel is sufficiently small (for example, a 96-well plate or culture in 200 ⁇ L or less of culture medium), the start of culture at concentration Y is interpreted as the point in time when the medium addition operation, half-volume medium replacement operation, or full-volume medium replacement operation described below is performed to achieve concentration Y.
- the concentration of substance X in the medium is Y
- exogenous substance X in the absence of substance X means a medium to which exogenous substance X has not been added, a medium that does not contain exogenous substance X, or a state in which exogenous substance X is not present.
- human protein X means that protein X has the amino acid sequence of protein X that is naturally expressed in the human body.
- isolated protein X means that an operation has been performed to remove the target component or factors other than the cell, and that the protein is no longer in a naturally occurring state. Therefore, "isolated protein X" does not include endogenous protein X that is produced from the cells or tissues to be cultured and is contained in the cells, tissues, and medium.
- the purity of "isolated protein X" (the percentage of protein X weight in the total protein weight) is usually 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably 99% or more, and most preferably 100%.
- a “derivative” refers to a group of compounds that are produced by replacing a part of the molecule of a specific compound with another functional group or another atom.
- a "modified form" of a protein refers to a protein that has been mutated by deletion, addition, substitution, or other such amino acid residues to the extent that the properties of the original protein can be maintained.
- the number of mutated amino acids is not particularly limited, but may be 1-4, 1-3, 1-2, or 1.
- a "modified form" of a protein may be a protein that has an amino acid sequence that shows at least 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more, or 99.5% or more identity with the original protein.
- the mutated amino acids in the modified form may be non-natural.
- time A includes time A (day A) and refers to anything after time A (day A).
- time B includes time B (day B) and refers to anything before time B (day B).
- feeder cells refers to cells other than stem cells that are allowed to coexist when the stem cells are cultured.
- feeder cells used in the culture of pluripotent stem cells to maintain their undifferentiated state include mouse fibroblasts (MEF), human fibroblasts, and SNL cells.
- the feeder cells are preferably subjected to a growth-inhibiting treatment.
- the growth-inhibiting treatment include treatment with a growth inhibitor (e.g., mitomycin C) and UV irradiation.
- the feeder cells used in the culture of pluripotent stem cells to maintain their undifferentiated state contribute to maintaining the undifferentiated state of pluripotent stem cells by secreting humoral factors (preferably factors for maintaining their undifferentiated state) and creating a scaffold (extracellular matrix) for cell adhesion.
- in the absence of feeder cells refers to culturing in the absence of feeder cells.
- examples of “in the absence of feeder cells” include conditions where no feeder cells are added, or conditions where feeder cells are substantially not included (for example, the ratio of the number of feeder cells to the total number of cells is 3% or less).
- a "cell population” refers to a group of cells composed of two or more cells.
- a cell population may be composed of one type of cell, or may be composed of multiple types of cells.
- the cells that compose the cell population may be suspended in a medium, or may be adhered to a culture vessel or the like.
- the cells that compose the cell population may be single cells, or at least a portion of the cell population may be formed by cell adhesion between the cells.
- the term “single cells” refers to, for example, cells that have almost no cell-to-cell adhesion (e.g., surface adhesion).
- being dispersed into single cells refers to a state in which cell-to-cell bonds (e.g., adherens junctions) are almost completely lost.
- dispersed cells are preferably single cells, but may also include cell clumps consisting of a small number of cells, for example, between 2 and 100, or may include cell clumps consisting of between 2 and 50 cells.
- Dispersed cells may, for example, include 70% or more single cells and 30% or less cell clumps, and preferably include 80% or more single cells and 20% or less cell clumps.
- the population of dispersed single cells may contain minute aggregates of 5 or fewer cells.
- a cell population may include cell aggregates and two-dimensional cell sheets.
- cell aggregate refers to a mass formed by the gathering of cells, in which the cells are adhered to each other. Embryoid bodies, spheres, spheroids, and organoids are also included in the cell aggregates.
- the cells are preferably adhered to each other on their surfaces.
- the cells are adhered to each other, for example, forming an adhesion junction.
- two or more cell aggregates can be further artificially adhered or aggregated to each other. Masses in which cell populations are further adhered or aggregated to each other, and assembloids are also included in the cell aggregates.
- the shape of the cell aggregates is not limited to spherical, but may be, for example, bisphere-like, bead-like, a collection of spheres, string-like or branched (shapes described in Scientific reports, 11:21421 (2021), Patent Application No. 2021-078154), etc.
- the term "two-dimensional cell sheet" refers to a monolayer or multilayer structure composed of a single or multiple cells having intercellular junctions in the two-dimensional direction.
- Uniform cell aggregates means that when multiple cell aggregates are cultured, the size of each cell aggregate is constant, and when the size of a cell aggregate is evaluated by the length of its maximum diameter, uniform cell aggregates means that the variance of the length of the maximum diameter is small. More specifically, this means that the maximum diameter of 75% or more of the multiple cell aggregates is within ⁇ 100% of the average maximum diameter of the multiple cell aggregates, preferably within ⁇ 50% of the average maximum diameter, and more preferably within ⁇ 20% of the average maximum diameter.
- tissue refers to a structure made up of a group of cells in which multiple types of cells with different morphologies and properties are arranged three-dimensionally in a specific pattern.
- Ectoderm refers to the outermost of the three germ layers formed after fertilization of an egg during the early development of an organism. As development progresses, the ectoderm divides into the neuroectoderm and the superficial ectoderm, and the neuroectoderm further divides into the neural tube and neural crest. Various organs of the body are formed from these ectoderm, and organs formed from the ectoderm are said to be derived from the ectoderm.
- the neural tube forms organs or tissues of the central nervous system, such as the brain and spinal cord. For example, some central nervous system cells, facial bones and cartilage, sensory neurons, autonomic neurons, pigment cells, mesenchymal cells, etc.
- pluripotent stem cells express genes known as ectoderm markers, such as Pax3, Otx2, and Sox1.
- Endoderm refers to the innermost of the three germ layers formed after fertilization of an egg during the early development of an organism. For example, the digestive system, urinary tract, pharynx, trachea, bronchi, and lungs are formed from the endoderm. In the process of differentiating into endoderm, pluripotent stem cells express genes known as endoderm markers, such as SOX17, HNF-3 ⁇ /FoxA2, Klf5, GATA4, GATA6, and PDX-1.
- Endoderm refers to the germ layer formed between the ectoderm and endoderm. Organs or tissues such as circulatory system, skeleton, and muscle are formed from the mesoderm.
- pluripotent stem cells express genes known as mesoderm markers, such as T/Brachury, SMA, ABCA4, Nkx2.5, and PDGFR ⁇ .
- Nevous tissue refers to tissue composed of nervous system cells such as the developing or adult cerebrum, midbrain, cerebellum, spinal cord, retina, sensory nerves, peripheral nerves, etc.
- neuroepithelial tissue refers to nervous tissue that has formed an epithelial structure having a layered structure, and the amount of neuroepithelial tissue in nervous tissue can be evaluated by bright field observation using an optical microscope or the like.
- central nervous system refers to the area where nervous tissue is accumulated and which is the center of information processing. In vertebrates, the central nervous system includes the brain and spinal cord. The central nervous system is derived from the ectoderm.
- Neural cells refers to cells of ectoderm-derived tissues other than epidermal cells.
- neural cells include neural progenitor cells, neurons (nerve cells), glial cells, neural stem cells, neuronal progenitor cells, glial progenitor cells, and other cells.
- Neural cells also include cells that make up retinal tissue (retinal cells), retinal progenitor cells, retinal layer-specific nerve cells, neural retina cells, and retinal pigment epithelial cells, which will be described later.
- Neural cells can be identified using markers such as nestin, ⁇ III tubulin (Tuj1), PSA-NCAM, and N-cadherin.
- Neurons are functional cells that form neural circuits and contribute to signal transmission, and can be identified using the expression of immature neuronal markers such as TuJ1, Dcx, and HuC/D, and/or mature neuronal markers such as Map2 and NeuN.
- immature neuronal markers such as TuJ1, Dcx, and HuC/D
- mature neuronal markers such as Map2 and NeuN.
- Neural precursor cells are a collection of precursor cells, including neural stem cells, neuronal precursor cells, and glial precursor cells, and have the ability to proliferate and produce neurons and glia. Neural precursor cells can be identified using markers such as Nestin, GLAST, Sox2, Sox1, Musashi, and Pax6. Cells that are positive for neural cell markers and proliferation markers (Ki67, pH3, MCM) can also be identified as neural precursor cells.
- ventricle refers to a cavity formed by central nervous tissue. In living organisms, it is an acellular structure that is usually filled with tissue fluid such as cerebrospinal fluid, with the apical side of the nervous tissue facing the ventricle.
- tissue fluid such as cerebrospinal fluid
- the periventricular layer surrounding the ventricle is an area where neural stem cells exist and where cell proliferation and neuronal production occur during development.
- ventricles can be detected by techniques such as immunohistochemistry using central nervous tissue markers (Bf1, Pax6, etc.) and apical surface markers (PKC-zeta, etc.).
- the term "diencephalon” refers to the neural tissue of the central nervous system adjacent to the third ventricle.
- the diencephalon includes tissues such as the epithalamus, thalamus, hypothalamus, and pituitary gland.
- hypothalamus refers to a region of the diencephalon adjacent to the pituitary gland.
- the hypothalamus is further regionalized into the dorsal hypothalamus and the ventral hypothalamus.
- the hypothalamus before regionalization can be identified using markers such as Rx, Vax1, and Six3.
- the dorsal hypothalamus can be identified using markers such as Otp, Brn2, vasopressin, and Pax6.
- the ventral hypothalamus can be identified using markers such as Nkx2.1 and SF1.
- hypothalamic neuroepithelial tissue refers to neuroepithelial tissue expressing a hypothalamic marker.
- the ventral hypothalamic neuroepithelial tissue is Rx-positive, Chx10-negative, and Nkx2.1-positive neuroepithelial tissue.
- the dorsal hypothalamic neuroepithelial tissue is Rx-positive, Chx10-negative, and Pax6-positive neuroepithelial tissue.
- the term "hypothalamus” may include "ependymal cells.” Ependymal cells are a type of epithelial cell, and refer to cells localized in the ventricular wall.
- ependymal cells tanycytes
- Elongated ependymal cells refer to cells with few cilia that are particularly localized in the third ventricle of the hypothalamus. Elongated ependymal cells can be identified using markers such as Nestin, Vimentin, Lhx2, and Sox2.
- non-neuroepithelial tissue refers to tissues having an epithelial structure other than neuroepithelial tissue.
- Epithelial tissue is formed from any of the germ layers, ectoderm, mesoderm, endoderm, and trophectoderm.
- Epithelial tissue includes epithelium, mesothelium, and endothelium.
- tissues included in non-neuroepithelial tissue include epidermis, corneal epithelium, nasal epithelium, oral epithelium, tracheal epithelium, bronchial epithelium, airway epithelium, kidney epithelium, kidney cortical epithelium, and placental epithelium.
- Epithelial tissues are usually connected by various intercellular junctions to form tissues with a single layer or stratified layer structure. The presence or absence of these epithelial tissues and the amount of them present can be confirmed by observation with an optical microscope or by immunohistochemistry using antibodies against epithelial cell markers (anti-E-Cadherin antibody, anti-N-Cadherin antibody, anti-EpCAM antibody, etc.).
- epithelial polarity refers to the bias in the distribution of spatially formed components and cell functions within epithelial cells.
- corneal epithelial cells are localized in the outermost layer of the eyeball, and express apical-specific proteins such as membrane-bound mucins (MUC-1, 4, 16) for retaining tears on the apical side, and express basal-specific proteins such as ⁇ 6 integrin and ⁇ 1 integrin for adhering to the basement membrane on the basal side.
- apical-specific proteins such as membrane-bound mucins (MUC-1, 4, 16) for retaining tears on the apical side
- basal-specific proteins such as ⁇ 6 integrin and ⁇ 1 integrin for adhering to the basement membrane on the basal side.
- epithelial cell polarity in epithelial cells and epithelial tissues in tissues derived from a living body and in cell populations produced by the production methods of the present invention can be detected by techniques such as immunohistochemistry using phalloidin, apical markers (anti-MUC-1 antibody, anti-PKC-zeta antibody, etc.), and basal markers (anti- ⁇ 6 integrin antibody, anti- ⁇ 1 integrin antibody, etc.).
- plaque refers to the primordium of an organ that is formed by the thickening of a portion of the epidermal ectoderm, primarily during the developmental process of vertebrates.
- tissues derived from the placode include the lens, nose, inner ear, trigeminal nerve, and adenohypophysis.
- Markers of the placode or its precursor tissue, the preplacode region include Six1, Six4, Dlx5, Eya2, Emx2, and Bf1.
- pituitary placode refers to a thickened structure formed in the epidermal ectoderm region during embryonic development, which expresses pituitary progenitor cell markers.
- pituitary progenitor cell markers include Lim3 (Lhx3), Pitx1/2, Islet1/2, etc.
- the pituitary placode expresses at least one, and preferably all, pituitary progenitor cell markers selected from the group consisting of Lim3, Pitx1/2, and Islet1/2.
- the pituitary placode invaginates to form Rathke's pouch, a sac-like structure in the developing stage, and as development progresses further, it forms the adenohypophysis.
- adenohypophysis refers to tissue that contains at least one type of pituitary cell of the anterior or intermediate lobe.
- Pituitary cells include pituitary hormone-producing cells that produce hormones that regulate physiological functions, and non-hormone-producing cells.
- Pituitary hormone-producing cells include cells that make up the anterior lobe, such as growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, adrenocorticotropic hormone (ACTH)-producing cells, thyroid-stimulating hormone (TSH)-producing cells, follicle-stimulating hormone (FSH)-producing cells, and luteinizing hormone (LH)-producing cells; and cells that make up the intermediate lobe, such as melanocyte-stimulating hormone (MSH)-producing cells.
- Non-hormone-producing cells include vascular endothelial cells, pericytes, follicle-stellate cells, pituitary stem cells, and pituitary progenitor cells.
- the adenohypophysis contains at least one, preferably two, more preferably three types of pituitary hormone-producing cells selected from the group consisting of growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, and adrenocorticotropic hormone (ACTH)-producing cells.
- GH growth hormone
- PRL prolactin
- ACTH adrenocorticotropic hormone
- the adenohypophysis contains at least one, preferably two or more (2, 3, 4, 5, or 6) types of pituitary hormone-producing cells selected from the group consisting of growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, adrenocorticotropic hormone (ACTH)-producing cells, thyroid stimulating hormone (TSH)-producing cells, follicle stimulating hormone (FSH)-producing cells, and luteinizing hormone (LH)-producing cells.
- GH growth hormone
- PRL prolactin
- ACTH adrenocorticotropic hormone
- TSH thyroid stimulating hormone
- LH luteinizing hormone
- adenohypophysis and pituitary hormone-producing cells are present in tissue derived from a living body and in a cell population produced by the production method of the present invention can be detected by techniques such as immunohistochemistry using pituitary hormone-producing cell markers and ELISA for the secreted hormones.
- Pituitary hormone-producing cell markers include growth hormone (GH)-producing cell markers (anti-Pit1 antibody, anti-GH antibody, etc.), prolactin (PRL)-producing cell markers (anti-Pit1 antibody, anti-PRL antibody, etc.), adrenocorticotropic hormone (ACTH)-producing cell markers (anti-T-Pit antibody, anti-NeuroD1 antibody, anti-ACTH antibody, etc.), thyroid stimulating hormone (TSH)-producing cell markers (anti-GATA2 antibody, anti-ACTH antibody, etc.), and markers of follicle stimulating hormone (FSH)-producing cells and luteinizing hormone (LH)-producing cells (anti-GATA2 antibody, anti-SF1 antibody, anti-FSH antibody, anti-LH antibody, etc.). These can be detected by immunohistochemistry using these markers, and ELISA for the secreted hormones.
- GH growth hormone
- PRL prolactin
- ACTH adrenocorticotropic hormone
- pituitary stem cells refers to undifferentiated multipotent stem cells or progenitor cells that exist in the pituitary gland and contribute to the regeneration of pituitary tissue and the supply of pituitary hormone-producing cells.
- pituitary stem cells Whether or not pituitary stem cells are present in the cell population and tissue produced by the production method of the present invention can be determined by detecting pituitary stem cell markers such as Sox2, Sox9, E-Cadherin, Nestin, S100 ⁇ , GFR ⁇ 2, Prop1, CD133, ⁇ -Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE, as well as Ki67, Ribosomal protein markers such as ri ...
- pituitary stem cell markers such as Sox2, Sox9, E-Cadherin, Nestin, S100 ⁇ , GFR ⁇ 2, Prop1, CD133, ⁇ -Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE, as well as Ki67, Ribosomal
- Detection can be achieved by a variety of techniques, including immunohistochemistry using antibodies against cell proliferation markers such as phosphorylated histone H3 and MCM, proliferative cell labeling assays using nucleic acid analogs such as BrdU, EdU, and IdU, fluorescently labeled dipeptide ( ⁇ -alanyl-lysyl-N-7-amino-4-methylcoumarin-3-acetic acid) uptake assays, and pituitary sphere formation assays.
- cell proliferation markers such as phosphorylated histone H3 and MCM
- proliferative cell labeling assays using nucleic acid analogs such as BrdU, EdU, and IdU
- fluorescently labeled dipeptide ⁇ -alanyl-lysyl-N-7-amino-4-methylcoumarin-3-acetic acid
- oral epithelium refers to the epithelial tissue and cells that form the oral cavity.
- oral epithelium include oral mucosal epithelium, salivary gland epithelium, and odontogenic epithelium.
- Oral mucosal epithelium is usually a mucosal tissue made of stratified squamous epithelium, and contains basal cells, Merkel cells, melanin-producing cells, etc. on the basement membrane in contact with connective tissue, with spinous cells, granular cells, and a stratum corneum formed on the upper layer.
- Oral mucosal epithelium can be detected as tissue that is positive for cytokeratin 7, 8, 13, 14, and 19, for example.
- stem cell niche refers to a microenvironment involved in the proliferation, differentiation, and maintenance of properties of stem cells.
- niches in the body include the hematopoietic stem cell niche, hair follicle stem cell niche, intestinal epithelial stem cell niche, muscle stem cell niche, and pituitary niche.
- tissue-specific stem cells and supporting cells that provide the niche exist, and the stem cells are maintained by cytokines, chemokines, extracellular matrix, cell adhesion factors, intercellular signaling factors, and the like provided by the supporting cells.
- pituitary niche refers to the microenvironment involved in the proliferation, differentiation, and maintenance of properties of pituitary stem cells.
- pituitary niches include the Marginal Cell Layer (MCL) niche that exists around the residual cavity (Rathke's cleft) that remains between the anterior and intermediate lobes of the pituitary gland as a vestige of the hollow part of the sac-like Rathke's pouch during development, and the parenchymal niche that is scattered throughout the anterior pituitary gland.
- MCL Marginal Cell Layer
- mesenchymal cells refer to non-epithelial cells that are derived primarily from the mesoderm and neural crest and form connective tissue. Some of these cells are multipotent somatic stem cells known as mesenchymal stem cells. Whether or not mesenchymal cells are present in the cell populations and tissues produced by the production method of the present invention can be detected by techniques such as immunohistochemistry using antibodies against mesenchymal cell markers such as nestin, vimentin, cadherin-11, laminin, and CD44.
- mesenchymal stem cells Whether or not mesenchymal stem cells are present can be detected by techniques such as immunohistochemistry using antibodies against mesenchymal stem cell markers such as CD9, CD13, CD29, CD44, CD55, CD59, CD73, CD105, CD140b, CD166, VCAM-1, STRO-1, c-Kit, Sca-1, Nucleostemin, CDCP1, BMPR2, BMPR1A, and BPMR1B.
- mesenchymal stem cell markers such as CD9, CD13, CD29, CD44, CD55, CD59, CD73, CD105, CD140b, CD166, VCAM-1, STRO-1, c-Kit, Sca-1, Nucleostemin, CDCP1, BMPR2, BMPR1A, and BPMR1B.
- a cell population comprising pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be produced by inducing differentiation using a method comprising the following steps: (A) A step of culturing pluripotent stem cells in a differentiation-inducing medium to obtain a cell population, and then culturing the cell population in suspension in a medium containing a substance that activates the bone morphogenetic protein signaling pathway and a substance that acts on the Shh signal pathway.
- the cell population is preferably a cell aggregate.
- the method includes the following steps: (A') A step of suspension-culturing cell aggregates obtained by suspension-culturing pluripotent stem cells in a medium containing an activator of bone morphogenetic protein signaling pathway and a substance acting on the Shh signal pathway.
- the bone morphogenetic protein signaling pathway activator and the Shh signal pathway acting substance, their concentrations, and the culture period in their presence can be performed according to the methods described in WO 2016/013669, WO 2019/103129, Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351), etc., or the culture conditions described in step (2) of "4. Method 1 for producing a cell population containing pituitary tissue".
- the culture period of the step (A) or (A') may be any culture condition and culture period that can obtain cell aggregates containing 1) hypothalamic neuroepithelial tissue, and 2) pituitary placode and/or Rathke's pouch. In one embodiment, the step (A) or (A') is performed until 10% or more, preferably 30% or more, more preferably 50% or more of the cell aggregates in the culture contain pituitary placode and/or Rathke's pouch.
- the culture period of the step (A) or (A') may be, for example, 6 days or more, 8 days or more, 10 days or more, and 20 days or less, 18 days or less, or 16 days or less, and may be, for example, 10 days or more and 16 days or less.
- the hypothalamic neuroepithelial tissue contained in the cell aggregate obtained in step (A) or (A') is preferably ventral hypothalamic neuroepithelial tissue.
- the concentration of the bone morphogenetic protein signaling pathway activator may be varied during the period of addition; for example, the concentration may be gradually reduced by half every 2 to 4 days.
- the culture may be continued in a medium not containing the bone morphogenetic protein signaling pathway activator.
- the cells may be continuously cultured in a medium that does not contain a substance that activates the bone morphogenetic protein signaling pathway but contains a substance that acts on the Shh signaling pathway.
- the cells may be cultured under high oxygen partial pressure conditions.
- High oxygen partial pressure conditions refer to conditions in which the oxygen partial pressure exceeds the oxygen partial pressure in air (20%).
- the oxygen partial pressure is 30 to 60%, preferably 35 to 60%, and more preferably 40 to 60%.
- the culture after completion of step (A) or (A') may be performed for a period of time sufficient to induce differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells, for example, until 5% or more, 10% or more, 30% or more, or 50% or more of the cell aggregates being cultured contain pituitary hormone-producing cells.
- the period may be, for example, 10 days or more, 20 days or more, 30 days or more, 40 days or more, and 90 days or less, 80 days or less, 70 days or less, 60 days or less, or 50 days or less.
- the cell aggregates obtained by suspension culture of pluripotent stem cells in the step (A') are cell aggregates formed by culturing dispersed pluripotent stem cells under non-adhesive conditions in a differentiable medium.
- the method for producing the cell aggregates includes the SFEBq method (Serum-Free Floating culture of Embryoid Body-like aggregates with quick reaggregation), and the cell aggregates are formed within 12 to 72 hours from the start of differentiation induction (start of floating culture of pluripotent stem cells).
- the differentiable medium used here can be appropriately selected from the above-mentioned media.
- the medium may appropriately contain a differentiation inducer suitable for inducing differentiation of pituitary hormone-producing cells, and examples of the differentiation inducer include a ROCK inhibitor, a substance acting on the Shh signaling pathway, a substance inhibiting the JNK signaling pathway, and a substance inhibiting the Wnt signaling pathway.
- the cell aggregates can also be produced according to the methods described in WO 2016/013669, WO 2019/103129, Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351), WO 2023/054396, etc., or the culture conditions described in step (1) or (1') of "4.
- Method for producing a cell population containing pituitary tissue 1 when performing the culture conditions described in step (1) or (1') of "4. Method for producing a cell population containing pituitary tissue 1", the method may be performed in consideration of the method described in the above-mentioned publication (e.g., WO 2023/054396, etc.).
- Method for producing a cell population containing pituitary tissue 1 One embodiment of a method for producing pituitary hormone-producing cells or a cell population containing said cells included in the therapeutic agent of the present invention is a method for producing a cell population containing pituitary hormone-producing cells, comprising the following steps (1') to (2) (hereinafter also referred to as "production method 1").
- step (1') a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor to form a cell population; (2) A second step of culturing the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue.
- a substance inhibiting the JNK signaling pathway is used in combination with a substance inhibiting the Wnt signaling pathway.
- the pituitary tissue in step (2) may contain precursor cells of pituitary hormone-producing cells and/or pituitary hormone-producing cells, depending on the culture period in step (2).
- a more preferred embodiment of the production method of the present invention is a method for producing a cell population containing pituitary tissue, comprising the following steps (a) and (1') to (2): (a) a step a of culturing pluripotent stem cells in a medium containing 1) a TGF ⁇ family signaling pathway inhibitor and/or a Sonic Hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells; (1') a first step of culturing (preferably in suspension) the cells obtained in step a in the presence of a substance inhibiting the JNK signaling pathway; (2) A second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue.
- the first step is a step of forming cell aggregates
- the cell population obtained in the first step that is subjected to the second step may be cell aggregates.
- step (1') preferably, a substance inhibiting the JNK signaling pathway is used in combination with a substance inhibiting the Wnt signaling pathway.
- a more preferred embodiment of the production method of the present invention is a method for producing a cell population containing pituitary tissue, comprising the following step (a) and the following steps (1) to (3): (a) a step a of culturing pluripotent stem cells in a medium containing 1) a TGF ⁇ family signaling pathway inhibitor and/or a Sonic Hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells; (1) a first step of culturing (preferably in suspension) the cells obtained in step a in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor; (2) a second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway; (3) A third step in which the cell population obtained in the second step is cultured (preferably in suspension) in the absence of a substance acting on the Sonic Hedgehog signaling pathway to obtain
- the first step is a step of forming cell aggregates
- the cell population obtained in the first step that is subjected to the second step, and the cell population obtained in the second step that is subjected to the third step may each be a cell aggregate.
- Step a is described below, in which pluripotent stem cells are cultured in a medium containing 1) a TGF ⁇ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and 2) an undifferentiated maintenance factor, in the absence of feeder cells.
- pluripotent stem cells are treated with a TGF ⁇ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and then cultured (preferably in suspension) in the first step, thereby changing the state of the pluripotent stem cells, improving the efficiency of non-neuroepithelial tissue formation, improving the quality of the resulting cell population (aggregates), making them more likely to differentiate and less likely to suffer cell death, and improving the efficiency of producing pituitary tissue cells.
- Step (a) is carried out in the absence of feeder cells.
- feeder-free refers to conditions substantially free of feeder cells (for example, the ratio of the number of feeder cells to the total number of cells is 3% or less).
- the pluripotent stem cells are preferably embryonic stem cells or induced pluripotent stem cells.
- Induced pluripotent stem cells are available from a designated institution, and can also be purchased commercially.
- human induced pluripotent stem cell lines 201B7, 201B7-Ff, 253G1, 253G4, 1201C1, 1205D1, 1210B2, and 1231A3 are available from Kyoto University and iPS Academia Japan, Inc.
- As established induced pluripotent stem cell lines for example, Ff-I01, Ff-I14, and QHJI01s04 cells established at Kyoto University are available from Kyoto University.
- HC-6 #10, 1231A3, and 1383D2 lines are available from the National Institute of Physical and Chemical Research.
- the TGF ⁇ family signaling pathway (i.e., the TGF ⁇ superfamily signaling pathway) is a signaling pathway that uses transforming growth factor ⁇ (TGF ⁇ ), Nodal/Activin, or BMP as ligands and is transmitted intracellularly by the Smad family.
- TGF ⁇ transforming growth factor ⁇
- Nodal/Activin Nodal/Activin
- BMP BMP
- TGF ⁇ family signaling pathway inhibitor refers to a substance that inhibits the TGF ⁇ family signaling pathway, i.e., the signaling pathway transmitted by the Smad family, and specific examples include TGF ⁇ signaling pathway inhibitors, Nodal/Activin signaling pathway inhibitors, and BMP signaling pathway inhibitors. As the TGF ⁇ family signaling pathway inhibitor, TGF ⁇ signaling pathway inhibitors are preferred.
- the TGF ⁇ signaling pathway inhibitor is not particularly limited as long as it inhibits the signaling pathway caused by TGF ⁇ , and may be any of nucleic acids, proteins, and low molecular weight organic compounds.
- examples of such substances include substances that act directly on TGF ⁇ (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes that code for TGF ⁇ (e.g., antisense oligonucleotides, siRNA, etc.), substances that inhibit the binding between TGF ⁇ receptors and TGF ⁇ , and substances that inhibit physiological activities caused by signaling via TGF ⁇ receptors (e.g., TGF ⁇ receptor inhibitors, Smad inhibitors, etc.).
- An example of a protein known as an inhibitor of the TGF ⁇ signaling pathway is Lefty.
- SB431542 (sometimes abbreviated as "SB431") (4-[4-(3,4-Methylenedioxyphenyl)-5-(2-pyridyl)-1H-imidazol-2-yl]benzoamide), SB505124 (2-[4-(1,3-Benzodioxol-5-yl)-2-(1,1-dimethylethyl)-1H-imidazol-5-yl]-6-methylpyridine), S B525334 (6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline), LY2157299 (4-[5,6-Dihydro-2-(6 -methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-6-quinolinecarboxamide
- SB431542 is a compound known as an inhibitor of the TGF ⁇ receptor (ALK5) and Activin receptor (ALK4/7) (i.e., a TGF ⁇ R inhibitor).
- SIS3 is a TGF ⁇ signaling pathway inhibitor that inhibits phosphorylation of SMAD3, an intracellular signaling factor under the control of the TGF ⁇ receptor.
- ITD-1 is a promoter of proteasomal degradation of the TGF- ⁇ type II receptor.
- the TGF ⁇ signaling pathway inhibitor preferably includes an Alk5/TGF ⁇ R1 inhibitor.
- the Alk5/TGF ⁇ R1 inhibitor is preferably SB431542, SB505124, SB525334, LY2157299, GW788388, LY364947, SD-208, EW-7197, A83-01, RepSox, SM16, R268712, IN1130, Galunisertib, AZ1 2799734, A77-01, KRCA 0008, GSK 1838705, Crizotinib, Certinib, ASP 3026, TAE684, AZD3463, TP0427736, and more preferably SB431542 or A83-01.
- the concentration of the TGF ⁇ signaling pathway inhibitor in the medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects.
- SB431542 is used as the TGF ⁇ signaling pathway inhibitor in step (a)
- it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 100 ⁇ M, more preferably about 10 nM to about 50 ⁇ M, even more preferably about 100 nM to about 50 ⁇ M, and particularly preferably about 1 ⁇ M to about 10 ⁇ M.
- TGF ⁇ signaling pathway inhibitor other than SB431542 when used, it is desirably used at a concentration that exhibits TGF ⁇ signaling pathway inhibitory activity equivalent to that of SB431542 at the above-mentioned concentration.
- the TGF ⁇ signaling pathway inhibitory activity of SB431542 and the like can be determined by methods well known to those skilled in the art, for example, by detecting phosphorylation of Smads by Western blotting (Mol Cancer Ther. (2004) 3, 737-45.).
- a substance acting on the Shh signaling pathway is a substance that can enhance signal transduction mediated by Shh.
- substances acting on the Shh signaling pathway include proteins belonging to the Hedgehog family (e.g., Shh, Ihh) or fragments or mutants thereof (SHH C25II N-terminus, SHH C24II N-terminus), Shh receptors, Shh receptor agonists, Smo agonists, Purmorphamine (9-cyclohe xyl-N-[4-(morpholinyl)phenyl]-2-(1-naphthalenyloxy)-9H-purin-6-amine), GSA-10(Propyl 4-(1-hexyl-4-hydroxy-2-oxo-1,2-dihydr) oquinoline-3-carboxamide)benzoate), Hh-Ag1.5, 20 (S)-Hydroxycholesterol, SAG (Smoothened Agonist: N-Methyl-N'-(3-pyridinylbenz
- the substance acting on the Shh signaling pathway preferably includes at least one selected from the group consisting of SAG, Purmorphamine, and GSA-10, and more preferably includes SAG.
- the concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range in which the above-mentioned effects can be achieved.
- SAG is usually used at a concentration of about 1 nM to about 2000 nM, preferably about 10 nM to about 1000 nM, more preferably about 10 nM to about 700 nM, even more preferably about 50 nM to about 700 nM, particularly preferably about 100 nM to about 600 nM, and most preferably about 100 nM to about 500 nM.
- the Shh signaling promoting activity can be determined by methods well known to those skilled in the art, for example, a reporter gene assay focusing on the expression of the Gli1 gene (Oncogene (2007) 26, 5163-5168).
- the medium used in step (a) contains an undifferentiated state maintenance factor to enable culture to maintain the undifferentiated state.
- the undifferentiated state maintenance factor is not particularly limited as long as it is a substance that has the effect of suppressing the differentiation of pluripotent stem cells.
- examples of undifferentiated state maintenance factors commonly used by those skilled in the art include FGF signaling pathway acting substances, TGF ⁇ family signaling pathway acting substances, insulin, etc.
- Specific examples of FGF signaling pathway acting substances include fibroblast growth factors (e.g., bFGF, FGF4, and FGF8).
- TGF ⁇ family signaling pathway acting substances examples include TGF ⁇ signaling pathway acting substances and Nodal/Activin signaling pathway acting substances.
- TGF ⁇ signaling pathway acting substances examples include TGF ⁇ 1 and TGF ⁇ 2.
- substances acting on the Nodal/Activin signaling pathway include Nodal, Activin A, and Activin B. These substances may be used alone or in combination.
- the medium in step (a) preferably contains bFGF as an undifferentiated maintenance factor.
- the undifferentiation maintenance factor is usually a mammalian undifferentiation maintenance factor. Examples of mammals include those mentioned above. Since the undifferentiation maintenance factor may have cross-reactivity between mammalian species, any mammalian undifferentiation maintenance factor may be used as long as it can maintain the undifferentiated state of the pluripotent stem cells to be cultured.
- the undifferentiation maintenance factor is preferably an undifferentiation maintenance factor of the same mammalian species as the cells to be cultured.
- human undifferentiation maintenance factors e.g., bFGF, FGF4, FGF8, EGF, Nodal, Activin A, Activin B, TGF ⁇ 1, TGF ⁇ 2, etc.
- the undifferentiation maintenance factor is preferably isolated.
- the undifferentiation maintenance factor may be one produced by any host or one artificially synthesized, so long as it has the ability to maintain the undifferentiation state of the pluripotent stem cells to be cultured.
- the undifferentiation maintenance factor used in the present invention is preferably one that has been modified in the same manner as that occurring in a living body, and more preferably one that has been produced in cells of the same type as the pluripotent stem cells to be cultured under conditions that do not contain any xenogeneic components.
- One embodiment of the production method according to the present invention includes a step of providing an isolated factor for maintaining undifferentiation.
- One embodiment of the production method according to the present invention includes a step of exogenously (or extrinsically) adding the isolated factor for maintaining undifferentiation to the medium used in step (a).
- the factor for maintaining undifferentiation may be added in advance to the medium used in step (a).
- the concentration of the undifferentiation maintenance factor in the medium used in step (a) is a concentration that can maintain the undifferentiated state of the pluripotent stem cells being cultured, and can be appropriately set by a person skilled in the art.
- the concentration is usually about 4 ng/ml to about 500 ng/ml, preferably about 10 ng/ml to about 200 ng/ml, and more preferably about 30 ng/ml to about 150 ng/ml.
- Step (a) is carried out in the absence of feeder cells.
- the culture of pluripotent stem cells in step (a) may be carried out under either suspension culture or adherent culture conditions, but is preferably carried out by adherent culture.
- an appropriate matrix may be used as a scaffold to provide the pluripotent stem cells with a scaffold in place of feeder cells.
- the pluripotent stem cells are cultured in an adherent manner in a culture vessel whose surface is coated with a matrix scaffold.
- Matrices that can be used as scaffolds include laminin (Nat Biotechnol. 28, 611-615 (2010)), laminin fragments (Nat Commun 3, 1236 (2012)), basement membrane preparations (Nat Biotechnol 19, 971-974 (2001)), gelatin, collagen, heparan sulfate proteoglycan, entactin, and vitronectin.
- the matrix used is preferably laminin 511 (Nat Biotechnol 28, 611-615 (2010)).
- the laminin fragment is not particularly limited as long as it has adhesive properties to pluripotent stem cells and enables the maintenance culture of pluripotent stem cells under feeder-free conditions, but is preferably an E8 fragment.
- the laminin E8 fragment was identified as a fragment with strong cell adhesive activity among fragments obtained by digesting laminin 511 with elastase (EMBO J., 3: 1463-1468, 1984; J. Cell Biol., 105: 589-598, 1987).
- the E8 fragment of laminin 511 is preferably used (Nat Commun 3, 1236 (2012); Scientific Reports 4, 3549 (2014)).
- the laminin E8 fragment does not need to be a product of elastase digestion of laminin, and may be a recombinant product. It may be produced in a genetically modified animal (such as a silkworm). In order to avoid contamination with unidentified components, recombinant laminin fragments are preferably used.
- the E8 fragment of laminin 511 is commercially available and can be purchased from, for example, Nippi Corporation.
- the laminin or laminin fragment used in this specification is preferably isolated.
- the pluripotent stem cells are cultured in an adherent manner in a culture vessel whose surface is preferably coated with isolated laminin 511 or the E8 fragment of laminin 511, more preferably with the E8 fragment of laminin 511.
- the medium used in step (a) is not particularly limited as long as it is a medium that allows the maintenance of undifferentiated culture of pluripotent stem cells under feeder-free conditions (feeder-free medium).
- the medium used in step (a) may be a serum medium or a serum-free medium. From the viewpoint of avoiding contamination with chemically undefined components, the medium used in step (a) is preferably a serum-free medium.
- the medium may contain a serum substitute.
- the culture time of the pluripotent stem cells in step (a) is not particularly limited as long as it is within a range that can achieve the effect of improving the quality of the cell population (aggregates) that can be formed in the subsequent first step, but is usually 0.5 to 144 hours, preferably 2 to 96 hours, more preferably 6 to 48 hours, even more preferably 12 to 48 hours, and particularly preferably 18 to 28 hours, for example 24 hours.
- step (a) is started 0.5 to 144 hours, preferably 18 to 28 hours, before the start of the first step, and the first step is continued after step (a) is completed.
- human pluripotent stem cells are cultured as adherent cells in a serum-free medium containing bFGF in the absence of feeder cells.
- the adherent culture is preferably performed in a culture vessel whose surface is coated with laminin 511, E8 fragment of laminin 511, or vitronectin.
- the adherent culture is preferably performed using StemFit as a feeder-free medium.
- StemFit medium contains bFGF as a component for maintaining undifferentiated state (Scientific Reports (2014) 4, 3594).
- human pluripotent stem cells are cultured in suspension in a serum-free medium containing bFGF in the absence of feeder cells.
- the human pluripotent stem cells may form aggregates of human pluripotent stem cells.
- culture conditions such as culture temperature and CO2 concentration can be appropriately set.
- the culture temperature is, for example, about 30° C. to about 40° C., preferably about 37° C.
- the CO2 concentration is, for example, about 1% to about 10%, preferably about 5%, when a bicarbonate buffered medium is used.
- step (1') pluripotent stem cells are cultured in the presence of an inhibitor of the c-Jun N-terminal kinase (JNK) signaling pathway to obtain a cell population.
- JNK c-Jun N-terminal kinase
- JNK is a kinase belonging to the MAPK family and is involved in intracellular signal transduction in response to various environmental stresses, inflammatory cytokines, growth factors, and stimulation by GPCR agonists.
- JNK signaling pathway inhibitor refers to any substance that can suppress signal transduction transmitted by JNK, but is not limited thereto.
- JNK signaling pathway inhibitors include substances that have the activity of inhibiting signal transduction by mechanisms such as inhibiting factors upstream or downstream of the JNK signaling mechanism, or the enzyme activity, multimerization, and binding to other factors or nucleic acids of JNK itself, and promoting degradation.
- JNK signaling pathway inhibitors include, but are not limited to, JNK inhibitors, Rac inhibitors, MKK inhibitors, MEK inhibitors, Src inhibitors, receptor tyrosine kinase (RTK) inhibitors, and ASK inhibitors.
- JNK inhibitors include, for example, JNK-IN-8 ((E)-3-(4-(dimethylamino)but-2-enamido)-N-(3-methyl-4-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)phenyl)benzamide), SP600125 (Anthra[1-9-cd]pyrazol-6(2H)-one), DB07268 (2-[[2-[(3-Hydroxyphenyl)amino]-4-pyrimidinyl]amino]benzamide), and Ta nzisertib (trans-4-[[9-[(3S)-Tetrahydro-3-furanyl]-8-[(2,4,6-trifluorophenyl)amino]-9H-purin-2-yl]amino]cyclohexanol), Be ntamapimod (1,3-Benzothi
- Rac inhibitors include, for example, EHT1864 (5-(5-(7-(Trifluoromethyl)quinolin-4-ylthio)pentyloxy)-2-(morpholinomethyl)-4H-pyran-4-one dihydrochloride), NSC23766 (N6-[2-[[4-(Diethylamino)-1-methylbutyl]amino]-6-methyl-4-pyrimidinyl nyl]-2-methyl-4,6-quinolinediaminetrihydrochloride), EHop-016(N4-(9-Ethyl-9H-carbazol-3-yl)-N2-[3-(4-morpholinyl)propy l]-2,4-pyrimidinediamine), 1A-116(N-(3,5-Dimethylphenyl)-N'-[2-(trifluoromethyl)phenyl]guan idine), ZCL278 (2-(4-bromo-2-chlorophenoxy)-N-(4
- the timing of addition of the JNK signaling pathway inhibitor in the present invention is not limited as long as the effect of improving the efficiency of production of pituitary tissue from human pluripotent stem cells is exhibited, but it is preferable that it is added already at the time of adding the BMP signaling pathway active substance in step (2) described below, within 72 hours from the start of differentiation induction.
- the more preferable timing of addition of the JNK inhibitor is simultaneous with the start of differentiation induction.
- step (1) is a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor.
- the Wnt signaling pathway is a signaling pathway that uses Wnt family proteins as ligands and mainly Frizzled as a receptor. Examples of the signaling pathway include the canonical Wnt pathway and the non-canonical Wnt pathway.
- the canonical Wnt pathway is transmitted by ⁇ -Catenin.
- non-canonical Wnt pathway examples include the Planar Cell Polarity (PCP) pathway, the Wnt/JNK pathway, the Wnt/Calcium pathway, the Wnt-RAP1 pathway, the Wnt-Ror2 pathway, the Wnt-PKA pathway, the Wnt-GSK3MT pathway, the Wnt-aPKC pathway, the Wnt-RYK pathway, and the Wnt-mTOR pathway.
- PCP Planar Cell Polarity
- Wnt/JNK pathway the Wnt/Calcium pathway
- Wnt-RAP1 pathway the Wnt-Ror2 pathway
- the Wnt-PKA pathway the Wnt-GSK3MT pathway
- Wnt-aPKC pathway the Wnt-RYK pathway
- Wnt-mTOR pathway examples include the non-canonical Wnt pathway.
- the Wnt signaling pathway inhibitor is not limited as long as it can suppress signaling induced by Wnt family proteins.
- the inhibitor may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit Wnt processing and extracellular secretion, substances that act directly on Wnt (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes encoding Wnt (e.g., antisense oligonucleotides, siRNA, CRISPRi, etc.), substances that inhibit the binding between Wnt receptors and Wnt, and substances that inhibit physiological activity resulting from signaling by Wnt receptors.
- substances that inhibit Wnt processing and extracellular secretion e.g., proteins, antibodies, aptamers, etc.
- substances that suppress the expression of genes encoding Wnt e.g., antisense oligonucleotides, siRNA, CRISPRi, etc.
- Proteins known to inhibit the Wnt signaling pathway include proteins belonging to the secreted frizzled related protein (sFRP) class (sFRP1-5, Wnt inhibitory factor-1 (WIF-1), Cerberus), proteins belonging to the Dickkopf (Dkk) class (Dkk1-4, Kremen), APCDD1, APCDD1L, proteins belonging to the Draxin family, IGFBP-4, Notum, and proteins belonging to the SOST/Sclerostin family.
- sFRP secreted frizzled related protein
- WIF-1 Wnt inhibitory factor-1
- Cerberus proteins belonging to the Dickkopf (Dkk) class (Dkk1-4, Kremen)
- APCDD1, APCDD1L proteins belonging to the Draxin family
- IGFBP-4 IGFBP-4
- Notum proteins belonging to the SOST/Sclerostin family.
- Wnt signaling pathway inhibitor compounds well known to those skilled in the art can be used.
- Wnt signaling pathway inhibitors include Frizzled inhibitors, Dishevelled (Dvl) inhibitors, Tankyrase (TANK) inhibitors, casein kinase 1 inhibitors, catenin-responsive transcription inhibitors, p300 inhibitors, CREB-binding protein (CBP) inhibitors, BCL-9 inhibitors, and TCF degradation inducers (Am J Cancer Res. 2015; 5 (8): 2344-2360).
- Inhibitors of the non-classical Wnt pathway include, for example, porcupine (PORCN) inhibitors, calcium/calmodulin-dependent protein kinase II (CaMKII) inhibitors, TGF- ⁇ -activated kinase 1 (TAK1) inhibitors, Nemo-Like Kinase (NLK) inhibitors, LIM Kinase inhibitors, mammalian target of These include rapamycin (mTOR) inhibitors, Rac inhibitors, c-Jun NH 2-terminal kinase (JNK) inhibitors, protein kinase C (PKC) inhibitors, methionine aminopeptidase 2 (MetAP2) inhibitors, calcineurin inhibitors, and nuclear factor of activated T cells (NFAT) inhibitors.
- PORCN porcupine
- CaMKII calcium/calmodulin-dependent protein kinase II
- NLK Nemo-Like Kinas
- Wnt signaling pathway inhibitors include KY02111 (N-(6-chloro-2-benzothiazolyl)-3,4-dimethoxybenzenepropanamide) and KY03-I (2-(4-(3,4-dimethoxyphenyl)butanamide)-6-iodobenzothiazole). These substances may be used alone or in combination.
- PORCN inhibitors include, for example, IWP-2 (N-(6-methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthioeno[3,2-d]pyrimidin-2-yl)thio]acetamide), IWP-3 (2-[[3-(4-fluorophenyl)-3, IWP-4 (N-(6-methyl-2-b) enzothiazolyl)-2-[[3,4,6,7-tetrahydro-3-(2-methoxy phenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl]thio]acetamide), IWP-L6(N-(5-phenyl-2-pyridinyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-p) henylthieno[3,2-d]pyrimidin-2-yl)thio]acetami de), IWP
- the Wnt signaling pathway inhibitor preferably includes at least one selected from the group consisting of a PORCN inhibitor, KY02111, and KY03-I, and more preferably includes a PORCN inhibitor. It is also preferable that the Wnt signaling pathway inhibitor includes a substance having inhibitory activity against the non-classical Wnt pathway of Wnt. More preferably, the Wnt signaling pathway inhibitor includes a substance having inhibitory activity against the Wnt/Planar Cell Polarity (PCP) pathway.
- PCP Planar Cell Polarity
- the PORCN inhibitor used in the present invention preferably includes at least one selected from the group consisting of IWP-2, IWP-3, IWP-4, IWP-L6, IWP-12, LGK-974, Wnt-C59, ETC-159, and GNF-6231, more preferably includes IWP-2 or Wnt-C59, and even more preferably includes IWP-2.
- the concentration of the Wnt signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of improving the production efficiency of cells that constitute the pituitary gland, for example, when IWP-2, a type of PORCN inhibitor, is used as the Wnt signaling pathway inhibitor, its concentration is usually about 10 nM to about 50 ⁇ M, preferably about 10 nM to about 30 ⁇ M, more preferably about 100 nM to about 10 ⁇ M, and most preferably about 0.5 ⁇ M.
- Wnt-C59 a type of PORCN inhibitor
- its concentration is usually about 10 pM to about 1 ⁇ M, preferably about 100 pM to about 500 nM, and more preferably about 50 nM.
- KY02111 its concentration is usually about 10 nM to about 50 ⁇ M, preferably about 10 nM to about 30 ⁇ M, more preferably about 100 nM to about 10 ⁇ M, and even more preferably about 5 ⁇ M.
- Wnt signaling pathway inhibitor other than the above, it is desirable to use it at a concentration that shows the same Wnt signaling pathway inhibitory activity as the above concentrations.
- a TGF ⁇ signaling pathway inhibitor is further present in the medium in the first step (step (1) or step (1')).
- the TGF ⁇ signaling pathway inhibitor used in the first step may be the same as that exemplified in step (a).
- the TGF ⁇ signaling pathway inhibitors in step (a) and the first step may be the same or different, but are preferably the same.
- the concentration of the TGF ⁇ signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used, within a range that can achieve the above-mentioned effects.
- SB431542 is used as the TGF ⁇ signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 100 ⁇ M, more preferably about 100 nM to about 50 ⁇ M, and even more preferably about 500 nM to about 10 ⁇ M.
- a TGF ⁇ signaling pathway inhibitor other than SB431542 it is desirable to use it at a concentration that shows the same TGF ⁇ signaling pathway inhibitory activity as SB431542 at the above-mentioned concentration.
- TAK1 is a serine-threonine protein kinase of the MAP kinase kinase kinase (MAPKKK) family that mediates signal transduction activated by TGF ⁇ , bone morphogenetic protein (BMP), interleukin 1 (IL-1), TNF- ⁇ , etc.
- the TAK1 inhibitor is not limited as long as it can suppress signal transduction mediated by TAK1. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit the binding of TAK1 to a substrate, substances that inhibit the phosphorylation of TAK1, substances that promote the dephosphorylation of TAK1, substances that inhibit the transcription or translation of TAK1, and substances that promote the degradation of TAK1.
- TAK1 inhibitor for example, (5Z)-7-Oxozeaenol ((3S, 5Z, 8S, 9S, 11E)-3,4,9,10-tetrahydro-8,9,16-trihydroxy-14-methoxy-3 -methyl-1H-2-benzoxacyclotetradecine-1,7(8H)-dione), N-Des(aminocarbonyl)AZ-TAK1 inhibitor(3-Amino-5-[4-(4-morpholinyl) methyl)phenyl]-2-thiophenecarbox amide), Takinib (N1-(1-Propyl-1H-benzimidazol-2-yl)-1,3-benzenedicarboxamide), NG25 (N-[4-[(4-Ethyl-1-piperazinyl)methyl]-3- (trifluoromethyl)phenyl]-4-methyl-3-(1H-pyrrolo[2,3-b]pyridin-4-yl
- the TAK1 inhibitor is preferably (5Z)-7-Oxozeaenol.
- (5Z)-7-Oxozeaenol is used as the TAK1 inhibitor in the first step, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 50 ⁇ M, more preferably about 100 nM to about 25 ⁇ M, and even more preferably about 500 nM to about 10 ⁇ M.
- a TAK1 inhibitor other than (5Z)-7-Oxozeaenol it is preferably used at a concentration that exhibits TAK1 inhibitory activity equivalent to that of (5Z)-7-Oxozeaenol at the above concentration.
- TAK1 inhibitory activity can be determined, for example, by a method such as the kinase assay described in Cell chemical biology 24.8 (2017): 1029-1039. From the viewpoint of controlling the proportion of cells contained in the pituitary tissue, the TAK1 inhibitor can be added at any stage of the first step or any subsequent steps and then removed. In a preferred embodiment, the TAK1 inhibitor is added at the start of step (b) described below.
- the medium used in the first step is not particularly limited as long as it is as described above.
- the medium used in the first step may be a serum medium or a serum-free medium.
- a serum-free medium is preferably used in this specification.
- the amount of KSR added to the serum-free medium is usually about 1% to about 30%, and preferably about 2% to about 20%, in the case of human ES cells, for example.
- serum-free media examples include a medium in which 5% KSR, 450 ⁇ M 1-monothioglycerol, and 1x Chemically Defined Lipid Concentrate are added to a 1:1 mixture of IMDM and F-12, or a medium in which 5% to 20% KSR, NEAA, pyruvic acid, and 2-mercaptoethanol are added to GMEM.
- the cells may be in either an adherent state or a floating state.
- the pluripotent stem cells are dispersed into single cells and then re-aggregated to form floating cell aggregates.
- the "dispersed cells” obtained by the dispersion operation are preferably single cells, but may also include cell clumps consisting of a small number of cells, for example, 2 to 100 cells, or may include cell clumps consisting of 2 to 50 cells.
- the "dispersed cells” may, for example, include 70% or more single cells and 30% or less cell clumps, and preferably include 80% or more single cells and 20% or less cell clumps.
- Methods for dispersing pluripotent stem cells include mechanical dispersion treatment, cell dispersion treatment, and cell protective agent addition treatment, and these treatments may be performed in combination.
- a preferred method for dispersing cells is to perform cell dispersion treatment simultaneously with cell protective agent addition treatment, followed by mechanical dispersion treatment.
- Cytoprotective agents used in the cytoprotective agent addition treatment include substances acting on the FGF signaling pathway, heparin, Rho-associated protein kinase (ROCK) inhibitors, myosin inhibitors, polyamines, integrated stress response (ISR) inhibitors, caspase inhibitors, cell adhesion promoters, serum, or serum substitutes.
- a preferred cytoprotective agent is a ROCK inhibitor.
- a ROCK inhibitor In order to suppress cell death of pluripotent stem cells (especially human pluripotent stem cells) induced by dispersion, it is preferable to add a ROCK inhibitor from the start of the culture in the first step.
- ROCK inhibitors include Y-27632 ((R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide, dihydrochloride), Fasudil (HA1077) (1-(5-isoquinolinylsulfonyl) homopiperazine, hydrochloride), ride), H-1152 (5-[[(2S)-hexahydro-2-methyl-1H-1,4-diazepin-1-yl]sulfonyl]-4-methyl-isoquinoline, dihydrochloride), HA-110 0(Hydroxyfasudil)(1-(1-Hydroxy-5-isoquinolinesulfonyl)h omopiperazine, hydrochloride), Chroman 1 ((3S)-N-[2-[2-(dimethylamino)ethoxy]-4-(1H-pyrazol-4-yl
- a prepared cell protective agent can also be used.
- prepared cell protective agents include RevitaCell Supplement (manufactured by Thermo Fisher Scientific), CloneR, and CloneR2 (manufactured by Stemcell Technologies). These substances may be used alone or in combination.
- RevitaCell Supplement manufactured by Thermo Fisher Scientific
- CloneR CloneR2
- these substances may be used alone or in combination.
- the ROCK inhibitor Y-27632 when added as the cell protective agent, it is usually added to the culture environment at a concentration of about 10 nM to about 10 mM, preferably about 100 nM to about 1 mM, and more preferably about 1 ⁇ M to about 100 ⁇ M.
- the ROCK inhibitor Chroman 1 when added as the cell protective agent, it is usually added to the culture environment at a concentration of about 10 pM to about 1 mM, preferably about 100 pM to about 100 ⁇ M, and more preferably about 1 nM to about 10 ⁇ M.
- the concentration of the ROCK inhibitor in the culture medium may be maintained for the period during which cell death of pluripotent stem cells needs to be suppressed (for example, 1 to 3 days, preferably 1 or 2 days), and may be gradually reduced after the period has elapsed by performing a half-volume medium replacement operation, etc., with a medium that does not contain a ROCK inhibitor, as described below, when performing a medium replacement operation. Also, after the period has elapsed, a full-volume medium replacement operation may be performed with a medium that does not contain a ROCK inhibitor.
- the cell dispersion liquid used in the cell dispersion treatment may be a solution containing at least one of an enzyme such as trypsin, collagenase, hyaluronidase, elastase, pronase, DNase, papain, etc., and a chelating agent such as ethylenediaminetetraacetic acid.
- an enzyme such as trypsin, collagenase, hyaluronidase, elastase, pronase, DNase, papain, etc.
- a chelating agent such as ethylenediaminetetraacetic acid.
- Commercially available cell dispersion liquids such as TripLE Select (Thermo Fisher Scientific), TripLE Express (Thermo Fisher Scientific), and Accumax (Innovative Cell Technologies), may also be used.
- a preferred cell dispersion liquid for treating the pluripotent stem cells obtained after step (a) is TrypLE Select, but is not limited to this.
- Mechanical dispersion methods include pipetting or scraping with a scraper.
- the dispersed cells are suspended in the above-mentioned medium.
- One method for dispersing pluripotent stem cells is, for example, treating a colony of pluripotent stem cells with ethylenediaminetetraacetic acid or Accumax in the presence of a ROCK inhibitor, and then dispersing the colony by pipetting.
- suspension culture is performed in the first step, a suspension of dispersed pluripotent stem cells is seeded into a non-adhesive cultureware. If the cultureware is non-adhesive, the cells are cultured in suspension, and multiple pluripotent stem cells gather together to form cell aggregates.
- dispersed pluripotent stem cells may be seeded in a relatively large culture vessel such as a 10 cm dish to simultaneously form multiple cell aggregates in one culture vessel.
- a relatively large culture vessel such as a 10 cm dish
- U-bottom, V-bottom such as a non-cell-adhesive 96-well microplate.
- the culture vessel can be made non-cell-adhesive by processing such as coating the surface of the culture vessel with a superhydrophilic polymer.
- a non-cell-adhesive multi-well plate is the PrimeSurface 96V-bottom plate (MS-9096V, manufactured by Sumitomo Bakelite Co., Ltd.). Centrifugation may be performed to form cell aggregates more quickly.
- a uniform population of cell aggregates can be obtained by collecting the cell aggregates formed in each well from multiple wells. If the cell aggregates are uniform, the production efficiency for each well and each repeated experiment can be more stable in the subsequent steps, and cells that make up the pituitary gland can be produced with greater reproducibility.
- a culture vessel in which one well is divided into a plurality of microwells and two or more cell aggregates are formed can be used.
- the suspension culture of any one or more of the first step, the second step, the b step, and the third step can be performed in a culture vessel in which at least one well is formed, and the well is divided into a plurality of microwells, and the suspension culture is performed so that one cell aggregate is formed in each of the microwells, and the number of cell aggregates corresponding to the number of divided microwells can be prepared for each well.
- a culture vessel in which a plurality of mortars, downward pyramids, concaves, grids, protuberances, etc. are formed on the bottom surface, which allows cells to settle in one place and promotes the formation of aggregates, or a culture vessel in which only a part of the bottom surface is processed to allow cells to adhere to the bottom surface so that aggregates can be easily formed can be used.
- the culture area per well of a culture device having microwells is not particularly limited, but from the viewpoint of efficient production of cell masses, the bottom area is preferably greater than 1 cm2 (equivalent to a 48-well plate), more preferably greater than 2 cm2 (equivalent to a 24-well plate), and even more preferably greater than 4 cm2 (equivalent to a 12-well plate).
- Examples of the culture equipment include, but are not limited to, the embryoid body formation plate AggreWell (manufactured by StemCell Technologies), PAMCELL (manufactured by ANK), spheroid microplate (manufactured by Corning), NanoCulture Plate/Dish (manufactured by Organogenix), Cell-able (manufactured by Toyo Gosei), EZSPHERE (manufactured by AGC Technoglass), SPHERICALPLATE 5D (manufactured by Mito Kogyo Co., Ltd.), TASCL (manufactured by Sims Bio), and microwell bag (e.g., those described in Scientific reports, 2022, 12.1: 1-11.).
- AggreWell manufactured by StemCell Technologies
- PAMCELL manufactured by ANK
- spheroid microplate manufactured by Corning
- NanoCulture Plate/Dish manufactured by Organogenix
- Cell-able manufactured by Toyo Gosei
- a three-dimensional cell culture vessel that allows the culture medium to be replaced for the entire plate at once while the cell aggregates remain in each well.
- An example of such a three-dimensional cell culture vessel is the PrimeSurface 96 slit well plate (manufactured by Sumitomo Bakelite Co., Ltd.). This plate has narrow openings (slits) at the top of each of the 96 wells through which the culture medium can enter and exit. The slits are set to a width that makes it difficult for cell aggregates to pass through, so the culture medium for the entire plate can be replaced at once while preventing adhesion between the cell aggregates, improving the efficiency of the operation and the quality of the cell aggregates.
- the concentration of pluripotent stem cells in the first step can be appropriately set so as to form cell aggregates more uniformly and efficiently.
- human pluripotent stem cells e.g., human iPS cells obtained from step (a)
- a liquid prepared so as to obtain about 1 x 10 3 to about 1 x 10 5 cells, preferably about 3 x 10 3 to about 5 x 10 4 cells, more preferably about 4 x 10 3 to about 2 x 10 4 cells, even more preferably about 4 x 10 3 to about 1.6 x 10 4 cells, and particularly preferably about 8 x 10 3 to about 1.2 x 10 4 cells per well is added to each well, and the plate is left to form cell aggregates.
- human pluripotent stem cells e.g., human iPS cells obtained in step (a)
- a cultureware having about 260 microwells per dish, a liquid prepared so that typically about 1 x 104 to about 1 x 108 cells, preferably about 3 x 104 to about 5 x 107 cells, more preferably about 4 x 104 to about 2 x 107 cells, even more preferably about 4 x 104 to about 1.6 x 107 cells, and particularly preferably about 8 x 104 to about 1.2 x 107 cells per dish is added to the dish, and the dish is allowed to stand to form cell aggregates.
- human pluripotent stem cells e.g., human iPS cells obtained from step (a)
- a solution prepared so that each well contains usually about 1 ⁇ 10 4 to about 1 ⁇ 10 8 cells, preferably about 3 ⁇ 10 4 to about 5 ⁇ 10 7 cells, more preferably about 4 ⁇ 10 4 to about 2 ⁇ 10 7 cells, even more preferably about 4 ⁇ 10 4 to about 1.6 ⁇ 10 7 cells, and particularly preferably about 8 ⁇ 10 4 to about 1.2 ⁇ 10 7 cells is added to each well, and the plate is centrifuged to form cell aggregates.
- the number of cells can be determined by counting with a hemocytometer.
- the time required for suspension culture to form cell aggregates can be appropriately determined depending on the pluripotent stem cells used, but it is desirable that the time be as short as possible in order to form uniform cell aggregates.
- the process from the dispersed cells until they form cell aggregates can be divided into a process in which the cells gather and a process in which the gathered cells form aggregates.
- the time from the time the dispersed cells are seeded i.e., the time when suspension culture begins
- the cells gather is preferably within about 24 hours, more preferably within about 12 hours.
- the time from the time the dispersed cells are seeded i.e., the time when suspension culture begins
- the time until the cell aggregates are formed is preferably within about 72 hours, more preferably within about 48 hours.
- the time until the cell aggregates are formed can be appropriately adjusted by adjusting the tool for aggregating the cells, the centrifugation conditions, etc.
- epithelial-like structures can be reproducibly formed in cells induced to differentiate from the formed aggregates.
- Examples of experimental procedures for forming cell aggregates include a method of confining cells in a small space using a small well plate (e.g., a plate with a well bottom area of about 0.1 to 2.0 cm2 calculated as a flat bottom) or a micropore, and a method of aggregating cells by centrifuging for a short time using a small centrifuge tube.
- Examples of small well plates include 24-well plates (area of about 1.88 cm2 calculated as a flat bottom), 48-well plates (area of about 1.0 cm2 calculated as a flat bottom), 96-well plates (area of about 0.35 cm2 calculated as a flat bottom, inner diameter of about 6 to 8 mm), and 384-well plates.
- a preferred example is a 96-well plate.
- Examples of the shape of the small well plate, when viewed from above, include polygonal, rectangular, elliptical, and perfect circle as the shape of the bottom of the well.
- a preferred example is a perfect circle.
- the shape of the bottom of the well when viewed from the side is preferably a structure with a high outer periphery and a low inner recess, for example, a U-bottom, a V-bottom, or an M-bottom, preferably a U-bottom or a V-bottom, and most preferably a V-bottom.
- a cell culture dish e.g., a 60 mm to 150 mm dish, culture flask
- the bottom of the small well plate is preferably a non-cell-adhesive bottom, preferably a bottom coated with a non-cell-adhesive material.
- a cell population of the desired shape can be prepared by suspending a dispersed single cell or a spheroid composed of multiple cells in a biocompatible ink (bioink) and printing it out using a bio 3D printer (e.g., Celllink's BIO X, etc.), or by piercing a cell population with a needle and stacking it (Cyfuse's Spike, etc.).
- a biocompatible ink bioink
- the formation of cell aggregates can be determined based on the size and cell number of the cell aggregates, the macroscopic morphology, the microscopic morphology and its uniformity determined by tissue staining analysis, the expression and uniformity of differentiated and undifferentiated markers, the control of expression of differentiation markers and their synchronicity, and the reproducibility of differentiation efficiency between aggregates.
- adherent culture is performed.
- the pluripotent stem cells on the culture vessel after step (a) may be used as they are in the first step, or the pluripotent stem cells may be dispersed into single cells and then seeded again on the adherent culture vessel.
- an appropriate extracellular matrix or synthetic cell adhesion molecule may be used as a scaffold.
- the scaffold allows adherent culture of the pluripotent stem cells in the culture vessel whose surface is coated.
- the extracellular matrix is preferably matrigel or laminin.
- synthetic cell adhesion molecules include synthetic peptides containing a cell adhesive domain such as poly-D-lysine and RGD sequence.
- the number of cells seeded is not particularly limited as long as differentiation into the pituitary gland occurs, but from the viewpoint of reproducing adhesion and interaction between cells, it is also preferable that the cell density reaches semi-confluence, which corresponds to more than 60% of the culture space of the vessel, within 72 hours after seeding on the culture vessel.
- the micropattern on the cultureware can be composed of a cell adhesive region and a cell non-adhesive region, and it is preferable that cells are cultured in the cell adhesive region.
- the shape of the cell adhesive region and the cell non-adhesive region is not limited as long as they can be developed on the cultureware.
- the cell adhesive region and the cell non-adhesive region may be formed as a single region on one cultureware, or multiple regions may be formed. It is preferable that the cell adhesive region is artificially treated for the purpose of improving adhesion.
- Examples of cultureware with a micropattern include CYTOOChip (manufactured by CYTOO), ibidi Micropatterning (manufactured by ibidi), etc.
- the cultureware can also be prepared using a PDMS mold and a matrix, etc.
- a cultureware coated with an extracellular matrix or a substrate that promotes cell adhesion may be processed with a laser or the like using, for example, a cell processing device (Model: CPD-017, Kataoka Seisakusho Co., Ltd.) to create cell adhesive and non-cell adhesive regions of any shape.
- step (a) When culturing the pluripotent stem cells obtained in step (a) on a cultureware with a micropattern, this can be carried out by referring to, for example, a previously reported method (Nature protocols, 11(11), 2223-2232.).
- the culture equipment has a flow path (micro-flow path) for perfusing the medium, and cells may be cultured in a perfusion environment in the first step and subsequent steps.
- a culture equipment is also called a microfluidic chip.
- the culture equipment e.g., microfluidic chip
- may be connected by a flow path to other culture equipment e.g., microfluidic chip for culturing cells or tissues other than the cells to be cultured in the manufacturing method of the present invention. This makes it possible to reproduce the interaction between the pituitary gland and other cells or tissues.
- Examples of other cells or tissues to be co-cultured with the pituitary gland include, but are not limited to, tissues that are regulated by hormones secreted from the pituitary gland, and tissues that promote the growth, differentiation, maturation, and survival of the pituitary gland, such as cells or tissues of the brain, blood vessels, bone, muscle, fat, thyroid gland, liver, adrenal gland, testis, ovary, and breast.
- Examples of methods for perfusing the medium include, but are not limited to, the use of a magnetic stirrer, a peristaltic pump, etc.
- the culture equipment may have a membrane that is permeable to oxygen or culture medium.
- the culture equipment may be capable of forming a concentration gradient of compounds, growth factors, etc.
- the membrane is, for example, a porous membrane.
- cells can be cultured by the manufacturing method of the present invention on one side separated by the membrane, and other cells or tissues, feeder cells, etc. can be cultured on the other side. This makes it possible to culture cells that constitute the pituitary gland or their precursor cells and cell populations containing these without contamination with other cells or tissues.
- examples include an operation in which new medium is added without discarding the original medium (medium addition operation), an operation in which about half the original medium (about 30-90% of the volume of the original medium, for example about 40-60%) is discarded and about half the new medium (about 30-90% of the volume of the original medium, for example about 40-60%) is added (half medium exchange operation), and an operation in which about the entire amount of the original medium (90% or more of the volume of the original medium) is discarded and about the entire amount of new medium (90% or more of the volume of the original medium) is added (full medium exchange operation).
- an operation may be performed in which about half of the original medium is discarded and about half of a new medium containing the specific component at a concentration higher than the final concentration is added (half medium exchange operation).
- the medium exchange operation may be performed multiple times a day, preferably multiple times within an hour (e.g., 2 to 3 times).
- the cells or cell aggregates may be transferred to another culture vessel.
- the tools used for the medium exchange operation are not particularly limited, but examples include a pipetter, Pipetman (registered trademark), a multichannel pipette, and a continuous dispenser.
- a pipetter for example, when a 96-well plate is used as the culture equipment, a multichannel pipette may be used.
- the culture time in the first step is usually about 8 hours to 6 days, preferably about 12 hours to 60 hours.
- a compound that promotes differentiation into the placode region in order to improve the efficiency of pituitary production.
- compounds that have the above-mentioned effect include BRL-54443, phenanthrolin, and parthenolide, which are described in US Patent US20160326491A1.
- BRL-54443 is used as a compound that promotes differentiation into the placode region, it is usually used at a concentration of about 10 nM to about 100 ⁇ M
- phenanthrolin when phenanthrolin is used, it is usually used at a concentration of about 10 nM to about 100 ⁇ M
- parthenolide it is usually used at a concentration of about 10 nM to about 100 ⁇ M.
- the culture can be performed in the presence of a substance acting on the Sonic Hedgehog signaling pathway.
- a substance acting on the Sonic Hedgehog signaling pathway used in the first step, the same substances as those exemplified in step (a) can be used.
- the substances acting on the Shh signaling pathway in step (a) and the first step may be the same or different, but are preferably the same, and are preferably SAG.
- the concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range that can achieve the above-mentioned effects.
- SAG When SAG is used as the substance acting on the Shh signaling pathway in the first step, it is usually used at a concentration of about 1 nM to about 3 ⁇ M, preferably about 10 nM to about 2 ⁇ M, more preferably about 30 nM to about 1 ⁇ M, and even more preferably about 50 nM to about 500 nM.
- Step (2) the cell population obtained in the first step is cultured in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway. If the cells are cultured in suspension in the first step, the formed cell aggregates may be continuously cultured in suspension in step (2). If the cells are cultured in adhesion in the first step, the cells may be continuously cultured in adhesion in step (2). After the cells are cultured in suspension in the first step, they may be cultured in adhesion in step (2).
- a substance acting on the BMP signaling pathway is a substance that can enhance the signaling pathway mediated by BMP.
- substances that can enhance the signaling pathway mediated by BMP include substances that stabilize BMP ligands in a culture environment and improve their potency, substances that bind to type I BMP receptors ALK-1, ALK-2, ALK-3, and ALK-6 and activate and induce intracellular signaling downstream of the receptor, substances that induce phosphorylation of Smad-1, Smad-5, Smad-8, and Smad-9 involved in intracellular BMP signaling, and substances that induce and enhance functions such as activation and inhibition of gene transcription by Smad-1/5/8/9.
- a substance acting on the BMP signaling pathway examples include BMP proteins such as BMP2, BMP4, and BMP7, GDF proteins such as GDF5, 6, and 7, anti-BMP receptor antibodies, and BMP partial peptides. These substances may be used alone or in combination.
- a substance acting on the BMP signal transduction pathway can be defined as a substance that has the ability to induce differentiation into osteoblast-like cells and the ability to induce alkaline phosphatase production in cells such as mouse precursor chondrocyte cell line ATDC5, mouse calvaria-derived cell line MC3T3-E1, and mouse striated muscle-derived cell line C2C12.
- substances having the above activities include BMP2, BMP4, BMP5, BMP6, BMP7, BMP9, BMP10, BMP13/GDF6, BMP14/GDF5, GDF7, etc.
- BMP2 protein and BMP4 protein are available, for example, from R&D Systems, BMP7 protein is available, for example, from Biolegend, GDF5 protein is available, for example, from Peprotech, GDF6 protein is available, for example, from Primegene, and GDF7 protein is available, for example, from Fujifilm Wako Pure Chemical Industries, Ltd.
- the BMP signaling pathway active substance preferably includes at least one protein selected from the group consisting of BMP2, BMP4, BMP7, BMP13, and GDF7, and more preferably includes BMP4.
- the concentration of the substance acting on the BMP signaling pathway in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of improving the production efficiency of cells that constitute the pituitary gland, when BMP4 is used as the substance acting on the BMP signaling pathway, it is usually used at a concentration of about 1 pM to about 100 nM, preferably about 10 pM to about 50 nM, more preferably about 25 pM to about 25 nM, even more preferably about 25 pM to about 5 nM, particularly preferably about 100 pM to about 5 nM, and most preferably about 500 pM to about 2 nM.
- a substance acting on the BMP signaling pathway other than BMP4 when used, it is desirably used at a concentration that exhibits the same BMP signaling pathway promoting activity as BMP4 at the above-mentioned concentration.
- a person skilled in the art can easily determine the concentration of the substance acting on the BMP signaling pathway to be added by comparing the activity described in the product insert, such as the ED50 value of the ability to induce alkaline phosphatase production in the mouse precursor chondrocyte cell line ATDC5, with the concentration and activity of the above-mentioned BMP4.
- BMP signaling pathway active substances Compounds well known to those skilled in the art can also be used as BMP signaling pathway active substances.
- BMP signaling pathway active substances include Smurf1 inhibitors, Chk1 inhibitors, and phosphorylated Smad stabilizers.
- compounds having the above-mentioned activity include A-01 ([4-[[4-Chloro-3-(trifluoromethyl)phenyl]sulfonyl]-1-piperazinyl][4-(5-methyl-1H-pyrazol-1-yl)phenyl]methane), PD 407824 (9-Hydroxy-4-phenyl-pyr Examples include rolo[3,4-c]carbazole-1,3(2H,6H)-dione, SB4 (2-[[(4-bromophenyl)methyl]thio]benzoxazole), SJ000291942 (2-(4-ethylphenoxy)-N-(4-fluoro-3-nitrophenyl
- the substance acting on the Shh signaling pathway used in step (2) may be the same as those exemplified in step (a).
- the substances acting on the Shh signaling pathway in steps (a) and (2), and optionally the substance acting on the Shh signaling pathway in step (1), may be the same or different, but are preferably the same, and are preferably SAGs.
- the concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range that can achieve the above-mentioned effects.
- SAG When SAG is used as the substance acting on the Shh signaling pathway in step (2), it is usually used at a concentration of about 1 nM to about 5 ⁇ M, preferably about 10 nM to about 4.5 ⁇ M, more preferably about 50 nM to about 4 ⁇ M, and even more preferably about 100 nM to about 3 ⁇ M.
- the medium used in step (2) is not particularly limited as long as it contains a substance acting on the Shh signaling pathway and a substance acting on the BMP signaling pathway.
- Examples of the medium used in step (2) include the medium listed in the first step.
- the start time of step (2) is preferably 0.5 hours to 6 days after the start of the culture in the first step, more preferably 0.5 hours to 72 hours, and even more preferably 24 hours to 60 hours.
- step (2) is started during the above period in the presence of a Wnt signal pathway inhibitor, non-neuroepithelial-like tissue is formed on the surface of the cell aggregates, and pituitary glands are formed extremely efficiently.
- the start time of step (2) is preferably the time when at least 10% of the cells on the surface layer of the cell aggregate formed in the first step, more preferably at least 30%, and even more preferably at least 50% of the cells form tight junctions with each other.
- the start time of step (2) is preferably the time when at least 10% of the cells on the surface layer of the cell aggregate formed in the first step, more preferably at least 30%, and even more preferably at least 50% of the cells form tight junctions with each other.
- step (2) the start of culture in the presence of a substance acting on the BMP signaling pathway may be performed by carrying out the above-mentioned medium replacement operation (e.g., medium addition operation, half-medium replacement operation, full-medium replacement operation, etc.) using the culture vessel in which the first step was carried out, or the cells may be transferred to a different culture vessel.
- medium replacement operation e.g., medium addition operation, half-medium replacement operation, full-medium replacement operation, etc.
- the period of culture in the medium containing the substance acting on the BMP signaling pathway in step (2) can be appropriately set.
- the period of culture in step (2) may be any period during which the concentration of the substance acting on the BMP signaling pathway in the culture medium is maintained for a period necessary for inducing pituitary tissue (specifically, a cell population containing pituitary progenitor cells), and is usually 8 hours or more, preferably 10 hours or more, more preferably 12 hours or more, even more preferably 14 hours or more, and most preferably 16 hours or more.
- the concentration of the substance acting on the BMP signaling pathway may be gradually reduced after the lapse of the period by performing the above-mentioned half-volume medium replacement operation with a medium not containing a substance acting on the BMP signaling pathway. Also, after the lapse of the period, a full-volume medium replacement operation may be performed with a medium not containing a substance acting on the BMP signaling pathway. Even in this case, as long as the culture is continued in a medium containing a substance acting on the Shh signaling pathway, it is included as part of step (2) in the present specification.
- the period of culture in the medium containing the substance acting on the Shh signaling pathway in step (2) can be set appropriately.
- the culture period in the presence of the substance acting on the Shh signaling pathway in steps (1) and (2) is 20 to 40 days, preferably 25 to 35 days, and more preferably about 30 days.
- FGF signaling pathway active substance is not particularly limited as long as it is a substance that can enhance the signaling pathway mediated by FGF (fibroblast growth factor).
- FGF signaling pathway active substances include FGF proteins such as FGF1, FGF2 (also called bFGF), FGF3, FGF8, and FGF10, anti-FGF receptor antibodies, and FGF partial peptides. These substances may be used alone or in combination.
- FGF2 protein and FGF8 protein are available, for example, from Fujifilm Wako Pure Chemical Industries, Ltd.
- the FGF signaling pathway active substance preferably includes at least one selected from the group consisting of FGF2, FGF3, FGF8, FGF10, and modified forms thereof, more preferably includes FGF2, and even more preferably includes recombinant human FGF2.
- the concentration of the FGF signaling pathway active substance in the medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of differentiation into cells that constitute the pituitary gland and promotion of cell survival and proliferation, when FGF2 is used as the FGF signaling pathway active substance, it is usually used at a concentration of about 1 pg/ml to about 100 ⁇ g/ml, preferably about 10 pg/ml to about 50 ⁇ g/ml, more preferably about 100 pg/ml to about 10 ⁇ g/ml, even more preferably about 500 pg/ml to about 1 ⁇ g/ml, and most preferably about 1 ng/ml to about 200 ng/ml.
- FGF signaling pathway active substance other than FGF2 When using an FGF signaling pathway active substance other than FGF2, it is desirable to use it at a concentration that shows the same FGF signaling pathway promoting activity as FGF2 at the above concentration.
- the FGF signaling pathway promoting activity of the added substance can be measured, for example, by a method such as a cell proliferation test using 3T3 cells.
- Heparin is available as a sodium salt, for example from Fujifilm Wako Pure Chemical Industries, Ltd.
- the concentration of heparin or heparan sulfate in the medium can be appropriately set within a range that can achieve the above-mentioned effects.
- the concentration of heparin sodium in the medium is usually about 1 ng/ml to about 100 mg/ml, preferably about 10 ng/ml to about 50 mg/ml, more preferably about 100 ng/ml to about 10 mg/ml, even more preferably about 500 ng/ml to about 1 mg/ml, and most preferably about 1 ⁇ g/ml to about 200 ⁇ g/ml.
- concentration has the same FGF protein protection activity as the above-mentioned concentration of heparin.
- modified FGF such as Thermostable FGF2 described in US Patent No. US8772460B2, or FGF2 sustained-release beads such as StemBeads FGF2 in which FGF2 is bound to a biodegradable polymer.
- Thermostable FGF2 is available, for example, from HumanZyme.
- StemBeads FGF2 is available, for example, from StemCulture.
- the timing of addition of the substance acting on the FGF signaling pathway in step (2) and the subsequent steps can be set as appropriate.
- the substance acting on the FGF signaling pathway is added 6 hours or more, more preferably 12 hours or more, and even more preferably 18 hours or more after the addition of the substance acting on the BMP signaling pathway in step (2).
- step (2) it is also preferable to continue adding the additives used in step (a) or the first step, such as a JNK signaling pathway inhibitor, a Wnt signaling pathway inhibitor, a TGF ⁇ signaling pathway inhibitor, a TAK1 inhibitor, etc.
- the JNK signaling pathway inhibitor, Wnt signaling pathway inhibitor, or TGF ⁇ signaling pathway inhibitor added in step (2) may be different from the substance used in the previous step, but is preferably the same.
- the concentration and type of additives can be adjusted as appropriate.
- the timing of addition of these substances may be simultaneous with the start of step (2) or may be different.
- Step (b) the cell population obtained in step (2) is cultured under conditions in which a substance inhibiting the BMP signaling pathway is added. If the cells are cultured in suspension in step (2), the cell aggregates formed may be cultured in suspension in step (b). If the cells are cultured in adhesion in step (2), the cells may be cultured in adhesion in step (b).
- the BMP signaling pathway inhibitor is not limited as long as it can suppress signaling induced by BMP family proteins. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit BMP processing and extracellular secretion, substances that act directly on BMP (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes encoding BMP (e.g., antisense oligonucleotides, siRNA, etc.), substances that inhibit the binding of BMP receptors to BMP, and substances that inhibit physiological activity resulting from signaling by BMP receptors.
- substances that inhibit BMP processing and extracellular secretion e.g., proteins, antibodies, aptamers, etc.
- substances that suppress the expression of genes encoding BMP e.g., antisense oligonucleotides, siRNA, etc.
- substances that inhibit the binding of BMP receptors to BMP e.g., antisense oligonucle
- type I BMP receptors and type II BMP receptors There are type I BMP receptors and type II BMP receptors, and BMPR1A, BMPR1B, and ACVR are known as type I BMP receptors, while TGF-beta R-II, ActR-II, ActR-IIB, BMPR2, and MISR-II are known as type II BMP receptors.
- BMP signaling pathway inhibitors include, for example, Noggin, Chordin, Follistatin, Gremlin, Inhibin, Twisted Gastrulation, Coco, and secreted proteins belonging to the DAN family. Since a substance acting on the BMP signaling pathway is added to the culture medium in step (2) above, from the viewpoint of more effectively inhibiting the subsequent BMP signaling pathway, it is preferable that the BMP signaling pathway inhibitor in step (b) includes a substance that inhibits the signaling pathway subsequent to the secretion of BMP outside the cell, such as a substance that inhibits the binding between BMP receptors and BMP, or a substance that inhibits physiological activity resulting from signaling by BMP receptors, and more preferably includes an inhibitor of type I BMP receptor.
- BMP signaling pathway inhibitors include inhibitors of type I BMP receptors.
- Compounds having the above-mentioned activity include, for example, K02288 (3-[(6-Amino-5-(3,4,5-trimethoxyphenyl)-3-pyridinyl]phenol), Dorsomorphin (6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)pyrazolo[1,5-a]pyrimid), and ine), LDN-193189 (4-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride), LDN-212854 (5 -[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-
- the BMP signaling pathway inhibitor is preferably a type I BMP receptor inhibitor, more preferably at least one selected from the group consisting of K02288, Dorsomorphin, LDN-193189, LDN-212854, LDN-214117, ML347, DMH1 and DMH2, and even more preferably K02288 to LDN-193189.
- the concentration of the BMP signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of the efficiency of pituitary tissue formation, when K02288 is used as the BMP signaling pathway inhibitor in step (b), it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 50 ⁇ M, more preferably about 100 nM to about 50 ⁇ M, and even more preferably about 500 nM to about 25 ⁇ M.
- LDN-193189 When LDN-193189 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 10 ⁇ M, more preferably about 25 nM to about 1 ⁇ M, and even more preferably about 100 nM to about 500 nM.
- LDN-212854 When LDN-212854 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 10 ⁇ M, more preferably about 25 nM to about 5 ⁇ M, and even more preferably about 250 nM to about 3 ⁇ M.
- ML347 When ML347 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 50 ⁇ M, more preferably about 100 nM to about 50 ⁇ M, and even more preferably about 1 ⁇ M to about 25 ⁇ M.
- DMH2 When used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 ⁇ M, preferably about 10 nM to about 10 ⁇ M, more preferably about 25 nM to about 5 ⁇ M, and even more preferably about 250 nM to about 3 ⁇ M.
- K02288 when using a BMP signaling pathway inhibitor other than K02288, it is desirable to use it at a concentration that exhibits the same BMP signaling pathway inhibitory activity as K02288 at the above concentration.
- the timing for starting step (b) after carrying out step (2) can be set appropriately.
- the timing for starting step (b) is usually at least 8 hours and within 15 days, preferably at least 10 hours and within 12 days, more preferably at least 12 hours and within 9 days, even more preferably at least 14 hours and within 8 days, and most preferably at least 16 hours and within 7 days after starting step (2).
- the cell population may be treated with corticosteroids by adding corticosteroids to the medium.
- Treatment with corticosteroids promotes differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells other than ACTH-producing cells (i.e., GH-producing cells, PRL-producing cells, TSH-producing cells, LH-producing cells, FSH-producing cells, etc.).
- corticosteroids examples include, but are not limited to, natural glucocorticoids such as hydrocortisone, cortisone acetate, and fludrocortisone acetate; and artificially synthesized glucocorticoids such as dexamethasone, betamethasone, prednisolone, methylprednisolone, and triamcinolone.
- natural glucocorticoids such as hydrocortisone, cortisone acetate, and fludrocortisone acetate
- artificially synthesized glucocorticoids such as dexamethasone, betamethasone, prednisolone, methylprednisolone, and triamcinolone.
- the concentration of the adrenal cortical hormones in the medium is not particularly limited as long as it can promote differentiation from the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells), and can be appropriately set depending on the type of adrenal cortical hormone.
- hydrocortisone it is usually 100 ng/ml or more, preferably 1 ⁇ g/ml or more.
- hydrocortisone concentration it is usually 1000 ⁇ g/ml or less, preferably 100 ⁇ g/ml or less.
- the hydrocortisone concentration in the medium is usually about 100 ng/ml to about 1000 ⁇ g/ml, preferably about 1 to about 100 ⁇ g/ml.
- its concentration in the medium can be about 1/25 of that of hydrocortisone.
- the timing of adding the corticosteroids to the medium is not particularly limited as long as it can promote differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells).
- the corticosteroids may be added to the medium from the start of the second step, or the corticosteroids may be added to the medium after a certain period of culture in a medium without corticosteroids after the start of the second step.
- the corticosteroids are added to the medium when the appearance of ACTH-producing cells is confirmed in the cell population after the start of the second step.
- the cell aggregate is cultured in a medium without corticosteroids, and after the appearance of ACTH-producing cells is confirmed, the second step and the subsequent steps are continued in a medium containing corticosteroids.
- the appearance of ACTH-producing cells can be confirmed by immunohistological staining using an antibody against ACTH.
- the appearance of ACTH-producing cells can generally be expected 30 days or more after the start of the first step, so in one embodiment, corticosteroids are added to the medium 30 days or more after the start of the first step.
- the period for treating the cell aggregate with the corticosteroids is not particularly limited as long as it is possible to promote differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells), but the cell aggregate is usually treated with the corticosteroids until promotion of differentiation into pituitary hormone-producing cells (excluding ACTH-producing cells) is confirmed in the corticosteroid-treated group compared to the corticosteroid-untreated group.
- the treatment period is usually 7 days or more, preferably 12 days or more.
- the upper limit of the treatment period is not particularly limited, but the corticosteroids may be removed from the medium at the stage when promotion of differentiation into pituitary hormone-producing cells (excluding ACTH-producing cells) is confirmed in the corticosteroid-treated group compared to the corticosteroid-untreated group.
- step (2) it is also preferable to carry out step (2) and the subsequent steps in the presence of a substance that acts on the retinoic acid signaling pathway.
- substances that act on the retinoic acid signaling pathway include substances that bind to retinoic acid receptors (RAR) or retinoid X receptors (RXR) and activate downstream transcription.
- Examples of compounds that have the above-mentioned effects include all-trans retinoic acid, isotretinoin, 9-cis retinoic acid, TTNPB (4-[(E)-2-[(5,5,8,8-Tetramethyl-5,6,7,8-tetrahydronaphthalene)-2-yl]-1-propenyl]benzoic acid), Ch55 (4-[(E)-3-(3,5 -di-tert-butylphenyl)-3-oxo-1-propenyl]benzoic acid), EC19(3-[2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)e thynyl]benzoic acid), EC23(4-[2-(5,6,7,8-Tetrahydro-5,5,8 , 8-tetramethyl-2-naphthalenyl)ethynyl]-benzoicacid), Fenretinide (4-
- the retinoic acid pathway acting substance in step (2) and the subsequent steps preferably includes all-trans retinoic acid or EC23.
- the concentration of the retinoic acid pathway acting substance in the medium is not particularly limited as long as it is within a range in which the above-mentioned effects can be achieved, but when EC23 is used as the retinoic acid pathway acting substance, the concentration of EC23 is, for example, about 10 pM to about 30 ⁇ M, preferably about 100 pM to about 20 ⁇ M, more preferably about 10 nM to about 10 ⁇ M, and even more preferably about 100 nM to about 5 ⁇ M.
- a retinoic acid pathway acting substance other than EC23 it is desirable to use it at a concentration that exhibits retinoic acid pathway acting activity equivalent to that of EC23 at the above-mentioned concentration.
- the Notch signaling pathway refers to a signaling pathway that is activated by direct interaction between the Notch protein, which is a receptor expressed on the cell membrane, and the Notch ligand (Delta, Jagged, etc.) expressed on the membrane of an adjacent cell.
- the Notch protein is processed in stages, and the intracellular domain cut out on the membrane is transported into the nucleus to control the expression of downstream genes.
- the Notch signaling pathway inhibitor is not particularly limited as long as it can suppress signaling mediated by Notch. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include functionally defective Notch receptors and ligands, substances that inhibit Notch processing (S1 cleavage), substances that inhibit glycosylation of Notch and Notch ligands, substances that inhibit cell membrane translocation, substances that inhibit processing (S2 cleavage, S3 cleavage) of the intracellular domain of Notch (NICD) (gamma secretase inhibitors), substances that degrade NICD, and substances that inhibit NICD-dependent transcription.
- Notch gamma secretase inhibitors
- Notch signaling pathway inhibitors Compounds well known to those skilled in the art can also be used as Notch signaling pathway inhibitors.
- Examples of compounds having activity as Notch signaling pathway inhibitors include DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester), DBZ ((2S)-2-[[2-(3,5-difluorophenyl)acetyl]amino]-N-[( 7S)-5-methyl-6-oxo-7H-benzo[d][1]benzazepin-7-yl]propanamide), MDL28170 (benzyl N-[(2S)-3-methyl-1-oxo-1-[[(2S)-1-o xo-3-phenylpropan-2-yl]amino]butan-2-yl]carbamate), FLI- 06 (cyclohexyl 2,7,7-trimethyl
- the concentration of the Notch signaling pathway inhibitor in the medium is not particularly limited as long as it is within a range in which the above-mentioned effects can be achieved, but for example, when DAPT is used as the Notch signaling pathway inhibitor, the concentration of DAPT is, for example, about 100 pM to about 50 ⁇ M, preferably about 1 nM to about 30 ⁇ M, more preferably about 100 nM to about 20 ⁇ M, and even more preferably about 1 ⁇ M to about 10 ⁇ M.
- a Notch signaling pathway inhibitor other than DAPT it is desirable to use it at a concentration that exhibits Notch signaling pathway inhibitory activity equivalent to that of DAPT at the above-mentioned concentration.
- Step (3) the cell population cultured in step (2) or step (b) is cultured under conditions in which a substance acting on the Sonic Hedgehog signaling (Shh) pathway is not added.
- the third step of culturing the cell population obtained in the second step in the absence of a substance acting on the Shh signaling pathway to obtain a cell population containing pituitary tissue is called step (3)
- the third step of culturing the cell population obtained in step b in the absence of a substance acting on the Shh signaling pathway to obtain a cell population containing pituitary tissue is called step (3').
- the formed cell aggregates may be continuously cultured in suspension in the third step.
- the cells When the cells are cultured in adhesion in step (2) or step (b), the cells may be continuously cultured in adhesion in the third step. After the cells are cultured in suspension in step (2) or step (b), they may be cultured in adhesion in the third step.
- the medium used in the third step is not particularly limited as long as it does not contain a substance acting on the Shh signaling pathway.
- the condition of not adding a substance acting on the Shh signaling pathway in this step refers to a condition in which a substance acting on the Shh signaling pathway is not intentionally added to the culture environment of the cell population, and also includes a case in which a substance acting on the Shh signaling pathway is unintentionally included in the culture environment due to autocretion by the cell population, etc.
- Examples of the medium used in the third step include the medium listed in the first step and gfCDM medium containing 10% to 20% KSR.
- the third and subsequent steps may include a step of embedding the cell aggregates in a gel and culturing them in order to promote the survival and differentiation/maturation of the cells that make up the pituitary gland.
- gels include gels made of agarose, methylcellulose, collagen, matrigel, etc., and it is preferable to use matrigel.
- the cell aggregates may be embedded as they are, or the cells may be seeded in the gel after being dispersed and isolated. Specific cell types such as basal cells may be separated using a cell sorter or the like and then seeded.
- co-culture with cells other than the pituitary gland such as fibroblasts, mesenchymal cells, and vascular cells, may also be performed.
- the gel-embedded culture described above may be performed by referring to, for example, Nature 501, 373-379 (2013), Nature, 499, 481-484 (2013), Nat Protoc 14, 518-540 (2019), Genes 2020, 11, 603, etc.
- a culture method in which the cells are physically shaken in order to improve the supply of nutrients and oxygen to the cells and to improve material exchange.
- Such culture methods include methods other than stationary culture, such as shaking culture, rotary culture, and agitation culture.
- the means for carrying out shaking culture, rotary culture, agitation culture, and the like are not particularly limited, but for example, shaking culture, rotary culture, agitation culture, and the like can be carried out by placing the culture equipment in which the cells are cultured on a rotator, shaker, or the like, or placing the cells in an environment in which a stirrer or the like is rotating.
- the shaking speed range can be set, for example, to 5 to 60 rpm, preferably 5 to 40 rpm, and more preferably 5 to 20 rpm.
- the shaking speed range can be set, for example, in the range of 15 to 60 rpm, preferably 15 to 50 rpm, and more preferably 15 to 45 rpm.
- the shaking speed range can be set, for example, in the range of 5 to 50 rpm, preferably 5 to 40 rpm, and more preferably 5 to 30 rpm.
- a spinner flask e.g., 3152, manufactured by Corning
- culture can be performed at a rotation speed at which the cell aggregates do not settle with the naked eye.
- Culture can also be performed using a three-dimensional rotary suspension culture device (e.g., CellPet CUBE, manufactured by J-Tech; Clinostar, manufactured by Celvivo). From the viewpoint of suppressing physical damage to the cells, such as friction, it is also preferable to subject the cell aggregates embedded in the gel to shaking culture, rotation culture, or stirring culture.
- High-oxygen conditions during the culture process can be achieved, for example, by connecting an oxygen tank to the incubator in which the cells are cultured and artificially supplying oxygen.
- the oxygen concentration for this purpose is usually 25% to 80%, and more preferably 30% to 60%.
- culture equipment with high gas exchange efficiency can be used to increase the amount of oxygen supplied to the medium in which the cell aggregates are cultured.
- Examples of such culture equipment include cell culture dishes, Lumox dishes (manufactured by Sarstedt Co., Ltd.) with a gas-permeable film on the bottom of the plate, and VECELL 96-well plates (manufactured by Vecel Co., Ltd.). It is also preferable to use this in combination with the culture under high oxygen concentration conditions described above.
- a cell protective agent may be added to the medium from the viewpoint of maintaining the structure of the non-neuroepithelial tissue in the cell aggregate.
- cell protective agents used in the third and subsequent steps include the above-mentioned FGF signaling pathway active substances, heparin, ROCK inhibitors, basement membrane preparations, myosin inhibitors, polyamines, ISR inhibitors, caspase inhibitors, serum, or serum substitutes.
- myosin inhibitors include blebbistatin, which is an inhibitor of nonmuscle myosin II ATPase, and ML-7, ML-9, W-7, MLCK inhibitor peptide 18, and derivatives thereof, which are inhibitors of myosin light chain kinase (MLCK).
- the cell protective agent added may be different from that added in the first step, but is preferably the same.
- a preferred cell protective agent is a ROCK inhibitor.
- the ROCK inhibitor Y-27632 when added as a cell protective agent, it is usually added to the culture environment at a concentration of about 10 nM to about 10 mM, preferably about 100 nM to about 1 mM, and more preferably about 1 ⁇ M to about 100 ⁇ M.
- the ROCK inhibitor Chroman 1 when added, it is usually added to the culture environment at a concentration of about 10 pM to about 1 mM, preferably about 100 pM to about 100 ⁇ M, and more preferably about 1 nM to about 10 ⁇ M.
- nonmuscle myosin II ATPase inhibitor Blebbistatin When added as a cell protective agent, it is usually added to the culture environment at a concentration of about 10 nM to about 10 mM, preferably about 100 nM to about 1 mM, and more preferably about 1 ⁇ M to about 100 ⁇ M.
- a substance other than a cytoprotectant that has the effect of maintaining the structure of non-neuroepithelial tissue can also be added.
- examples of such substances include substances that promote cell adhesion, substances that promote the synthesis of basement membrane components, and substances that inhibit the decomposition of basement membrane components.
- the substance that promotes cell adhesion may promote cell-cell adhesion, cell-basement membrane adhesion, or cell-culture equipment adhesion, or may promote the production of factors involved in cell adhesion.
- substances that promote cell adhesion include adhesamine, adhesamine-RGDS derivatives, pyrintegrin, biotin tripeptide-1, acetyl tetrapeptide-3, RGDS peptide, and derivatives thereof.
- substances that promote the synthesis of basement membrane components include ascorbic acid derivatives.
- ascorbic acid derivatives include sodium ascorbyl phosphate, magnesium ascorbyl phosphate, ascorbyl 2-glucoside, 3-O-ethyl ascorbic acid, ascorbyl tetrahexyldecanoate, ascorbyl palmitate, ascorbyl stearate, ascorbyl 2-phosphate-6 palmitate, and glyceryl octyl ascorbic acid.
- substances that inhibit the decomposition of basement membrane components include inhibitors of matrix metalloproteases and serine proteases.
- ascorbic acid 2-phosphate which is a type of ascorbic acid derivative that promotes the synthesis of basement membrane components
- it is usually added to the culture environment at a concentration of 10 ⁇ g/ml or more and 1000 ⁇ g/ml or less, preferably 30 ⁇ g/ml or more and 500 ⁇ g/ml or less, and more preferably 50 ⁇ g/ml or more and 300 ⁇ g/ml or less.
- adding other ascorbic acid or ascorbic acid derivatives, etc. add them so that the molar equivalents are approximately the same as the above concentrations.
- a substance that has the effect of reducing oxidative stress examples include antioxidants, substances with free radical scavenging action, NADPH oxidase inhibitors, cyclooxygenase inhibitors, lipoxygenase (LOX) inhibitors, superoxide dismutase (SOD)-like substances, Nrf2 activators, etc.
- substances having the above-mentioned activity include ascorbic acid, N-acetyl-L-cysteine, ( ⁇ )- ⁇ -tocopherol acetate, apocynin (4'-hydroxy-3'-methoxyacetophenone), nicotinamide, taurine (2-aminoethanesulfonic acid), IM-93 (1-isopropyl-3-(1-methyl-1H-indole-3-yl)-4-(N,N-dimethyl-1,3-pr Celastrol (3-Hydroxy-24-nor-2-oxo-1 (10), 3,5, 7-friedelatetraen-29-oic Acid; Tripterin), Ebselen (2-Phenyl-1, 2-benzisoselenazol-3 (2H)-one), (-)-Epigallocatechin Gallate ((2R,3R)-2-(3,4,5-Trihydroxyphenyl)-3,4-dihydro-1[2H]-benzopyr
- Reagents already prepared for cell culture e.g., antioxidant supplements, Sigma Aldrich, A1345
- the substance having the effect of reducing oxidative stress used in the present invention preferably includes at least one selected from the group consisting of ascorbic acid, N-acetyl-L-cysteine and their derivatives.
- Ascorbic acid can be added to the medium, for example, in the form of its derivative ascorbic acid 2-phosphate, at a concentration of about 1 nM to about 1 M, preferably about 10 nM to about 100 mM, more preferably about 100 nM to about 10 mM, and even more preferably about 1 ⁇ M to about 3 mM, and N-acetyl-L-cysteine can be added to the medium at a concentration of, for example, about 1 nM to about 1 M, preferably about 10 nM to about 100 mM, more preferably about 100 nM to about 10 mM, and even more preferably about 1 ⁇ M to about 5 mM.
- the stress-responsive MAP kinase pathway (stress-activated protein kinase: SAPK) is one of the main intracellular signaling mechanisms in response to stress.
- inhibitors of the stress-responsive MAP kinase pathway include MAP3K inhibitors, MAP2K inhibitors, ASK inhibitors, MEK inhibitors, Akt inhibitors, Rho family kinase inhibitors, JNK inhibitors, p38 inhibitors, MSK inhibitors, STAT inhibitors, NF- ⁇ B inhibitors, and CAMK inhibitors.
- MEK inhibitors include Selumetinib (AZD6244, 6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide), Mirdametinib (PD0325901, N-[(2R)-2,3-dihydroxypropoxy]-3,4-difluoro-2-(2-fluoro-4-iodoanilino)benzamide), Trametinib (GSK1120212, N -[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide), U012 6(1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene), PD
- Examples of p38 inhibitors include SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine), Doramapimod (BIRB 796, 1-[5-tert-butyl-2-(4-methylphenyl)pyrazo l-3-yl]-3-[4-(2-morpholin-4-ylethoxy)naphthalen-1-yl]urea), SB202190 (FHPI,4-[4-(4-fluorophenyl)-5-pyridin-4-yl-1H-imidaz ol-2-yl]phenol), Ralimetinib dimesylate (5-[2-t ert-butyl-4-(4-fluorophenyl)-1H-imidazol-5-yl]-3-(2,2-dimethylpropyl)imidazo[4,5-b]pyridin-2-amine; methanesulfonic
- JNK inhibitors include those described in the first step.
- the substance used in the present invention that inhibits the intracellular signal transduction mechanism in response to stress is preferably one or more selected from the group consisting of MEK inhibitors, p38 inhibitors, and JNK inhibitors.
- SB203580 When SB203580 is used as the p38 inhibitor, it can be added to the medium at a concentration of usually about 1 nM to about 1 mM, preferably about 10 nM to about 100 ⁇ M, more preferably about 100 nM to about 10 ⁇ M, and even more preferably about 500 nM to about 5 ⁇ M.
- PD0325901 When PD0325901 is used as the MEK inhibitor, it can be added to the medium at a concentration of usually about 1 nM to about 1 mM, preferably about 10 nM to about 100 ⁇ M, more preferably about 100 nM to about 10 ⁇ M, and even more preferably about 500 nM to about 5 ⁇ M.
- JNK-IN-8 When JNK-IN-8 is used as the JNK inhibitor, it can be added to the medium at a concentration similar to that described in the first step.
- the cell populations comprising pituitary hormone-producing cells of the present invention may be cell aggregates comprising pituitary hormone-producing cells.
- the cell aggregate may be a cell aggregate containing pituitary hormone-producing cells throughout, and in one embodiment, containing them evenly.
- the cell aggregate may be a cell aggregate containing both a partial structure containing many pituitary hormone-producing cells (also referred to as pituitary tissue in this specification) and a partial structure containing a small amount of pituitary hormone-producing cells or no pituitary hormone-producing cells (also referred to as a cell population containing pituitary tissue and tissue other than the pituitary gland).
- the therapeutic agent of the present invention may contain any of the cell aggregates described above.
- the cell population containing pituitary tissue contained in the therapeutic agent of the present invention may be a cell aggregate containing pituitary tissue and not containing any tissue other than the pituitary gland.
- the cells contained in the cell aggregate are as described in 1 above.
- the cell population comprising pituitary tissue contained in the therapeutic agent of the present invention may be a cell population comprising both pituitary tissue and tissue other than the pituitary gland, and the cell population may be a cell population comprising 1) nervous cells or nervous tissue and 2) pituitary tissue.
- the cell population can be obtained by the production method 1 described above.
- the 1) nervous system cells or nervous tissue in the cell population of the present invention obtained by the above-mentioned production method 1 are cells or tissues of the central nervous system, or precursor tissues thereof.
- the cells or tissues of the central nervous system include the diencephalon (specifically the hypothalamus) and cells (including precursor cells) contained in these tissues, more preferably the hypothalamus or its precursor tissues, or cells contained therein, and even more preferably the hypothalamus having a ventricle-like structure in the tissue, or its precursor tissues, or cells contained therein.
- the nervous system cells or nervous tissue is, for example, N-Cadherin-positive neuroepithelial tissue.
- pituitary tissue in the cell population of the present invention obtained by the above-mentioned Production Method 1 is formed continuously with non-neuroepithelial tissue, and it is further preferred that the non-neuroepithelial tissue and pituitary tissue cover 1) nervous cells or nervous tissue.
- the non-neural epithelial tissue obtained by the above-mentioned production method 1 is oral epithelium or a precursor tissue thereof.
- the pituitary tissue preferably contains pituitary hormone-producing cells or their precursor cells, that is, pituitary precursor cells, and may contain pituitary stem cells or folliculostellate cells, or may contain all of pituitary hormone-producing cells, pituitary precursor cells, pituitary stem cells, and folliculostellate cells.
- a pituitary niche is formed in the pituitary tissue, and it is also preferable that the pituitary niche is an MCL niche-like structure around the residual cavity remaining between the anterior and intermediate lobes of the pituitary gland, and it is also preferable that the pituitary niche is a parenchymal layer niche-like structure, and it is even more preferable that it contains both an MCL niche-like structure and a parenchymal layer niche-like structure.
- Mesenchymal cells contained in the cell population of the present invention obtained by the above-mentioned Production Method 1 express at least one mesenchymal cell marker selected from the group consisting of, for example, nestin, vimentin, cadherin-11, laminin, CD44, CD90, and CD105.
- Non-neuroepithelial tissues that can be included in the present invention express at least one non-neuroepithelial tissue marker selected from the group consisting of, for example, cytokeratin, E-Cadherin, and EpCAM.
- Pituitary stem cells that may be contained in the cell population contained in the therapeutic agent of the present invention express at least one pituitary stem cell marker selected from the group consisting of, for example, Sox2, Sox9, E-Cadherin, Nestin, S100 ⁇ , GFR ⁇ 2, Prop1, CD133, ⁇ -Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE.
- a preferred embodiment of the cell population of the present invention contains pituitary stem cells that are positive for a pituitary stem cell marker (e.g., CXADR).
- the proportion of pituitary stem cells in the cell population may be 1% or more, preferably 3% or more or 5% or more.
- the pituitary tissue may be collected from the cell population containing pituitary tissue obtained by the above-mentioned "2.
- Method for Producing a Cell Population Containing Pituitary Tissue includes the following steps (1), (2) and (4).
- step a (1) a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor to obtain a cell population; (2) a second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue; (4) A fourth step of recovering pituitary tissue from the cell population obtained in the second step.
- the first and second steps can be carried out in the same manner as the first and second steps in "2. Method for producing a cell population containing pituitary tissue" above. If desired, step a may be carried out before step 1.
- step 3 may be carried out between steps 2 and 4.
- the pituitary tissue can be recovered by a method such as physically peeling off the pituitary tissue using a needle under a microscope.
- the formed cell population is a three-dimensional tissue such as a cell mass
- the pituitary tissue formed on the outside of the cell mass (Rathke's pouch portion) is peeled off and recovered using tweezers under a microscope.
- the pituitary tissue can be identified as a semi-transparent thin epithelium on the surface of the obtained cell mass, as described in Nature communications, 2016, 7.
- freezing and thawing can also be used as a method for recovering pituitary tissue from a cell population (cell mass).
- the outer pituitary tissue is peeled off from the cell mass without physical treatment by freezing and thawing a cell mass having pituitary tissue on the outside and mesenchymal nerve or neuroepithelial tissue on the inside.
- the pituitary hormone-producing cells, cell populations containing pituitary hormone-producing cells, or cell aggregates containing pituitary hormone-producing cells, cell populations containing pituitary tissue, or pituitary tissue recovered from cell populations containing pituitary tissue, contained in the therapeutic agent of the present invention can be produced by the production method described above.
- Method for producing purified product In order to obtain a cell population (purified product) that does not contain unintended cells (cells other than pituitary hormone-producing cells) or has a reduced proportion of unintended cells, as described in the above "1.
- Therapeutic drug and cell transplantation of the present invention the following steps may be performed. Specifically, after the second step (step (2)) described in the above "4. Method for producing a cell population containing pituitary tissue 1", the following step may be performed: selecting and recovering pituitary hormone-producing cells from the cell population containing pituitary hormone-producing cells obtained in the second step. This step may be performed with reference to a method known per se (e.g., International Publication No. WO 2023/054395).
- cells expressing EpCAM are separated from the cell population containing pituitary tissue obtained in the second step.
- a step of dispersing the cell population containing pituitary tissue obtained in the second step may be carried out prior to carrying out.
- the obtained cell population containing pituitary tissue may first be pretreated with a ROCK inhibitor.
- a ROCK inhibitor In order to suppress cell death induced by the subsequent dispersion of the cell population containing pituitary tissue, it is preferable to add the ROCK inhibitor before the start of the pretreatment.
- the ROCK inhibitor is added, for example, at least 24 hours, at least 12 hours, at least 6 hours, at least 3 hours, at least 2 hours, or at least 1 hour before the start of the treatment.
- ROCK inhibitors include Y-27632 ((R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide, dihydrochloride).
- the concentration of the ROCK inhibitor used in this treatment is a concentration that can suppress cell death induced by the subsequent dispersion of the cell population containing the pituitary tissue.
- concentration of the ROCK inhibitor may be varied during the period of addition, for example, the concentration may be reduced by half in the latter half of the period.
- the medium used in step (2) can be used as the medium for carrying out this pretreatment. Note that if the ROCK inhibitor has already been added to the medium at the desired concentration, there is no need to carry out this pretreatment step.
- the cell population containing the pituitary tissue that has been pretreated as described above is dispersed by enzyme treatment.
- the cell population that has been pretreated as described above is first transferred to an incubator containing PBS and washed with the same medium.
- the enzyme for dispersion is not particularly limited as long as it can disperse the cells, but examples include enzymes such as papain, EDTA, trypsin, collagenase (collagenase types I to VII), metalloprotease, hyaluronidase, elastase, dispase, deoxyribonuclease, and mixtures thereof.
- a preferred enzyme is papain.
- the conditions for the enzyme treatment (temperature, time, etc.) can be appropriately set depending on the enzyme used, etc.
- a step of physically cutting the cell population containing the pituitary tissue e.g., scalpel, scissors, etc.
- a step of physically cutting the cell population e.g., scalpel, scissors, etc.
- a step of physically cutting the cell population e.g., scalpel, scissors, etc.
- a step of physically cutting the cell population may be performed before the treatment.
- the floating cells may be collected and the enzyme treatment may be carried out again. This may be repeated multiple times.
- the enzyme treatment may be carried out using the enzymes described above, and preferred enzymes include EDTA; trypsin, collagenase (collagenase types I to VIII), metalloprotease, hyaluronidase, elastase, dispase, deoxyribonuclease, and mixtures thereof.
- Preferred enzymes include collagenase, more preferably collagenase type I.
- the conditions for the enzyme treatment may be set appropriately depending on the enzyme used.
- the enzyme treatment can be carried out with the enzymes mentioned above, and preferred enzymes include EDTA; trypsin, and more preferably EDTA; trypsin and deoxyribonuclease. Commercially available products such as TrypLE (Invitrogen) may be used in place of EDTA; trypsin.
- the enzyme treatment conditions can be appropriately set depending on the enzymes used.
- a single-cell suspension can be prepared by the above series of enzyme treatments. When preparing a single-cell suspension, dead cells may be removed by a method known per se.
- cells expressing EpCAM are separated from the dispersed cell population obtained as described above.
- Methods for separating desired cells expressing EpCAM from dispersed cell populations include methods using flow cytometry or mass cytometry, magnetic cell separation methods, etc., and these methods can be performed using methods known per se.
- cells expressing EpCAM can be separated by a method including a step of contacting the cells with a substance (e.g., antibody, etc.) that specifically binds to EpCAM molecules.
- a substance e.g., antibody, etc.
- the above-mentioned substances include those that have a detectable label (e.g., GFP, PE) attached thereto, and those that have no label attached thereto.
- the separation can be achieved by further using a substance that has a detectable label attached thereto that directly or indirectly recognizes the substance.
- a substance that has a detectable label attached thereto that directly or indirectly recognizes the substance.
- the substance is an antibody
- a fluorescent dye, a metal isotope, or beads (e.g., magnetic beads) can be directly or indirectly supported on the antibody, thereby labeling a marker on the cell surface, and the cells can be separated based on the label.
- only one type of antibody may be used, or two or more types of antibodies may be used.
- the above-mentioned step (3) may be carried out. The timing of carrying out this step is not limited to after the second step (step (2)).
- step (b) may be carried out after step (2) and then this step may be carried out.
- step (b) may be carried out after step (2), and then step (3) may be carried out, and then this step may be carried out.
- step (3) may be carried out after carrying out this step.
- the pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be administered in a therapeutically effective amount for a disease based on a pituitary disorder for which cell transplantation therapy is desired, and may vary depending on factors such as the age, weight, and severity of the disease of the transplantation subject, and are not particularly limited, and may be, for example, 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells, preferably 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells, and more preferably 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells.
- the therapeutic agent of the present invention contains pituitary hormone-producing cells as a cell population containing pituitary hormone-producing cells, a cell aggregate containing pituitary tissue, or pituitary tissue recovered from a cell population containing pituitary tissue, it is sufficient to administer a therapeutically effective amount for a disease due to a pituitary disorder for which cell transplantation therapy is desired, and this can be varied appropriately depending on factors such as the age, weight, and severity of the disease of the transplantation subject.
- the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention are used in cell transplantation therapy, it is desirable to use cells derived from iPS cells established from somatic cells with the same or substantially the same HLA genotype of the recipient individual, from the viewpoint of preventing rejection reactions.
- substantially the same means that the HLA genotype matches the transplanted cells to such an extent that the immune response can be suppressed by an immunosuppressant, for example, somatic cells with HLA types matching the three loci of HLA-A, HLA-B, and HLA-DR, or four loci including HLA-C, or six loci including HLA-DP and HLA-DQ. If sufficient cells cannot be obtained due to age, constitution, etc., they can also be transplanted in a state where they are embedded in a capsule or porous container made of polyethylene glycol or silicone to avoid rejection reactions.
- the therapeutic agent of the present invention can be prepared by mixing with a medicamentously acceptable aqueous liquid, for example.
- a method for producing a therapeutic agent for a disease due to a disorder of the pituitary gland which contains pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, and includes a step of formulating the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention.
- Such a production method may include a step of preparing the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention. It may also include a step of preserving the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention.
- the pharma- ceutically acceptable aqueous liquid that may be contained in the therapeutic agent of the present invention may contain, for example, an appropriate selection of a buffer, an isotonicity agent, a pH adjuster, an antioxidant, a chelating agent, etc., within a range that does not affect the viability and physiological activity of the transplanted pituitary hormone-producing cells.
- a buffer include phosphate buffer, borate buffer, citrate buffer, tartrate buffer, acetate buffer, amino acid, epsilon-aminocaproic acid, etc.
- isotonicity agent examples include sugars such as D-sorbitol, D-glucose, D-mannitol, etc., polyhydric alcohols such as glycerin, propylene glycol, etc., salts such as sodium chloride, boric acid, etc.
- Chelating agents include sodium edetate and citric acid.
- pH adjusters include sodium hydroxide, potassium hydroxide, sodium carbonate, sodium bicarbonate, boric acid or a salt thereof (borax), hydrochloric acid, citric acid or a salt thereof (sodium citrate, sodium dihydrogen citrate, etc.), phosphoric acid or a salt thereof (disodium hydrogen phosphate, potassium dihydrogen phosphate, etc.), acetic acid or a salt thereof (sodium acetate, ammonium acetate, etc.), tartaric acid or a salt thereof (sodium tartrate, etc.), and the like.
- antioxidants include ascorbic acid, glutathione, sodium hydrogen sulfite, dry sodium sulfite, sodium pyrosulfite, and tocopherol.
- pharmaceutical acceptable aqueous liquid include aqueous liquids such as physiological saline, isotonic solutions containing glucose and other auxiliary drugs (eg, D-sorbitol, D-mannitol, sodium chloride, etc.).
- the therapeutic agent of the present invention may be compounded with, for example, a soothing agent (eg, benzalkonium chloride, procaine hydrochloride, etc.), a stabilizer (eg, human serum albumin, polyethylene glycol, etc.), a preservative, an antioxidant, and the like.
- a soothing agent eg, benzalkonium chloride, procaine hydrochloride, etc.
- a stabilizer eg, human serum albumin, polyethylene glycol, etc.
- a preservative e.g, benzalkonium chloride, procaine hydrochloride, etc.
- the therapeutic agent of the present invention may be combined with an angiogenesis promoter to form a combination drug (combination drug).
- the angiogenesis promoter is not particularly limited as long as it is medicamentically acceptable, and examples thereof include vascular endothelial growth factor (VEGF), fibroblast growth factor: acidic (aFGF) and basic (bFGF), epidermal growth factor (EGF), granulocyte macrophage-colony stimulating factor (GM-CSF), hepatocyte growth factor (HGF), and sphingosine-1-phosphate (S1P).
- VEGF vascular endothelial growth factor
- aFGF fibroblast growth factor: acidic (aFGF) and basic (bFGF)
- EGF epidermal growth factor
- GM-CSF granulocyte macrophage-colony stimulating factor
- HGF hepatocyte growth factor
- S1P sphingosine-1-phosphate
- the combination drug of the present invention may contain one or more of each formulation.
- the agent When the agent is formulated separately, there is no particular limitation, but the agent may be contained in a biocompatible gel (particularly, a hydrogel, etc.), film, etc., with the intention of sustained release of the angiogenesis promoter, etc.
- the biocompatible gel, film, etc. may be bioabsorbable.
- the above-mentioned gel, film, etc. are not particularly limited, but examples thereof include amniotic membrane, polyvinylidene fluoride (PVDF), polytetrafluoroethylene (PTFE), polyurethane, polypropylene, polyester, vinyl chloride, polycarbonate, acrylic, silicone, MPC (2-methacryloyloxyethyl phosphorylcholine), polylactic acid, polyglycolic acid, lactic acid-glycolic acid copolymer (PLGA), fibrin, gelatin, collagen, collagen sodium alginate, poly(N-isopropyl acrylate), ...
- the gel may be a temperature-responsive gel in which polyacrylamide (PIPAAm) is crosslinked with polyethylene glycol (PEG), hyaluronic acid, glycosaminoglycan, proteoglycan, chondroitin, cellulose, agarose, carboxymethylcellulose, chitin, chitosan, gelatin, atelocollagen, elastin, fibronectin, pronectin, laminin, tenascin, fibroin, entactin, thrombospondin, retronectin, dextrin, trehalose, etc.
- PIPAAm polyacrylamide
- PEG polyethylene glycol
- hyaluronic acid glycosaminoglycan
- glycosaminoglycan proteoglycan
- chondroitin cellulose
- agarose carboxymethylcellulose
- chitin chitosan
- gelatin atelocollagen
- elastin fibronectin
- pronectin laminin
- each formulation can be administered (transplanted) to a subject simultaneously or at different times, via the same route or via different routes.
- the administration (transplantation) amount and number of administrations (transplants) of each formulation can be appropriately determined depending on various conditions such as the type of angiogenesis promoter, the symptoms, age, weight, and drug tolerance of the subject to be administered.
- One example is a method in which a gelatin hydrogel sustained release device containing basic fibroblast growth factor (FGF) is applied to the transplant site in advance, and the therapeutic agent of the present invention is transplanted after a certain period of time has passed.
- FGF basic fibroblast growth factor
- the therapeutic agent of the present invention is provided in a frozen state under conditions normally used for cryopreservation of cells, and can be thawed when needed.
- it may further contain serum or a substitute thereof, an organic solvent (e.g., DMSO), etc.
- an organic solvent e.g., DMSO
- the concentration of serum or a substitute thereof is not particularly limited, but may be about 1 to about 30% (v/v), preferably about 5 to about 20% (v/v).
- the concentration of the organic solvent is not particularly limited, but may be 0 to about 50% (v/v), preferably about 5 to about 20% (v/v).
- cell populations such as pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be transplanted into the subcutaneous tissue and/or muscle tissue of a subject to treat diseases caused by disorders of the pituitary gland that contain pituitary hormone-producing cells. Therefore, the present invention also provides a treatment method characterized by treating diseases caused by disorders of the pituitary gland by administering a therapeutically effective amount of pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells to the subcutaneous tissue and/or muscle tissue.
- the pituitary hormone-producing cells, cell populations containing pituitary hormone-producing cells, cell aggregates containing pituitary hormone-producing cells, cell populations containing pituitary tissue, or pituitary tissue recovered from cell populations containing pituitary tissue contained in the therapeutic agent of the present invention may be produced using pluripotent stem cells whose genes have been manipulated by genome editing as a raw material.
- Genes that are the subject of genome editing include, but are not limited to, genes involved in the differentiation of pituitary tissue by the above-mentioned method for producing pituitary hormone-producing cells, etc. of the present invention, genes involved in the differentiation into unintended cells other than the pituitary gland that are by-produced by the production method, hormone-related genes secreted from the pituitary gland, genes involved in the infection of diseases, etc.
- Model Animals An embodiment of the present invention includes a method for producing a non-human animal model bearing human pituitary tissue, which comprises administering human pituitary hormone-producing cells into the subcutaneous tissue and/or muscle tissue of the non-human animal.
- the non-human animal is not particularly limited as long as it is a mammal other than a human, and specifically includes a mammal used for evaluating the efficacy or safety of a substance, such as a mouse, a rat, a hamster or a guinea pig, a dog, a rabbit, or a lemur, a loris, a tree shrew, or a monkey.
- the human pituitary hormone-producing cells include a cell population containing any of the above-mentioned human pituitary hormone-producing cells.
- the subcutaneous tissue and muscle tissue into which the human pituitary hormone-producing cells are transplanted are as described above in 1.
- the human pituitary hormone-producing cells may be transplanted into a non-human animal in accordance with the Examples of the present specification.
- the model non-human animal of the present invention may be an animal in which part or all of the pituitary gland has been surgically removed.
- the present invention includes a non-human animal model bearing human pituitary tissue obtained by the above-mentioned production method.
- the present invention encompasses a method for evaluating the efficacy or safety of a test substance, which comprises administering the test substance to the non-human animal model bearing human pituitary tissue.
- the method of administering the test substance to the non-human animal is not particularly limited, and the substance can be administered orally or parenterally (including, but not limited to, intravenous administration, subcutaneous administration, intradermal administration, etc.).
- the effect of the test substance on the secretion or in vivo action of a pituitary hormone can be examined by administering the test substance to the non-human animal and measuring the concentration of the pituitary hormone in the non-human animal (e.g., blood concentration).
- the evaluation method of the present invention may comprise the following steps: (i) administering a test substance to the non-human animal model bearing human pituitary gland tissue; (ii) examining the amount of pituitary hormone or activity of pituitary hormone in a non-human animal administered the test substance; and (iii) evaluating the effect of the test substance on the secretion or activity of the pituitary hormone based on the results of (ii).
- the evaluation method of the present invention is a method for screening for a preventive or therapeutic agent for a disease caused by a pituitary gland disorder, and may comprise the following steps: (i) administering a test substance to the non-human animal model bearing human pituitary gland tissue; (ii) examining the amount or activity of the pituitary hormone in the non-human animal administered the test substance; and (iii) selecting the test substance in which the amount or activity of the pituitary hormone is increased/decreased as a candidate for a preventive or therapeutic drug for a disease based on a disorder of the pituitary gland.
- test substances used in the screening method of the present invention include, for example, cell extracts, cell culture supernatants, microbial fermentation products, extracts from marine organisms, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic small molecule compounds, and natural compounds.
- the test substances can also be obtained using any of the many approaches in combinatorial library methods known in the art, including (1) biological libraries, (2) synthetic library methods using deconvolution, (3) "one-bead one-compound” library methods, and (4) synthetic library methods using affinity chromatography selection.
- Example 1 Examination of transplantation sites of pituitary hormone-producing cells ⁇ Materials and methods> (1) Maintenance and differentiation culture of human embryonic stem cells (hESC) (pituitary organoids) hESCs (KhES-1) were obtained from the RIKEN BioResource Research Center and used in accordance with the Japanese government's hESC research guidelines. All experimental protocols and procedures were approved by the Ethics Committee of the Nagoya University graduate School of Medicine (approval ES-001). hESC maintenance and differentiation cultures were cultured under feeder-free conditions according to the method described in Scientific Reports, 4, 3594 (2014).
- hESC human embryonic stem cells
- KhES-1 epithelial organoids
- StemFit medium (AK03, Ajinomoto Co., Inc.) was used as the feeder-free medium, and Laminin511-E8 (Nippi Co., Ltd.) was used as the feeder-free scaffold.
- a specific maintenance culture operation first, subconfluent hESC (KhES-1 strain) was washed with PBS and dispersed into single cells using TrypLE Select (Life Technologies). Then, the human ES cells dispersed into the single cells were seeded on a plastic culture dish coated with Laminin511-E8 and cultured feeder-free in StemFit medium in the presence of Y27632 (ROCK inhibitor, 10 ⁇ M).
- the seeded cell number of the human ES cells dispersed into the single cells was 2.4 ⁇ 10 4.
- the entire medium was replaced with StemFit medium not containing Y27632.
- the entire medium was replaced with StemFit medium not containing Y27632 once every 1 to 2 days, and then the cells were cultured until they became subconfluent (about 60% of the culture area was covered with cells) 7 days after seeding.
- human ES cells (KhES-1 strain) were cultured for 24 hours using StemFit medium, 6 days after seeding, with the addition of SB431542 (5 ⁇ M) and SAG (300 nM) at the same time as changing the medium to StemFit medium.
- the prepared subconfluent human ES cells were treated with cell dispersion liquid using TrypLE Select (Life Technologies), and further dispersed into single cells by pipetting.
- the human ES cells dispersed into the single cells were suspended in 100 ⁇ L of serum-free medium so that the number of cells was 1.0 ⁇ 10 4 per well of a non-cell-adhesive 96-well culture plate (PrimeSurface 96V bottom plate, MS-9096V, Sumitomo Bakelite Co., Ltd.), and the suspension culture was performed under the conditions of 37 ° C. and 5% CO 2 .
- the serum-free medium (gfCDM+KSR) used was a 1:1 mixture of F-12 medium and IMDM medium, to which 5% KSR, 450 ⁇ M 1-monothioglycerol, and 1x chemically defined lipid concentrate had been added.
- Y27632 final concentration 20 ⁇ M
- IWP-2 0.5 ⁇ M
- SB431542 1 ⁇ M
- JNK-IN-8 1 ⁇ M
- SAG 100 nM
- the cell aggregates were transferred to 10 cm suspension culture dishes at 60 cell aggregates per dish, and suspension culture was continued using serum-free medium under conditions of 40% O2 and 5% CO2. From the 29th to 50th day after the start of differentiation induction, serum-free medium supplemented with 10% KSR was used for culture, and from the 50th day after the start of differentiation induction, serum-free medium supplemented with 20% KSR was used for culture.
- the cell aggregates on the 100th day after the start of differentiation induction were fixed with 4% paraformaldehyde and frozen sections were prepared. These frozen sections were immunostained using adrenocorticotropic hormone (ACTH) (anti-ACTH antibody, Fitzgerald, mouse), which is a pituitary hormone-producing cell marker, E-cadherin (anti-E-cadherin antibody, TAKARA, rat), which is an epithelial cell marker, and Lhx3 (anti-Lhx3 antibody, homemade (Non-Patent Documents 1 and 2), rabbit), which is a pituitary precursor marker.
- ACTH adrenocorticotropic hormone
- E-cadherin anti-E-cadherin antibody, TAKARA, rat
- Lhx3 anti-Lhx3 antibody, homemade (Non-Patent Documents 1 and 2), rabbit
- the cell nuclei were stained with DAPI.
- mice Male severe combined immunodeficiency mice (SCID) mice (CB-17/Icr-Hsd-Prkdc scid , Japan SLC Co., Ltd., Shizuoka, Japan) aged 8-9 weeks were subjected to transhypopituitary glandectomy. Mice were anesthetized with an intraperitoneal (i.p.) injection of a mixture of three drugs (medetomidine 0.75 mg/kg, midazolam 4 mg/kg, butorphanol 5 mg/kg) (Kawai S, et al.
- i.p. intraperitoneal
- mice Effect of three types of mixed anesthetic agents alternate to ketamine in mice. Exp Anim. 2011; 60: 481-487), and pituitary tissue was aspirated from the sella turcica via the ear canal using a needle (KN-390, Natsume Seisakusho Co., Ltd., Tokyo, Japan) attached to a 1 ml syringe containing 0.2 ml saline. After the procedure, mice were injected with the medetomidine antagonist atipamezole (0.75 mg/kg, i.p.).
- Plasma ACTH assay was performed using an ACTH ELISA kit (MD bioproducts, Oakdale, MN), and absorbance (ACTH concentration) was read using a Cytation 5 (Biotek, Winooski, VT).
- CRH loading tests were performed 1 week after hypophysectomy and 1/4 weeks after PO transplantation treatment, including sham-operated mice, up to 6 months later (Figure 2A). Mice with plasma ACTH levels below 10 pg/ml after CRH stimulation were classified as hypopituitaric and used as subjects.
- avascular area (AR) transplantation group In the avascular area (AR) transplantation group, a vertical skin incision was made in the left dorsal skin, and five POs were placed. All hypopituitarized mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
- Antigen targets include ACTH (mouse, 1:200, 10C-CR1096M1; Fitzgerald), LHX3 (LIM homeobox protein 3, rabbit, 1:3000, in-house (Non-Patent Documents 1 and 2)), human nucleus (mouse, 1:1000, MAB4383; Millipore), E-cadherin (rat, 1:50, M108; Takara Bio Inc.), and SMA (smooth muscle actin, mouse, 1:200, M0851; DAKO).
- RNA extraction and cDNA synthesis from PO and undifferentiated hESC RNA was extracted from PO and undifferentiated hESC using RNeasy Mini Kit (Qiagen, Hilden, Germany) according to the manufacturer's instructions. RNA quality was assessed using TapeStation 4150 (Agilent Technologies, Santa Clara, CA). cDNA was synthesized using ReverTra Ace qPCR RT Master Mix with gDNA Remover (Toyobo, Osaka, Japan).
- Quantitative PCR Quantitative PCR was performed on five POs and five transplanted POs (separated from 10,000 ESCs/sample) using a LightCycler 480 system (Roche Diagnostics, Rotnch, Switzerland). Data were normalized to that of GAPDH as an endogenous control and relative quantification based on a standard curve was determined.
- the primers used are shown in Table 1 below.
- the AR group served as a control for the study of the highly vascularized ISWAT, as described above.
- basal plasma ACTH levels (“basal”, Figure 4A) were consistently higher in the ISWAT group than in the sham group. At 2, 4, 8, and 17 weeks after transplantation, ACTH values were significantly different between groups (p ⁇ 0.001-0.05, Figure 4A).
- CRH-stimulated plasma ACTH levels (“stimulated”, Figure 4A) were also higher in the ISWAT group than in the sham group, with statistically significant differences between groups at 2, 4, 8, 21, and 26 weeks (p ⁇ 0.001-0.005, Figure 4A).
- VEGF Vascular endothelial growth factor
- FGF2 basic fibroblast growth factor
- ANGPT2 angiopoietin 2
- hESC-derived POs can be transplanted into the subcutaneous tissue of hypopituitaric SCID mice and are functional.
- Transplanted mice responded to CRH with the release of ACTH into the circulation, indicating that the injected CRH stimulated the transplanted ACTH-producing cells.
- Comparison of ISWAT and AR transplants demonstrated that the vascularized subcutaneous adipose tissue was superior to the relatively vascularized space under the skin on the back, resulting in more sustained ACTH secretion.
- PSC-derived PO transplantation improves physical activity levels and body weight (Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells. Nat Commun.
- a key aspect of transplantation of PSC-derived cells is the removal of the graft in the event of tumor development.
- Subcutaneous transplantation as employed in this invention allows for relatively simple and non-invasive removal.
- transplantation into adipose tissue is more effective than transplantation into an avascular site (AR), which is important for clinical application.
- AR avascular site
- Example 2 Further investigation of transplantation sites of pituitary hormone-producing cells (inguinal subcutaneous, axillary subcutaneous, muscle) Materials and Methods hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
- mice were hypophysectomized, and blood samples were taken and ACTH was measured.
- mice were subjected to inguinal ISWAT transplantation (Inguinal).
- ISWAT transplantation Inguinal
- mice were placed in a supine position and the left axillary region was shaved under isoflurane inhalation anesthesia.
- a 4 mm vertical skin incision was made, a pocket was made in the subcutaneous adipose tissue, and five POs taken from cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Nylon suture closure on the PO was followed by skin closure.
- mice For gluteal intramuscular transplantation (Muscle), mice were placed in a prone position and the left gluteal region was shaved under isoflurane inhalation anesthesia. A 4 mm vertical skin incision was made, a pocket was made in the gluteal muscle, and five POs taken from cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed followed by nylon suture closure over the PO. All hypopituitaric mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
- axillary subcutaneous white adipose tissue transplantation group and intramuscular transplantation group were comparable when compared with the ISWAT group.
- axillary subcutaneous white adipose tissue transplantation and intramuscular transplantation are also suitable transplantation sites.
- n number was increased to confirm reproducibility.
- Two animals were added to the Axilla group, one to the Inguinal group, and two to the Muscle group.
- Example 3 Examination of transplantation of purified cell aggregates (purified bodies) (subcutaneously in the groin) Materials and Methods (1) Purification of pituitary hormone-producing cells (purified bodies) from hESC-derived PO Cell sorting (purification) using anti-EpCAM antibody was carried out on hESC-derived PO on days 64-76 after the start of differentiation induction, which was obtained by the same differentiation induction method as in Example 1. The cell sorting was carried out in three steps: 1. pretreatment, 2. dispersion of cell aggregates, and 3. separation of EpCAM-positive cells.
- Y27632 (20 ⁇ M) was added to the medium on the day before the operation to pretreat the cell aggregates. In the subsequent operations before cell sorting, 20 ⁇ M Y27632 was added to all solutions to which the cells were exposed.
- the cell aggregates were chopped or cut with a scalpel. Next, the cell aggregates were washed with PBS, and then the dispersion liquid of the nerve cell dispersion kit (manufactured by Wako) was added, and the enzyme treatment was performed at 37°C for 30 minutes, and the cell aggregates were further loosened by pipetting. After the reaction was completed, the cells were washed with DMEM/F12 (manufactured by Wako), and a collagenase solution was added and the cells were swirled and shaken (140-150 rpm) at 37°C for 30 minutes.
- DMEM/F12 manufactured by Wako
- the composition of the collagenase solution was the above-mentioned DMEM/F12 to which 0.2% collagenase type I (manufactured by Wako) and 0.1% BSA (manufactured by Thermofisher) were added. After collagenase treatment, the cells were washed with PBS, and 10x TrypLE Select solution (Thermofisher) + 0.2 mg/ml DNase I (Roche) was added and treated with the enzyme at 37°C for 10 minutes, after which the cells were dispersed into single cells by pipetting.
- the cells were suspended and neutralized in serum-free medium containing 20% KSR, centrifuged, and then resuspended in serum-free medium containing 10 ⁇ g/ml DNase I and 20% KSR.
- the single cells were passed through a 70 ⁇ m cell strainer to remove aggregates.
- EpCAM-positive cells Separation of EpCAM-positive cells by FACS
- the single cells obtained above were suspended in a DMEM/F-12 solution containing 1 mM EDTA and 1% FBS after centrifugation.
- PE-labeled anti-EpCAM antibody (Miltenyi) was added, incubated at 4°C for 10 minutes, and fluorescence-activated cell sorting (FACS) was performed to separate and collect EpCAM-positive cells.
- FACS fluorescence-activated cell sorting
- the purified EpCAM-positive cells were reaggregated in 200 ⁇ L of serum-free medium supplemented with 20% KSR and 20 ⁇ M Y27632 to give 10 ⁇ 10 4 cells per well of a non-adhesive 96-well culture plate, and cultured in suspension at 37° C., 40% O 2 , and 5% CO 2. From the third day after the start of suspension culture, serum-free medium supplemented with 20% KSR without Y27632 was used, and half of the medium was replaced every 3 to 4 days for long-term culture.
- Example 4 Further investigation of transplantation sites of pituitary hormone-producing cells (inguinal subcutaneous, axillary subcutaneous, muscle) Materials and Methods hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
- mice were hypophysectomized in the same manner as in Example 2, and then transplanted with ISWAT in the groin (Inguinal), subcutaneous white adipose tissue in the axilla (Axilla), or intramuscular transplantation in the gluteal region (Muscle).
- Example 5 Examination of the transplantation effect by angiogenesis induction ⁇ Materials and methods> hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
- the skin incision during pretreatment was used, and five POs taken from the cell culture medium were placed in the pocket using a wide-bore tip under a microscope, since Surgicel is bioabsorbable and does not need to be removed. Skin closure was performed following nylon suture closure on the PO.
- the skin incision during pretreatment was used, and five POs taken from the cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed following nylon suture closure on the PO.
- avascular area (AR) transplantation group a linear skin incision was made in the midline skin of the back, and five POs were placed. All hypopituitaric mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
- Example 6 Subcutaneous transplantation of human ES cell-derived pituitary organoids into primates under immunosuppressive drug conditions
- Materials and Methods hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 103 to 117 days after differentiation was used for transplantation into monkeys.
- Anesthesia was induced by intramuscular administration of atropine (50 ⁇ g/kg) as preoperative medication. Ketamine (5 mg/kg) and xylazine (1 mg/kg) were administered intramuscularly. A tracheal tube was inserted and fixed to the left side of the monkey's mouth. Dexamethasone (120 ⁇ g), thyroxine (10 ⁇ g/body), desmopressin (0.5 ⁇ g), and ceftriaxone (20 mg/kg) were administered intravenously immediately before surgery as steroid replacement and antibiotic therapy, respectively. Anesthesia during surgery was administered by inhalation of isoflurane at a concentration of 1-3%. An endoscopic transoral transsphenoidal approach was used for sphenoidal hypophysectomy.
- the exact direction of the pituitary gland was confirmed by an intraoperative navigation system.
- the monkey was placed prone and the head was rotated slightly toward the surgeon.
- the maxillary canines were pulled upward with a thread to open the monkey's mouth as wide as possible.
- the soft palate was incised linearly in the midline, and then the soft palate was retracted laterally with a thread.
- the mucosa in front of the sphenoid bone was incised linearly in the midline to expose the sphenoid bone.
- a hole was drilled in the sphenoid bone with a high-speed drill with a diamond burr. Because there are no surgical landmarks inside the petrous sphenoid sinus, the surgical direction was confirmed many times during this operation by the navigation system.
- the dural sella was cut with scissors.
- the pituitary gland and stalk were removed, and the pathological specimens were frozen and preserved. Close examination with an angled endoscope confirmed that no pituitary gland remained.
- the saddle was reconstructed with a fat graft taken from the abdominal subcutaneous fat layer, and fibrin glue was used to prevent postoperative cerebrospinal fluid (CSF) leakage. No bleeding or CSF leakage was observed after reconstruction.
- the soft palate was sutured to complete the operation. The infusion was discontinued immediately after the monkey regained consciousness. After awakening, the monkey was able to eat food and drink water freely.
- CSF cerebrospinal fluid
- Ceftriaxone (20 mg/kg) was administered intramuscularly for 7 days after surgery. In the event of postoperative pain, intramuscular administration of buprenorphine (4 ⁇ g/kg) was considered. For hypopituitarism, hormone replacement was administered intramuscularly daily.
- the doses of dexamethasone, thyroxine, and vasopressin were determined based on the clinical doses for hypopituitarism in humans.
- Dexamethasone was gradually tapered from 120 ⁇ g/day to 40 ⁇ g/day, and then changed to oral administration of 1.5 mg of cortryl from 66 days after surgery. Thyroxine (10 ⁇ g/day) was administered intramuscularly immediately after surgery.
- Diabetes insipidus was treated with vasopressin (0.5 ⁇ g/day) and free drinking of water.
- the monkeys' water balance was evaluated by body weight and serum sodium (Na) value, because it was difficult to accurately measure water intake and urine volume.
- Water and electrolyte imbalance was evaluated by blood tests. Infection was evaluated by the surgical wound, body temperature, and food intake. A hormone stimulation test was performed under sedation on the 17th day after surgery. Anterior pituitary function was measured after intravenous administration of corticotropin-releasing hormone (CRH) (1.5 ⁇ g/kg) and growth hormone-releasing factor (GRF) (1 ⁇ g/kg). Blood was collected 60 minutes after injection and posterior pituitary function was evaluated based on the presence or absence of DI and blood AVP concentration.
- CH corticotropin-releasing hormone
- GRF growth hormone-releasing factor
- ACTH/Cortisol concentrations were measured weekly until one month after transplantation treatment. Thereafter, ACTH/Cortisol concentrations were measured every two weeks. CRH loading tests were performed 17 days after hypophysectomy and 4 weeks after PO implantation treatment. The negative feedback of the graft on cortisol was evaluated. Blood samples were collected via the dorsal vein of the hand before and 2 hours after administration of 1 mg dexamethasone. After blood collection, the blood was heparinized and centrifuged to obtain plasma.
- ACTH concentrations in the blood supernatant were determined using an electrochemiluminescence immunoassay (ECLIA) kit (SRL Co., Ltd., Tokyo, Japan), which is used clinically in Japan.
- ELIA electrochemiluminescence immunoassay
- the sheet placed during cell transplantation was removed using the skin incision made during pretreatment, and 1440 POs (5 pieces/20 g) washed with HBSS under visual observation were placed in the pocket. Following nylon suture closure on the PO, skin closure was performed. To prevent postoperative adrenal crisis, the patient was given an intravenous injection of 4.5 mg hydrocortone.
- the present invention is useful because it makes it possible to provide a therapeutic drug for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used to transplant into a transplantation site (subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue) that is less invasive and easy to resect when the transplanted cells become tumorous.
- a transplantation site subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue
- the therapeutic drug of the present invention is also useful because it makes it possible to realize clinical applications of pituitary hormone-producing cells.
- the method of the present invention is also useful because it makes it possible to create a model animal having human pituitary tissue that maintains hormone secretion ability for a long period of time, which is useful for evaluating the efficacy and safety of compounds.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biomedical Technology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Cell Biology (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Developmental Biology & Embryology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Epidemiology (AREA)
- General Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Endocrinology (AREA)
- Diabetes (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Environmental Sciences (AREA)
- Genetics & Genomics (AREA)
- Virology (AREA)
- Reproductive Health (AREA)
- Molecular Biology (AREA)
- Gynecology & Obstetrics (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Urology & Nephrology (AREA)
- General Engineering & Computer Science (AREA)
- Hematology (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
Abstract
Description
本発明は、下垂体ホルモン産生細胞が対象の皮下組織及び/又は筋肉組織に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬に関する。 The present invention relates to a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized in that the pituitary hormone-producing cells are used to be transplanted into the subcutaneous tissue and/or muscle tissue of a subject.
下垂体は頭部に存在する内分泌器官であり、副腎皮質刺激ホルモン(ACTH)、成長ホルモンといった生体の維持、成長に重要な各種の下垂体ホルモンを産生する。下垂体の形成不全や下垂体機能低下症、下垂体腺腫等の疾患により下垂体の機能不全が生じた場合、成長障害、生殖器関連の異常、副腎や甲状腺の異常に類する重篤な症状が生じる。一般に、障害を受けた下垂体組織が自然に再生し機能を回復することは稀である。 The pituitary gland is an endocrine organ located in the head that produces various pituitary hormones, such as adrenocorticotropic hormone (ACTH) and growth hormone, which are important for the maintenance and growth of the body. When pituitary gland malfunctions due to diseases such as pituitary aplasia, hypopituitarism, or pituitary adenoma, serious symptoms such as growth disorders, reproductive organ abnormalities, and abnormalities of the adrenal gland or thyroid gland occur. In general, it is rare for damaged pituitary tissue to regenerate naturally and regain function.
例えば、下垂体機能低下症の現在の治療法は、通常、不足していると特定されたホルモンの投与に限定されているが、その用量調節は難しいものである。また、このようなホルモンの投与は、ヒューマンエラーによる投与忘れ等の問題もある。この点、下垂体ホルモン産生細胞を臨床的に展開できれば、ホルモン補充療法の抱えるこれらの問題が解消される可能性がある。このような背景を基に、これまで多能性幹細胞由来の下垂体ホルモン産生細胞の製造方法が、本発明者らにより確立、改良され、高品質な細胞集団を製造することが可能となった(特許文献1~4、非特許文献1及び2)。
For example, current treatments for hypopituitarism are usually limited to administering the hormone identified as deficient, but adjusting the dosage is difficult. Furthermore, administering such hormones can be problematic, such as forgetting to administer the hormone due to human error. In this regard, if pituitary hormone-producing cells could be deployed clinically, these problems associated with hormone replacement therapy could be resolved. Based on this background, the present inventors have established and improved a method for producing pituitary hormone-producing cells derived from pluripotent stem cells, making it possible to produce high-quality cell populations (
従来、下垂体ホルモン産生細胞をマウスに移植してホルモン分泌効果等を確認する際、腎臓の被膜下に移植されてきたが、侵襲性が高く移植対象への負担が大きいという課題があった。一方、皮下組織や筋肉組織への移植は、侵襲性が低く、移植した細胞が腫瘍化するなど、切除する必要が生じた場合でも外科的に切除することが容易であることから、簡便でかつ安全性の高い方法であるとされる。しかしながら、皮下組織自体に血管が少ないことから移植された細胞の生着が困難であり、皮下組織は当該細胞の移植には適さないという考えが一般的であった。 Traditionally, when pituitary hormone-producing cells were transplanted into mice to confirm the hormone secretion effect, etc., they were transplanted under the kidney capsule, but this was highly invasive and placed a heavy burden on the recipient, which was an issue. On the other hand, transplantation into subcutaneous tissue or muscle tissue is considered to be a simple and safe method, as it is less invasive and can be easily removed surgically even if the transplanted cells become tumorous and require removal. However, subcutaneous tissue itself has few blood vessels, making it difficult for transplanted cells to take root, and it was generally thought that subcutaneous tissue was not suitable for transplanting such cells.
従って、本発明の課題は、下垂体ホルモン産生細胞の臨床応用を実現するために、侵襲性が低く、移植した細胞が腫瘍化するなど、切除する必要が生じた場合でも外科的に切除することが容易であるような移植部位(皮下組織(特に皮下脂肪組織)及び/又は筋肉組織)に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬を提供することである。 The objective of the present invention is therefore to provide a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used for transplantation into a transplantation site (subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue) that is less invasive and can be easily surgically removed even if the transplanted cells become tumorous and require removal, in order to realize clinical application of the pituitary hormone-producing cells.
本発明者らは、皮下組織自体に血管が少ないことから細胞の生着が困難であり、皮下組織は当該細胞の移植には適さないという既存の考えにも関わらず、多能性幹細胞からインビトロで分化誘導させた下垂体ホルモン産生細胞が皮下組織に移植された場合に生着可能であることを見出した。すなわち、本発明者らが製造した多能性幹細胞由来の下垂体ホルモン産生細胞について、鋭意検討の結果、驚くべきことに、当該下垂体ホルモン産生細胞をマウスにおいて2種類の皮下組織、即ち、背中の皮膚の直下の無血管領域(AR:avascular region)と白色脂肪組織に富んだ鼠蹊部の皮下組織(ISWAT:inguinal subcutaneous white adipose tissue)に移植したところ、当該細胞が6か月以上生着し、ホルモン分泌機能を示すことを見出した。さらに、本発明者らは、これらの領域のうち、特に白色脂肪に富んだ鼠蹊部、腋下、腹、背中、鼠径部、大腿部、臀部、上腕部等に移植する方がその効果が高いこと、及び細胞の生着には血管新生が関与していることも見出した。また、本発明者らは、皮下組織下に存在する筋肉組織においても、皮下組織と同様に、当該下垂体ホルモン産生細胞の移植に適していることを見出した。これらの知見に基づいてさらに研究を重ねた結果、下垂体ホルモン産生細胞が皮下組織及び/又は筋肉組織に移植されるように用いられることを特徴とする本発明を完成するに至った。 The present inventors have found that pituitary hormone-producing cells induced to differentiate in vitro from pluripotent stem cells can be engrafted when transplanted into subcutaneous tissue, despite the existing belief that subcutaneous tissue is unsuitable for transplantation of cells because of the lack of blood vessels in the subcutaneous tissue itself. That is, as a result of extensive investigations into the pituitary hormone-producing cells derived from pluripotent stem cells produced by the present inventors, they surprisingly found that when the pituitary hormone-producing cells were transplanted into two types of subcutaneous tissue in mice, namely the avascular region (AR) directly beneath the skin of the back and the subcutaneous tissue of the groin rich in white adipose tissue (ISWAT), the cells survived for more than six months and exhibited hormone secretion function. Furthermore, the inventors have found that transplantation into these regions, particularly into the groin, armpits, abdomen, back, groin, thighs, buttocks, upper arms, etc., which are rich in white fat, is more effective, and that angiogenesis is involved in cell engraftment. The inventors have also found that muscle tissue present under the subcutaneous tissue is suitable for transplantation of the pituitary hormone-producing cells, just like subcutaneous tissue. As a result of further research based on these findings, the inventors have completed the present invention, which is characterized in that the pituitary hormone-producing cells are used for transplantation into subcutaneous tissue and/or muscle tissue.
すなわち、本発明は以下の通りのものである。
[1]下垂体ホルモン産生細胞が対象の皮下組織及び/又は筋肉組織に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬。
[2]前記皮下組織が、鼠蹊部、腋下、背中、腹部、大腿部、臀部、上腕部及び頭皮から選択される少なくとも1種類の部位に存在する皮下組織である[1]に記載の治療薬。
[3]前記皮下組織が、皮下脂肪組織である[1]又は[2]に記載の治療薬。
[4]前記脂肪組織が、白色脂肪組織である[3]に記載の治療薬。
[5]前記筋肉組織が、背中、腹部、大腿部、臀部及び上腕部から選択される少なくとも1種類の部位に存在する筋肉組織である[1]に記載の治療薬。
[6]下垂体ホルモン産生細胞が、前記組織中の血管付近に移植されるように用いられることを特徴とする[1]~[5]のいずれか一つに記載の治療薬。
[7]下垂体ホルモン産生細胞が、前記組織中の人工的に設けられた空間に移植されるように用いられることを特徴とする[1]~[6]のいずれか一つに記載の治療薬。
[8]前記下垂体の障害に基づく疾患が、下垂体機能低下症、下垂体性小人症、副腎皮質機能低下症、部分的下垂体機能低下症、下垂体前葉ホルモン単独欠損症及び頭蓋咽頭腫から選択される少なくとも1種類の疾患である[1]~[7]のいずれか一つに記載の治療薬。
[9]前記下垂体ホルモン産生細胞が、下垂体ホルモン産生細胞を含む細胞凝集体である[1]~[8]のいずれか一つに記載の治療薬。
[10]前記下垂体ホルモン産生細胞が、血管新生因子を発現する下垂体ホルモン産生細胞である、[1]~[9]のいずれか一つに記載の治療薬。
[11]前記血管新生因子がVEGFA、VEGFB、VEGFC及びANGPT2からなる群から選択される1以上である、[10]に記載の治療薬。
[12]前記下垂体ホルモン産生細胞が、以下の工程を含む方法によって製造される下垂体ホルモン産生細胞である[1]~[11]のいずれか一つに記載の治療薬;
(1)多能性幹細胞をc-jun N末キナーゼ(JNK)シグナル伝達経路阻害物質及びWntシグナル伝達経路阻害物質の存在下で培養し、細胞集団を得る第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質及びソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養し、下垂体ホルモン産生細胞を含む細胞集団を得る第二工程。
[13]前記工程が第一工程の前にさらに下記のa工程を行うことを特徴とする[12]に記載の治療薬;
(a)多能性幹細胞を、フィーダー細胞非存在下で、1)TGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質、並びに2)未分化維持因子を含む培地で培養するa工程。
[14]
前記工程が第二工程の後にさらに下記工程を行うことを特徴とする[12]又は[13]に記載の治療薬;
第二工程で得られた下垂体ホルモン産生細胞を含む細胞集団から下垂体ホルモン産生細胞を選別し、回収する工程。
[15]移植後のヒト対象において、移植された下垂体ホルモン産生細胞の周辺において血管新生が生じる、[1]~[14]のいずれか一つに記載の治療薬。
[16]血管新生促進剤と組み合わせて用いられることを特徴とする、[1]~[15]のいずれか一つに記載の治療薬。
[17]前記下垂体ホルモン産生細胞が、移植後のヒト対象において、皮下組織及び/又は筋肉組織内に生着する、[1]~[16]のいずれか一つに記載の治療薬。
[18]移植後のヒト対象において、血中に下垂体ホルモンが分泌されることを特徴とする、[1]~[17]のいずれか一つに記載の治療薬。
[19]ヒト対象の皮下組織及び/又は筋肉組織へ下垂体ホルモン産生細胞を投与することを含む、下垂体の障害に基づく疾患の治療方法。
[20]非ヒト動物の皮下組織及び/又は筋肉組織へヒト下垂体ホルモン産生細胞を投与することを特徴とする、担ヒト下垂体組織モデル非ヒト動物の作製方法。
[21]皮下組織及び/又は筋肉組織に、ヒト下垂体ホルモン産生細胞を含み、当該ヒト下垂体ホルモン産生細胞が産生する下垂体ホルモンが血中に分泌され得る、担ヒト下垂体組織モデル非ヒト動物。
[22][21]に記載の担ヒト下垂体組織モデル非ヒト動物に、被験物質を投与することを含む、当該被験物質の薬効及び安全性を評価する方法。
That is, the present invention is as follows.
[1] A therapeutic agent for a disease caused by a disorder of the pituitary gland, comprising pituitary hormone-producing cells, characterized in that the pituitary hormone-producing cells are used to be transplanted into the subcutaneous tissue and/or muscle tissue of a subject.
[2] The therapeutic agent according to [1], wherein the subcutaneous tissue is subcutaneous tissue present in at least one area selected from the groin, armpit, back, abdomen, thigh, buttocks, upper arm, and scalp.
[3] The therapeutic agent described in [1] or [2], wherein the subcutaneous tissue is subcutaneous adipose tissue.
[4] The therapeutic agent described in [3], wherein the adipose tissue is white adipose tissue.
[5] The therapeutic agent described in [1], wherein the muscle tissue is muscle tissue present in at least one area selected from the back, abdomen, thighs, buttocks, and upper arms.
[6] The therapeutic agent according to any one of [1] to [5], wherein the pituitary hormone-producing cells are used so as to be transplanted near blood vessels in the tissue.
[7] The therapeutic agent according to any one of [1] to [6], characterized in that the pituitary hormone-producing cells are used so as to be transplanted into an artificially created space in the tissue.
[8] The therapeutic agent according to any one of [1] to [7], wherein the disease caused by a pituitary disorder is at least one disease selected from hypopituitarism, pituitary dwarfism, adrenal insufficiency, partial hypopituitarism, isolated anterior pituitary hormone deficiency, and craniopharyngioma.
[9] The therapeutic agent according to any one of [1] to [8], wherein the pituitary hormone-producing cells are cell aggregates containing pituitary hormone-producing cells.
[10] The therapeutic agent according to any one of [1] to [9], wherein the pituitary hormone-producing cells are pituitary hormone-producing cells that express an angiogenic factor.
[11] The therapeutic agent described in [10], wherein the angiogenic factor is one or more selected from the group consisting of VEGFA, VEGFB, VEGFC and ANGPT2.
[12] The therapeutic agent according to any one of [1] to [11], wherein the pituitary hormone-producing cells are pituitary hormone-producing cells produced by a method comprising the steps of:
(1) a first step of culturing pluripotent stem cells in the presence of a c-jun N-terminal kinase (JNK) signaling pathway inhibitor and a Wnt signaling pathway inhibitor to obtain a cell population;
(2) A second step of culturing the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary hormone-producing cells.
[13] The therapeutic agent according to [12], wherein the process further comprises the following step a before the first step:
(a) A step a of culturing pluripotent stem cells in a medium containing 1) a TGFβ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells.
[14]
The therapeutic agent according to [12] or [13], wherein the step further comprises the following step after the second step:
A step of selecting and recovering pituitary hormone-producing cells from the cell population containing pituitary hormone-producing cells obtained in the second step.
[15] The therapeutic agent according to any one of [1] to [14], wherein angiogenesis occurs around the transplanted pituitary hormone-producing cells in a human subject after transplantation.
[16] The therapeutic agent according to any one of [1] to [15], which is used in combination with an angiogenesis promoter.
[17] The therapeutic agent according to any one of [1] to [16], wherein the pituitary hormone-producing cells are engrafted in subcutaneous tissue and/or muscle tissue of a human subject after transplantation.
[18] The therapeutic agent according to any one of [1] to [17], characterized in that a pituitary hormone is secreted into the blood in a human subject after transplantation.
[19] A method for treating a disease caused by a pituitary gland disorder, comprising administering pituitary hormone-producing cells to the subcutaneous tissue and/or muscle tissue of a human subject.
[20] A method for producing a non-human animal model bearing human pituitary tissue, which comprises administering human pituitary hormone-producing cells into the subcutaneous tissue and/or muscle tissue of the non-human animal.
[21] A non-human animal model bearing human pituitary gland tissue, which comprises human pituitary hormone-producing cells in subcutaneous tissue and/or muscle tissue, and which is capable of secreting pituitary hormones produced by the human pituitary hormone-producing cells into blood.
[22] A method for evaluating the efficacy and safety of a test substance, comprising administering the test substance to the non-human animal model bearing human pituitary tissue according to [21].
本発明によれば、侵襲性が低く、移植した細胞が腫瘍化するなど、切除する必要が生じた場合でも外科的に切除することが容易であるような皮下組織及び/又は筋肉組織に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬を提供することが可能となる。また、本発明の治療薬により、下垂体ホルモン産生細胞の臨床応用の実現が可能となる。また、本発明の方法により、化合物の薬効及び安全性評価に有用な、ヒトホルモン分泌能が長期間維持されるヒト下垂体組織を有するモデル動物の作出が可能となる。 The present invention makes it possible to provide a therapeutic agent for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used for transplantation into subcutaneous tissue and/or muscle tissue that is less invasive and can be easily surgically removed even if the transplanted cells become tumorous and require removal. Furthermore, the therapeutic agent of the present invention makes it possible to realize clinical applications of pituitary hormone-producing cells. Furthermore, the method of the present invention makes it possible to create model animals having human pituitary tissue that maintains the ability to secrete human hormones for a long period of time, which is useful for evaluating the efficacy and safety of compounds.
1.本発明の治療薬及び細胞移植
本発明は、下垂体ホルモン産生細胞が皮下組織、好ましくは皮下脂肪組織、更に好ましくは皮下の白色脂肪組織及び/又は筋肉組織に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬に関する。本明細書において「下垂体ホルモン産生細胞」とは、成長ホルモン(GH)、プロラクチン(PRL)、副腎皮質刺激ホルモン(ACTH)、甲状腺刺激ホルモン(TSH)、卵胞刺激ホルモン(FSH)、黄体化ホルモン(LH)、又はメラニン細胞刺激ホルモン(MSH)等を産生する細胞をいい、これらの下垂体ホルモンの少なくとも1つを産生する細胞の総称である。
1. Therapeutic Agent and Cell Transplantation of the Present Invention The present invention relates to a therapeutic agent for diseases due to disorders of the pituitary gland, comprising pituitary hormone-producing cells, characterized in that the pituitary hormone-producing cells are used to be transplanted into subcutaneous tissue, preferably subcutaneous adipose tissue, more preferably subcutaneous white adipose tissue and/or muscle tissue. In this specification, "pituitary hormone-producing cells" refers to cells that produce growth hormone (GH), prolactin (PRL), adrenocorticotropic hormone (ACTH), thyroid stimulating hormone (TSH), follicle stimulating hormone (FSH), luteinizing hormone (LH), melanocyte stimulating hormone (MSH), or the like, and is a general term for cells that produce at least one of these pituitary hormones.
本発明の治療薬に含有される下垂体ホルモン産生細胞は、その由来や製造方法等に関して、上述のような下垂体ホルモンを産生し得る限り特に限定されないが、好ましくは、多能性幹細胞から分化誘導された下垂体ホルモン産生細胞である。
一態様において、本明細書における下垂体ホルモン産生細胞として、副腎皮質刺激ホルモン(ACTH)産生細胞、成長ホルモン(GH)産生細胞及びプロラクチン(PRL)産生細胞からなる群から選ばれる少なくとも1種が挙げられる。
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞は、副腎皮質刺激ホルモン(ACTH)産生細胞、成長ホルモン(GH)産生細胞及びプロラクチン(PRL)産生細胞からなる群から選ばれる少なくとも1種(1種、2種又は3種)を含む。更に、一態様において、本明細書における下垂体ホルモン産生細胞を含む細胞集団は、以下のような特徴を有する。
The pituitary hormone-producing cells contained in the therapeutic agent of the present invention are not particularly limited in terms of their origin, production method, etc., as long as they are capable of producing the pituitary hormones described above, but are preferably pituitary hormone-producing cells induced to differentiate from pluripotent stem cells.
In one embodiment, the pituitary hormone-producing cells in the present specification include at least one type selected from the group consisting of adrenocorticotropic hormone (ACTH)-producing cells, growth hormone (GH)-producing cells, and prolactin (PRL)-producing cells.
In one embodiment, the pituitary hormone-producing cells contained in the therapeutic agent of the present invention include at least one type (one, two or three types) selected from the group consisting of adrenocorticotropic hormone (ACTH)-producing cells, growth hormone (GH)-producing cells and prolactin (PRL)-producing cells. Furthermore, in one embodiment, the cell population containing the pituitary hormone-producing cells in the present specification has the following characteristics.
具体的には、上記下垂体ホルモン産生細胞を含む細胞集団は、好ましくは、血管新生因子を発現する細胞を含む。ここにおいて血管新生因子とは、血管新生に関与する因子、好ましくは血管新生の促進に関与する因子であれば特に限定はなく、アンジオゲニン、アンジオポエチン-1、Del-1、線維芽細胞増殖因子:酸性(aFGF)及び塩基性(bFGF)、上皮成長因子(EGF)、血管内皮細胞増殖因子(VEGF)、フォリスタチン、顆粒球マクロファージ-コロニー刺激因子(GM-CSF)、肝細胞増殖因子(HGF)、単球走化性タンパク質-1(MCP-1)、スフィンゴシン-1-リン酸(S1P)、血小板由来増殖因子(PDGF)、トランスフォーミング増殖因子α(TGF-α)、トランスフォーミング増殖因子β(TGF-β)、腫瘍壊死因子α(TNF-α)等が挙げられる。
一態様において、該血管新生因子は、血管内皮細胞増殖因子A(VEGFA)、血管内皮細胞増殖因子B(VEGFB)、血管内皮細胞増殖因子C(VEGFC)、及びアンジオポエチン-2(ANGPT2)からなる群から選択される1以上である。また、本発明において、移植細胞の観点から、該細胞が血管新生因子を発現することで、移植後、本発明の治療薬に含まれる下垂体ホルモン産生細胞の周辺において血管新生が生じ得るという特徴を有することを見出した。さらに、下垂体ホルモン産生細胞の周辺において血管新生が生じることで、下垂体ホルモン産生細胞が皮下組織内や筋肉組織内に生着し得る。
Specifically, the cell population containing the pituitary hormone-producing cells preferably contains cells expressing an angiogenic factor. Here, the angiogenic factor is not particularly limited as long as it is a factor involved in angiogenesis, preferably a factor involved in promoting angiogenesis, and examples thereof include angiogenin, angiopoietin-1, Del-1, fibroblast growth factors: acidic (aFGF) and basic (bFGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), follistatin, granulocyte macrophage-colony stimulating factor (GM-CSF), hepatocyte growth factor (HGF), monocyte chemotactic protein-1 (MCP-1), sphingosine-1-phosphate (S1P), platelet-derived growth factor (PDGF), transforming growth factor α (TGF-α), transforming growth factor β (TGF-β), tumor necrosis factor α (TNF-α), and the like.
In one embodiment, the angiogenic factor is one or more selected from the group consisting of vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor B (VEGFB), vascular endothelial growth factor C (VEGFC), and angiopoietin-2 (ANGPT2). In addition, in the present invention, from the viewpoint of transplanted cells, it has been found that the cells have a characteristic that, by expressing an angiogenic factor, angiogenesis can occur around the pituitary hormone-producing cells contained in the therapeutic agent of the present invention after transplantation. Furthermore, angiogenesis can occur around the pituitary hormone-producing cells, allowing the pituitary hormone-producing cells to be engrafted in subcutaneous tissue or muscle tissue.
また、具体的には、上記下垂体ホルモン産生細胞を含む細胞集団は、下垂体ホルモン産生細胞へ分化し得る細胞を含んでいても良い。当該細胞として、下垂体幹細胞又は下垂体前駆細胞(下垂体ホルモン産生細胞の前駆細胞)等が挙げられる。すなわち、本発明の下垂体ホルモン産生細胞を含む細胞集団には、下垂体ホルモン産生細胞、並びに、下垂体幹細胞及び/又は下垂体前駆細胞を含む細胞集団が含まれる。
すなわち、当該細胞集団は下垂体幹細胞マーカーを発現する細胞を含んでいてもよい。下垂体幹細胞マーカーとしては、Sox2、Sox9、E-Cadherin、Nestin、S100β、GFRα2、Prop1、CD133、β-Catenin、Klf4、Oct4、Pax6、コクサッキーウイルス・アデノウイルス共通受容体(CXADR)、PRRX1/2、Ephrin-B2及びACEの少なくとも1つが挙げられる。一態様として、下垂体幹細胞マーカーは、SOX2及び/又はCXADRであってよい。
Specifically, the cell population containing the pituitary hormone-producing cells may contain cells that can differentiate into pituitary hormone-producing cells. Examples of such cells include pituitary stem cells or pituitary progenitor cells (precursor cells of pituitary hormone-producing cells). That is, the cell population containing the pituitary hormone-producing cells of the present invention includes cell populations containing pituitary hormone-producing cells, and pituitary stem cells and/or pituitary progenitor cells.
That is, the cell population may contain cells expressing a pituitary stem cell marker. Examples of the pituitary stem cell marker include at least one of Sox2, Sox9, E-Cadherin, Nestin, S100β, GFRα2, Prop1, CD133, β-Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE. In one embodiment, the pituitary stem cell marker may be SOX2 and/or CXADR.
本発明の治療薬に含有される下垂体ホルモン産生細胞は、少なくとも1種類の下垂体ホルモン産生細胞として含有されていてもよく、単一の又は複数種の下垂体ホルモン産生細胞を含む細胞集団であってよい。
当該細胞集団としては、分散された細胞集団、下垂体ホルモン産生細胞を含む細胞凝集体、又は二次元の細胞シート等が挙げられる。
また一態様において、当該細胞集団として、下垂体ホルモン産生細胞を含む下垂体組織と下垂体組織以外の組織(例えば視床下部組織等)を含む細胞凝集体が挙げられる。
また、一態様において、当該細胞集団として、下垂体ホルモン産生細胞を含む下垂体組織と、下垂体組織以外の組織(例えば視床下部組織等)を含む細胞集団から回収される下垂体組織が挙げられる。
本明細書において、下垂体ホルモン産生細胞を含む細胞凝集体を、下垂体オルガノイド(PO)ともいう。
一態様において当該細胞集団は細胞凝集体である。
The pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be contained as at least one type of pituitary hormone-producing cells, or may be a cell population containing a single type or multiple types of pituitary hormone-producing cells.
The cell population may be a dispersed cell population, a cell aggregate containing pituitary hormone-producing cells, or a two-dimensional cell sheet.
In one embodiment, the cell population includes a cell aggregate containing pituitary tissue containing pituitary hormone-producing cells and tissue other than pituitary tissue (for example, hypothalamic tissue, etc.).
In one embodiment, the cell population includes pituitary tissue containing pituitary hormone-producing cells, and pituitary tissue recovered from a cell population containing tissue other than pituitary tissue (e.g., hypothalamic tissue, etc.).
As used herein, cell aggregates containing pituitary hormone-producing cells are also referred to as pituitary organoids (PO).
In one embodiment, the cell population is a cell aggregate.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞集団は、少なくとも全細胞数の5%以上、好ましくは10%以上の下垂体ホルモン産生細胞を含有する細胞集団であることが好ましい。
本発明の治療薬に含有される下垂体ホルモン産生細胞は、後述する下垂体ホルモン産生マーカーを指標として同定することができる。
In one aspect, the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention is preferably a cell population containing pituitary hormone-producing cells at least 5% or more, preferably 10% or more of the total cell number.
The pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be identified using the pituitary hormone-producing markers described below as indicators.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞は、少なくともACTH産生細胞を含有する。
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞集団は、全細胞数の3%以上、好ましくは5%以上のACTH陽性細胞を含有する細胞集団であることが好ましい。
In one embodiment, the pituitary hormone-producing cells contained in the therapeutic agent of the present invention contain at least ACTH-producing cells.
In one embodiment, the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention is preferably a cell population containing ACTH-positive cells at 3% or more, preferably 5% or more of the total cell number.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞凝集体の長径(又は、円相当径)は、例えば、0.1~7mmであってよく、好ましくは0.4~5mmであってよい。
該細胞凝集体の長径、短径及び高さを測定する方法は、特に限定されず、例えば、顕微鏡下で撮像した画像から測定すればよい。例えば、96ウェル培養プレートで培養時の細胞凝集体を、キーエンス倒立顕微鏡10倍レンズで撮像し、撮像した画像から測定できる。ここで、長径とは、撮像画像において、該細胞凝集体の2つの端点を結ぶ線分のうち、最も長い線分及びその長さを意味する。円相当径とは、二次元面に投影したときに得られる図形(円形又は楕円形)の面積に相当する、真円の直径を意味する。後述する通り、当該凝集体はスフェア状の凝集体であり、長径及び円相当径は近似し、真球に近いスフェア状の凝集体に近づくほど、長径及び円相当径の差は小さくなる。
In one embodiment, the major axis (or equivalent circle diameter) of the cell aggregate containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be, for example, 0.1 to 7 mm, and preferably 0.4 to 5 mm.
The method for measuring the major axis, minor axis and height of the cell aggregate is not particularly limited, and may be measured from an image captured under a microscope. For example, the cell aggregate during culture in a 96-well culture plate can be imaged with a 10x lens of a Keyence inverted microscope, and the height can be measured from the image. Here, the major axis refers to the longest line segment and its length among the line segments connecting the two end points of the cell aggregate in the captured image. The circle equivalent diameter refers to the diameter of a perfect circle, which corresponds to the area of a figure (circular or elliptical) obtained when projected onto a two-dimensional surface. As described later, the aggregate is a spherical aggregate, and the major axis and the circle equivalent diameter are approximate, and the difference between the major axis and the circle equivalent diameter becomes smaller as the aggregate approaches a spherical aggregate close to a perfect sphere.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞集団は、視床下部組織の細胞を含んでいてもよい。当該細胞としては、Rx陽性、Pax6陽性且つNkx2.1陰性の細胞(背側視床下部)、Rx陽性、Pax6陰性且つNkx2.1陽性(腹側視床下部)が挙げられる。 In one embodiment, the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may contain cells of hypothalamic tissue. Such cells include Rx-positive, Pax6-positive and Nkx2.1-negative cells (dorsal hypothalamus) and Rx-positive, Pax6-negative and Nkx2.1-positive cells (ventral hypothalamus).
一態様において、本発明の治療薬に包含される下垂体ホルモン産生細胞を含む細胞集団は、下垂体幹細胞を含んでいてもよい。当該細胞集団は、少なくとも全細胞数の1%以上、好ましくは2%以上の下垂体幹細胞、すなわち上記の下垂体幹細胞マーカーのいずれかが陽性の細胞を含む。当該下垂体幹細胞マーカーとして、例えば、SOX2又はCXADRが挙げられる。 In one embodiment, the cell population containing pituitary hormone-producing cells included in the therapeutic agent of the present invention may contain pituitary stem cells. The cell population contains at least 1% or more, preferably 2% or more, of the total number of cells that are pituitary stem cells, i.e., cells that are positive for any of the above-mentioned pituitary stem cell markers. Examples of the pituitary stem cell marker include SOX2 and CXADR.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞集団は、分化誘導工程で生じる下垂体ホルモン産生細胞以外の目的外細胞が除かれた細胞集団であっても良い。目的外細胞が含まれない、又は目的外細胞が含まれる割合を低下させた細胞集団は、下垂体ホルモン産生細胞又はその中間体(下垂体ホルモン産生細胞の前駆細胞)に発現し目的外細胞で発現しない細胞表面に発現するマーカータンパク質の発現を指標として下垂体ホルモン産生細胞を選別し、得ることができる。前記マーカータンパク質として、EpCAM等が挙げられる(国際公開第2021/201175号を参照)。 In one embodiment, the cell population containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be a cell population from which non-target cells other than pituitary hormone-producing cells generated in the differentiation induction step have been removed. A cell population that does not contain non-target cells or has a reduced proportion of non-target cells can be obtained by selecting pituitary hormone-producing cells using as an index the expression of a marker protein expressed on the cell surface that is expressed in pituitary hormone-producing cells or their intermediates (precursor cells of pituitary hormone-producing cells) and not expressed in non-target cells. Examples of the marker protein include EpCAM (see International Publication No. WO 2021/201175).
上記の目的外細胞が含まれない、又は目的外細胞が含まれる割合を低下させた細胞集団(以下、「精製体」と称する場合がある)は、一例として、後述の「7.精製体の製造方法」に記載の方法により得ることができる。当該細胞集団(精製体)の特徴は、一態様において、目的外細胞が含まれる割合を低下させた細胞集団(細胞凝集体)におけるEpCAM及び/又はE-cadherin陽性細胞の存在割合は80%以上であってもよく、好ましくは85%以上、90%以上、95%以上であってもよい。 The cell population (hereinafter sometimes referred to as "purified body") that does not contain the above-mentioned undesired cells or in which the proportion of undesired cells is reduced can be obtained, for example, by the method described below in "7. Method for producing purified body". In one embodiment, the cell population (purified body) is characterized in that the proportion of EpCAM and/or E-cadherin positive cells in the cell population (cell aggregate) in which the proportion of undesired cells is reduced may be 80% or more, and preferably 85% or more, 90% or more, or 95% or more.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団における神経系細胞の存在割合は、20%以下であってもよく、好ましくは15%以下、10%以下、5%以下であってもよい。 In one embodiment, the proportion of neural cells in a cell population in which the proportion of non-target cells is reduced may be 20% or less, and preferably 15% or less, 10% or less, or 5% or less.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団におけるACTH陽性細胞の存在割合は、5%以上であってもよく、好ましくは10%以上であってもよい。ACTH陽性細胞は、NeuroD1及びTbx19を同時に発現する。従って、当該細胞集団中に存在する総細胞数に対するNeuroD1陽性及び/又はTbx19陽性細胞数の割合も、ACTH陽性細胞数の存在割合と同等の割合であってもよい。 In one embodiment, the proportion of ACTH-positive cells in a cell population in which the proportion of non-target cells is reduced may be 5% or more, and preferably 10% or more. ACTH-positive cells simultaneously express NeuroD1 and Tbx19. Therefore, the proportion of NeuroD1-positive and/or Tbx19-positive cells to the total number of cells present in the cell population may be the same as the proportion of ACTH-positive cells.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団におけるACTH分泌能は、200~1,000,000pg/mlであってもよく、1,000~500,000pg/ml、5,000~300,000pg/ml、10,000~100,000pg/mlであってもよい。 In one embodiment, the ACTH secretion ability in a cell population in which the proportion of non-target cells is reduced may be 200 to 1,000,000 pg/ml, 1,000 to 500,000 pg/ml, 5,000 to 300,000 pg/ml, or 10,000 to 100,000 pg/ml.
ACTH分泌能は、ACTH細胞数及び培養条件などの影響を受ける。ACTH分泌能は、特定の培養条件における、細胞集団(細胞凝集体)1個から培地中に分泌されたACTHの濃度で表され得る。例えば、特定の培養条件とは、(i)下垂体ホルモン産生細胞の培養に用いることが可能な培地、好ましくはKSRを含む無血清培地(例:IMDM、DMEM、EMEM、αMEM、GMEM、F-12培地、DMEM/F12、IMDM/F12、上皮細胞用培地(例:CnT-Prime epithelial culture medium)、又はこれらの混合培地等)を用い、(ii)外部からACTH産生に影響を与える物質(例:ACTH産生を上昇させるNotchシグナル阻害剤)を添加せず、(iii)培地量を0.18~20mlに設定し、(iv)3~4日間培養する条件を意味する。当該条件における培養後の培地を回収し、当該培地中のACTHの濃度を測定すればよい。当業者であれば、ELISA等の公知の手法を用いて、ACTHの濃度を測定することが可能である。 ACTH secretion ability is affected by the number of ACTH cells and culture conditions. ACTH secretion ability can be expressed as the concentration of ACTH secreted into the medium from one cell population (cell aggregate) under specific culture conditions. For example, the specific culture conditions refer to the following conditions: (i) a medium that can be used for culturing pituitary hormone-producing cells, preferably a serum-free medium containing KSR (e.g., IMDM, DMEM, EMEM, αMEM, GMEM, F-12 medium, DMEM/F12, IMDM/F12, epithelial cell medium (e.g., CnT-Prime epithelial culture medium), or a mixed medium thereof, is used; (ii) no substance that affects ACTH production from the outside is added (e.g., Notch signal inhibitors that increase ACTH production); (iii) the medium volume is set to 0.18 to 20 ml; and (iv) the culture is performed for 3 to 4 days. After culturing under these conditions, the medium is collected and the concentration of ACTH in the medium is measured. Those skilled in the art can measure the concentration of ACTH using known techniques such as ELISA.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団に存在する総細胞数に対する下垂体幹細胞数の割合(下垂体幹細胞数の存在割合)は、1%以上、好ましくは3%以上又は5%以上であってもよい。下垂体幹細胞は、上述のようにSox2、Sox9、E-Cadherin、Nestin、S100β、GFRα2、Prop1、CD133、β-Catenin、Klf4、Oct4、Pax6、コクサッキーウイルス・アデノウイルス共通受容体(CXADR)、PRRX1/2、Ephrin-B2、ACEといった下垂体幹細胞マーカーを同時に発現する。従って、下垂体幹細胞数の存在割合は、該細胞集団中に存在する総細胞数に対する上記の下垂体幹細胞マーカー(例えば、Sox2、CXADR)陽性細胞数の割合を測定することによって決定することができる。 In one embodiment, the ratio of the number of pituitary stem cells to the total number of cells present in a cell population in which the proportion of non-target cells is reduced (the proportion of pituitary stem cells present) may be 1% or more, preferably 3% or more or 5% or more. As described above, pituitary stem cells simultaneously express pituitary stem cell markers such as Sox2, Sox9, E-Cadherin, Nestin, S100β, GFRα2, Prop1, CD133, β-Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE. Therefore, the proportion of pituitary stem cells present can be determined by measuring the proportion of cells positive for the above pituitary stem cell markers (e.g., Sox2, CXADR) to the total number of cells present in the cell population.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団の細胞密度が3,000~20,000 cells/mm2であってもよく、好ましくは5,000~10,000 cells/mm2、より好ましくは6,000~9,000 cells/mm2であってもよい。 In one embodiment, the cell density of the cell population in which the proportion of non-target cells is reduced may be 3,000 to 20,000 cells/mm 2 , preferably 5,000 to 10,000 cells/mm 2 , and more preferably 6,000 to 9,000 cells/mm 2 .
本願明細書において、目的外細胞が含まれる割合を低下させた細胞集団の細胞密度は、細胞集団の切断面、例えば、中心から200μm以内の位置における、単位面積当たりの細胞数で表し得る。当業者であれば、公知の手法を用いて細胞集団の細胞密度を測定することができる。具体的には、細胞集団を切断した切片を準備し、DAPI、抗E-cadherin抗体などを用いた免疫染色などの手法により細胞数を測定し、当該切片の面積により除すればよい。 In the present specification, the cell density of a cell population in which the proportion of non-target cells has been reduced can be expressed as the number of cells per unit area on the cut surface of the cell population, for example, at a position within 200 μm from the center. A person skilled in the art can measure the cell density of a cell population using known methods. Specifically, a slice is prepared by cutting the cell population, and the number of cells is measured by a method such as immunostaining using DAPI or anti-E-cadherin antibody, and then divided by the area of the slice.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団の直径は、200~3,000μmであり、好ましくは300~2,000μm、より好ましくは400~1,500μmであってもよい。 In one embodiment, the diameter of the cell population in which the proportion of non-target cells is reduced may be 200 to 3,000 μm, preferably 300 to 2,000 μm, and more preferably 400 to 1,500 μm.
更なる態様において、目的外細胞が含まれる割合を低下させた細胞集団は、スフェア状の細胞凝集体であってもよい。本明細書における「スフェア状の細胞凝集体」は、球状に近い立体的な形を有する細胞凝集体を意味する。球状に近い立体的な形とは、三次元構造を有する形であって、二次元面に投影したときに、例えば、円形又は楕円形を示す球状形が挙げられる。 In a further embodiment, the cell population in which the proportion of non-target cells is reduced may be a spherical cell aggregate. In this specification, "spherical cell aggregate" refers to a cell aggregate having a three-dimensional shape close to a sphere. A three-dimensional shape close to a sphere is a shape having a three-dimensional structure, and examples of such shapes include a spherical shape that is circular or elliptical when projected onto a two-dimensional surface.
当該細胞凝集体は、好ましくは真球に近いスフェア状の細胞凝集体である。真球に近いスフェア状の細胞凝集体とは、真球に近い立体的な形を有する細胞凝集体を意味する。真球に近い立体的な形とは、複数の方向から二次元面に投影したときに得られる複数の円形が、当該円形の中心から外周への長さが全て同じ、あるいは、最も長い線分と最も短い線分の差が10%以下、5%以下、3%以下、2%以下又は1%以下である球上形を意味する。 The cell aggregate is preferably a spherical cell aggregate close to a perfect sphere. A spherical cell aggregate close to a perfect sphere means a cell aggregate having a three-dimensional shape close to a perfect sphere. A three-dimensional shape close to a perfect sphere means a spherical shape in which the multiple circles obtained when projected onto a two-dimensional surface from multiple directions all have the same length from the center to the circumference of the circles, or the difference between the longest and shortest line segments is 10% or less, 5% or less, 3% or less, 2% or less, or 1% or less.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団は、その表面において、EpCAM陽性細胞及びE-cadherinの少なくとも1つが陽性の細胞が互いに面接着し、上皮構造を形成していてもよい。 In one embodiment, a cell population in which the proportion of non-target cells has been reduced may have, on its surface, EpCAM-positive cells and cells positive for at least one of E-cadherin that adhere to each other to form an epithelial structure.
本明細書において「面接着」とは、隣り合う細胞同士が面で接着することを意味する。より詳細には、面接着とは、ある細胞の表面積のうち別の細胞の表面と接着している割合が、例えば、1%以上、好ましくは3%以上、より好ましくは5%以上、さらに好ましくは10%以上であることをいう。一態様では、細胞の表面とは、細胞膜表面のことをいう。細胞の表面は、細胞接着因子(例えば、E-cadherinやEpCAM等)の免疫染色により、観察できる。面接着ではない状態として、点接着が挙げられる。点接着とは、細胞と細胞が点で接着することをいう。 As used herein, "surface adhesion" means that adjacent cells adhere to each other at their surfaces. More specifically, surface adhesion means that the proportion of the surface area of a cell that is adhered to the surface of another cell is, for example, 1% or more, preferably 3% or more, more preferably 5% or more, and even more preferably 10% or more. In one embodiment, the cell surface means the cell membrane surface. The cell surface can be observed by immunostaining for cell adhesion factors (e.g., E-cadherin, EpCAM, etc.). Point adhesion is an example of a state that is not surface adhesion. Point adhesion means that cells adhere to each other at points.
本明細書において「上皮構造」とは、頂端面又は基底膜などの、上皮組織が特徴的に有する構造を意味する。後述する頂端面及び基底膜のマーカーを用いて免疫染色することにより確認することができる。 In this specification, "epithelial structure" refers to a structure that epithelial tissue has characteristically, such as the apical surface or basement membrane. This can be confirmed by immunostaining using markers for the apical surface and basement membrane, which will be described later.
上皮組織は極性化して「頂端面(apical surface)」と「基底膜」ができる。「基底膜」とは、基底膜のマーカーであるラミニン及びIV型コラーゲンを多く含む50~100nmの、上皮細胞が産生した基底(basal)側の層(基底膜)が存在する基底膜のことをいう。「頂端面」は、「基底膜」と反対側に形成される表面(表層面)のことをいう。また、このような頂端面は、頂端面のマーカー(例:atypical-PKC(以下、「aPKC」と略記する場合がある)、E-cadherin、N-cadherin)に対する抗体を用いて、当業者に周知の免疫染色法等で同定することができる。 Epithelial tissue is polarized to form an "apical surface" and a "basement membrane." The term "basement membrane" refers to a basement membrane that contains a 50-100 nm basal layer (basement membrane) produced by epithelial cells that is rich in laminin and type IV collagen, which are markers of the basement membrane. The term "apical surface" refers to the surface (superficial surface) that is formed on the opposite side of the "basement membrane." Such apical surfaces can be identified by immunostaining methods well known to those skilled in the art using antibodies against apical surface markers (e.g., atypical-PKC (hereinafter sometimes abbreviated as "aPKC"), E-cadherin, and N-cadherin).
「上皮組織」とは、体表面、管腔(消化管など)、体腔(心膜腔など)などの表面を細胞が隙間なく覆うことで形成される組織である。上皮組織を形成している細胞を上皮細胞という。上皮細胞は、細胞が頂端(apical)-基底(basal)方向の極性を持つ。上皮細胞は、接着結合(adherence junction)及び/又は密着結合(tight junction)により上皮細胞同士で強固な結合をつくり、細胞の層を形成できる。この細胞層が、1ないし十数層重なってできた組織が上皮組織である。 "Epithelial tissue" is tissue formed when cells tightly cover the surface of the body, lumen (such as the digestive tract), and body cavity (such as the pericardial cavity). The cells that form epithelial tissue are called epithelial cells. Epithelial cells have apical-basal polarity. Epithelial cells form strong bonds with each other through adherens junctions and/or tight junctions, and can form cell layers. Epithelial tissue is a tissue formed when one to a dozen or so cell layers are stacked on top of each other.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団は、EpCAM陽性細胞及び/又はE-cadherin陽性細胞が互いに面接着することで形成される島状構造を複数有してもよい。また、更なる態様において、当該細胞集団では、複数の島状構造が集まってスポンジ状構造が形成されてもよい。 In one embodiment, the cell population in which the proportion of non-target cells is reduced may have a plurality of island-like structures formed by surface adhesion of EpCAM-positive cells and/or E-cadherin-positive cells to each other. In a further embodiment, the cell population may have a sponge-like structure formed by a collection of a plurality of island-like structures.
「島状構造(islet-like strucure)」とは、2個~20個程度(例:3個、4個、5個、6個、7個、8個、9個、10個、15個、20個)のEpCAM陽性細胞及び/又はE-cadherin陽性細胞が互いに面接着することによって形成される構造であり得る。各島状構造に含まれる当該細胞の個数は、それぞれの島状構造ごとに異なっていてもよい。面接着とは、隣り合う細胞同士が面で接着することを意味する。より詳細には、面接着とは、細胞同士が面で接着している割合が、例えば、1%以上、好ましくは3%以上、より好ましくは5%以上、さらに好ましくは10%以上であることを言う。島状構造に存在する細胞の長径は、例えば2~50μm、好ましくは4~30μm、短径は、例えば0.5~20μm、好ましくは1~10μmであってよい。 The term "islet-like structure" refers to a structure formed by surface adhesion of about 2 to 20 (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 15, 20) EpCAM-positive cells and/or E-cadherin-positive cells to each other. The number of cells contained in each island-like structure may differ for each island-like structure. Surface adhesion means that adjacent cells are adhered to each other by their surfaces. More specifically, surface adhesion means that the rate at which cells are adhered to each other by their surfaces is, for example, 1% or more, preferably 3% or more, more preferably 5% or more, and even more preferably 10% or more. The major axis of the cells present in the island-like structure may be, for example, 2 to 50 μm, preferably 4 to 30 μm, and the minor axis may be, for example, 0.5 to 20 μm, preferably 1 to 10 μm.
当該島状構造には、下垂体ホルモン産生細胞を含んでいてもよい。具体的には、ACTH陽性細胞、PRL陽性細胞、FSH陽性細胞、LH陽性細胞、GH陽性細胞、TSH陽性細胞、MSH陽性細胞などが挙げられる。 The island structure may contain pituitary hormone-producing cells. Specific examples include ACTH-positive cells, PRL-positive cells, FSH-positive cells, LH-positive cells, GH-positive cells, TSH-positive cells, and MSH-positive cells.
「スポンジ状構造」とは、複数の島状構造が集まって形成され、島状構造の間に空隙(void)が存在する構造である。必ずしも全ての島状構造の間に空隙が存在していなくてもよい。空隙の存在によって培養液の栄養や酸素が当該細胞集団の内部にまで到達可能であるため、当該細胞集団の中心部においてもネクローシス細胞の割合は少ない。 A "sponge-like structure" is a structure formed by a group of multiple island structures with voids between the island structures. It is not necessary that there are voids between all of the island structures. The presence of voids allows nutrients and oxygen from the culture medium to reach the inside of the cell mass, so the proportion of necrotic cells is low even in the center of the cell mass.
「島状構造」及び「スポンジ状構造」は、典型的には、EpCAMによるソーティング前、すなわち多能性幹細胞を浮遊培養することによって得られる細胞凝集体には認められない。「島状構造」及び「スポンジ状構造」は、典型的には、EpCAMによるソーティング後の再凝集により初めて確認される構造である。 "Island-like structures" and "sponge-like structures" are typically not observed before sorting with EpCAM, i.e., in cell aggregates obtained by culturing pluripotent stem cells in suspension. "Island-like structures" and "sponge-like structures" are typically structures that are first identified upon re-aggregation after sorting with EpCAM.
一態様において、目的外細胞が含まれる割合を低下させた細胞集団のGH分泌能が、0.05~100ng/mlであってもよく、一態様として、0.1~50ng/ml、0.3~30ng/ml、0.5~10ng/mlであってもよい。 In one embodiment, the GH secretion ability of the cell population in which the proportion of non-target cells is reduced may be 0.05 to 100 ng/ml, and in another embodiment, may be 0.1 to 50 ng/ml, 0.3 to 30 ng/ml, or 0.5 to 10 ng/ml.
GH分泌能は、GH細胞数及び培養条件などの影響を受ける。GH分泌能は、特定の培養条件における、細胞集団(細胞凝集体)1個から培地中に分泌されたGHの濃度で表され得る。例えば、特定の培養条件とは、(i)下垂体ホルモン産生細胞の培養に用いることが可能な培地、好ましくはKSRを含む無血清培地(例:DMEM、MEM、GMEM、上皮細胞用培地(例:CnT-Prime epithelial culture medium)を用い、(ii)外部からGH産生に影響を与える物質(例:GH産生を上昇させる副腎皮質ホルモン類(例:デキサメサゾン))を添加せず、(iii)培地量を0.18~20mlに設定し、(iv)3~4日間培養する条件を意味する。当該条件における培養後の培地を回収し、当該培地中のGHの濃度を測定すればよい。当業者であれば、ELISA等の公知の手法を用いて、GHの濃度を測定することが可能である。 GH secretion ability is affected by the number of GH cells and culture conditions. GH secretion ability can be expressed as the concentration of GH secreted into the medium from one cell group (cell aggregate) under specific culture conditions. For example, the specific culture conditions refer to the following conditions: (i) a medium that can be used for culturing pituitary hormone-producing cells, preferably a serum-free medium containing KSR (e.g., DMEM, MEM, GMEM, epithelial cell medium (e.g., CnT-Prime epithelial culture medium) is used, (ii) no substances that affect GH production from the outside are added (e.g., adrenal cortical hormones that increase GH production (e.g., dexamethasone)), (iii) the medium volume is set to 0.18 to 20 ml, and (iv) the culture is performed for 3 to 4 days. The medium after culture under the above conditions is collected, and the GH concentration in the medium is measured. A person skilled in the art can measure the GH concentration using a known method such as ELISA.
目的外細胞が含まれる割合を低下させた細胞集団について、当該細胞集団の使用目的によって、特定の物質の存在下において培養することにより、特定の下垂体ホルモン産生の分泌量を増加させてもよい。 A cell population with a reduced proportion of non-target cells may be cultured in the presence of a specific substance depending on the intended use of the cell population to increase the amount of secretion of a specific pituitary hormone.
例えば、上記の方法において、(ii)の条件を、副腎皮質ホルモン類を含む培地で培養するという条件に変更することにより、GH分泌能は向上する。副腎皮質ホルモン類として、デキサメサゾン、ベタメタゾン、プレドニゾロン、メチルプレドニゾロン、トリアムシノロン等の人工的に合成された糖質コルチコイド;ハイドロコルチゾン、酢酸コルチゾン、酢酸フルドロコルチゾン等の天然糖質コルチコイド等が挙げられる。当業者であれば副腎皮質ホルモン類の濃度を適宜設定することが可能であるが、一態様として、デキサメサゾンの濃度は4ng/ml~40μg/ml、好ましくは40ng/ml~4μg/mlであってもよい。 For example, in the above method, the GH secretion ability is improved by changing the condition (ii) to a condition of culturing in a medium containing adrenal cortical hormones. Examples of adrenal cortical hormones include artificially synthesized glucocorticoids such as dexamethasone, betamethasone, prednisolone, methylprednisolone, and triamcinolone; and natural glucocorticoids such as hydrocortisone, cortisone acetate, and fludrocortisone acetate. A person skilled in the art can appropriately set the concentration of the adrenal cortical hormones, but in one embodiment, the concentration of dexamethasone may be 4 ng/ml to 40 μg/ml, preferably 40 ng/ml to 4 μg/ml.
副腎皮質ホルモン類存在下で培養した後の下垂体ホルモン産生細胞を含む細胞集団のGH分泌能は、一態様として、0.1~10,000ng/ml、0.5~5,000ng/ml、5~1,000ng/ml、20~500ng/mlであってよい。この場合、ACTH分泌能は、10~500,000pg/ml、好ましくは100~100,000pg/ml、より好ましくは500~30,000pg/ml、さらに好ましくは1,000~10,000pg/mlである。 In one embodiment, the GH secretion ability of the cell population containing pituitary hormone-producing cells after culturing in the presence of adrenal cortical hormones may be 0.1 to 10,000 ng/ml, 0.5 to 5,000 ng/ml, 5 to 1,000 ng/ml, or 20 to 500 ng/ml. In this case, the ACTH secretion ability is 10 to 500,000 pg/ml, preferably 100 to 100,000 pg/ml, more preferably 500 to 30,000 pg/ml, and even more preferably 1,000 to 10,000 pg/ml.
この様に、副腎皮質ホルモン類存在下で培養した後の下垂体ホルモン産生細胞を含む細胞集団は、該培養前より多くの下垂体ホルモン産生細胞を含む。一態様において、副腎皮質ホルモン類存在下で培養した場合、当該細胞集団中に存在する総細胞数に対するACTH陽性(産生)細胞数の割合(ACTH陽性細胞数の存在割合)は減ってもよい。その場合のACTH陽性細胞数の存在割合は、2%以上、好ましくは3%以上、より好ましくは5%以上である。 In this way, the cell population containing pituitary hormone-producing cells after culturing in the presence of corticosteroids contains more pituitary hormone-producing cells than before the culturing. In one embodiment, when cultured in the presence of corticosteroids, the ratio of the number of ACTH-positive (producing) cells to the total number of cells present in the cell population (the proportion of ACTH-positive cells present) may decrease. In that case, the proportion of ACTH-positive cells present is 2% or more, preferably 3% or more, and more preferably 5% or more.
上述のように、当該目的外細胞が含まれる割合を低下させた細胞集団(精製体)は、一態様において、以下の(i)~(vi)の特徴の少なくとも1つを有してもよい。
(i)当該細胞集団におけるEpCAM及びE-cadherinの少なくとも1つが陽性の細胞の存在割合が80%以上である。
(ii)当該細胞集団における神経系細胞の存在割合が20%以下である。
(iii)当該細胞集団におけるACTH陽性細胞の存在割合が5%以上である。
(iv)当該細胞集団の細胞密度が、3,000~20,000cells/mm2である。
(v)当該細胞集団の長径が200~3,000μmである。
(vi)当該細胞集団の表面において、EpCAM及びE-cadherinの少なくとも1つが陽性の細胞が互いに面接着し、上皮構造を形成している。
(vii)当該細胞集団が、スポンジ状構造を形成している。
As described above, the cell population (purified product) in which the proportion of undesired cells is reduced may, in one embodiment, have at least one of the following characteristics (i) to (vi).
(i) The proportion of cells positive for at least one of EpCAM and E-cadherin in the cell population is 80% or more.
(ii) The proportion of neural cells in the cell population is 20% or less.
(iii) The proportion of ACTH-positive cells in the cell population is 5% or more.
(iv) The cell density of the cell population is 3,000 to 20,000 cells/ mm2 .
(v) the major axis of the cell population is 200 to 3,000 μm.
(vi) On the surface of the cell population, cells positive for at least one of EpCAM and E-cadherin adhere to each other to form an epithelial structure.
(vii) The cell population forms a sponge-like structure.
更なる態様において、当該(i)~(vi)の特徴の少なくとも1つを有する細胞集団(精製体)は、以下の(viii)~(x)の特徴の少なくとも1つを有してもよい。
(viii)当該細胞集団における下垂体幹細胞の存在割合が3%以上である。(ix)当該細胞集団のACTH分泌能が、200~1,000,000pg/mlである。
(x)当該細胞集団のGH分泌能が、0.05~100ng/mlである。
In a further embodiment, the cell population (purified product) having at least one of the characteristics (i) to (vi) may also have at least one of the characteristics (viii) to (x) below.
(viii) the proportion of pituitary stem cells in the cell population is 3% or more, (ix) the ACTH secretion ability of the cell population is 200 to 1,000,000 pg/ml.
(x) The cell population has a GH secretion ability of 0.05 to 100 ng/ml.
当該目的外細胞が含まれる割合を低下させた細胞集団(精製体)は、一態様において、EpCAM陽性細胞及びE-cadherin陽性細胞の存在割合が80%以上であり、神経系細胞の存在割合が20%以下であり、かつ当該細胞集団のACTH分泌能が200~1,000,000pg/mlであるという特徴を有していてもよい。 In one embodiment, the cell population (purified body) in which the proportion of non-target cells is reduced may be characterized in that the proportion of EpCAM-positive cells and E-cadherin-positive cells is 80% or more, the proportion of nervous system cells is 20% or less, and the ACTH secretion ability of the cell population is 200 to 1,000,000 pg/ml.
本発明の治療薬は、皮下組織及び/又は筋肉組織に移植されるように用いられる。本明細書において、「皮下組織」とは、皮膚の3層構造の最も下方(すなわち内臓側)にある組織で、表皮と真皮を支持する組織をいう。また、「皮下組織」は、真皮と筋肉(筋膜を含む)に挟まれた組織ともいえる。本明細書において、「筋肉組織」とは、皮下組織下にある組織であり、筋肉と筋膜を含む組織をいう。筋肉は、横紋筋である骨格筋と心筋、及び平滑筋に大別される。本明細書において、「皮下組織及び/又は筋肉組織」という部位は、「真皮よりも深層に位置する部位」あるいは「皮下組織から筋肉までを含む部位」と互換的に用い得る。 The therapeutic agent of the present invention is used to be implanted into subcutaneous tissue and/or muscle tissue. In this specification, "subcutaneous tissue" refers to the tissue located at the bottom of the three-layer structure of the skin (i.e., on the visceral side) and supporting the epidermis and dermis. "Subcutaneous tissue" can also be said to be tissue sandwiched between the dermis and muscle (including fascia). In this specification, "muscle tissue" refers to tissue located below the subcutaneous tissue and including muscle and fascia. Muscle is broadly divided into skeletal muscle, which is striated muscle, cardiac muscle, and smooth muscle. In this specification, the site "subcutaneous tissue and/or muscle tissue" can be used interchangeably with "site located deeper than the dermis" or "site including from the subcutaneous tissue to the muscle."
本発明の治療薬は、皮下組織及び/又は筋肉組織に移植されるように用いられれば、皮下組織や筋肉組織の種類や移植部位は特に限定されない。本発明の治療薬が皮下組織に移植されるように用いられる場合、好ましくは、皮下脂肪組織、具体的には、褐色脂肪組織又は白色脂肪組織、より好ましくは、白色脂肪組織に用いられる。また、該皮下脂肪組織は、外科的に細胞の移植が可能な部位の皮下脂肪組織であれば特に限定はないが、好ましくは、鼠蹊部、腋下、背中及び腹部から選択される少なくとも1種類の部位に存在する皮下脂肪組織である。一態様として、本発明の治療薬は、鼠蹊部の白色脂肪組織に移植されるように用いられる。また、一態様として、本発明の治療薬は、腋下の白色脂肪組織に移植されるように用いられる。
皮下組織、特に皮下脂肪組織、より特に白色脂肪組織では、血管網が形成されているため、移植部位の観点からも、本発明の治療薬の移植後、下垂体ホルモン産生細胞の周辺において血管新生が生じ得る。また、下垂体ホルモン産生細胞の周辺において血管新生が生じることで、下垂体ホルモン産生細胞の皮下組織内への生着が促進され得る。該生着により、下垂体ホルモンが血管に分泌され得る。該生着の観点等から、一態様では、本発明の治療薬において、下垂体ホルモン産生細胞が、皮下組織中の血管付近に移植されるように用いられる。
The therapeutic agent of the present invention is not particularly limited to the type of subcutaneous tissue or muscle tissue or the site of transplantation, as long as it is used to be transplanted into subcutaneous tissue and/or muscle tissue. When the therapeutic agent of the present invention is used to be transplanted into subcutaneous tissue, it is preferably used in subcutaneous adipose tissue, specifically brown adipose tissue or white adipose tissue, more preferably white adipose tissue. In addition, the subcutaneous adipose tissue is not particularly limited as long as it is a subcutaneous adipose tissue at a site where cells can be surgically transplanted, but is preferably subcutaneous adipose tissue present at at least one site selected from the groin, armpit, back, and abdomen. In one embodiment, the therapeutic agent of the present invention is used to be transplanted into white adipose tissue in the groin. In another embodiment, the therapeutic agent of the present invention is used to be transplanted into white adipose tissue in the armpit.
Since a vascular network is formed in subcutaneous tissue, particularly in subcutaneous adipose tissue, more particularly in white adipose tissue, from the viewpoint of the transplantation site, angiogenesis may occur around the pituitary hormone-producing cells after transplantation of the therapeutic agent of the present invention. In addition, angiogenesis around the pituitary hormone-producing cells may promote the survival of the pituitary hormone-producing cells in the subcutaneous tissue. This survival allows the pituitary hormones to be secreted into the blood vessels. From the viewpoint of survival, in one embodiment, the therapeutic agent of the present invention is used such that the pituitary hormone-producing cells are transplanted near the blood vessels in the subcutaneous tissue.
また、本発明の治療薬が筋肉組織に移植されるように用いられる場合、横紋筋及び平滑筋のいずれに移植されるように用いられてもよく、特に限定されないが、一態様として、横紋筋、特に骨格筋が挙げられる。骨格筋は、典型的には、両端に比較的長い腱組織を有し、骨への付着面積が比較的少ない表層部に位置する筋肉(表層筋(superficial muscle))と、広範囲に渡って骨に直接付着し、少ない腱組織を有する深層部に位置する筋肉(深層筋(deep muscle))から構成される。本発明の治療薬を骨格筋に用いる場合、表層筋又は深層筋のいずれに移植してもよく、深層筋と表層筋の間に移植してもよい。本発明の治療薬が筋肉組織に移植されるように用いられる場合、移植部位としては、例えば、背中(腰部を含む)、腹部、大腿部、臀部、上腕部、胸部などの部位に存在する筋肉組織が挙げられる。筋肉組織は、上述する皮下組織と同様に血管網が形成されているため、移植部位の観点からも、本発明の治療薬の移植後、下垂体ホルモン産生細胞の周辺において血管新生が生じ得る。また、下垂体ホルモン産生細胞の周辺において血管新生が生じることで、下垂体ホルモン産生細胞の筋肉組織内への生着が促進され得る。該生着により、下垂体ホルモンが血管に分泌され得る。生着の観点等から、一態様では、本発明の治療薬において、下垂体ホルモン産生細胞が、筋肉組織中の血管付近に移植されるように用いられる。 In addition, when the therapeutic agent of the present invention is used to be transplanted into muscle tissue, it may be used to be transplanted into either striated muscle or smooth muscle, and is not particularly limited, but one embodiment is striated muscle, particularly skeletal muscle. Skeletal muscle is typically composed of muscles (superficial muscles) located in the superficial layer, which have relatively long tendon tissue at both ends and a relatively small attachment area to bone, and muscles (deep muscles) located in the deep layer, which directly attach to bone over a wide area and have little tendon tissue. When the therapeutic agent of the present invention is used to be transplanted into skeletal muscle, it may be transplanted into either superficial muscle or deep muscle, or between deep muscle and superficial muscle. When the therapeutic agent of the present invention is used to be transplanted into muscle tissue, examples of the transplant site include muscle tissue present in areas such as the back (including the lower back), abdomen, thighs, buttocks, upper arms, and chest. Since muscle tissue has a vascular network formed in the same manner as the subcutaneous tissue described above, from the viewpoint of the transplantation site, angiogenesis may occur around the pituitary hormone-producing cells after transplantation of the therapeutic agent of the present invention. Furthermore, angiogenesis around the pituitary hormone-producing cells may promote the engraftment of the pituitary hormone-producing cells into the muscle tissue. This engraftment may cause the pituitary hormones to be secreted into the blood vessels. From the viewpoint of engraftment, in one embodiment, the therapeutic agent of the present invention is used such that the pituitary hormone-producing cells are transplanted near the blood vessels in the muscle tissue.
本発明の治療薬を皮下組織及び/又は筋肉組織に移植されるように用いる場合、一態様では、下垂体ホルモン産生細胞が、該組織中の人工的に設けられた空間(ポケット)に移植されるように用いられることを特徴とする。該空間は、本発明の治療薬に含まれる細胞凝集体(細胞集団)の大きさや個数に応じて、適宜、その大きさを変更してもよい。人工的に設けられた空間の大きさとしては、典型的には、0.001~50cm3が挙げられる。より具体的には、例えば、下垂体オルガノイド(例:2~5mm径)5個の移植を企図する場合、空間の大きさとしては、1~15cm3が挙げられる。また、例えば、上述の精製体(例:0.5~1mm径)の1又は複数個の移植を企図する場合、人工的に設けられた空間は、所望する数の精製体だけが入る程度の空間が好ましく、皮下組織や筋肉組織中の(小)血管の直下であることも好ましい。さらに本発明の治療薬は、「8.下垂体ホルモン産生細胞を含む組成物及び治療方法」に後述するように、血管新生促進剤(例:bFGFなど)と組み合わせてもよい。 When the therapeutic agent of the present invention is used to be transplanted into subcutaneous tissue and/or muscle tissue, in one embodiment, the pituitary hormone-producing cells are used to be transplanted into an artificially created space (pocket) in the tissue. The size of the space may be changed appropriately depending on the size and number of cell aggregates (cell populations) contained in the therapeutic agent of the present invention. The size of the artificially created space is typically 0.001 to 50 cm 3. More specifically, for example, when transplanting five pituitary organoids (e.g., 2 to 5 mm diameter), the size of the space may be 1 to 15 cm 3. In addition, for example, when transplanting one or more of the above-mentioned purified bodies (e.g., 0.5 to 1 mm diameter), the artificially created space is preferably a space that can accommodate only the desired number of purified bodies, and is also preferably directly under a (small) blood vessel in the subcutaneous tissue or muscle tissue. Furthermore, the therapeutic agent of the present invention may be combined with an angiogenesis promoter (e.g., bFGF, etc.) as described below in "8. Compositions containing pituitary hormone-producing cells and therapeutic methods."
本明細書において、「下垂体の障害に基づく疾患」とは、下垂体の障害に基づく動物の疾患であってよく、下垂体の障害に基づく非ヒト動物の疾患であってもよい。「下垂体の障害に基づく疾患」としては、具体的には、汎下垂体機能低下症、下垂体性小人症、成人成長ホルモン分泌不全症、副腎皮質機能低下症、部分的下垂体機能低下症、下垂体前葉ホルモン単独欠損症、下垂体性性腺機能低下症、自己免疫性視床下部下垂体炎や、下垂体腫瘍・その他腫瘍などの手術後の下垂体機能・ホルモン分泌不全等が挙げられる。
動物としては、哺乳動物が好ましく、げっ歯類、有蹄類、ネコ目、ウサギ目、霊長類等が包含される。げっ歯類には、マウス、ラット、ハムスター、モルモット等が包含される。有蹄類には、ブタ、ウシ、ヤギ、ウマ、ヒツジ等が包含される。ネコ目には、イヌ、ネコ等が包含される。ウサギ目には、ウサギ等が含包される。霊長類とは、霊長目に属する哺乳動物をいい、霊長類としては、キツネザル、ロリス、ツバイ等の原猿亜目、及びサル、類人猿、ヒト等の真猿亜目が含まれる。
In the present specification, the term "diseases caused by pituitary gland disorders" may refer to animal diseases caused by pituitary gland disorders, or non-human animal diseases caused by pituitary gland disorders. Specific examples of "diseases caused by pituitary gland disorders" include panhypopituitarism, pituitary dwarfism, adult growth hormone deficiency, adrenal insufficiency, partial hypopituitarism, isolated anterior pituitary hormone deficiency, pituitary hypogonadism, autoimmune hypothalamic hypophysitis, and pituitary function and hormone secretion deficiency after surgery for pituitary tumors and other tumors.
The animal is preferably a mammal, and includes rodents, ungulates, felines, lagomorphs, primates, etc. Rodents include mice, rats, hamsters, guinea pigs, etc. Ungulates include pigs, cows, goats, horses, sheep, etc. Felidae include dogs, cats, etc. Lagomorphs include rabbits, etc. Primates refer to mammals belonging to the order Primates, and include prosimians such as lemurs, lorises, and tree shrews, and anthropoids such as monkeys, apes, and humans.
本発明の治療薬に含有される下垂体ホルモン産生細胞、下垂体ホルモン産生細胞を含む細胞凝集体、又は二次元細胞シートは、上述の通り、その由来や製造方法等に関して、上述のようなホルモンを産生し得る限り特に限定されないが、例えば、国際公開第2013/065763号、国際公開第2016/013669号、国際公開第2019/103129号、PCT/JP2022/36019、Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351)、Kasai T, et al. Hypothalamic contribution to pituitary functions is recapitulated in vitro using 3D-cultured human iPS cells (Cell Rep. 2020; 30: 18-24)、Suga H, et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature (2011; 480: 57-62)、Taga S, et al. Cell aggregate including pituitary hormone-producing cell, and method (JP Patent applications 2022-116715)、Nakano T, et al. Method for producing cell mass including pituitary tissue, and cell mass thereof (JP Patent applications 2022-116716)、Eiraku M, et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals (Cell Stem Cell. 2008; 3: 519-532)等に記載の公知の方法やこれらの方法の組み合わせ等により製造し得る。 As described above, the pituitary hormone-producing cells, cell aggregates containing pituitary hormone-producing cells, or two-dimensional cell sheets contained in the therapeutic agent of the present invention are not particularly limited in terms of their origin or production method, etc., as long as they are capable of producing the hormones described above. For example, the pituitary hormone-producing cells, cell aggregates containing pituitary hormone-producing cells, and two-dimensional cell sheets described in WO 2013/065763, WO 2016/013669, WO 2019/103129, PCT/JP2022/36019, Ozone C, et al. Functional anterior pituitary generated in self-organizing cult ure of human embryonic stem cells (Nat Commun. 2016; 7: 10351), Kasai T, et al. 4), Suga H, et al. Self-formation of functional ade nohypophysis in three-dimensional culture. Nature (2011; 480: 57-62), Taga S, et al. Cell aggregate including pituitary hormone-producing cell, and method (JP Patent applications 2022-116715), Nakano T, et al. Method for producing cell mass including pituitary tis They can be produced by known methods such as those described in (JP Patent applications 2022-116716), Eiraku M, et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals (Cell Stem Cell. 2008; 3: 519-532), or a combination of these methods.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞は、後述するようにPCT/JP2022/36019に記載の方法により製造してもよい。 In one embodiment, the pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be produced by the method described in PCT/JP2022/36019, as described below.
2.細胞培養
本明細書において、「幹細胞」とは、分化能及び増殖能(特に自己複製能)を有する未分化な細胞を意味する。幹細胞には、分化能力に応じて、多能性幹細胞(pluripotent stem cell)、複能性幹細胞(multipotent stemcell)、単能性幹細胞(unipotent stem cell)等が含まれる。「多能性幹細胞」とは、インビトロにおいて培養することが可能で、かつ、生体を構成する全ての細胞に分化し得る能力(分化多能性:pluripotency)を有する幹細胞をいう。全ての細胞とは、外胚葉、中胚葉及び内胚葉の三胚葉由来の細胞である。「複能性幹細胞」とは、全ての種類ではないが、複数種の組織や細胞へ分化し得る能力を有する幹細胞を意味する。「単能性幹細胞」とは、特定の組織や細胞へ分化し得る能力を有する幹細胞を意味する。
2. Cell Culture In this specification, "stem cells" refer to undifferentiated cells that have differentiation and proliferation capabilities (particularly self-renewal capabilities). Stem cells include pluripotent stem cells, multipotent stem cells, unipotent stem cells, etc., depending on their differentiation capabilities. "Pluripotent stem cells" refer to stem cells that can be cultured in vitro and have the ability to differentiate into all cells that constitute a living body (pluripotency). All cells are cells derived from the three germ layers of ectoderm, mesoderm, and endoderm. "Multipotent stem cells" refer to stem cells that have the ability to differentiate into multiple types of tissues and cells, although not all types. "Unipotent stem cells" refer to stem cells that have the ability to differentiate into specific tissues and cells.
多能性幹細胞は、受精卵、クローン胚、生殖幹細胞、組織内幹細胞、体細胞等から誘導することができる。多能性幹細胞としては、胚性幹細胞(ES細胞:Embryonic stem cell)、EG細胞(Embryonic germ cell)、人工多能性幹細胞(iPS細胞:induced pluripotent stem cell)等を挙げることができる。間葉系幹細胞(mesenchymal stem cell:MSC)から得られるMuse細胞(Multi-lineage differentiating Stress Enduring cell)、及び生殖細胞(例えば精巣)から作製されるGS細胞も多能性幹細胞に包含される。なお、ヒト胚性幹細胞は、受精14日以内のヒト胚から樹立されたものである。 Pluripotent stem cells can be derived from fertilized eggs, cloned embryos, germline stem cells, tissue stem cells, somatic cells, etc. Examples of pluripotent stem cells include embryonic stem cells (ES cells), EG cells (embryonic germ cells), and induced pluripotent stem cells (iPS cells). Muse cells (multi-lineage differentiating stress ending cells) obtained from mesenchymal stem cells (MSCs), and GS cells produced from germ cells (e.g. testes) are also included in pluripotent stem cells. Human embryonic stem cells are established from human embryos within 14 days of fertilization.
胚性幹細胞は、1981年に初めて樹立され、1989年以降ノックアウトマウス作製にも応用されている。1998年にはヒト胚性幹細胞が樹立されており、再生医学にも利用されつつある。ES細胞は、内部細胞集団をフィーダー細胞上又はleukemia inhibitory factor(LIF)を含む培地中で培養することにより製造することができる。ES細胞の製造方法は、例えば国際公開第96/22362号、国際公開第02/101057号、米国特許第5843780号明細書、米国特許第6200806号明細書、米国特許第6280718号明細書等に記載されている。胚性幹細胞は、所定の機関より入手でき、市販品を購入することもできる。例えばヒト胚性幹細胞であるKhES-1、KhES-2及びKhES-3は、京都大学再生医科学研究所より入手可能である。いずれもマウス胚性幹細胞である、EB5細胞は国立研究開発法人理化学研究所より、D3株はAmerican Type Culture Collection(ATCC)より、入手可能である。ES細胞の1つである核移植ES細胞(ntES細胞)は、細胞核を取り除いた卵子に体細胞の細胞核を移植して作ったクローン胚から樹立することができる。 Embryonic stem cells were first established in 1981, and have been used to create knockout mice since 1989. Human embryonic stem cells were established in 1998, and are also being used in regenerative medicine. ES cells can be produced by culturing the inner cell population on feeder cells or in a medium containing leukemia inhibitory factor (LIF). Methods for producing ES cells are described in, for example, International Publication No. 96/22362, International Publication No. 02/101057, U.S. Patent No. 5,843,780, U.S. Patent No. 6,200,806, and U.S. Patent No. 6,280,718, etc. Embryonic stem cells are available from designated institutions, and can also be purchased commercially. For example, human embryonic stem cells KhES-1, KhES-2, and KhES-3 are available from the Institute for Frontier Medical Sciences, Kyoto University. Both are mouse embryonic stem cells; EB5 cells are available from the RIKEN Institute, a national research and development agency, and the D3 line is available from the American Type Culture Collection (ATCC). Nuclear transfer ES cells (ntES cells), which are a type of ES cell, can be established from a cloned embryo created by transplanting the nucleus of a somatic cell into an egg from which the nucleus has been removed.
EG細胞は、始原生殖細胞をマウス幹細胞因子(mSCF)、LIF及び塩基性線維芽細胞増殖因子(bFGF)を含む培地中で培養することにより製造することができる(Cell,70:841-847,1992)。 EG cells can be produced by culturing primordial germ cells in a medium containing mouse stem cell factor (mSCF), LIF, and basic fibroblast growth factor (bFGF) (Cell, 70:841-847, 1992).
「人工多能性幹細胞」とは、体細胞を公知の方法等により初期化(reprogramming)することにより、多能性を誘導した細胞である。人工多能性幹細胞としては、具体的には線維芽細胞、末梢血単核球等に分化した体細胞をOct3/4、Sox2、Klf4、Myc(c-Myc、N-Myc、L-Myc)、Glis1、Nanog、Sall4、lin28、Esrrb等を含む初期化遺伝子群から選ばれる複数の遺伝子の発現により初期化して多分化能を誘導した細胞が挙げられる。2006年、山中らによりマウス細胞で人工多能性幹細胞が樹立された(Cell,2006,126(4)pp.663-676)。人工多能性幹細胞は、2007年にヒト線維芽細胞でも樹立され、胚性幹細胞と同様に多能性と自己複製能を有する(Cell,2007,131(5)pp.861-872;Science,2007,318(5858)pp.1917-1920;Nat.Biotechnol.,2008,26(1)pp.101-106)。人工多能性幹細胞として、遺伝子発現による直接初期化で製造する方法以外に、化合物の添加などにより体細胞より人工多能性幹細胞を誘導することもできる(Science,2013,341,pp.651-654)。 "Induced pluripotent stem cells" are cells in which pluripotency has been induced by reprogramming somatic cells using known methods. Specific examples of induced pluripotent stem cells include cells in which pluripotency has been induced by reprogramming somatic cells differentiated into fibroblasts, peripheral blood mononuclear cells, etc., through the expression of multiple genes selected from a group of reprogramming genes including Oct3/4, Sox2, Klf4, Myc (c-Myc, N-Myc, L-Myc), Glis1, Nanog, Sall4, lin28, Esrrb, etc. In 2006, Yamanaka et al. established induced pluripotent stem cells in mouse cells (Cell, 2006, 126(4) pp.663-676). In 2007, artificial pluripotent stem cells were established from human fibroblasts, and like embryonic stem cells, they have pluripotency and the ability to self-replicate (Cell, 2007, 131(5) pp.861-872; Science, 2007, 318(5858) pp.1917-1920; Nat. Biotechnol., 2008, 26(1) pp.101-106). In addition to producing artificial pluripotent stem cells by direct reprogramming through gene expression, artificial pluripotent stem cells can also be induced from somatic cells by adding compounds (Science, 2013, 341, pp.651-654).
人工多能性幹細胞を製造する際に用いられる体細胞としては、特に限定は無いが、組織由来の線維芽細胞、血球系細胞(例えば末梢血単核球、T細胞等)、肝細胞、膵臓細胞、腸上皮細胞、平滑筋細胞等が挙げられる。 Somatic cells used in producing induced pluripotent stem cells are not particularly limited, but include tissue-derived fibroblasts, blood cells (e.g., peripheral blood mononuclear cells, T cells, etc.), liver cells, pancreatic cells, intestinal epithelial cells, smooth muscle cells, etc.
人工多能性幹細胞を製造する際に、数種類の遺伝子(例えばOct3/4、Sox2、Klf4及びMycの4因子)の発現により初期化する場合、遺伝子を発現させるための手段は特に限定されない。遺伝子を発現させるための手段としては、例えばウイルスベクター(例えば、レトロウイルスベクター、レンチウイルスベクター、センダイウイルスベクター、アデノウイルスベクター、アデノ随伴ウイルスベクター)を用いた感染法、プラスミドベクター(例えばプラスミドベクター、エピソーマルベクター)を用いた遺伝子導入法(例えばリン酸カルシウム法、リポフェクション法、レトロネクチン法、エレクトロポレーション法)、RNAベクターを用いた遺伝子導入法(例えばリン酸カルシウム法、リポフェクション法、エレクトロポレーション法)、タンパク質の直接注入法等が挙げられる。 When producing induced pluripotent stem cells, if initialization is performed by expression of several types of genes (e.g., four factors: Oct3/4, Sox2, Klf4, and Myc), the means for expressing the genes is not particularly limited. Examples of means for expressing genes include infection methods using viral vectors (e.g., retroviral vectors, lentiviral vectors, Sendai virus vectors, adenoviral vectors, and adeno-associated virus vectors), gene transfer methods using plasmid vectors (e.g., plasmid vectors, episomal vectors) (e.g., calcium phosphate method, lipofection method, retronectin method, and electroporation method), gene transfer methods using RNA vectors (e.g., calcium phosphate method, lipofection method, and electroporation method), and direct protein injection methods.
また、株化された人工多能性幹細胞を入手する事も可能であり、例えば、京都大学で樹立された201B7細胞、201B7-Ff細胞、253G1細胞、253G4細胞、1201C1細胞、1205D1細胞、1210B2細胞、1231A3細胞等のヒト人工多能性細胞株が、京都大学及びiPSアカデミアジャパン株式会社より入手可能である。株化された人工多能性幹細胞として、例えば、京都大学で樹立されたFf-I01細胞、Ff-I14細胞及びQHJI01s04細胞が、京都大学より入手可能である。 It is also possible to obtain established induced pluripotent stem cell lines; for example, human induced pluripotent cell lines such as 201B7 cells, 201B7-Ff cells, 253G1 cells, 253G4 cells, 1201C1 cells, 1205D1 cells, 1210B2 cells, and 1231A3 cells established at Kyoto University are available from Kyoto University and iPS Academia Japan Inc. As established induced pluripotent stem cell lines, for example, Ff-I01 cells, Ff-I14 cells, and QHJI01s04 cells established at Kyoto University are available from Kyoto University.
多能性幹細胞は、遺伝子改変されていてもよい。遺伝子改変された多能性幹細胞は、例えば相同組換え技術を用いることにより作製できる。改変される染色体上の遺伝子としては、例えば細胞マーカー遺伝子、組織適合性抗原の遺伝子、神経系細胞の障害に基づく疾患関連遺伝子などが挙げられる。染色体上の標的遺伝子の改変は、Manipulating the Mouse Embryo,A Laboratory Manual,Second Edition,Cold Spring Harbor Laboratory Press(1994)、Gene Targeting,A Practical Approach,IRL Press at Oxford University Press(1993)、バイオマニュアルシリーズ8、ジーンターゲッティング、ES細胞を用いた変異マウスの作製、羊土社(1995)等に記載の方法を用いて行うことができる。
Pluripotent stem cells may be genetically modified. Genetically modified pluripotent stem cells can be produced, for example, by using homologous recombination techniques. Genes on chromosomes that can be modified include, for example, cell marker genes, histocompatibility antigen genes, and disease-related genes based on disorders of nervous system cells. Modification of target genes on chromosomes can be carried out using methods described in Manipulating the Mouse Embryo, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1994), Gene Targeting, A Practical Approach, IRL Press at Oxford University Press (1993),
具体的には、例えば改変する標的遺伝子(例えば細胞マーカー遺伝子、組織適合性抗原の遺伝子や疾患関連遺伝子等)のゲノム遺伝子を単離し、単離されたゲノム遺伝子を用いて標的遺伝子を相同組換えするためのターゲットベクターを作製する。作製されたターゲットベクターを幹細胞に導入し、標的遺伝子とターゲットベクターの間で相同組換えを起こした細胞を選択することにより、染色体上の遺伝子が改変された幹細胞を作製することができる。 Specifically, for example, the genomic gene of the target gene to be modified (e.g., a cell marker gene, a gene for a histocompatibility antigen, or a disease-related gene) is isolated, and a target vector is prepared using the isolated genomic gene for homologous recombination of the target gene. The target vector thus prepared is introduced into stem cells, and cells in which homologous recombination has occurred between the target gene and the target vector are selected, thereby producing stem cells in which genes on chromosomes have been modified.
標的遺伝子のゲノム遺伝子を単離する方法としては、Molecular Cloning,A Laboratory Manual,Second Edition,Cold Spring Harbor Laboratory Press(1989)やCurrent Protocols in Molecular Biology,John Wiley&Sons(1987-1997)等に記載された公知の方法が挙げられる。ゲノムDNAライブラリースクリーニングシステム(Genome Systems製)やUniversal GenomeWalker Kits(Clontech製)などを用いることができる。 Methods for isolating the genomic gene of the target gene include known methods described in Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989) and Current Protocols in Molecular Biology, John Wiley & Sons (1987-1997). A genomic DNA library screening system (manufactured by Genome Systems) or Universal GenomeWalker Kits (manufactured by Clontech) can also be used.
標的遺伝子を相同組換えするためのターゲットベクターの作製、及び相同組換え体の効率的な選別は、Gene Targeting,A Practical Approach,IRL Press at Oxford University Press(1993)、バイオマニュアルシリーズ8、ジーンターゲッティング、ES細胞を用いた変異マウスの作製、羊土社(1995)等に記載の方法に従って行うことができる。ターゲットベクターは、リプレースメント型又はインサーション型のいずれでも用いることができる。選別方法としては、ポジティブ選択、プロモーター選択、ネガティブ選択又はポリA選択などの方法を用いることができる。選別した細胞株の中から目的とする相同組換え体を選択する方法としては、ゲノムDNAに対するサザンハイブリダイゼーション法やPCR法等が挙げられる。
The preparation of a target vector for homologous recombination of a target gene and efficient selection of a homologous recombinant can be performed according to the methods described in Gene Targeting, A Practical Approach, IRL Press at Oxford University Press (1993),
多能性幹細胞として、ゲノム編集を行った多能性幹細胞を用いることもできる。「ゲノム編集」とは、ヌクレアーゼを用いた部位特異的なゲノムDNA鎖の切断、又は塩基の化学的変換等の原理により標的遺伝子もしくはゲノム領域を意図的に改変する技術である。部位特異的ヌクレアーゼとしては、ジンクフィンガーヌクレアーゼ(ZFN)、TALEN、CRISPR/Cas9等が挙げられる。ゲノム編集技術を用いることにより、特定の遺伝子を欠失したノックアウト細胞株、特定の遺伝子座に人工的に別の配列を挿入したノックイン細胞株等を作製することができる。 Pluripotent stem cells that have undergone genome editing can also be used as pluripotent stem cells. "Genome editing" is a technique for intentionally modifying target genes or genomic regions using principles such as site-specific cleavage of genomic DNA strands using nucleases or chemical conversion of bases. Examples of site-specific nucleases include zinc finger nucleases (ZFNs), TALENs, and CRISPR/Cas9. Using genome editing technology, it is possible to create knockout cell lines in which specific genes have been deleted, knock-in cell lines in which a different sequence has been artificially inserted into a specific gene locus, etc.
一態様において、本発明に用いる多能性幹細胞は、哺乳動物の多能性幹細胞であり、本発明の治療薬に用いる下垂体ホルモン産生細胞を製造する場合には、移植する対象と同一の哺乳動物の多能性幹細胞であることが望ましい。例えば、ヒトを対象に移植される下垂体ホルモン産生細胞を製造するためには、ヒトの多能性幹細胞を使用する。
一態様において、本発明における多能性幹細胞は、担ヒト下垂体組織モデル非ヒト動物を作製する場合には、ヒトの多能性幹細胞を使用する。
「細胞接着(Cell adhesion)」には、細胞と細胞との接着(細胞-細胞接着)及び細胞と細胞外マトリクス(基質)との接着(細胞-基質接着)が含まれる。インビトロの人工培養環境下で生じる、細胞の培養器材等への接着も細胞接着に含有される。細胞-細胞接着において形成される結合は細胞-細胞結合(cell-cell junction)であり、細胞-基質接着において形成される結合は細胞-基質結合(cell-substratum junction)である。細胞接着の種類として、例えば固定結合(anchoring junction)、連絡結合(communicating junction)、閉鎖結合(occluding junction)が挙げられる。
In one embodiment, the pluripotent stem cells used in the present invention are mammalian pluripotent stem cells, and when producing pituitary hormone-producing cells for use in the therapeutic agent of the present invention, it is preferable that the pluripotent stem cells are the same mammalian pluripotent stem cells as the subject to be transplanted. For example, to produce pituitary hormone-producing cells to be transplanted into a human subject, human pluripotent stem cells are used.
In one embodiment, when a human pituitary tissue-bearing non-human animal model is produced, the pluripotent stem cells used in the present invention are human pluripotent stem cells.
"Cell adhesion" includes adhesion between cells (cell-cell adhesion) and adhesion between cells and extracellular matrix (substrate) (cell-substrate adhesion). Cell adhesion also includes adhesion of cells to culture equipment and the like that occurs in an in vitro artificial culture environment. The bond formed in cell-cell adhesion is a cell-cell junction, and the bond formed in cell-substrate adhesion is a cell-substrate junction. Types of cell adhesion include, for example, anchoring junction, communicating junction, and occluding junction.
細胞-細胞結合として、「密着結合(tight junction)」、「接着結合(adherence junction)」が挙げられる。密着結合は比較的強い細胞-細胞結合であり、上皮細胞で生じ得る。細胞間に密着結合が存在しているかどうかは、例えば密着接合の構成成分に対する抗体(抗クローディン抗体、抗ZO-1抗体等)を用いた免疫組織化学等の手法により検出することができる。 Cell-cell junctions include "tight junctions" and "adherence junctions." Tight junctions are relatively strong cell-cell junctions that can occur in epithelial cells. The presence of tight junctions between cells can be detected by techniques such as immunohistochemistry using antibodies against components of tight junctions (anti-claudin antibodies, anti-ZO-1 antibodies, etc.).
「浮遊培養」とは、細胞が培養液に浮遊して存在する状態を維持しつつ培養することをいう。すなわち浮遊培養は、細胞を培養器材及び培養器材上のフィーダー細胞等(以下、「培養器材等」と記す。)に接着させない条件で行われ、培養器材等に接着させる条件で行われる培養(接着培養)とは区別される。より詳細には、浮遊培養とは、細胞と培養器材等との間に、強固な細胞-基質結合を作らせない条件での培養をいう。培養している細胞が浮遊培養状態であるか接着培養であるかの判別は、例えば顕微鏡観察時の培養器材の揺動等によって当業者であれば容易に可能である。 "Suspension culture" refers to culturing cells while maintaining a state in which they exist suspended in the culture solution. In other words, suspension culture is carried out under conditions that do not allow cells to adhere to the cultureware and feeder cells, etc. on the cultureware (hereinafter referred to as "cultureware, etc."), and is distinguished from culture carried out under conditions that allow cells to adhere to the cultureware, etc. (adherent culture). More specifically, suspension culture refers to culture under conditions that do not allow strong cell-substrate bonds to be formed between the cells and the cultureware, etc. Those skilled in the art can easily distinguish whether the cultured cells are in a suspension culture state or an adherent culture state, for example, by rocking the cultureware during microscopic observation.
「接着培養」とは、細胞が培養器材等に接着して存在する状態を維持しつつ培養することをいう。ここで、細胞が培養器材等に接着するとは、例えば細胞と培養器材等との間に細胞接着の一種である強固な細胞-基質結合ができることをいう。 "Adherent culture" refers to culturing cells while maintaining a state in which the cells are attached to culture equipment, etc. Here, cells adhering to culture equipment, etc. refers to, for example, the formation of strong cell-substrate bonds, which is a type of cell adhesion, between the cells and the culture equipment, etc.
浮遊培養中の細胞凝集体においては、細胞と細胞とが面接着する。浮遊培養中の細胞凝集体では、細胞と培養器材等との間に、強固な細胞-基質結合は形成されておらず、細胞-基質結合はほとんど形成されないか、形成されていてもその寄与が小さい。浮遊培養中の細胞凝集体の内部には、内在の細胞-基質結合が存在してもよい。「細胞と細胞とが面接着(plane attachment)する」とは、細胞と細胞とが面で接着することをいう。より詳細には、「細胞と細胞とが面接着する」とは、ある細胞の表面積のうち別の細胞の表面と接着している割合が、例えば1%以上、好ましくは3%以上、より好ましくは5%以上であることをいう。細胞の表面は、膜を染色する試薬(例えばDiI)による染色、細胞接着因子(例えば、E-cadherin、N-cadherin等)の免疫染色等により観察できる。 In cell aggregates during suspension culture, cells adhere to each other through a plane. In cell aggregates during suspension culture, strong cell-substrate bonds are not formed between the cells and the cultureware, and cell-substrate bonds are hardly formed, or even if they are formed, their contribution is small. Internal cell-substrate bonds may exist inside cell aggregates during suspension culture. "Plane attachment between cells" refers to cells adhering to each other through a plane. More specifically, "plane attachment between cells" refers to the proportion of the surface area of a cell that is attached to the surface of another cell, for example, 1% or more, preferably 3% or more, and more preferably 5% or more. The cell surface can be observed by staining with a membrane-staining reagent (e.g., DiI) or immunostaining with cell adhesion factors (e.g., E-cadherin, N-cadherin, etc.).
接着培養を行う際に用いられる培養器材は、接着培養することが可能なものであれば特に限定されず、当業者であれば適宜決定することが可能である。このような培養器材としては、例えばフラスコ、組織培養用フラスコ、ディッシュ、組織培養用ディッシュ、マルチディッシュ、マイクロプレート、マイクロウェルプレート、マイクロポア、マルチプレート、マルチウェルプレート、チャンバースライド、シャーレ、チューブ、トレイ、培養バック及びオーガンオンチップ等の生体機能チップが挙げられる。細胞接着性の培養器材としては、細胞との接着性を向上させる目的で培養器材の表面が人工的に処理されているもの等を使用できる。人工的な処理とは、例えば細胞外マトリクス、高分子等によるコーティング処理、及びガスプラズマ処理、正電荷処理等の表面加工が挙げられる。細胞が接着される細胞外マトリクスとしては、例えば基底膜標品、ラミニン、エンタクチン、コラーゲン、ゼラチン等が挙げられる。高分子としては、ポリリジン、ポリオルニチン等が挙げられる。培養器材の培養面は、平底でもよいし、凹凸があってもよい。 The cultureware used for adhesion culture is not particularly limited as long as it is capable of adhesion culture, and a person skilled in the art can appropriately determine the cultureware. Examples of such cultureware include flasks, flasks for tissue culture, dishes, dishes for tissue culture, multi-dishes, microplates, microwell plates, micropores, multi-plates, multi-well plates, chamber slides, petri dishes, tubes, trays, culture bags, and biofunctional chips such as organ-on-chips. Examples of cell-adhesive cultureware that can be used include cultureware whose surface has been artificially treated for the purpose of improving adhesion to cells. Examples of artificial treatments include coating treatments with extracellular matrices, polymers, etc., and surface treatments such as gas plasma treatment and positive charge treatment. Examples of extracellular matrices to which cells are attached include basement membrane preparations, laminin, entactin, collagen, gelatin, etc. Examples of polymers include polylysine, polyornithine, etc. The culture surface of the cultureware may be flat or uneven.
「ラミニン」とは、α、β、γ鎖からなるヘテロ三量体分子であり、サブユニット鎖の組成が異なるアイソフォームが存在する細胞外マトリクスタンパク質である。具体的には、ラミニンは、5種のα鎖、4種のβ鎖及び3種のγ鎖のヘテロ三量体の組合せで約15種類のアイソフォームを有する。α 鎖(α1~α5)、β鎖(β1~β4)及びγ鎖(γ1~γ3)のそれぞれの数字を組み合わせて、ラミニンの名称が定められている。例えばα5鎖、β1鎖、γ1鎖の組合せによるラミニンをラミニン511という。 Laminin is a heterotrimeric molecule consisting of α, β, and γ chains, and is an extracellular matrix protein with isoforms that differ in the composition of the subunit chains. Specifically, laminin has approximately 15 isoforms, which are heterotrimeric combinations of five types of α chains, four types of β chains, and three types of γ chains. The names of laminins are determined by combining the numbers of the α chains (α1-α5), β chains (β1-β4), and γ chains (γ1-γ3). For example, a laminin made up of a combination of α5, β1, and γ1 chains is called laminin 511.
浮遊培養を行う際に用いられる培養器材は、浮遊培養することが可能なものであれば特に限定されず、当業者であれば適宜決定することが可能である。このような培養器材としては、例えばフラスコ、組織培養用フラスコ、ディッシュ、ペトリデッシュ、組織培養用ディッシュ、マルチディッシュ、マイクロプレート、マイクロウェルプレート、マイクロポア、マルチプレート、マルチウェルプレート、チャンバースライド、シャーレ、チューブ、トレイ、培養バック、スピナーフラスコ及びローラーボトルが挙げられる。これらの培養器材は、浮遊培養を可能とするために、細胞非接着性であることが好ましい。細胞非接着性の培養器材としては、培養器材の表面に、細胞との接着性を向上させる目的で行われる上述の人工的な処理がされていないもの等を使用できる。細胞非接着性の培養器材として、細胞との接着性を低下させる目的で培養器材の表面が人工的に処理されたものを使用することもできる。培養器材の培養面は、平底、U底又はV底でもよいし、凹凸があってもよい。細胞との接着性を低下させる処理としては、例えば2-methacryloyloxyethyl phosphorylcholine(MPC)ポリマー、Poly(2-hydroxyethyl methacrylate)(Poly-HEMA)、polyethylene glycol(PEG)等のコーティングによる超親水性処理、タンパク低吸着処理等が挙げられる。 The cultureware used for suspension culture is not particularly limited as long as it is capable of suspension culture, and a person skilled in the art can appropriately determine the cultureware. Examples of such cultureware include flasks, flasks for tissue culture, dishes, Petri dishes, dishes for tissue culture, multi-dishes, microplates, microwell plates, micropores, multi-plates, multi-well plates, chamber slides, petri dishes, tubes, trays, culture bags, spinner flasks, and roller bottles. These cultureware are preferably non-adhesive to cells in order to enable suspension culture. As non-adhesive cultureware, those whose surfaces have not been artificially treated to improve adhesion to cells can be used. As non-adhesive cultureware, those whose surfaces have been artificially treated to reduce adhesion to cells can also be used. The culture surface of the cultureware may be flat, U- or V-bottom, or may be uneven. Examples of treatments that reduce adhesion to cells include superhydrophilic treatments using coatings such as 2-methacryloyloxyethyl phosphorylcholine (MPC) polymer, Poly(2-hydroxyethyl methacrylate) (Poly-HEMA), and polyethylene glycol (PEG), as well as low protein adsorption treatments.
「振盪培養」とは、培養器材を揺動させることにより培養液を攪拌し、培養液中への酸素供給、細胞の周囲との物質交換等を促進する培養法である。攪拌培養、流路培養等を行うこともできる。 "Shaking culture" is a culture method in which the culture medium is stirred by shaking the culture equipment, promoting the supply of oxygen to the culture medium and the exchange of materials with the surrounding cells. Agitation culture, flow path culture, etc. can also be performed.
浮遊培養を行う際に生じる剪断力等の物理的ストレスから細胞凝集体を保護し、また細胞が分泌する増殖因子及びサイトカイン類の局所濃度を高め、組織の発達を促進する目的から、細胞凝集体をゲルに包埋、又は物質透過性のあるカプセルに封入したのちに浮遊培養を実施することもできる(Nature,2013,501.7467:373)。上記封入した細胞凝集体を振盪培養しても良い。包埋に用いるゲル又はカプセルは、生体由来又は合成高分子製のいずれであってもよい。このような目的に用いるゲル又はカプセルとしては、例えばマトリゲル(Corning社製)、PuraMatrix(3D Matrix社製)、VitroGel 3D(TheWell Bioscience社製)、コラーゲンゲル(新田ゼラチン社製)、アルギン酸ゲル(PGリサーチ社製)、Cell-in-a-Box(Austrianova社製)等が挙げられる。 In order to protect the cell aggregates from physical stresses such as shear forces that occur during suspension culture, and to increase the local concentrations of growth factors and cytokines secreted by the cells and promote tissue development, the cell aggregates can be embedded in a gel or encapsulated in a substance-permeable capsule before suspension culture (Nature, 2013, 501.7467:373). The encapsulated cell aggregates may be cultured with shaking. The gel or capsule used for embedding may be made of either a biological or synthetic polymer. Examples of gels or capsules used for such purposes include Matrigel (manufactured by Corning), PuraMatrix (manufactured by 3D Matrix), VitroGel 3D (manufactured by TheWell Bioscience), collagen gel (manufactured by Nitta Gelatin), alginate gel (manufactured by PG Research), and Cell-in-a-Box (manufactured by Austrianova).
細胞の培養に用いられる培地は、動物細胞の培養に通常用いられる培地を基礎培地として調製することができる。基礎培地としては、例えばBasal Medium Eagle(BME)、BGJb培地、CMRL 1066培地、Glasgow Minimum Essential Medium(Glasgow MEM)、Improved MEM Zinc Option、Iscove’s Modified Dulbecco ’s Medium(IMDM)、Medium 199、Eagle Minimum Essential Medium(Eagle MEM)、Alpha Modified Eagle Minimum Essential Medium(αMEM)、Dulbecco’s Modified Eagle Medium(DMEM)、F-12培地、DMEM/F12、IMDM/F12、ハム培地、RPMI 1640、Fischer’s培地、又はこれらの混合培地等が挙げられる。 The medium used for cell culture can be prepared as a basal medium using a medium commonly used for culturing animal cells. Examples of basal media include Basal Medium Eagle (BME), BGJb medium, CMRL 1066 medium, Glasgow Minimum Essential Medium (Glasgow MEM), Improved MEM Zinc Option, Iscove's Modified Dulbecco's Medium (IMDM), Medium 199, Eagle Minimu Examples of such medium include Eagle MEM, Alpha Modified Eagle Minimum Essential Medium (αMEM), Dulbecco's Modified Eagle Medium (DMEM), F-12 medium, DMEM/F12, IMDM/F12, Ham's medium, RPMI 1640, Fischer's medium, and mixtures of these.
多能性幹細胞の維持培養には、上記基礎培地をベースとした多能性幹細胞培養用の培地、好ましく公知の胚性幹細胞又は人工多能性幹細胞用の培地、フィーダーフリー下で多能性幹細胞を培養するための培地(フィーダーフリー培地)等を用いることができる。フィーダーフリー培地として、多くの合成培地が開発・市販されており、例えばEssential 8培地が挙げられる。Essential 8培地は、DMEM/F12培地に、添加剤として、L-ascorbic acid-2-phosphate magnesium(64mg/l)、sodium selenium(14μg/1)、insulin(19.4mg/l)、NaHCO3(543mg/l)、transferrin(10.7mg/l)、bFGF(100ng/mL)、及びTGFβファミリーシグナル伝達経路作用物質(TGFβ 1(2ng/mL)又はNodal(100ng/mL))を含む(Nature Methods,8,424-429(2011))。市販のフィーダーフリー培地としては、例えばEssential 8(Thermo Fisher Scientific社製)、S-medium(DSファーマバイオメディカル社製)、StemPro(Thermo Fisher Scientific社製)、hESF9、mTeSR1(STEMCELL Technologies社製)、mTeSR2(STEMCELL Technologies社製)、TeSR-E8(STEMCELL Technologies社製)、mTeSR Plus(STEMCELL Technologies社製)、StemFit(味の素社製)、ReproMed iPSC Medium(リプロセル社製)、NutriStem XF(Biological Industries社製)、NutriStem V9(Biological Industries社製)、Cellartis DEF-CS Xeno-Free Culture Medium(タカラバイオ社製)、Stem-Partner SF(極東製薬社製)、PluriSTEM Human ES/iPS Cell Medium(メルク社製)、StemSure hPSC MediumΔ(富士フイルム和光純薬社製)等が挙げられる。 For the maintenance culture of pluripotent stem cells, a medium for pluripotent stem cell culture based on the above-mentioned basal medium, a medium for preferably known embryonic stem cells or induced pluripotent stem cells, a medium for culturing pluripotent stem cells under feeder-free conditions (feeder-free medium), etc. Many synthetic media have been developed and are commercially available as feeder-free media, for example, Essential 8 medium. Essential 8 medium contains, in DMEM/F12 medium, the following additives: L-ascorbic acid-2-phosphate magnesium (64 mg/l), sodium selenium (14 μg/l), insulin (19.4 mg/l), NaHCO 3 (543 mg/l), transferrin (10.7 mg/l), bFGF (100 ng/mL), and a TGFβ family signaling pathway active substance (TGFβ 1 (2 ng/mL) or Nodal (100 ng/mL)) (Nature Methods, 8, 424-429 (2011)). Commercially available feeder-free media include, for example, Essential 8 (manufactured by Thermo Fisher Scientific), S-medium (manufactured by DS Pharma Biomedical), StemPro (manufactured by Thermo Fisher Scientific), hESF9, mTeSR1 (manufactured by STEMCELL Technologies), mTeSR2 (manufactured by STEMCELL Technologies), TeSR-E8 (manufactured by STEMCELL Technologies), mTeSR Plus (manufactured by STEMCELL Technologies), StemFit (manufactured by Ajinomoto Co., Inc.), and ReproMed iPSC. Examples of such medium include Cellartis DEF-CS Xeno-Free Culture Medium (manufactured by ReproCELL), NutriStem XF (manufactured by Biological Industries), NutriStem V9 (manufactured by Biological Industries), Cellartis DEF-CS Xeno-Free Culture Medium (manufactured by Takara Bio Inc.), Stem-Partner SF (manufactured by Kyokuto Pharmaceutical Co., Ltd.), PluriSTEM Human ES / iPS Cell Medium (manufactured by Merck), and StemSure hPSC Medium Δ (manufactured by Fujifilm Wako Pure Chemical Industries).
無血清培地は、血清代替物を含有していてもよい。血清代替物としては、例えばアルブミン、トランスフェリン、脂肪酸、コラーゲン前駆体、微量元素、2-メルカプトエタノール又は3’-チオールグリセロール又はこれらの均等物等を適宜含有するものを挙げることができる。かかる血清代替物は、例えば、国際公開第98/30679号に記載の方法により調製することができる。血清代替物としては市販品を利用してもよい。市販の血清代替物としては、例えばKnockout Serum Replacement(Thermo Fisher Scientific社製)(以下、「KSR」と記すこともある。)、Chemically-defined Lipid concentrated(Thermo Fisher Scientific社製)、Glutamax(Thermo Fisher Scientific社製)、B27 Supplement(Thermo Fisher Scientific社製)、N2 Supplement(Thermo Fisher Scientific社製)等が挙げられる。 The serum-free medium may contain a serum substitute. Examples of serum substitutes include those that contain albumin, transferrin, fatty acids, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, or equivalents thereof, as appropriate. Such serum substitutes can be prepared, for example, by the method described in WO 98/30679. Commercially available products may be used as serum substitutes. Examples of commercially available serum substitutes include Knockout Serum Replacement (manufactured by Thermo Fisher Scientific) (hereinafter sometimes referred to as "KSR"), Chemically-defined Lipid Concentrated (manufactured by Thermo Fisher Scientific), Glutamax (manufactured by Thermo Fisher Scientific), B27 Supplement (manufactured by Thermo Fisher Scientific), and N2 Supplement (manufactured by Thermo Fisher Scientific).
浮遊培養及び接着培養で用いる無血清培地は、適宜、脂肪酸又は脂質、アミノ酸(例えば非必須アミノ酸)、ビタミン、増殖因子、サイトカイン、抗酸化剤、2-メルカプトエタノール、ピルビン酸、緩衝剤、無機塩類等を含有してもよい。 The serum-free medium used in suspension culture and adherent culture may contain fatty acids or lipids, amino acids (e.g., non-essential amino acids), vitamins, growth factors, cytokines, antioxidants, 2-mercaptoethanol, pyruvic acid, buffers, inorganic salts, etc., as appropriate.
調製の煩雑さを回避するために、無血清培地として、市販のKSR(Thermo Fisher Scientific社製)を適量(例えば約0.5%から約30%、好ましくは約1%から約20%)添加した無血清培地(例えばF-12培地とIMDM培地の1:1混合液に1×chemically-defined Lipid concentrated、5%KSR及び450μM 1-モノチオグリセロールを添加した培地)を使用してもよい。また、KSR同等品として特表2001-508302号公報に開示された培地が挙げられる。 To avoid the complication of preparation, a serum-free medium containing an appropriate amount (e.g., about 0.5% to about 30%, preferably about 1% to about 20%) of commercially available KSR (manufactured by Thermo Fisher Scientific) may be used as the serum-free medium (e.g., a medium containing 1x chemically defined lipid concentrated, 5% KSR and 450 μM 1-monothioglycerol added to a 1:1 mixture of F-12 medium and IMDM medium) to avoid the complication of preparation. An example of a KSR equivalent is the medium disclosed in JP2001-508302A.
「血清培地」とは、無調整又は未精製の血清を含む培地を意味する。当該培地は、脂肪酸又は脂質、アミノ酸(例えば非必須アミノ酸)、ビタミン、増殖因子、サイトカイン、抗酸化剤、2-メルカプトエタノール、1-モノチオグリセロール、ピルビン酸、緩衝剤、無機塩類等を含有してもよい。 "Serum medium" refers to a medium containing unconditioned or unpurified serum. The medium may contain fatty acids or lipids, amino acids (e.g., non-essential amino acids), vitamins, growth factors, cytokines, antioxidants, 2-mercaptoethanol, 1-monothioglycerol, pyruvic acid, buffers, inorganic salts, etc.
本発明における培養は、好ましくはゼノフリー条件で行われる。「ゼノフリー」とは、培養対象の細胞の生物種とは異なる生物種由来の成分が排除された条件を意味する。 The culture in the present invention is preferably carried out under xeno-free conditions. "Xeno-free" refers to conditions in which components derived from organisms other than the organism of the cells to be cultured are excluded.
本発明に用いる培地は、化学的に未決定な成分の混入を回避する観点から、好ましくは、含有成分が化学的に決定された培地(Chemicallydefined medium;CDM)である。 The medium used in the present invention is preferably a medium whose components are chemically defined (CDM) in order to avoid contamination with chemically undefined components.
「基底膜(Basement membrane)」とは、細胞外マトリクスより構成される薄い膜状の構造体を意味する。生体においては、基底膜は上皮細胞の基底側(basal)に形成される。基底膜の成分としては、IV型コラーゲン、ラミニン、ヘパラン硫酸プロテオグリカン(パールカン)、エンタクチン/ニドゲン、サイトカイン、成長因子等が挙げられる。生体由来の組織中並びに本発明の製造方法等で作製された細胞集団中に基底膜が存在しているかどうかは、例えばPAM染色等の組織染色、並びに基底膜の構成成分に対する抗体(抗ラミニン抗体、抗IV型コラーゲン抗体等)を用いた免疫組織化学等の手法により検出することができる。 "Basement membrane" refers to a thin membrane-like structure composed of extracellular matrix. In living organisms, basement membranes are formed on the basal side of epithelial cells. Components of basement membranes include type IV collagen, laminin, heparan sulfate proteoglycan (perlecan), entactin/nidogen, cytokines, growth factors, etc. Whether or not basement membrane is present in tissues derived from living organisms and in cell populations produced by the production method of the present invention can be detected by, for example, tissue staining such as PAM staining, and immunohistochemistry using antibodies against components of basement membranes (anti-laminin antibodies, anti-type IV collagen antibodies, etc.).
「基底膜標品」とは、その上に基底膜形成能を有する所望の細胞を播種して培養した場合に、上皮細胞様の細胞形態、分化、増殖、運動、機能発現等を制御する機能を有する基底膜構成成分を含むものをいう。本明細書において、細胞の接着培養を行う際には、基底膜標品存在下で培養することができる。ここで、「基底膜構成成分」とは、動物の組織において、上皮細胞層と間質細胞層等との間に存在する薄い膜状をした細胞外マトリクス分子をいう。基底膜標品は、例えば、基底膜を介して支持体上に接着している基底膜形成能を有する細胞を、該細胞の脂質溶解能を有する溶液やアルカリ溶液などを用いて支持体から除去することで作製することができる。基底膜標品としては、基底膜調製物として市販されている商品、例えば、マトリゲル(Corning社製)、Geltrex(Thermo Fisher Scientific社製)、基底膜成分として公知の細胞外マトリクス分子(例えば、ラミニン、IV型コラーゲン、ヘパラン硫酸プロテオグリカン、エンタクチン等)を含むものが挙げられる。 The term "basement membrane preparation" refers to a preparation containing basement membrane components that have the function of controlling epithelial cell-like cell morphology, differentiation, proliferation, movement, functional expression, etc., when desired cells having basement membrane formation ability are seeded and cultured thereon. In this specification, when cells are cultured for adhesion, they can be cultured in the presence of a basement membrane preparation. Here, "basement membrane components" refers to thin membrane-like extracellular matrix molecules that exist between epithelial cell layers and interstitial cell layers, etc., in animal tissues. A basement membrane preparation can be prepared, for example, by removing cells having basement membrane formation ability that are adhered to a support via a basement membrane from the support using a solution capable of dissolving the lipids of the cells, an alkaline solution, or the like. Examples of basement membrane preparations include products commercially available as basement membrane preparations, such as Matrigel (manufactured by Corning) and Geltrex (manufactured by Thermo Fisher Scientific), and those that contain extracellular matrix molecules known as basement membrane components (e.g., laminin, type IV collagen, heparan sulfate proteoglycan, entactin, etc.).
細胞又は組織の培養には、Engelbreth-Holm-Swarm(EHS)マウス肉腫等の組織又は細胞から抽出、可溶化されたマトリゲル(Corning社製)等の基底膜標品を用いることができる。同様に細胞培養に用いる基底膜成分として、ヒト可溶化羊膜(生物資源応用研究所社製)、HEK293細胞に産生させたヒト組み換えラミニン(BioLamina社製)、ヒト組み換えラミニン断片(ニッピ社製)、ヒト組み換えビトロネクチン(Thermo Fisher Scientific社製)等も用いることができる。異なる生物種由来の成分混入を回避する観点、及び感染症のリスクを回避する観点から、基底膜標品は、好ましくは成分の明らかな組み換えタンパク質を用いる。 For cell or tissue culture, basement membrane preparations such as Matrigel (Corning) extracted and solubilized from tissues or cells such as Engelbreth-Holm-Swarm (EHS) mouse sarcoma can be used. Similarly, as basement membrane components for cell culture, human solubilized amniotic membrane (BioResource Application Research Institute), human recombinant laminin produced in HEK293 cells (BioLamina), human recombinant laminin fragments (Nippi), human recombinant vitronectin (Thermo Fisher Scientific), etc. can also be used. From the viewpoint of avoiding contamination with components from different biological species and from the viewpoint of avoiding the risk of infection, it is preferable to use recombinant proteins with known components as basement membrane preparations.
本明細書において、「物質Xを含む培地」及び「物質Xの存在下」とは、それぞれ外来性(exogenous)の物質Xが添加された培地もしくは外来性の物質Xを含む培地、及び外来性の物質Xの存在下を意味する。外来性の物質Xは、例えば培地中に存在する細胞又は組織が当該物質Xを内在的(endogenous)に発現、分泌又は産生する内在的な物質Xと区別される。培地中の物質Xは、物質Xの分解又は培地の蒸発による微量な濃度の変化が起こっていてもよい。 In this specification, "medium containing substance X" and "in the presence of substance X" respectively mean a medium to which exogenous substance X has been added or a medium containing exogenous substance X, and in the presence of exogenous substance X. Exogenous substance X is distinguished from endogenous substance X, for example, in which cells or tissues present in the medium endogenously express, secrete, or produce the substance X. Substance X in the medium may have undergone slight changes in concentration due to decomposition of substance X or evaporation of the medium.
本明細書において、物質Xの濃度がYである培地中での培養開始時とは、好ましくは培地中の物質Xの濃度がYで均一となった時点を指すが、培養容器が十分に小さい(例えば、96ウェルプレートや、培養液が200μL以下での培養)場合、濃度がYとなるように後述する培地添加操作、半量培地交換操作又は全量培地交換操作を行った時点を濃度Yでの培養開始時と解釈する。また、培地中の物質Xの濃度がYであるとは、一定の培養期間を通じた物質Xの平均の濃度がYである場合、物質XをYの濃度で含む期間が培養期間の50%以上である場合、物質XをYの濃度で含む期間が各工程において想定される培養期間のうち最も短い期間以上である場合等を含む。 In this specification, the start of culture in a medium in which the concentration of substance X is Y preferably refers to the point in time when the concentration of substance X in the medium becomes uniform at Y, but if the culture vessel is sufficiently small (for example, a 96-well plate or culture in 200 μL or less of culture medium), the start of culture at concentration Y is interpreted as the point in time when the medium addition operation, half-volume medium replacement operation, or full-volume medium replacement operation described below is performed to achieve concentration Y. In addition, when the concentration of substance X in the medium is Y, this includes the case where the average concentration of substance X throughout a certain culture period is Y, the period during which substance X is contained at concentration Y is 50% or more of the culture period, the period during which substance X is contained at concentration Y is longer than the shortest culture period expected in each process, etc.
本明細書において、「物質Xの非存在下」とは、外来性(exogenous)の物質Xが添加されていない培地もしくは外来性の物質Xを含まない培地、又は外来性の物質Xの存在しない状態を意味する。 In this specification, "in the absence of substance X" means a medium to which exogenous substance X has not been added, a medium that does not contain exogenous substance X, or a state in which exogenous substance X is not present.
本明細書において、「ヒトタンパク質X」とは、タンパク質Xが、ヒト生体内で天然に発現するタンパク質Xのアミノ酸配列を有することを意味する。 As used herein, "human protein X" means that protein X has the amino acid sequence of protein X that is naturally expressed in the human body.
「単離」とは、目的とする成分や細胞以外の因子を除去する操作がなされ、天然に存在する状態を脱していることを意味する。従って、「単離されたタンパク質X」には、培養対象の細胞や組織から産生され細胞や組織及び培地中に含まれている内在性のタンパク質Xは包含されない。「単離されたタンパク質X」の純度(総タンパク質重量に占めるタンパク質Xの重量の百分率)は、通常70%以上、好ましくは80%以上、より好ましくは90%以上、さらに好ましくは99%以上、最も好ましくは100%である。 "Isolated" means that an operation has been performed to remove the target component or factors other than the cell, and that the protein is no longer in a naturally occurring state. Therefore, "isolated protein X" does not include endogenous protein X that is produced from the cells or tissues to be cultured and is contained in the cells, tissues, and medium. The purity of "isolated protein X" (the percentage of protein X weight in the total protein weight) is usually 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably 99% or more, and most preferably 100%.
本明細書において、「誘導体」とは、特定の化合物に対して、当該化合物の分子内の一部が、他の官能基又は他の原子と置換されることにより生じる化合物群を意図する。本明細書において、タンパク質の「改変体」とは、もとのタンパク質の特性を維持できる範囲でアミノ酸残基の欠失、付加、置換等の変異がされているタンパク質をいう。変異のアミノ酸の数としては、特に制限されないが、1~4、1~3、1~2、又は1個が挙げられる。タンパク質の「改変体」は、もとのタンパク質と少なくとも90%以上、91%以上、92%以上、93%以上、94%以上、95%以上、96%以上、97%以上、98%以上、99%以上、又は99.5%以上の同一性を示すアミノ酸配列を有するタンパク質であってもよい。改変体において変異したアミノ酸は、非天然型であっても良い。 In this specification, the term "derivative" refers to a group of compounds that are produced by replacing a part of the molecule of a specific compound with another functional group or another atom. In this specification, a "modified form" of a protein refers to a protein that has been mutated by deletion, addition, substitution, or other such amino acid residues to the extent that the properties of the original protein can be maintained. The number of mutated amino acids is not particularly limited, but may be 1-4, 1-3, 1-2, or 1. A "modified form" of a protein may be a protein that has an amino acid sequence that shows at least 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more, or 99.5% or more identity with the original protein. The mutated amino acids in the modified form may be non-natural.
本明細書において、「A時間(A日)以降」とはA時間(A日)を含み、A時間(A日)から後のことをいう。「B時間(B日)以内」とは、B時間(B日)を含み、B時間(B日)から前のことをいう。 In this specification, "after time A (day A)" includes time A (day A) and refers to anything after time A (day A). "Within time B (day B)" includes time B (day B) and refers to anything before time B (day B).
「フィーダー細胞」とは、幹細胞を培養するときに共存させる当該幹細胞以外の細胞を指す。多能性幹細胞の未分化維持培養に用いられるフィーダー細胞としては、例えばマウス線維芽細胞(MEF)、ヒト線維芽細胞、SNL細胞等が挙げられる。フィーダー細胞は、増殖抑制処理されていることが好ましい。増殖抑制処理としては、増殖抑制剤(例えばマイトマイシンC)処理、UV照射等が挙げられる。多能性幹細胞の未分化維持培養に用いられるフィーダー細胞は、液性因子(好ましくは未分化維持因子)の分泌、細胞接着用の足場(細胞外基質)の作製により、多能性幹細胞の未分化維持に貢献する。 "Feeder cells" refers to cells other than stem cells that are allowed to coexist when the stem cells are cultured. Examples of feeder cells used in the culture of pluripotent stem cells to maintain their undifferentiated state include mouse fibroblasts (MEF), human fibroblasts, and SNL cells. The feeder cells are preferably subjected to a growth-inhibiting treatment. Examples of the growth-inhibiting treatment include treatment with a growth inhibitor (e.g., mitomycin C) and UV irradiation. The feeder cells used in the culture of pluripotent stem cells to maintain their undifferentiated state contribute to maintaining the undifferentiated state of pluripotent stem cells by secreting humoral factors (preferably factors for maintaining their undifferentiated state) and creating a scaffold (extracellular matrix) for cell adhesion.
本明細書において、フィーダー細胞非存在下(フィーダーフリー)とは、フィーダー細胞非存在下にて培養することである。フィーダー細胞非存在下とは、例えばフィーダー細胞を添加していない条件、又はフィーダー細胞を実質的に含まない(例えば全細胞数に対するフィーダー細胞数の割合が3%以下)の条件が挙げられる。 In this specification, "in the absence of feeder cells" refers to culturing in the absence of feeder cells. Examples of "in the absence of feeder cells" include conditions where no feeder cells are added, or conditions where feeder cells are substantially not included (for example, the ratio of the number of feeder cells to the total number of cells is 3% or less).
「細胞集団」とは、2以上の細胞から構成される細胞群をいう。細胞集団は、一種の細胞から構成されていてもよいし、複数種の細胞から構成されていてもよい。細胞集団を構成する細胞は、培地中に浮遊していてもよいし、培養器材等に接着していてもよい。また、細胞集団を構成する細胞は、単一細胞であってもよいし、細胞集団の少なくとも一部において、細胞同士が細胞接着し、細胞集団を形成していてもよい。
ここで「単一細胞」とは、例えば、細胞同士の接着(例えば面接着)がほとんどなくなった細胞をいう。一部の態様において、単一細胞に分散されるとは、細胞―細胞間結合(例えば接着結合)がほとんどなくなった状態が挙げられる。
本明細書において、「分散された細胞」は、好ましくは単一細胞であるが、例えば2以上100以下の少数の細胞からなる細胞の塊を含んでもよく、2以上50以下の細胞からなる細胞の塊を含んでもよい。「分散された細胞」は、例えば単一細胞を7割以上及び細胞の塊を3割以下含んでいてもよく、好ましくは単一細胞を8割以上及び細胞の塊を2割以下含む。
一態様において、単一細胞が分散された細胞集団には5個以下の細胞が形成する微小な凝集体が含まれていてもよい。
本明細書において、細胞集団は、細胞凝集体や二次元の細胞シートを含んでよい。
A "cell population" refers to a group of cells composed of two or more cells. A cell population may be composed of one type of cell, or may be composed of multiple types of cells. The cells that compose the cell population may be suspended in a medium, or may be adhered to a culture vessel or the like. Furthermore, the cells that compose the cell population may be single cells, or at least a portion of the cell population may be formed by cell adhesion between the cells.
Here, the term "single cells" refers to, for example, cells that have almost no cell-to-cell adhesion (e.g., surface adhesion). In some embodiments, being dispersed into single cells refers to a state in which cell-to-cell bonds (e.g., adherens junctions) are almost completely lost.
As used herein, "dispersed cells" are preferably single cells, but may also include cell clumps consisting of a small number of cells, for example, between 2 and 100, or may include cell clumps consisting of between 2 and 50 cells. "Dispersed cells" may, for example, include 70% or more single cells and 30% or less cell clumps, and preferably include 80% or more single cells and 20% or less cell clumps.
In one embodiment, the population of dispersed single cells may contain minute aggregates of 5 or fewer cells.
As used herein, a cell population may include cell aggregates and two-dimensional cell sheets.
「細胞凝集体」(cell aggregate)とは、細胞が集合して形成された塊であって、細胞同士が接着している塊をいう。胚様体(Embryoid body)、スフェア(Sphere)、スフェロイド(Spheroid)、オルガノイド(Organoid)も細胞凝集体に包含される。細胞凝集体は、好ましくは細胞同士が面接着している。一部の態様において、細胞凝集体の一部分あるいは全部において、細胞同士が細胞接着し、例えば接着結合(adherence junction)を形成している。一部の態様において、二つ以上の細胞凝集体同士をさらに人工的に接着又は凝集させることもできる。細胞集団同士をさらに接着又は凝集させた塊、及びアセンブロイド(assembloid)も細胞凝集体に含まれる。細胞凝集体の形態は球状に限らず、例えば双球状、数珠状、球の集合体状、紐状・分枝状(Scientific reports, 11:21421(2021)、特願2021-078154に記載の形状)等の形態であってもよい。
本明細書において「二次元の細胞シート」とは、二次元の方向に細胞間の結合を有する単一又は複数の細胞から構成される単層又は重層の構造体をいう。
The term "cell aggregate" refers to a mass formed by the gathering of cells, in which the cells are adhered to each other. Embryoid bodies, spheres, spheroids, and organoids are also included in the cell aggregates. In the cell aggregates, the cells are preferably adhered to each other on their surfaces. In some embodiments, in a part or all of the cell aggregate, the cells are adhered to each other, for example, forming an adhesion junction. In some embodiments, two or more cell aggregates can be further artificially adhered or aggregated to each other. Masses in which cell populations are further adhered or aggregated to each other, and assembloids are also included in the cell aggregates. The shape of the cell aggregates is not limited to spherical, but may be, for example, bisphere-like, bead-like, a collection of spheres, string-like or branched (shapes described in Scientific reports, 11:21421 (2021), Patent Application No. 2021-078154), etc.
As used herein, the term "two-dimensional cell sheet" refers to a monolayer or multilayer structure composed of a single or multiple cells having intercellular junctions in the two-dimensional direction.
「均一な細胞凝集体」とは、複数の細胞凝集体を培養する際に各細胞凝集体の大きさが一定であることを意味し、細胞凝集体の大きさを最大径の長さで評価する場合、均一な細胞凝集体とは、最大径の長さの分散が小さいことを意味する。より具体的には、複数の細胞凝集体のうち75%以上の細胞凝集体において、その最大径が複数の細胞凝集体の最大径平均値の±100%以内、好ましくは最大径平均値の±50%以内、より好ましくは最大径平均値の±20%以内であることを意味する。 "Uniform cell aggregates" means that when multiple cell aggregates are cultured, the size of each cell aggregate is constant, and when the size of a cell aggregate is evaluated by the length of its maximum diameter, uniform cell aggregates means that the variance of the length of the maximum diameter is small. More specifically, this means that the maximum diameter of 75% or more of the multiple cell aggregates is within ±100% of the average maximum diameter of the multiple cell aggregates, preferably within ±50% of the average maximum diameter, and more preferably within ±20% of the average maximum diameter.
「組織」とは、形態や性質が異なる複数種類の細胞が一定のパターンで立体的に配置した構造を有する細胞集団の構造体をさす。 "Tissue" refers to a structure made up of a group of cells in which multiple types of cells with different morphologies and properties are arranged three-dimensionally in a specific pattern.
「外胚葉」とは生物の初期発生の過程において卵の受精後に形成される3つの胚葉のうち、最も外側に存在する胚葉を表す。外胚葉は発生の進行に従い神経外胚葉と表層外胚葉に分かれ、さらに神経外胚葉は神経管と神経堤に分かれる。これら外胚葉から身体の各種器官が形成され、外胚葉から形成された器官は外胚葉に由来すると称される。例えば神経管からは脳及び脊髄等の中枢神経系の器官又は組織が形成される。例えば神経堤からは一部の中枢神経系細胞、顔面の骨及び軟骨、感覚神経細胞、自律神経細胞、色素細胞、間葉系細胞等が形成される。表層外胚葉からは表皮、内耳、下垂体前葉、嗅上皮を含む上気道組織等が形成される。プラコード及びプラコード由来組織は表層外胚葉に由来する。多能性幹細胞は、外胚葉に分化する過程で、例えばPax3、Otx2、Sox1といった外胚葉マーカーとして知られている遺伝子を発現する。 "Ectoderm" refers to the outermost of the three germ layers formed after fertilization of an egg during the early development of an organism. As development progresses, the ectoderm divides into the neuroectoderm and the superficial ectoderm, and the neuroectoderm further divides into the neural tube and neural crest. Various organs of the body are formed from these ectoderm, and organs formed from the ectoderm are said to be derived from the ectoderm. For example, the neural tube forms organs or tissues of the central nervous system, such as the brain and spinal cord. For example, some central nervous system cells, facial bones and cartilage, sensory neurons, autonomic neurons, pigment cells, mesenchymal cells, etc. are formed from the neural crest. The epidermis, inner ear, anterior pituitary gland, upper respiratory tract tissue including olfactory epithelium, etc. are formed from the superficial ectoderm. Placodes and placode-derived tissues are derived from the superficial ectoderm. In the process of differentiating into ectoderm, pluripotent stem cells express genes known as ectoderm markers, such as Pax3, Otx2, and Sox1.
「内胚葉」とは生物の初期発生の過程において卵の受精後に形成される3つの胚葉のうち、最も内側に存在する胚葉を表す。内胚葉からは例えば消化器、尿路、咽頭、気管、気管支、肺が形成される。多能性幹細胞は、内胚葉に分化する過程で、例えばSOX17、HNF-3β/FoxA2、Klf5、GATA4、GATA6、PDX-1といった内胚葉マーカーとして知られている遺伝子を発現する。
「中胚葉」とは外胚葉と内胚葉の間に形成される胚葉を表す。中胚葉からは例えば循環器、骨格、筋肉といった器官又は組織が形成される。多能性幹細胞は、中胚葉に分化する過程で、例えばT/Brachury、SMA、ABCA4、Nkx2.5、PDGFRαといった中胚葉マーカーとして知られている遺伝子を発現する。
「神経組織」とは、発生期又は成体期の大脳、中脳、小脳、脊髄、網膜、感覚神経、末梢神経等の神経系細胞によって構成される組織を意味する。本明細書において、「神経上皮組織」とは、神経組織が層構造をもつ上皮構造を形成したものをいい、神経組織中の神経上皮組織は光学顕微鏡を用いた明視野観察等により存在量を評価することができる。
"Endoderm" refers to the innermost of the three germ layers formed after fertilization of an egg during the early development of an organism. For example, the digestive system, urinary tract, pharynx, trachea, bronchi, and lungs are formed from the endoderm. In the process of differentiating into endoderm, pluripotent stem cells express genes known as endoderm markers, such as SOX17, HNF-3β/FoxA2, Klf5, GATA4, GATA6, and PDX-1.
"Mesoderm" refers to the germ layer formed between the ectoderm and endoderm. Organs or tissues such as circulatory system, skeleton, and muscle are formed from the mesoderm. In the process of differentiating into the mesoderm, pluripotent stem cells express genes known as mesoderm markers, such as T/Brachury, SMA, ABCA4, Nkx2.5, and PDGFRα.
"Nervous tissue" refers to tissue composed of nervous system cells such as the developing or adult cerebrum, midbrain, cerebellum, spinal cord, retina, sensory nerves, peripheral nerves, etc. As used herein, "neuroepithelial tissue" refers to nervous tissue that has formed an epithelial structure having a layered structure, and the amount of neuroepithelial tissue in nervous tissue can be evaluated by bright field observation using an optical microscope or the like.
「中枢神経系」とは、神経組織が集積し、情報処理の中心をなす領域を表す。脊椎動物では、脳と脊髄が中枢神経系に含まれる。中枢神経系は外胚葉に由来する。 The "central nervous system" refers to the area where nervous tissue is accumulated and which is the center of information processing. In vertebrates, the central nervous system includes the brain and spinal cord. The central nervous system is derived from the ectoderm.
「神経系細胞(Neural cell)」とは、外胚葉由来組織のうち表皮系細胞以外の細胞を表す。すなわち、神経系細胞には、神経系前駆細胞、ニューロン(神経細胞)、グリア細胞、神経幹細胞、ニューロン前駆細胞、グリア前駆細胞等の細胞を含む。神経系細胞には、後述する網膜組織を構成する細胞(網膜細胞)、網膜前駆細胞、網膜層特異的神経細胞、神経網膜細胞、網膜色素上皮細胞も包含される。神経系細胞は、Nestin、βIIIチューブリン(Tuj1)、PSA-NCAM、N-cadherin等をマーカーとして同定することができる。 "Neural cells" refers to cells of ectoderm-derived tissues other than epidermal cells. In other words, neural cells include neural progenitor cells, neurons (nerve cells), glial cells, neural stem cells, neuronal progenitor cells, glial progenitor cells, and other cells. Neural cells also include cells that make up retinal tissue (retinal cells), retinal progenitor cells, retinal layer-specific nerve cells, neural retina cells, and retinal pigment epithelial cells, which will be described later. Neural cells can be identified using markers such as nestin, βIII tubulin (Tuj1), PSA-NCAM, and N-cadherin.
ニューロンは、神経回路を形成し情報伝達に貢献する機能的な細胞であり、TuJ1、Dcx、HuC/D等の幼若神経細胞マーカー、及び/又はMap2、NeuN等の成熟神経細胞マーカーの発現を指標に同定することができる。 Neurons are functional cells that form neural circuits and contribute to signal transmission, and can be identified using the expression of immature neuronal markers such as TuJ1, Dcx, and HuC/D, and/or mature neuronal markers such as Map2 and NeuN.
神経系前駆細胞(Neural Precursor cell)は、神経幹細胞、ニューロン前駆細胞及びグリア前駆細胞を含む前駆細胞の集合体であり、増殖能とニューロン及びグリア産生能をもつ。神経系前駆細胞はNestin、GLAST、Sox2、Sox1、Musashi、Pax6等をマーカーとして同定することができる。神経系細胞のマーカー陽性かつ増殖マーカー(Ki67、pH3、MCM)陽性の細胞を、神経系前駆細胞として同定することもできる。 Neural precursor cells are a collection of precursor cells, including neural stem cells, neuronal precursor cells, and glial precursor cells, and have the ability to proliferate and produce neurons and glia. Neural precursor cells can be identified using markers such as Nestin, GLAST, Sox2, Sox1, Musashi, and Pax6. Cells that are positive for neural cell markers and proliferation markers (Ki67, pH3, MCM) can also be identified as neural precursor cells.
本明細書において、「脳室」とは、中枢神経組織によって形成された腔所をさす。生体においては通常脳脊髄液等の組織液で満たされている無細胞性の構造であり、神経組織の頂端面側が脳室に面している。脳室を取り巻く脳室周囲層は神経幹細胞が存在し、発生期に細胞の増殖とニューロン産生が生じる領域である。本発明の製造方法で製造される細胞集団並びに組織中に脳室が含まれるかどうかは、例えば中枢神経組織マーカー(Bf1、Pax6等)及び頂端面マーカー(PKC-zeta等)を用いた免疫組織化学等の手法により検出することが出来る。 In this specification, the term "ventricle" refers to a cavity formed by central nervous tissue. In living organisms, it is an acellular structure that is usually filled with tissue fluid such as cerebrospinal fluid, with the apical side of the nervous tissue facing the ventricle. The periventricular layer surrounding the ventricle is an area where neural stem cells exist and where cell proliferation and neuronal production occur during development. Whether or not the cell population and tissue produced by the production method of the present invention contains ventricles can be detected by techniques such as immunohistochemistry using central nervous tissue markers (Bf1, Pax6, etc.) and apical surface markers (PKC-zeta, etc.).
本明細書において、「間脳」とは、第三脳室に接した中枢神経系の神経組織をさす。間脳には、視床上部、視床、視床下部、下垂体といった組織が含まれる。 In this specification, the term "diencephalon" refers to the neural tissue of the central nervous system adjacent to the third ventricle. The diencephalon includes tissues such as the epithalamus, thalamus, hypothalamus, and pituitary gland.
本明細書において、「視床下部」とは、下垂体に接した間脳の一領域を指す。視床下部はさらに背側視床下部及び腹側視床下部に領域化される。領域下される前の視床下部はRx、Vax1、Six3といったマーカーを用いて同定することが出来る。背側視床下部はOtp、Brn2、バゾプレシン、Pax6といったマーカーを用いて同定することが出来る。腹側視床下部はNkx2.1、SF1といったマーカーを用いて同定することが出来る。
本明細書において、「視床下部神経上皮組織」とは、視床下部マーカーを発現する神経上皮組織をいう。一態様において、腹側視床下部神経上皮組織は、Rx陽性、Chx10陰性、且つNkx2.1陽性の神経上皮組織である。一態様において、背側視床下部神経上皮組織は、Rx陽性、Chx10陰性、且つPax6陽性の神経上皮組織である。
本明細書において、「視床下部」は「上衣細胞」を含んでいてもよい。上位細胞は上皮細胞の一種であって、脳室壁に局在する細胞を表す。上衣細胞のうち、「伸長上衣細胞:タニサイト(Tanycytes)」は、特に視床下部の第三脳室に局在する、繊毛に乏しい細胞を表す。伸長上衣細胞は、Nestin、Vimentin、Lhx2、Sox2といったマーカーを用いて同定することが出来る。
As used herein, the term "hypothalamus" refers to a region of the diencephalon adjacent to the pituitary gland. The hypothalamus is further regionalized into the dorsal hypothalamus and the ventral hypothalamus. The hypothalamus before regionalization can be identified using markers such as Rx, Vax1, and Six3. The dorsal hypothalamus can be identified using markers such as Otp, Brn2, vasopressin, and Pax6. The ventral hypothalamus can be identified using markers such as Nkx2.1 and SF1.
As used herein, the term "hypothalamic neuroepithelial tissue" refers to neuroepithelial tissue expressing a hypothalamic marker. In one embodiment, the ventral hypothalamic neuroepithelial tissue is Rx-positive, Chx10-negative, and Nkx2.1-positive neuroepithelial tissue. In one embodiment, the dorsal hypothalamic neuroepithelial tissue is Rx-positive, Chx10-negative, and Pax6-positive neuroepithelial tissue.
In this specification, the term "hypothalamus" may include "ependymal cells." Ependymal cells are a type of epithelial cell, and refer to cells localized in the ventricular wall. Among ependymal cells, "elongated ependymal cells (tanycytes)" refer to cells with few cilia that are particularly localized in the third ventricle of the hypothalamus. Elongated ependymal cells can be identified using markers such as Nestin, Vimentin, Lhx2, and Sox2.
本明細書において、「非神経上皮組織」とは、上皮構造を有する組織のうち神経上皮組織以外の組織を表す。上皮組織は外胚葉、中胚葉、内胚葉、栄養外胚葉のいずれの胚葉からも形成される。上皮組織には上皮、中皮、内皮が含まれる。非神経上皮組織に含まれる組織の例としては、表皮、角膜上皮、鼻腔上皮、口腔上皮、気管上皮、気管支上皮、気道上皮、腎上皮、腎皮質上皮、胎盤上皮等が挙げられる。
上皮組織は通常種々の細胞間結合によりつながれており、単層又は重層化した層構造を有する組織を形成する。これら上皮組織の有無の確認、存在量の定量は光学顕微鏡による観察か、上皮細胞マーカーに対する抗体(抗E-Cadherin抗体、抗N-Cadherin抗体、抗EpCAM抗体等)を用いた免疫組織化学等の手法により可能である。
As used herein, the term "non-neuroepithelial tissue" refers to tissues having an epithelial structure other than neuroepithelial tissue. Epithelial tissue is formed from any of the germ layers, ectoderm, mesoderm, endoderm, and trophectoderm. Epithelial tissue includes epithelium, mesothelium, and endothelium. Examples of tissues included in non-neuroepithelial tissue include epidermis, corneal epithelium, nasal epithelium, oral epithelium, tracheal epithelium, bronchial epithelium, airway epithelium, kidney epithelium, kidney cortical epithelium, and placental epithelium.
Epithelial tissues are usually connected by various intercellular junctions to form tissues with a single layer or stratified layer structure. The presence or absence of these epithelial tissues and the amount of them present can be confirmed by observation with an optical microscope or by immunohistochemistry using antibodies against epithelial cell markers (anti-E-Cadherin antibody, anti-N-Cadherin antibody, anti-EpCAM antibody, etc.).
本明細書において、「上皮細胞極性(Epithelial polarity)」とは上皮細胞内に空間的に形成されている構成成分及び細胞機能の分布の偏りを表す。例えば、角膜上皮細胞は眼球の最も外層に局在し、頂端側(apical)では涙液を保持するための膜結合型ムチン(MUC-1、4、16)等の頂端側特異的なタンパク質を発現し、基底側(basal)では基底膜に接着するためのα6インテグリン、β1インテグリン等の基底側特異的なタンパク質を発現している。
生体由来の組織中並びに本発明の製造方法等で作製された細胞集団中の上皮細胞並びに上皮組織に上皮細胞極性が存在しているかどうかは、Phalloidin、頂端側マーカー(抗MUC-1抗体、抗PKC-zeta抗体等)並びに基底側マーカー(抗α6インテグリン抗体、抗β1インテグリン抗体等)を用いた免疫組織化学等の手法により検出することができる。
As used herein, the term "epithelial polarity" refers to the bias in the distribution of spatially formed components and cell functions within epithelial cells. For example, corneal epithelial cells are localized in the outermost layer of the eyeball, and express apical-specific proteins such as membrane-bound mucins (MUC-1, 4, 16) for retaining tears on the apical side, and express basal-specific proteins such as α6 integrin and β1 integrin for adhering to the basement membrane on the basal side.
The presence of epithelial cell polarity in epithelial cells and epithelial tissues in tissues derived from a living body and in cell populations produced by the production methods of the present invention can be detected by techniques such as immunohistochemistry using phalloidin, apical markers (anti-MUC-1 antibody, anti-PKC-zeta antibody, etc.), and basal markers (anti-α6 integrin antibody, anti-β1 integrin antibody, etc.).
本明細書において、「プラコード(placode)」とは、主に脊椎動物の発生過程において表皮外胚葉の一部が肥厚して形成される器官の原基のことを表す。プラコードに由来する組織としては、水晶体、鼻、内耳、三叉神経、腺性下垂体等が挙げられる。プラコード、又はその前駆組織である前プラコード領域(preplacode region)のマーカーとしては、Six1、Six4、Dlx5、Eya2、Emx2、Bf1等が挙げられる。 As used herein, the term "placode" refers to the primordium of an organ that is formed by the thickening of a portion of the epidermal ectoderm, primarily during the developmental process of vertebrates. Examples of tissues derived from the placode include the lens, nose, inner ear, trigeminal nerve, and adenohypophysis. Markers of the placode or its precursor tissue, the preplacode region, include Six1, Six4, Dlx5, Eya2, Emx2, and Bf1.
本明細書において、「下垂体プラコード」とは、胚発生の過程で表皮外胚葉の領域に形成される肥厚した構造であって、下垂体前駆細胞マーカーを発現するものをいう。下垂体前駆細胞マーカーとしては、Lim3(Lhx3)、Pitx1/2、Islet1/2等を挙げることができる。下垂体プラコードは、Lim3、Pitx1/2及びIslet1/2からなる群から選択される少なくとも1つ、好ましくは全ての下垂体前駆細胞マーカーを発現する。下垂体プラコードが陥入し、発生途中の袋状の構造であるラトケ嚢(Rathke’s pouch)を形成し、さらに発生が進むと腺性下垂体を形成する。 As used herein, the term "pituitary placode" refers to a thickened structure formed in the epidermal ectoderm region during embryonic development, which expresses pituitary progenitor cell markers. Examples of pituitary progenitor cell markers include Lim3 (Lhx3), Pitx1/2, Islet1/2, etc. The pituitary placode expresses at least one, and preferably all, pituitary progenitor cell markers selected from the group consisting of Lim3, Pitx1/2, and Islet1/2. The pituitary placode invaginates to form Rathke's pouch, a sac-like structure in the developing stage, and as development progresses further, it forms the adenohypophysis.
本明細書において、「腺性下垂体」とは、少なくとも1種の前葉又は中葉の下垂体細胞を含む組織をいう。下垂体細胞には、生理機能を調節するホルモンを産生する下垂体ホルモン産生細胞と、非ホルモン産生細胞が含まれる。下垂体ホルモン産生細胞としては、成長ホルモン(GH)産生細胞、プロラクチン(PRL)産生細胞、副腎皮質刺激ホルモン(ACTH)産生細胞、甲状腺刺激ホルモン(TSH)産生細胞、卵胞刺激ホルモン(FSH)産生細胞、黄体化ホルモン(LH)産生細胞等の前葉を構成する細胞;メラニン細胞刺激ホルモン(MSH)産生細胞等の中葉を構成する細胞が挙げられる。非ホルモン産生細胞としては、血管内皮細胞、周皮細胞、濾胞星状細胞、下垂体幹細胞、下垂体前駆細胞が含まれる。 As used herein, the term "adenohypophysis" refers to tissue that contains at least one type of pituitary cell of the anterior or intermediate lobe. Pituitary cells include pituitary hormone-producing cells that produce hormones that regulate physiological functions, and non-hormone-producing cells. Pituitary hormone-producing cells include cells that make up the anterior lobe, such as growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, adrenocorticotropic hormone (ACTH)-producing cells, thyroid-stimulating hormone (TSH)-producing cells, follicle-stimulating hormone (FSH)-producing cells, and luteinizing hormone (LH)-producing cells; and cells that make up the intermediate lobe, such as melanocyte-stimulating hormone (MSH)-producing cells. Non-hormone-producing cells include vascular endothelial cells, pericytes, follicle-stellate cells, pituitary stem cells, and pituitary progenitor cells.
一態様において、腺性下垂体は、成長ホルモン(GH)産生細胞、プロラクチン(PRL)産生細胞、及び副腎皮質刺激ホルモン(ACTH)産生細胞からなる群から選択される少なくとも1種、好ましくは2種、より好ましくは3種の下垂体ホルモン産生細胞を含む。更なる態様において、腺性下垂体は、成長ホルモン(GH)産生細胞、プロラクチン(PRL)産生細胞、副腎皮質刺激ホルモン(ACTH)産生細胞、甲状腺刺激ホルモン(TSH)産生細胞、卵胞刺激ホルモン(FSH)産生細胞、及び黄体化ホルモン(LH)産生細胞からなる群から選択される少なくとも1種、好ましくは2種以上(2、3、4、5又は6種)の下垂体ホルモン産生細胞を含む。
生体由来の組織中並びに本発明の製造方法等で作製された細胞集団中に腺性下垂体及び下垂体ホルモン産生細胞が含まれているかどうかは、下垂体ホルモン産生細胞マーカーを用いた免疫組織化学及び分泌されたホルモンに対するELISA等の手法により検出することが出来る。
下垂体ホルモン産生細胞マーカーとしては、成長ホルモン(GH)産生細胞マーカー(抗Pit1抗体、抗GH抗体等)、プロラクチン(PRL)産生細胞マーカー(抗Pit1抗体、抗PRL抗体等)、副腎皮質刺激ホルモン(ACTH)産生細胞マーカー(抗T-Pit抗体、抗NeuroD1抗体、抗ACTH抗体等)、甲状腺刺激ホルモン(TSH)産生細胞マーカー(抗GATA2抗体、抗ACTH抗体等)、並びに、卵胞刺激ホルモン(FSH)産生細胞及び黄体化ホルモン(LH)産生細胞のマーカー(抗GATA2抗体、抗SF1抗体、抗FSH抗体、抗LH抗体等)を用いた免疫組織化学及び分泌されたホルモンに対するELISA等の手法により検出することが出来る。
In one embodiment, the adenohypophysis contains at least one, preferably two, more preferably three types of pituitary hormone-producing cells selected from the group consisting of growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, and adrenocorticotropic hormone (ACTH)-producing cells. In a further embodiment, the adenohypophysis contains at least one, preferably two or more (2, 3, 4, 5, or 6) types of pituitary hormone-producing cells selected from the group consisting of growth hormone (GH)-producing cells, prolactin (PRL)-producing cells, adrenocorticotropic hormone (ACTH)-producing cells, thyroid stimulating hormone (TSH)-producing cells, follicle stimulating hormone (FSH)-producing cells, and luteinizing hormone (LH)-producing cells.
Whether or not adenohypophysis and pituitary hormone-producing cells are present in tissue derived from a living body and in a cell population produced by the production method of the present invention can be detected by techniques such as immunohistochemistry using pituitary hormone-producing cell markers and ELISA for the secreted hormones.
Pituitary hormone-producing cell markers include growth hormone (GH)-producing cell markers (anti-Pit1 antibody, anti-GH antibody, etc.), prolactin (PRL)-producing cell markers (anti-Pit1 antibody, anti-PRL antibody, etc.), adrenocorticotropic hormone (ACTH)-producing cell markers (anti-T-Pit antibody, anti-NeuroD1 antibody, anti-ACTH antibody, etc.), thyroid stimulating hormone (TSH)-producing cell markers (anti-GATA2 antibody, anti-ACTH antibody, etc.), and markers of follicle stimulating hormone (FSH)-producing cells and luteinizing hormone (LH)-producing cells (anti-GATA2 antibody, anti-SF1 antibody, anti-FSH antibody, anti-LH antibody, etc.). These can be detected by immunohistochemistry using these markers, and ELISA for the secreted hormones.
本明細書において「下垂体幹細胞」とは、下垂体に存在し、下垂体組織の再生や下垂体ホルモン産生細胞の供給に寄与する未分化な複能性幹細胞、前駆細胞のことをいう。本発明の製造方法で製造される細胞集団並びに組織中に下垂体幹細胞が含まれているかどうかは、例えばSox2、Sox9、E-Cadherin、Nestin、S100β、GFRα2、Prop1、CD133、β-Catenin、Klf4、Oct4、Pax6、コクサッキーウイルス・アデノウイルス共通受容体(CXADR)、PRRX1/2、Ephrin-B2、ACEといった下垂体幹細胞マーカー及びKi67、リン酸化ヒストンH3、MCMといった細胞増殖マーカーに対する抗体を用いた免疫組織化学、BrdU、EdU、IdU等の核酸アナログを用いた増殖細胞標識アッセイ、蛍光標識ジペプチド(β-alanyl-lysyl-N-7-amino-4-methylcoumarin-3-acetic acid)の取り込みアッセイ、下垂体スフェア(pitusphere)形成アッセイ等の手法により、検出することができる。 As used herein, "pituitary stem cells" refers to undifferentiated multipotent stem cells or progenitor cells that exist in the pituitary gland and contribute to the regeneration of pituitary tissue and the supply of pituitary hormone-producing cells. Whether or not pituitary stem cells are present in the cell population and tissue produced by the production method of the present invention can be determined by detecting pituitary stem cell markers such as Sox2, Sox9, E-Cadherin, Nestin, S100β, GFRα2, Prop1, CD133, β-Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE, as well as Ki67, Ribosomal protein markers such as ri ... Detection can be achieved by a variety of techniques, including immunohistochemistry using antibodies against cell proliferation markers such as phosphorylated histone H3 and MCM, proliferative cell labeling assays using nucleic acid analogs such as BrdU, EdU, and IdU, fluorescently labeled dipeptide (β-alanyl-lysyl-N-7-amino-4-methylcoumarin-3-acetic acid) uptake assays, and pituitary sphere formation assays.
本明細書において「口腔上皮」とは、口腔を形成する上皮組織及びその細胞を表す。口腔上皮としては、例えば口腔粘膜上皮、唾液腺上皮、歯原性上皮が挙げられる。口腔粘膜上皮は通常、重層扁平上皮からなる粘膜組織であり、結合組織と接した基底膜上に基底細胞、メルケル細胞、メラニン産生細胞等を含み、その上層に有刺細胞、顆粒細胞、角質層が形成される。口腔粘膜上皮は、例えばサイトケラチン7、8、13、14、19陽性の組織として検出され得る。
In this specification, "oral epithelium" refers to the epithelial tissue and cells that form the oral cavity. Examples of oral epithelium include oral mucosal epithelium, salivary gland epithelium, and odontogenic epithelium. Oral mucosal epithelium is usually a mucosal tissue made of stratified squamous epithelium, and contains basal cells, Merkel cells, melanin-producing cells, etc. on the basement membrane in contact with connective tissue, with spinous cells, granular cells, and a stratum corneum formed on the upper layer. Oral mucosal epithelium can be detected as tissue that is positive for
本明細書において、「ニッチ」、もしくは「幹細胞ニッチ」とは幹細胞の増殖、分化、性質の維持等に関わる微小環境をいう。生体におけるニッチの例としては、造血幹細胞ニッチ、毛包幹細胞ニッチ、腸管上皮幹細胞ニッチ、筋幹細胞ニッチ、下垂体ニッチなどが挙げられる。これらのニッチにおいては、それぞれの組織特有の幹細胞とニッチを提供する支持細胞とが存在し、支持細胞が提供するサイトカイン、ケモカイン、細胞外マトリクス、細胞接着因子、細胞間シグナル伝達因子等により幹細胞が維持されている。 In this specification, "niche" or "stem cell niche" refers to a microenvironment involved in the proliferation, differentiation, and maintenance of properties of stem cells. Examples of niches in the body include the hematopoietic stem cell niche, hair follicle stem cell niche, intestinal epithelial stem cell niche, muscle stem cell niche, and pituitary niche. In these niches, tissue-specific stem cells and supporting cells that provide the niche exist, and the stem cells are maintained by cytokines, chemokines, extracellular matrix, cell adhesion factors, intercellular signaling factors, and the like provided by the supporting cells.
本明細書において、「下垂体ニッチ」とは、下垂体幹細胞の増殖、分化、性質の維持等に関わる微小環境をいう。下垂体ニッチとしては、発生期の袋状のラトケ嚢の中空部の痕跡として下垂体前葉と中葉の間に残る遺残腔(ラトケ裂溝)周辺に存在するMarginal Cell Layer(MCL)ニッチ、及び下垂体前葉に散在する実質層(Parenchymal)ニッチが挙げられる。 As used herein, the term "pituitary niche" refers to the microenvironment involved in the proliferation, differentiation, and maintenance of properties of pituitary stem cells. Examples of pituitary niches include the Marginal Cell Layer (MCL) niche that exists around the residual cavity (Rathke's cleft) that remains between the anterior and intermediate lobes of the pituitary gland as a vestige of the hollow part of the sac-like Rathke's pouch during development, and the parenchymal niche that is scattered throughout the anterior pituitary gland.
本明細書において「間葉系細胞」とは、主に中胚葉並びに神経堤に由来し、結合組織を形成する非上皮性の細胞である。これらの細胞のうちの一部は、間葉系幹細胞と呼ばれる複能性を有した体性幹細胞である。本発明の製造方法で製造される細胞集団並びに組織中に間葉系細胞が含まれるかどうかは、Nestin、Vimentin、Cadherin-11、Laminin、CD44といった間葉系細胞マーカーに対する抗体を用いた免疫組織化学等の手法により検出することができる。間葉系幹細胞が含まれるかどうかは、CD9、CD13、CD29、CD44、CD55、CD59、CD73、CD105、CD140b、CD166、VCAM-1、STRO-1、c-Kit、Sca-1、Nucleostemin、CDCP1、BMPR2、BMPR1A及びBPMR1Bといった間葉系幹細胞マーカーに対する抗体を用いた免疫組織化学等の手法により検出することができる。 As used herein, "mesenchymal cells" refer to non-epithelial cells that are derived primarily from the mesoderm and neural crest and form connective tissue. Some of these cells are multipotent somatic stem cells known as mesenchymal stem cells. Whether or not mesenchymal cells are present in the cell populations and tissues produced by the production method of the present invention can be detected by techniques such as immunohistochemistry using antibodies against mesenchymal cell markers such as nestin, vimentin, cadherin-11, laminin, and CD44. Whether or not mesenchymal stem cells are present can be detected by techniques such as immunohistochemistry using antibodies against mesenchymal stem cell markers such as CD9, CD13, CD29, CD44, CD55, CD59, CD73, CD105, CD140b, CD166, VCAM-1, STRO-1, c-Kit, Sca-1, Nucleostemin, CDCP1, BMPR2, BMPR1A, and BPMR1B.
3.下垂体組織を含む細胞集団の製造方法
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞を含む細胞集団は、以下の工程を含む方法で分化誘導することにより製造することができる:
(A)多能性幹細胞を分化誘導可能な培地で培養して得られる細胞集団を、骨形成因子シグナル伝達経路活性化物質及びShhシグナル経路作用物質を含む培地中で浮遊培養する工程。
3. Method for Producing a Cell Population Comprising Pituitary Tissue In one embodiment, a cell population comprising pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be produced by inducing differentiation using a method comprising the following steps:
(A) A step of culturing pluripotent stem cells in a differentiation-inducing medium to obtain a cell population, and then culturing the cell population in suspension in a medium containing a substance that activates the bone morphogenetic protein signaling pathway and a substance that acts on the Shh signal pathway.
一態様において、前記細胞集団は、細胞凝集体であることが好ましい。具体的には、以下の工程を含む方法が挙げられる:
(A’)多能性幹細胞を浮遊培養して得られる細胞凝集体を、骨形成因子シグナル伝達経路活性化物質及びShhシグナル経路作用物質を含む培地中で浮遊培養する工程。
In one embodiment, the cell population is preferably a cell aggregate. Specifically, the method includes the following steps:
(A') A step of suspension-culturing cell aggregates obtained by suspension-culturing pluripotent stem cells in a medium containing an activator of bone morphogenetic protein signaling pathway and a substance acting on the Shh signal pathway.
前記工程(A)及び(A’)において、骨形成因子シグナル伝達経路活性化物質及びShhシグナル経路作用物質、その濃度、並びにそれらの存在下での培養期間については、国際公開第2016/013669号、国際公開第2019/103129号、Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351)等に記載された方法、又は、「4.下垂体組織を含む細胞集団の製造方法1」の工程(2)に記載の培養条件に準じて行うことができる。
In the steps (A) and (A'), the bone morphogenetic protein signaling pathway activator and the Shh signal pathway acting substance, their concentrations, and the culture period in their presence can be performed according to the methods described in WO 2016/013669, WO 2019/103129, Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351), etc., or the culture conditions described in step (2) of "4.
一態様において、前記工程(A)又は(A’)の培養期間は、1)視床下部神経上皮組織、及び2)下垂体プラコード及び/又はラトケ嚢を含む細胞凝集体を得ることができる培養条件及び培養期間であればよい。一態様として、培養中の細胞凝集体のうち10%以上、好ましくは30%以上、より好ましくは50%以上の細胞凝集体が下垂体プラコード及び/又はラトケ嚢を含むまで、工程(A)又は(A’)が実施される。工程(A)又は(A’)の培養期間は、例えば、6日以上、8日以上、10日以上、且つ、20日以下、18日以下、16日以下であってよく、一例として、10日以上16日以下であってよい。
工程(A)又は(A’)において得られる細胞凝集体に含まれる視床下部神経上皮組織は、好ましくは、腹側視床下部神経上皮組織である。
In one embodiment, the culture period of the step (A) or (A') may be any culture condition and culture period that can obtain cell aggregates containing 1) hypothalamic neuroepithelial tissue, and 2) pituitary placode and/or Rathke's pouch. In one embodiment, the step (A) or (A') is performed until 10% or more, preferably 30% or more, more preferably 50% or more of the cell aggregates in the culture contain pituitary placode and/or Rathke's pouch. The culture period of the step (A) or (A') may be, for example, 6 days or more, 8 days or more, 10 days or more, and 20 days or less, 18 days or less, or 16 days or less, and may be, for example, 10 days or more and 16 days or less.
The hypothalamic neuroepithelial tissue contained in the cell aggregate obtained in step (A) or (A') is preferably ventral hypothalamic neuroepithelial tissue.
骨形成因子シグナル伝達経路活性化物質の濃度は、添加する期間内で変動させてもよく、例えば、2~4日間毎に半分の割合で、段階的に濃度を低下させることができる。 The concentration of the bone morphogenetic protein signaling pathway activator may be varied during the period of addition; for example, the concentration may be gradually reduced by half every 2 to 4 days.
工程(A)又は(A’)終了後、骨形成因子シグナル伝達経路活性化物質を含まない培地での培養を継続してもよい。
また、工程(A)又は(A’)終了後、骨形成因子シグナル伝達経路活性化物質を含まずShhシグナル伝達経路作用物質を含む培地での培養を継続してもよい。
工程(A)又は(A’)終了後、高酸素分圧条件下で培養してもよい。高酸素分圧条件とは、空気中の酸素分圧(20%)を上回る酸素分圧条件を意味する。例えば、30~60%、好ましくは35~60%、より好ましくは40~60%である。
After completion of step (A) or (A'), the culture may be continued in a medium not containing the bone morphogenetic protein signaling pathway activator.
After completion of step (A) or (A'), the cells may be continuously cultured in a medium that does not contain a substance that activates the bone morphogenetic protein signaling pathway but contains a substance that acts on the Shh signaling pathway.
After completion of step (A) or (A'), the cells may be cultured under high oxygen partial pressure conditions. High oxygen partial pressure conditions refer to conditions in which the oxygen partial pressure exceeds the oxygen partial pressure in air (20%). For example, the oxygen partial pressure is 30 to 60%, preferably 35 to 60%, and more preferably 40 to 60%.
前記工程(A)又は(A’)終了後の培養は、下垂体プラコード及び/又はラトケ嚢から下垂体ホルモン産生細胞への分化が誘導されるのに十分な期間実施されればよく、例えば、培養中の細胞凝集体のうち5%以上、10%以上、30%以上又は50%以上の細胞凝集体が下垂体ホルモン産生細胞を含むまで実施されればよい。具体的には、例えば、10日以上、20日以上、30日以上、40日以上、且つ、90日以下、80日以下、70日以下、60日以下、50日以下であってよい。 The culture after completion of step (A) or (A') may be performed for a period of time sufficient to induce differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells, for example, until 5% or more, 10% or more, 30% or more, or 50% or more of the cell aggregates being cultured contain pituitary hormone-producing cells. Specifically, the period may be, for example, 10 days or more, 20 days or more, 30 days or more, 40 days or more, and 90 days or less, 80 days or less, 70 days or less, 60 days or less, or 50 days or less.
一態様において、前記工程(A’)において、多能性幹細胞を浮遊培養して得られる細胞凝集体は、分散された多能性幹細胞を、分化可能な培地中で非接着性の条件下で培養することにより、形成される細胞凝集体である。
一態様において、当該細胞凝集体の製造方法として、SFEBq法(Serum-Free Floating culture of Embryoid Body-like aggregates with quick reaggregation)が挙げられ、分化誘導開始(多能性幹細胞の浮遊培養開始)から12時間~72時間以内に形成される。ここで用いられる分化可能な培地は上述の培地より適宜選択することができる。当該培地には、下垂体ホルモン産生細胞の分化誘導に適した分化誘導剤等が、適宜含まれていてもよく、当該分化誘導剤等として、ROCK阻害剤、Shhシグナル伝達経路作用物質、JNKシグナル伝達経路阻害物質、Wntシグナル伝達経路阻害物質が挙げられる。当該細胞凝集体の製造についても、国際公開第2016/013669号、国際公開第2019/103129号、Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351)、国際公開第2023/054396号等に記載された方法、又は、「4.下垂体組織を含む細胞集団の製造方法1」の工程(1)又は(1’)に記載の培養条件に準じて行うことができる。また、「4.下垂体組織を含む細胞集団の製造方法1」の工程(1)又は(1’)に記載の培養条件を行う際には、上述したような公報(例えば、国際公開第2023/054396号等)に記載された方法を参酌した上で、行ってもよい。
In one aspect, the cell aggregates obtained by suspension culture of pluripotent stem cells in the step (A') are cell aggregates formed by culturing dispersed pluripotent stem cells under non-adhesive conditions in a differentiable medium.
In one embodiment, the method for producing the cell aggregates includes the SFEBq method (Serum-Free Floating culture of Embryoid Body-like aggregates with quick reaggregation), and the cell aggregates are formed within 12 to 72 hours from the start of differentiation induction (start of floating culture of pluripotent stem cells). The differentiable medium used here can be appropriately selected from the above-mentioned media. The medium may appropriately contain a differentiation inducer suitable for inducing differentiation of pituitary hormone-producing cells, and examples of the differentiation inducer include a ROCK inhibitor, a substance acting on the Shh signaling pathway, a substance inhibiting the JNK signaling pathway, and a substance inhibiting the Wnt signaling pathway. The cell aggregates can also be produced according to the methods described in WO 2016/013669, WO 2019/103129, Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells (Nat Commun. 2016; 7: 10351), WO 2023/054396, etc., or the culture conditions described in step (1) or (1') of "4. Method for producing a cell population containing
4.下垂体組織を含む細胞集団の製造方法1
本発明の治療薬に含まれる下垂体ホルモン産生細胞あるいは該細胞を含む細胞集団の製造方法の一態様は、下記工程(1’)~(2)を含む、下垂体ホルモン産生細胞を含む細胞集団の製造方法である(以下、「製造方法1」とも称する。)。
(1’)多能性幹細胞をJNKシグナル伝達経路阻害物質の存在下で培養し、細胞集団を形成させる第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質及びソニック・ヘッジホッグシグナル伝達経路作用物質の存在下に培養し、下垂体組織を含む細胞集団を得る第二工程。
工程(1’)において、好ましくは、JNKシグナル伝達経路阻害物質にWntシグナル伝達経路阻害物質を併用する。
また、工程(2)における下垂体組織は、工程(2)の培養期間によって、下垂体ホルモン産生細胞の前駆細胞及び/又は下垂体ホルモン産生細胞を含んでいてもよい。
工程(2)の終了後に、後述する工程(3)を行い、工程(2)で生成する当該前駆細胞を、下垂体ホルモン産生細胞へ分化させることができる。
4. Method for producing a cell population containing
One embodiment of a method for producing pituitary hormone-producing cells or a cell population containing said cells included in the therapeutic agent of the present invention is a method for producing a cell population containing pituitary hormone-producing cells, comprising the following steps (1') to (2) (hereinafter also referred to as "
(1') a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor to form a cell population;
(2) A second step of culturing the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue.
In step (1'), preferably, a substance inhibiting the JNK signaling pathway is used in combination with a substance inhibiting the Wnt signaling pathway.
Furthermore, the pituitary tissue in step (2) may contain precursor cells of pituitary hormone-producing cells and/or pituitary hormone-producing cells, depending on the culture period in step (2).
After completion of step (2), the below-described step (3) can be carried out to differentiate the precursor cells generated in step (2) into pituitary hormone-producing cells.
本発明の製造方法のより好ましい一態様は、下記工程(a)及び下記工程
(1’)~(2)を含む下垂体組織を含む細胞集団の製造方法である。
(a)多能性幹細胞を、フィーダー細胞非存在下で、1)TGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質、並びに2)未分化維持因子を含む培地で培養するa工程、
(1’)a工程で得られた細胞をJNKシグナル伝達経路阻害物質の存在下で培養(好ましくは浮遊培養)する第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質とソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養(好ましくは浮遊培養)し、下垂体組織を含有する細胞集団を得る第二工程。
好ましくは、第一工程は細胞の凝集体を形成させる工程であり、第二工程に付す第一工程で得られた細胞集団は細胞の凝集体であり得る。
工程(1’)において、好ましくは、JNKシグナル伝達経路阻害物質にWntシグナル伝達経路阻害物質を併用する。
A more preferred embodiment of the production method of the present invention is a method for producing a cell population containing pituitary tissue, comprising the following steps (a) and (1') to (2):
(a) a step a of culturing pluripotent stem cells in a medium containing 1) a TGFβ family signaling pathway inhibitor and/or a Sonic Hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells;
(1') a first step of culturing (preferably in suspension) the cells obtained in step a in the presence of a substance inhibiting the JNK signaling pathway;
(2) A second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue.
Preferably, the first step is a step of forming cell aggregates, and the cell population obtained in the first step that is subjected to the second step may be cell aggregates.
In step (1'), preferably, a substance inhibiting the JNK signaling pathway is used in combination with a substance inhibiting the Wnt signaling pathway.
本発明の製造方法の一層好ましい一態様は、下記工程(a)及び下記工程(1)~(3)を含む下垂体組織を含む細胞集団の製造方法である。
(a)多能性幹細胞を、フィーダー細胞非存在下で、1)TGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質、並びに2)未分化維持因子を含む培地で培養するa工程、
(1)a工程で得られた細胞をJNKシグナル伝達経路阻害物質及びWntシグナル伝達経路阻害物質の存在下で培養(好ましくは浮遊培養)する第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質とソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養(好ましくは浮遊培養)する第二工程、
(3)第二工程で得られた細胞集団を、ソニック・ヘッジホッグシグナル伝達経路作用物質の非存在下で培養(好ましくは浮遊培養)し、下垂体ホルモン産生細胞を含む細胞集団を得る第三工程。
好ましくは、第一工程は細胞の凝集体を形成させる工程であり、第二工程に付す第一工程で得られた細胞集団、及び第三工程に付す第二工程で得られた細胞集団は、それぞれ細胞の凝集体であり得る。
A more preferred embodiment of the production method of the present invention is a method for producing a cell population containing pituitary tissue, comprising the following step (a) and the following steps (1) to (3):
(a) a step a of culturing pluripotent stem cells in a medium containing 1) a TGFβ family signaling pathway inhibitor and/or a Sonic Hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells;
(1) a first step of culturing (preferably in suspension) the cells obtained in step a in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor;
(2) a second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway;
(3) A third step in which the cell population obtained in the second step is cultured (preferably in suspension) in the absence of a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary hormone-producing cells.
Preferably, the first step is a step of forming cell aggregates, and the cell population obtained in the first step that is subjected to the second step, and the cell population obtained in the second step that is subjected to the third step, may each be a cell aggregate.
<工程(a)>:a工程
多能性幹細胞を、フィーダー細胞非存在下で、1)TGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質、並びに2)未分化維持因子を含む培地で培養するa工程について説明する。
工程(a)において、多能性幹細胞をTGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質で処理してから、第一工程において培養(好ましくは浮遊培養)に付すことにより、多能性幹細胞の状態が変わり、非神経上皮組織の形成効率が改善し、得られる細胞集団(凝集体)の質が向上し、分化しやすく、細胞死が生じにくく、下垂体組織の細胞の製造効率が向上する。
<Step (a)>: Step a Step a is described below, in which pluripotent stem cells are cultured in a medium containing 1) a TGFβ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and 2) an undifferentiated maintenance factor, in the absence of feeder cells.
In step (a), pluripotent stem cells are treated with a TGFβ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and then cultured (preferably in suspension) in the first step, thereby changing the state of the pluripotent stem cells, improving the efficiency of non-neuroepithelial tissue formation, improving the quality of the resulting cell population (aggregates), making them more likely to differentiate and less likely to suffer cell death, and improving the efficiency of producing pituitary tissue cells.
工程(a)は、フィーダー細胞非存在下で実施する。
本明細書におけるフィーダー細胞非存在下(フィーダーフリー)とは、フィーダー細胞を実質的に含まない(例えば、全細胞数に対するフィーダー細胞数の割合が3%以下)条件を意味する。
Step (a) is carried out in the absence of feeder cells.
As used herein, the term "feeder-free" refers to conditions substantially free of feeder cells (for example, the ratio of the number of feeder cells to the total number of cells is 3% or less).
本発明に係る下垂体ホルモン産生細胞の製造方法において、多能性幹細胞は、好ましくは胚性幹細胞又は人工多能性幹細胞である。人工多能性幹細胞は、所定の機関より入手でき、市販品を購入することもできる。例えば、ヒト人工多能性幹細胞株201B7株、201B7-Ff細胞、253G1細胞、253G4細胞、1201C1細胞、1205D1細胞、1210B2細胞、1231A3細胞は京都大学及びiPSアカデミアジャパン株式会社より入手できる。株化された人工多能性幹細胞として、例えば、京都大学で樹立されたFf-I01細胞、Ff-I14細胞及びQHJI01s04細胞が、京都大学より入手可能である。また、HC-6 #10株、1231A3株及び1383D2株は国立研究開発法人理化学研究所より入手できる。 In the method for producing pituitary hormone-producing cells according to the present invention, the pluripotent stem cells are preferably embryonic stem cells or induced pluripotent stem cells. Induced pluripotent stem cells are available from a designated institution, and can also be purchased commercially. For example, human induced pluripotent stem cell lines 201B7, 201B7-Ff, 253G1, 253G4, 1201C1, 1205D1, 1210B2, and 1231A3 are available from Kyoto University and iPS Academia Japan, Inc. As established induced pluripotent stem cell lines, for example, Ff-I01, Ff-I14, and QHJI01s04 cells established at Kyoto University are available from Kyoto University. In addition, HC-6 #10, 1231A3, and 1383D2 lines are available from the National Institute of Physical and Chemical Research.
TGFβファミリーシグナル伝達経路(すなわち、TGFβスーパーファミリーシグナル伝達経路)とは、形質転換増殖因子β(TGFβ)、Nodal/Activin、又はBMPをリガンドとし、細胞内でSmadファミリーにより伝達されるシグナル伝達経路である。 The TGFβ family signaling pathway (i.e., the TGFβ superfamily signaling pathway) is a signaling pathway that uses transforming growth factor β (TGFβ), Nodal/Activin, or BMP as ligands and is transmitted intracellularly by the Smad family.
TGFβファミリーシグナル伝達経路阻害物質とは、TGFβファミリーシグナル伝達経路、すなわちSmadファミリーにより伝達されるシグナル伝達経路を阻害する物質を表し、具体的にはTGFβシグナル伝達経路阻害物質、Nodal/Activinシグナル伝達経路阻害物質及びBMPシグナル伝達経路阻害物質を挙げることができる。TGFβファミリーシグナル伝達経路阻害物質としては、TGFβシグナル伝達経路阻害物質が好ましい。 The term "TGFβ family signaling pathway inhibitor" refers to a substance that inhibits the TGFβ family signaling pathway, i.e., the signaling pathway transmitted by the Smad family, and specific examples include TGFβ signaling pathway inhibitors, Nodal/Activin signaling pathway inhibitors, and BMP signaling pathway inhibitors. As the TGFβ family signaling pathway inhibitor, TGFβ signaling pathway inhibitors are preferred.
TGFβシグナル伝達経路阻害物質としては、TGFβに起因するシグナル伝達経路を阻害する物質であれば特に限定は無く、核酸、タンパク質、低分子有機化合物のいずれであってもよい。当該物質として例えばTGFβに直接作用する物質(例えばタンパク質、抗体、アプタマー等)、TGFβをコードする遺伝子の発現を抑制する物質(例えばアンチセンスオリゴヌクレオチド、siRNA等)、TGFβ受容体とTGFβの結合を阻害する物質、TGFβ受容体によるシグナル伝達に起因する生理活性を阻害する物質(例えば、TGFβ受容体の阻害剤、Smadの阻害剤等)を挙げることができる。TGFβシグナル伝達経路阻害物質として知られているタンパク質として、Leftyが挙げられる。 The TGFβ signaling pathway inhibitor is not particularly limited as long as it inhibits the signaling pathway caused by TGFβ, and may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that act directly on TGFβ (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes that code for TGFβ (e.g., antisense oligonucleotides, siRNA, etc.), substances that inhibit the binding between TGFβ receptors and TGFβ, and substances that inhibit physiological activities caused by signaling via TGFβ receptors (e.g., TGFβ receptor inhibitors, Smad inhibitors, etc.). An example of a protein known as an inhibitor of the TGFβ signaling pathway is Lefty.
TGFβシグナル伝達経路阻害物質として、当業者に周知の化合物を使用することができる。具体的には、SB431542(「SB431」と略記する場合がある。)(4-[4-(3,4-Methylenedioxyphenyl)-5-(2-pyridyl)-1H-imidazol-2-yl]benzamide)、SB505124(2-[4-(1,3-Benzodioxol-5-yl)-2-(1,1-dimethylethyl)-1H-imidazol-5-yl]-6-methylpyridine)、SB525334(6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline)、LY2157299(4-[5,6-Dihydro-2-(6-methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-6-quinolinecarboxamide)、LY2109761(4-[5,6-dihydro-2-(2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-7-[2-(4-morpholinyl)ethoxy]-quinoline)、GW788388(4-{4-[3-(Pyridin-2-yl)-1H-pyrazol-4-yl]-pyridin-2-yl}-N-(tetrahydro-2H-pyran-4-yl)benzamide)、LY364947(4-[3-(2-Pyridinyl)-1H-pyrazol-4-yl]quinoline)、SD-208(2-[(5-Chloro-2-fluorophenyl)pteridin-4-yl]pyridin-4-yl-amine)、EW-7197(N-(2-fluorophenyl)-5-(6-methyl-2-pyridinyl)-4-[1,2,4]triazolo[1,5-a]pyridin-6-yl-1H-Imidazole-2-methanamine)、A83-01(3-(6-Methylpyridin-2-yl)-4-(4-quinolyl)-1-phenylthiocarbamoyl-1H-pyrazole)、RepSox(2-[5-(6-Methylpyridin-2-yl)-1H-pyrazol-4-yl]-1,5-naphthyridine)、SM16(4-[4-(1,3-Benzodioxol-5-yl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-2-yl]bicyclo[2.2.2]octane-1-carboxamide)、R268712(4-[2-Fluoro-5-[3-(6-methyl-2-pyridinyl)-1H-pyrazol-4-yl]phenyl]-1H-pyrazole-1-ethanol)、IN1130(3-[[5-(6-Methyl-2-pyridinyl)-4-(6-quinoxalinyl)-1H-imidazol-2-yl]methyl]benzamide)、Galunisertib(4-[5,6-Dihydro-2-(6-methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-6-quinolinecarboxamide)、AZ12799734(4-({4-[(2,6-dimethylpyridin-3-yl)oxy]pyridin-2-yl}amino)benzenesulfonamide)、A77-01(4-[3-(6-Methylpyridin-2-yl)-1H-pyrazol-4-yl]quinoline)、KRCA 0008(1,1-[(5-Chloro-2,4-pyrimidinediyl)bis[imino(3-methoxy-4,1-phenylene)-4,1-piperazinediyl]]bisethanone)、GSK 1838705(2-[[2-[[1-[(Dimethylamino)ethanoyl]-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino]-7H-pyrrolo[2,3-d]pyrimidin-4-yl]amino]-6-fluoro-N-methylbenzamide)、Crizotinib(3-[(1R)-1-(2,6-Dichloro-3-fluorophenyl)ethoxy]-5-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]-2-aminopyridine)、Ceritinib(5-Chloro-N2-[2-isopropoxy-5-methyl-4-(4-piperidyl)phenyl]-N4-(2-isopropylsulfonylphenyl)pyrimidine-2,4-diamine)、ASP 3026(N2-[2-Methoxy-4-[4-(4-methyl-1-piperazinyl)-1-piperidinyl]phenyl]-N4-[2-[(1-methylethyl)sulfonyl]phenyl]-1,3,5-triazine-2,4-diamine)、TAE684(5-Chloro-N2-[2-methoxy-4-[4-(4-methyl-1-piperazinyl)-1-piperidinyl]phenyl]-N4-[2-[(1-methylethyl)sulfonyl]phenyl]-2,4-pyrimidinediamine)、AZD3463(N-[4-(4-Amino-1-piperidinyl)-2-methoxyphenyl]-5-chloro-4-(1H-indol-3-yl)-2-pyrimidinamine)、TP0427736(6-[4-(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole)、TGFBR1-IN-1(5-(1,3-benzothiazol-6-yl)-N-(4-hydroxyphenyl)-1-(6-methylpyridin-2-yl)pyrazole-3-carboxamide)、TEW-7197(2-fluoro-N-[[5-(6-methylpyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-1H-imidazol-2-yl]methyl]aniline)、LY3200882(2-[4-[[4-[1-cyclopropyl-3-(oxan-4-yl)pyrazol-4-yl]oxypyridin-2-yl]amino]pyridin-2-yl]propan-2-ol)、BIBF-0775((3Z)-N-Ethyl-2,3-dihydro-N-methyl-2-oxo-3-[phenyl[[4-(1-piperidinylmethyl)phenyl]amino]methylene]-1H-indole-6-carboxamide)等のAlk5/TGFβR1阻害剤、SIS3(1-(3,4-dihydro-6,7-dimethoxy-2(1H)-isoquinolinyl)-3-(1-methyl-2-phenyl-1H-pyrrolo[2,3-b]pyridin-3-yl)-2-propen-1-one)等のSMAD3阻害剤、ITD-1(4-[1,1’-Biphenyl]-4-yl-1,4,5,6,7,8-hexahydro-2,7,7-trimethyl-5-oxo-3-quinolinecarboxylic acid ethyl ester)等の受容体分解促進剤及びこれら化合物の誘導体等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。SB431542は、TGFβ受容体(ALK5)及びActivin受容体(ALK4/7)の阻害剤(すなわちTGFβR阻害剤)として公知の化合物である。SIS3は、TGFβ受容体の制御下にある細胞内シグナル伝達因子であるSMAD3のリン酸化を阻害するTGFβシグナル伝達経路阻害物質である。ITD-1は、TGF-β type II receptorのプロテアソーム分解促進剤である。上記の化合物等がTGFβシグナル伝達経路阻害物質としての活性を有することは当業者にとって公知である(例えばExpert opinion on investigational drugs, 2010, 19.1: 77-91.等に記載されている)。 Compounds well known to those skilled in the art can be used as TGFβ signaling pathway inhibitors. Specifically, SB431542 (sometimes abbreviated as "SB431") (4-[4-(3,4-Methylenedioxyphenyl)-5-(2-pyridyl)-1H-imidazol-2-yl]benzoamide), SB505124 (2-[4-(1,3-Benzodioxol-5-yl)-2-(1,1-dimethylethyl)-1H-imidazol-5-yl]-6-methylpyridine), S B525334 (6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline), LY2157299 (4-[5,6-Dihydro-2-(6 -methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-6-quinolinecarboxamide), LY2109761(4- 5,6-dihydro-2-(2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-7-[2-(4-morpholinyl)ethoxy]-quinoline), GW788388 (4-{4-[3- (Pyridin-2-yl)-1H-pyrazol-4-yl]-pyridin-2-yl}-N-(tetrahydro-2H-pyran-4-yl)benzamide), LY364 947 (4-[3-(2-Pyridinyl)-1H-pyrazol-4-yl]quinoline), SD-208 (2-[(5-Chloro-2-fluorophenyl)pteridin-4-yl]pyridin-4-yl-amine ), EW-7197 (N-(2-fluorophenyl)-5-(6-methyl-2-pyridinyl)-4-[1,2,4]triazolo[1,5-a]pyridin-6-y l-1H-Imidazole-2-methanamine), A83-01 (3-(6-Methylpyridin-2-yl)-4-(4-quinolyl)-1-phenylthiocarbamoyl-1H-pyrazole), Rep Sox (2-[5-(6-Methylpyridin-2-yl)-1H-pyrazol-4-yl]-1,5-naphthyridine), SM16 (4-[4-(1,3-Benzodi oxol-5-yl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-2-yl]bicyclo[2.2.2]octane-1-carboxamide), R268712(4-[2-Fluoro-5-[3-(6-me thyl-2-pyridinyl)-1H-pyrazol-4-yl]phenyl]-1H-pyrazole-1-ethanol), IN1130(3-[[5-(6-Methyl -2-pyridinyl)-4-(6-quinoxalinyl)-1H-imidazol-2-yl]methyl]benzamide), Galunisertib (4-[5,6-Dihydro-2-(6-methyl-2-pyrid inyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-6-quinolinecarboxamide), AZ12799734(4-({4-[(2,6-dimethy lpyridin-3-yl)oxy]pyridin-2-yl}amino)benzenesulfonamide), A77-01(4-[3-(6-Methylpyridin-2-yl)-1H-pyrazol-4-yl]quino line), KRCA 0008(1,1-[(5-Chloro-2,4-pyrimidinediyl)bis[imino(3-methoxy-4,1-phenylene)-4,1-pipe razinediyl]]bisethanone), GSK 1838705 (2-[[2-[[1-[(Dimethylamino)ethanoyl]-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino] -7H-pyrrolo[2,3-d]pyrimidin-4-yl]amino]-6-fluoro-N-methylbenzamide), Crizotinib (3-[(1R) -1-(2,6-Dichloro-3-fluorophenyl)ethoxy]-5-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]-2-aminopyridine), Ceritinib (5-Chloro-N2-[ 2-isopropoxy-5-methyl-4-(4-piperidyl)phenyl]-N4-(2-isopropylsulfonylphenyl)pyrimidine-2 , 4-diamine), ASP 3026 (N2-[2-Methoxy-4-[4-(4-methyl-1-piperazinyl)-1-piperidinyl]phenyl]-N4-[2-[(1-methylethyl)sulfonyl]p henyl]-1,3,5-triazine-2,4-diamine), TAE684(5-Chloro-N2-[2-methoxy-4-[4-(4-methyl-1-pipe razinyl)-1-piperidinyl]phenyl]-N4-[2-[(1-methylethyl)sulfonyl]phenyl]-2,4-pyrimidinediamine), AZD3463(N-[4-(4-Amino-1- piperidinyl)-2-methoxyphenyl]-5-chloro-4-(1H-indol-3-yl)-2-pyrimidinamine), TP0427736(6-[4 -(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole), TGFBR1-IN-1(5-(1,3-benzothiazol-6-yl)-N-(4- hydroxyphenyl)-1-(6-methylpyridin-2-yl)pyrazole-3-carboxamide), TEW-7197(2-fluoro-N-[[5-(6-methyl pyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-1H-imidazol-2-yl]methyl]aniline), LY3200882(2-[4-[[4-[1-cycloprop yl-3-(oxan-4-yl)pyrazol-4-yl]oxypyridin-2-yl]amino]pyridin-2-yl]propan-2-ol), BIBF-0775((3 Z) -N-Ethyl-2,3-dihydro-N-methyl-2-oxo-3-[phenyl[[4-(1-piperidinylmethyl)phenyl]amino]methylene]-1H-indole-6-carboxamide ), SIS3 (1-(3,4-dihydro-6,7-dimethoxy-2(1H)-isoquinolinyl)-3-(1-methyl-2- Examples of such inhibitors include SMAD3 inhibitors such as 4-[1,1'-biphenyl]-4-yl-1,4,5,6,7,8-hexahydro-2,7,7-trimethyl-5-oxo-3-quinolinoleboxylic acid ethyl ester (ITD-1), and derivatives of these compounds. These substances may be used alone or in combination. SB431542 is a compound known as an inhibitor of the TGFβ receptor (ALK5) and Activin receptor (ALK4/7) (i.e., a TGFβR inhibitor). SIS3 is a TGFβ signaling pathway inhibitor that inhibits phosphorylation of SMAD3, an intracellular signaling factor under the control of the TGFβ receptor. ITD-1 is a promoter of proteasomal degradation of the TGF-β type II receptor. Those skilled in the art are aware that the above compounds have activity as TGFβ signaling pathway inhibitors (for example, see Expert Opinion on Investigational Drugs, 2010, 19.1: 77-91, etc.).
TGFβシグナル伝達経路阻害物質は、好ましくはAlk5/TGFβR1阻害剤を含む。Alk5/TGFβR1阻害剤は、好ましくはSB431542、SB505124、SB525334、LY2157299、GW788388、LY364947、SD-208、EW-7197、A83-01、RepSox、SM16、R268712、IN1130、Galunisertib、AZ12799734、A77-01、KRCA 0008、GSK 1838705、Crizotinib、Ceritinib、ASP 3026、TAE684、AZD3463、TP0427736からなる群より選ばれる少なくとも1つを含み、さらに好ましくはSB431542又はA83-01を含む。 The TGFβ signaling pathway inhibitor preferably includes an Alk5/TGFβR1 inhibitor. The Alk5/TGFβR1 inhibitor is preferably SB431542, SB505124, SB525334, LY2157299, GW788388, LY364947, SD-208, EW-7197, A83-01, RepSox, SM16, R268712, IN1130, Galunisertib, AZ1 2799734, A77-01, KRCA 0008, GSK 1838705, Crizotinib, Certinib, ASP 3026, TAE684, AZD3463, TP0427736, and more preferably SB431542 or A83-01.
培地中のTGFβシグナル伝達経路阻害物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。工程(a)におけるTGFβシグナル伝達経路阻害物質としてSB431542を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約100μM、より好ましくは約10nM~約50μM、さらに好ましくは約100nM~約50μM、特に好ましくは約1μM~約10μMの濃度で使用される。また、SB431542以外のTGFβシグナル伝達経路阻害物質を使用する場合、上記濃度のSB431542と同等のTGFβシグナル伝達経路阻害活性を示す濃度で用いられることが望ましい。なお、SB431542等のTGFβシグナル伝達経路阻害活性は、当業者に周知の方法、例えばSmadのリン酸化をウエスタンブロッティング法で検出することで決定できる(Mol Cancer Ther.(2004) 3,737-45.)。 The concentration of the TGFβ signaling pathway inhibitor in the medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. When SB431542 is used as the TGFβ signaling pathway inhibitor in step (a), it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 100 μM, more preferably about 10 nM to about 50 μM, even more preferably about 100 nM to about 50 μM, and particularly preferably about 1 μM to about 10 μM. Furthermore, when a TGFβ signaling pathway inhibitor other than SB431542 is used, it is desirably used at a concentration that exhibits TGFβ signaling pathway inhibitory activity equivalent to that of SB431542 at the above-mentioned concentration. The TGFβ signaling pathway inhibitory activity of SB431542 and the like can be determined by methods well known to those skilled in the art, for example, by detecting phosphorylation of Smads by Western blotting (Mol Cancer Ther. (2004) 3, 737-45.).
Shhシグナル伝達経路作用物質とは、Shhにより媒介されるシグナル伝達を増強し得る物質である。Shhシグナル伝達経路作用物質としては、例えばHedgehogファミリーに属するタンパク質(例えば、Shh、Ihh)又はそのフラグメント・変異体(SHH C25II N-terminus、SHH C24II N-terminus)、Shh受容体、Shh受容体アゴニスト、Smoアゴニスト、Purmorphamine(9-cyclohexyl-N-[4-(morpholinyl)phenyl]-2-(1-naphthalenyloxy)-9H-purin-6-amine)、GSA-10(Propyl 4-(1-hexyl-4-hydroxy-2-oxo-1,2-dihydroquinoline-3-carboxamido)benzoate)、Hh-Ag1.5、20(S)-Hydroxycholesterol、SAG(Smoothened Agonist:N-Methyl-N’-(3-pyridinylbenzyl)-N’-(3-chlorobenzo[b]thiophene-2-carbonyl)-1,4-diaminocyclohexane)、20(S)-hydroxy Cholesterol((3S,8S,9S,10R,13S,14S,17S)-17-[(2R)-2-hydroxy-6-methylheptan-2-yl]-10,13-dimethyl-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-3-ol)等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。上記の化合物等がShhシグナル伝達経路作用物質としての活性を有することは当業者にとって公知である(例えば、Molecular BioSystems,2010,6.1: 44-54.に記載されている)。 A substance acting on the Shh signaling pathway is a substance that can enhance signal transduction mediated by Shh. Examples of substances acting on the Shh signaling pathway include proteins belonging to the Hedgehog family (e.g., Shh, Ihh) or fragments or mutants thereof (SHH C25II N-terminus, SHH C24II N-terminus), Shh receptors, Shh receptor agonists, Smo agonists, Purmorphamine (9-cyclohe xyl-N-[4-(morpholinyl)phenyl]-2-(1-naphthalenyloxy)-9H-purin-6-amine), GSA-10(Propyl 4-(1-hexyl-4-hydroxy-2-oxo-1,2-dihydr) oquinoline-3-carboxamide)benzoate), Hh-Ag1.5, 20 (S)-Hydroxycholesterol, SAG (Smoothened Agonist: N-Methyl-N'-(3-pyridinylbenzyl)-N'-(3-chlorobenzo[b]thiophene-2-carbonyl )-1,4-diaminocyclohexane), 20(S)-hydroxy Cholester ol ((3S,8S,9S,10R,13S,14S,17S)-17-[(2R)-2-hydroxy-6-methylheptan-2-yl]-10,13-dimethyl-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-3-ol), etc. These substances may be used alone or in combination. Those skilled in the art are aware that the above compounds have activity as Shh signaling pathway acting substances (for example, as described in Molecular BioSystems, 2010, 6.1: 44-54.).
Shhシグナル伝達経路作用物質は、好ましくはSAG、Purmorphamine、GSA-10からなる群より選ばれる少なくとも1つを含み、より好ましくはSAGを含む。培地中のShhシグナル伝達経路作用物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。SAGは、工程(a)においては通常約1nM~約2000nM、好ましくは約10nM~約1000nM、より好ましくは約10nM~約700nM、さらに好ましくは約50nM~約700nM、特に好ましくは約100nM~約600nM、最も好ましくは約100nM~約500nMの濃度で使用される。また、SAG以外のShhシグナル伝達経路作用物質を使用する場合、上記濃度のSAGと同等のShhシグナル伝達促進活性を示す濃度で用いられることが望ましい。Shhシグナル伝達促進活性は、当業者に周知の方法、例えばGli1遺伝子の発現に着目したレポータージーンアッセイにて決定することができる(Oncogene(2007)26,5163-5168)。 The substance acting on the Shh signaling pathway preferably includes at least one selected from the group consisting of SAG, Purmorphamine, and GSA-10, and more preferably includes SAG. The concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range in which the above-mentioned effects can be achieved. In step (a), SAG is usually used at a concentration of about 1 nM to about 2000 nM, preferably about 10 nM to about 1000 nM, more preferably about 10 nM to about 700 nM, even more preferably about 50 nM to about 700 nM, particularly preferably about 100 nM to about 600 nM, and most preferably about 100 nM to about 500 nM. In addition, when a substance acting on the Shh signaling pathway other than SAG is used, it is desirable to use it at a concentration that shows the same Shh signaling promoting activity as SAG at the above-mentioned concentration. The Shh signaling promoting activity can be determined by methods well known to those skilled in the art, for example, a reporter gene assay focusing on the expression of the Gli1 gene (Oncogene (2007) 26, 5163-5168).
工程(a)において用いられる培地は、未分化維持培養を可能にするため、未分化維持因子を含む。未分化維持因子は、多能性幹細胞の分化を抑制する作用を有する物質であれば特に限定されない。当業者に汎用されている未分化維持因子としては、プライムド多能性幹細胞(Primed pluripotent stem cells)(例えば、ヒトES細胞、ヒトiPS細胞)の場合、FGFシグナル伝達経路作用物質、TGFβファミリーシグナル伝達経路作用物質、insulin等を挙げることができる。FGFシグナル伝達経路作用物質として具体的には、線維芽細胞増殖因子(例えば、bFGF、FGF4やFGF8)が挙げられる。また、TGFβファミリーシグナル伝達経路作用物質としては、TGFβシグナル伝達経路作用物質、Nodal/Activinシグナル伝達経路作用物質が挙げられる。TGFβシグナル伝達経路作用物質としては、例えば、TGFβ1、TGFβ2が挙げられる。Nodal/Activinシグナル伝達経路作用物質としては、例えば、Nodal、ActivinA、ActivinBが挙げられる。これらの物質は単独又は組み合わせて用いてもよい。ヒト多能性幹細胞(例えばヒトES細胞、ヒトiPS細胞)を培養する場合、工程(a)における培地は、好ましくは未分化維持因子として、bFGFを含む。 The medium used in step (a) contains an undifferentiated state maintenance factor to enable culture to maintain the undifferentiated state. The undifferentiated state maintenance factor is not particularly limited as long as it is a substance that has the effect of suppressing the differentiation of pluripotent stem cells. In the case of primed pluripotent stem cells (e.g., human ES cells, human iPS cells), examples of undifferentiated state maintenance factors commonly used by those skilled in the art include FGF signaling pathway acting substances, TGFβ family signaling pathway acting substances, insulin, etc. Specific examples of FGF signaling pathway acting substances include fibroblast growth factors (e.g., bFGF, FGF4, and FGF8). Examples of TGFβ family signaling pathway acting substances include TGFβ signaling pathway acting substances and Nodal/Activin signaling pathway acting substances. Examples of TGFβ signaling pathway acting substances include TGFβ1 and TGFβ2. Examples of substances acting on the Nodal/Activin signaling pathway include Nodal, Activin A, and Activin B. These substances may be used alone or in combination. When culturing human pluripotent stem cells (e.g., human ES cells, human iPS cells), the medium in step (a) preferably contains bFGF as an undifferentiated maintenance factor.
未分化維持因子は、通常哺乳動物の未分化維持因子である。哺乳動物としては、上記のものを挙げることができる。未分化維持因子は、哺乳動物の種間で交差反応性を有し得るので、培養対象の多能性幹細胞の未分化状態を維持可能な限り、いずれの哺乳動物の未分化維持因子を用いてもよい。未分化維持因子は、好ましくは培養する細胞と同一種の哺乳動物の未分化維持因子である。例えばヒト多能性幹細胞の培養には、ヒト未分化維持因子(例えば、bFGF、FGF4、FGF8、EGF、Nodal、ActivinA、ActivinB、TGFβ1、TGFβ2等)が用いられる。未分化維持因子は、好ましくは単離されている。 The undifferentiation maintenance factor is usually a mammalian undifferentiation maintenance factor. Examples of mammals include those mentioned above. Since the undifferentiation maintenance factor may have cross-reactivity between mammalian species, any mammalian undifferentiation maintenance factor may be used as long as it can maintain the undifferentiated state of the pluripotent stem cells to be cultured. The undifferentiation maintenance factor is preferably an undifferentiation maintenance factor of the same mammalian species as the cells to be cultured. For example, human undifferentiation maintenance factors (e.g., bFGF, FGF4, FGF8, EGF, Nodal, Activin A, Activin B, TGFβ1, TGFβ2, etc.) are used for culturing human pluripotent stem cells. The undifferentiation maintenance factor is preferably isolated.
未分化維持因子は、培養対象である多能性幹細胞の未分化維持能を有する限り、いずれの宿主によって生産されたもの又は人工合成されたものを用いることができる。本発明に用いる未分化維持因子は、生体内で生じるものと同様の修飾を受けたものが好ましく、培養対象である多能性幹細胞と同種の細胞において異種成分を含まない条件下で産生されたものがさらに好ましい。
本発明に係る製造方法の一態様は、単離された未分化維持因子を提供する工程を含む。本発明に係る製造方法の一態様は、工程(a)に用いる培地中へ、単離された未分化維持因子を外来的(又は外因的)に添加する工程を含む。工程(a)に用いる培地に予め未分化維持因子が添加されていてもよい。
The undifferentiation maintenance factor may be one produced by any host or one artificially synthesized, so long as it has the ability to maintain the undifferentiation state of the pluripotent stem cells to be cultured. The undifferentiation maintenance factor used in the present invention is preferably one that has been modified in the same manner as that occurring in a living body, and more preferably one that has been produced in cells of the same type as the pluripotent stem cells to be cultured under conditions that do not contain any xenogeneic components.
One embodiment of the production method according to the present invention includes a step of providing an isolated factor for maintaining undifferentiation. One embodiment of the production method according to the present invention includes a step of exogenously (or extrinsically) adding the isolated factor for maintaining undifferentiation to the medium used in step (a). The factor for maintaining undifferentiation may be added in advance to the medium used in step (a).
工程(a)において用いられる培地中の未分化維持因子濃度は、培養する多能性幹細胞の未分化状態を維持可能な濃度であり、当業者であれば適宜設定することができる。例えばフィーダー細胞非存在下で未分化維持因子としてbFGFを用いる場合、その濃度は、通常約4ng/ml~約500ng/ml、好ましくは約10ng/ml~約200ng/ml、より好ましくは約30ng/ml~約150ng/mlである。 The concentration of the undifferentiation maintenance factor in the medium used in step (a) is a concentration that can maintain the undifferentiated state of the pluripotent stem cells being cultured, and can be appropriately set by a person skilled in the art. For example, when bFGF is used as the undifferentiation maintenance factor in the absence of feeder cells, the concentration is usually about 4 ng/ml to about 500 ng/ml, preferably about 10 ng/ml to about 200 ng/ml, and more preferably about 30 ng/ml to about 150 ng/ml.
工程(a)は、フィーダー細胞非存在下で実施する。工程(a)における多能性幹細胞の培養は、浮遊培養及び接着培養のいずれの条件で行われてもよいが、好ましくは接着培養により行われる。フィーダー細胞非存在下での多能性幹細胞の培養においては、フィーダー細胞に代わる足場を多能性幹細胞に提供するため、適切なマトリクスを足場として用いてもよい。足場であるマトリクスにより、表面をコーティングした培養器材中で、多能性幹細胞を接着培養する。 Step (a) is carried out in the absence of feeder cells. The culture of pluripotent stem cells in step (a) may be carried out under either suspension culture or adherent culture conditions, but is preferably carried out by adherent culture. When culturing pluripotent stem cells in the absence of feeder cells, an appropriate matrix may be used as a scaffold to provide the pluripotent stem cells with a scaffold in place of feeder cells. The pluripotent stem cells are cultured in an adherent manner in a culture vessel whose surface is coated with a matrix scaffold.
足場として用いることのできるマトリクスとしては、ラミニン(Nat Biotechnol.28,611-615(2010))、ラミニン断片(Nat Commun 3,1236(2012))、基底膜標品(Nat Biotechnol 19,971-974(2001))、ゼラチン、コラーゲン、ヘパラン硫酸プロテオグリカン、エンタクチン、ビトロネクチン(Vitronectin)等が挙げられる。マトリクスは、好ましくはラミニン511が用いられる(Nat Biotechnol 28,611-615(2010))。
Matrices that can be used as scaffolds include laminin (Nat Biotechnol. 28, 611-615 (2010)), laminin fragments (
ラミニン断片は、多能性幹細胞への接着性を有しており、フィーダーフリー条件での多能性幹細胞の維持培養を可能とするものであれば特に限定されないが、好ましくはE8フラグメントである。ラミニンE8フラグメントは、ラミニン511をエラスターゼで消化して得られたフラグメントの中で、強い細胞接着活性をもつフラグメントとして同定されたものである(EMBO J.,3:1463-1468,1984、J. Cell Biol.,105:589-598,1987)。ラミニン断片としては、好ましくはラミニン511のE8フラグメントが用いられる(Nat Commun 3,1236(2012)、Scientific Reports 4,3549(2014))。ラミニンE8フラグメントは、ラミニンのエラスターゼ消化産物であることを要するものではなく、組換え体であってもよい。遺伝子組み換え動物(カイコ等)に産生させたものであってもよい。未同定成分の混入を回避する観点から、好ましくは組換え体のラミニン断片が用いられる。ラミニン511のE8フラグメントは市販されており、例えばニッピ株式会社等から購入可能である。
The laminin fragment is not particularly limited as long as it has adhesive properties to pluripotent stem cells and enables the maintenance culture of pluripotent stem cells under feeder-free conditions, but is preferably an E8 fragment. The laminin E8 fragment was identified as a fragment with strong cell adhesive activity among fragments obtained by digesting laminin 511 with elastase (EMBO J., 3: 1463-1468, 1984; J. Cell Biol., 105: 589-598, 1987). As the laminin fragment, the E8 fragment of laminin 511 is preferably used (
未同定成分の混入を回避する観点から、本明細書において用いられるラミニン又はラミニン断片は、単離されていることが好ましい。工程(a)におけるフィーダー細胞非存在下での多能性幹細胞の培養においては、好ましくは単離されたラミニン511又はラミニン511のE8フラグメントによって、より好ましくはラミニン511のE8フラグメントによって表面をコーティングした培養器材中で、多能性幹細胞を接着培養する。 In order to avoid contamination with unidentified components, the laminin or laminin fragment used in this specification is preferably isolated. In the culture of pluripotent stem cells in the absence of feeder cells in step (a), the pluripotent stem cells are cultured in an adherent manner in a culture vessel whose surface is preferably coated with isolated laminin 511 or the E8 fragment of laminin 511, more preferably with the E8 fragment of laminin 511.
工程(a)において用いられる培地は、フィーダーフリー条件下で、多能性幹細胞の未分化維持培養を可能にする培地(フィーダーフリー培地)であれば、特に限定されない。
工程(a)において用いられる培地は、血清培地であっても無血清培地であってもよい。化学的に未決定な成分の混入を回避する観点から、工程(a)において用いられる培地は、好ましくは無血清培地である。培地は、血清代替物を含んでいてもよい。
The medium used in step (a) is not particularly limited as long as it is a medium that allows the maintenance of undifferentiated culture of pluripotent stem cells under feeder-free conditions (feeder-free medium).
The medium used in step (a) may be a serum medium or a serum-free medium. From the viewpoint of avoiding contamination with chemically undefined components, the medium used in step (a) is preferably a serum-free medium. The medium may contain a serum substitute.
工程(a)における多能性幹細胞の培養時間は、続く第一工程において形成され得る細胞集団(凝集体)の質を向上させる効果が達成可能な範囲で特に限定されないが、通常0.5~144時間、好ましくは2~96時間、より好ましくは6~48時間、さらに好ましくは12~48時間、特に好ましくは18~28時間であり、例えば24時間である。すなわち、第一工程開始の0.5~144時間前、好ましくは18~28時間前に工程(a)を開始し、工程(a)を完了した後引き続き第一工程が行われる。 The culture time of the pluripotent stem cells in step (a) is not particularly limited as long as it is within a range that can achieve the effect of improving the quality of the cell population (aggregates) that can be formed in the subsequent first step, but is usually 0.5 to 144 hours, preferably 2 to 96 hours, more preferably 6 to 48 hours, even more preferably 12 to 48 hours, and particularly preferably 18 to 28 hours, for example 24 hours. In other words, step (a) is started 0.5 to 144 hours, preferably 18 to 28 hours, before the start of the first step, and the first step is continued after step (a) is completed.
工程(a)の好ましい一態様において、ヒト多能性幹細胞を、フィーダー細胞非存在下で、bFGFを含有する無血清培地中で、接着培養する。当該接着培養は、好ましくはラミニン511、ラミニン511のE8フラグメント又はビトロネクチンで表面をコーティングした培養器材中で実施される。当該接着培養は、好ましくはフィーダーフリー培地としてStemFitを用いて実施される。StemFit培地は未分化維持成分としてbFGFを含有する(Scientific Reports(2014)4,3594)。 In a preferred embodiment of step (a), human pluripotent stem cells are cultured as adherent cells in a serum-free medium containing bFGF in the absence of feeder cells. The adherent culture is preferably performed in a culture vessel whose surface is coated with laminin 511, E8 fragment of laminin 511, or vitronectin. The adherent culture is preferably performed using StemFit as a feeder-free medium. StemFit medium contains bFGF as a component for maintaining undifferentiated state (Scientific Reports (2014) 4, 3594).
工程(a)の好ましい一態様において、ヒト多能性幹細胞を、フィーダー細胞非存在下で、bFGFを含有する無血清培地中で、浮遊培養する。当該浮遊培養では、ヒト多能性幹細胞は、ヒト多能性幹細胞の凝集体を形成してもよい。 In a preferred embodiment of step (a), human pluripotent stem cells are cultured in suspension in a serum-free medium containing bFGF in the absence of feeder cells. In the suspension culture, the human pluripotent stem cells may form aggregates of human pluripotent stem cells.
工程(a)及び後述する工程において、培養温度、CO2濃度等の培養条件は適宜設定できる。培養温度は、例えば約30℃から約40℃、好ましくは約37℃である。CO2濃度は、重炭酸緩衝系の培地を用いる場合、例えば約1%から約10%、好ましくは約5%である。 In step (a) and the steps described below, culture conditions such as culture temperature and CO2 concentration can be appropriately set. The culture temperature is, for example, about 30° C. to about 40° C., preferably about 37° C. The CO2 concentration is, for example, about 1% to about 10%, preferably about 5%, when a bicarbonate buffered medium is used.
<工程(1)>、<工程(1’)>:第一工程
工程(1’)では、c-Jun N末キナーゼ(JNK)シグナル伝達経路阻害物質の存在下で多能性幹細胞を培養し、細胞集団を得る。
<Step (1)> and <Step (1')>: First Step In step (1'), pluripotent stem cells are cultured in the presence of an inhibitor of the c-Jun N-terminal kinase (JNK) signaling pathway to obtain a cell population.
JNKはMAPKファミリーに属するキナーゼであり、各種の環境ストレス、炎症性サイトカイン、成長因子、GPCRアゴニストによる刺激の細胞内シグナル伝達に関与する。 JNK is a kinase belonging to the MAPK family and is involved in intracellular signal transduction in response to various environmental stresses, inflammatory cytokines, growth factors, and stimulation by GPCR agonists.
本明細書においてJNKシグナル伝達経路阻害物質とは、JNKによって伝達されるシグナル伝達を抑制し得るものである限り限定されない。JNKシグナル伝達経路阻害物質として例えば、JNKシグナル伝達機構の上流ないし下流の因子、もしくはJNKそのものの酵素活性、多量体化、他の因子や核酸との結合を阻害する、分解を促進する等のメカニズムによりシグナル伝達を阻害する活性を有する物質がある。JNKシグナル伝達経路阻害物質としては、例えばJNK阻害剤、Rac阻害剤、MKK阻害剤、MEK阻害剤、Src阻害剤、受容体チロシンキナーゼ(RTK)阻害剤、ASK阻害剤等が挙げられるがこれに限定されない。 In this specification, the term "JNK signaling pathway inhibitor" refers to any substance that can suppress signal transduction transmitted by JNK, but is not limited thereto. Examples of JNK signaling pathway inhibitors include substances that have the activity of inhibiting signal transduction by mechanisms such as inhibiting factors upstream or downstream of the JNK signaling mechanism, or the enzyme activity, multimerization, and binding to other factors or nucleic acids of JNK itself, and promoting degradation. Examples of JNK signaling pathway inhibitors include, but are not limited to, JNK inhibitors, Rac inhibitors, MKK inhibitors, MEK inhibitors, Src inhibitors, receptor tyrosine kinase (RTK) inhibitors, and ASK inhibitors.
c-Jun N末キナーゼ(JNK)阻害剤としては、例えば、JNK-IN-8((E)-3-(4-(dimethylamino)but-2-enamido)-N-(3-methyl-4-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)phenyl)benzamide)、SP600125(Anthra[1-9-cd]pyrazol-6(2H)-one)、DB07268(2-[[2-[(3-Hydroxyphenyl)amino]-4-pyrimidinyl]amino]benzamide)、Tanzisertib(trans-4-[[9-[(3S)-Tetrahydro-3-furanyl]-8-[(2,4,6-trifluorophenyl)amino]-9H-purin-2-yl]amino]cyclohexanol)、Bentamapimod(1,3-Benzothiazol-2-yl)[2-[[4-[(morpholin-4-yl)methyl]benzyl]oxy]pyrimidin-4-yl]acetonitrile、TCS JNK 6o(N-(4-Amino-5-cyano-6-ethoxy-2-pyridinyl)-2,5-dimethoxybenzeneacetamide)、SU3327(5-[(5-Nitro-2-thiazolyl)thio]-1,3,4-thiadiazol-2-amine)、CEP1347((9S,10R,12R)-5-16-Bis[(ethylthio)methyl]-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3’,2’,1’-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester)、c-JUN peptide、AEG3482(6-Phenylimidazo[2,1-b]-1,3,4-thiadiazole-2-sulfonamide)、TCS JNK 5a(N-(3-Cyano-4,5,6,7-tetrahydrobenzo[b]thienyl-2-yl)-1-naphthalenecarboxamide)、BI-78D3(4-(2,3-Dihydro-1,4-benzodioxin-6-yl)-2,4-dihydro-5-[(5-nitro-2-thiazolyl)thio]-3H-1,2,4-triazol-3-one)、IQ-3(11H-Indeno[1,2-b]quinoxalin-11-one O-(2-furanylcarbonyl)oxime)、SR 3576(3-[4-[[[(3-Methylphenyl)amino]carbonyl]amino]-1H-pyrazol-1-yl]-N-(3,4,5-trimethoxyphenyl)benzamide)、IQ-1S(11H-Indeno[1,2-b]quinoxalin-11-one oxime sodium salt)、JIP-1(153-163)、CC-401(3-[3-[2-(1-Piperidinyl)ethoxy]phenyl]-5-(1H-1,2,4-triazol-5-yl)-1H-indazole dihydrochloride)、BI-87G3(2-(5-Nitrothiazol-2-ylthio)benzo[d]thiazole)、AS601245(2-(1,3-benzothiazol-2-yl)-2-[2-(2-pyridin-3-ylethylamino)pyrimidin-4-yl]acetonitrile)、CV-65(3,7-Dimethyl-1,9-dihydropyrido[3,2-g]quinoline-2,5,8,10-tetrone)、D-JNK1(CAS番号1445179-97-4)、ER-358063、ER-409903、ER-417258、CC-359((4S)-4-(2,4-difluoro-5-pyrimidin-5-ylphenyl)-4-methyl-5,6-dihydro-1,3-thiazin-2-amine)、CC-401(3-[3-(2-piperidin-1-ylethoxy)phenyl]-5-(1H-1,2,4-triazol-5-yl)-1H-indazole)、CC-930(4-[[9-[(3S)-oxolan-3-yl]-8-(2,4,6-trifluoroanilino)purin-2-yl]amino]cyclohexan-1-ol)、SB203580(4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine)及びこれらの誘導体等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。上記の化合物等がJNK阻害剤としての活性を有することは当業者にとって公知である(例えばJ Enzyme Inhib Med Chem.2020; 35(1): 574-583.に記載されている)。 c-Jun N-terminal kinase (JNK) inhibitors include, for example, JNK-IN-8 ((E)-3-(4-(dimethylamino)but-2-enamido)-N-(3-methyl-4-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)phenyl)benzamide), SP600125 (Anthra[1-9-cd]pyrazol-6(2H)-one), DB07268 (2-[[2-[(3-Hydroxyphenyl)amino]-4-pyrimidinyl]amino]benzamide), and Ta nzisertib (trans-4-[[9-[(3S)-Tetrahydro-3-furanyl]-8-[(2,4,6-trifluorophenyl)amino]-9H-purin-2-yl]amino]cyclohexanol), Be ntamapimod (1,3-Benzothiazol-2-yl)[2-[[4-[(morpholin-4-yl)methyl]benzyl]oxy]pyrimidin-4-yl]acetonitrile, TCS JNK 6o(N -(4-Amino-5-cyano-6-ethoxy-2-py ridinyl)-2,5-dimethoxybenzeneacetamide), SU3327(5-[(5-Nitro-2-thiazolyl)thio]-1,3,4-thiadiazol-2-amine), CEP1347((9S, 10R,12R)-5-16-Bis[(ethylthio)methyl]-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3 ',2',1'-kl]pyrrolo[3,4-i][1, 6] benzodiazocine-10-carboxylic acid methyl ester), c-JUN peptide, AEG3482 (6-Phenylimidazo[2,1-b]-1,3,4-thiadiazole- 2-sulfonamide), TCS JNK 5a (N-(3-Cyano-4,5,6,7-tetrahydrobenzo[b]thienyl-2-yl)-1-naphthalenecarboxamide), BI-78D3 (4-(2,3-D ihydro-1,4-benzodioxin-6-yl)-2,4- dihydro-5-[(5-nitro-2-thiazolyl)thio]-3H-1,2,4-triazol-3-one), IQ-3(11H-Indeno[1,2-b]quinoxalin-11-one O-(2-furanylca rbonyl)oxime), SR 3576 (3-[4-[[[(3-Methylphenyl)amino]carbonyl]amino]-1H-pyrazol-1-yl]-N-(3,4,5-trimethoxyphenyl)benzam ide), IQ-1S (11H-Indeno[1,2-b]qui noxalin-11-one oxime sodium salt), JIP-1 (153-163), CC-401 (3-[3-[2-(1-Piperidinyl)ethoxy]phenyl]-5-(1H-1,2,4-triazol-5-yl) -1H-indazole dihydrochloride), BI-87G3 (2-(5-Nitrothiazol-2-ylthio)benzo[d]thiazole), AS601245 (2-(1,3-benzothiazole) ol-2-yl)-2-[2-(2-pyridin-3-ylethylami no. 79-97-4), ER-358063, ER-409903, ER-417258, CC-359 ((4S)-4-(2,4-difluoro-5-pyrimidin-5-ylphenyl)-4-methyl-5,6-dihydro-1,3-thia zin-2-amine), CC-401 (3-[3 -(2-piperidin-1-ylethoxy)phenyl]-5-(1H-1,2,4-triazol-5-yl)-1H-indazole), CC-930 (4-[[9-[(3S)-oxolan-3-yl]-8-(2,4,6-trifluoroanilino)purin-2-yl]amino]cyclohexan-1-ol), SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine) and derivatives thereof. These substances may be used alone or in combination. It is known to those skilled in the art that the above compounds have activity as JNK inhibitors (for example, as described in J Enzyme Inhib Med Chem. 2020; 35(1): 574-583).
Rac阻害剤としては、例えば、EHT1864(5-(5-(7-(Trifluoromethyl)quinolin-4-ylthio)pentyloxy)-2-(morpholinomethyl)-4H-pyran-4-one dihydrochloride)、NSC23766(N6-[2-[[4-(Diethylamino)-1-methylbutyl]amino]-6-methyl-4-pyrimidinyl]-2-methyl-4,6-quinolinediaminetrihydrochloride)、EHop-016(N4-(9-Ethyl-9H-carbazol-3-yl)-N2-[3-(4-morpholinyl)propyl]-2,4-pyrimidinediamine)、1A-116(N-(3,5-Dimethylphenyl)-N’-[2-(trifluoromethyl)phenyl]guanidine)、ZCL278(2-(4-bromo-2-chlorophenoxy)-N-(4-(N-(4,6-dimethylpyrimidin-2-yl)sulfamoyl)phenylcarbamothioyl)acetamide)、MBQ-167(9-ethyl-3-(5-phenyl-1H-1,2,3-triazol-1-yl)-9H-carbazole)、KRpep-2d(actinium;[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(3R,8R,11S,14S,20S,23S,26S,29S,32S,35S,38S)-8-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-amino-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]carbamoyl]-29-[(2R)-butan-2-yl]-20-(carboxymethyl)-26-(hydroxymethyl)-23,32-bis[(4-hydroxyphenyl)methyl]-35-(2-methylpropyl)-2,10,13,19,22,25,28,31,34,37-decaoxo-11-propan-2-yl-5,6-dithia-1,9,12,18,21,24,27,30,33,36-decazatricyclo[36.3.0.014,18]hentetracontan-3-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]azanide)、ARS-853(1-[3-[4-[2-[[4-Chloro-2-hydroxy-5-(1-methylcyclopropyl)phenyl]amino]acetyl]-1-piperazinyl]-1-azetidinyl]-2-propen-1-one)、Salirasib(2-(((2E,6E)-3,7,11-Trimethyldodeca-2,6,10-trien-1-yl)thio)benzoic acid)、ML141(4-(5-(4-methoxyphenyl)-3-phenyl-4,5-dihydropyrazol-1-yl)benzenesulfonamide)及びこれらの誘導体等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。上記の化合物等がRac阻害剤としての活性を有することは当業者にとって公知である(例えばCancer research,2018,78.12: 3101-3111.に記載されている)。 Rac inhibitors include, for example, EHT1864 (5-(5-(7-(Trifluoromethyl)quinolin-4-ylthio)pentyloxy)-2-(morpholinomethyl)-4H-pyran-4-one dihydrochloride), NSC23766 (N6-[2-[[4-(Diethylamino)-1-methylbutyl]amino]-6-methyl-4-pyrimidinyl nyl]-2-methyl-4,6-quinolinediaminetrihydrochloride), EHop-016(N4-(9-Ethyl-9H-carbazol-3-yl)-N2-[3-(4-morpholinyl)propy l]-2,4-pyrimidinediamine), 1A-116(N-(3,5-Dimethylphenyl)-N'-[2-(trifluoromethyl)phenyl]guan idine), ZCL278 (2-(4-bromo-2-chlorophenoxy)-N-(4-(N-(4,6-dimethylpyrimidin-2-yl)sulfamoyl)phenylcarbamothioyl)acetamide), MBQ-167 (9-ethyl-3-(5-phenyl-1H-1,2,3-triazol-1-yl)-9H-carbazole), KRpep-2d (actinium; [(2S )-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(3R,8R,11S,14S,20S,23S,26S,29S,32S,35S,38S)-8-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-amino- 5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5- carbamimidamido-1-oxopentan-2-yl]-5-carbamimidamido-1-oxopentan-2-yl]carbamoyl]-29-[(2R)-butan-2-yl]-20-(carboxymet hyl)-26-(hydroxymethyl)-23,32-bis[(4-hydroxyphenyl)methyl]-35-(2-methylpropyl)-2,10,1 3,19,22,25,28,31,34,37-decaoxo-11-propan-2-yl-5,6-dithia-1,9,12,18,21,24,27,30,33,36-decazatrico[36.3.0.014,18]hente tracontan-3-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxop entan-2-yl]amino]-5-carbamimidamide-1-oxopentan-2-yl]azanide), ARS-853 (1-[3-[4 -[2-[[4-Chloro-2-hydroxy-5-(1-methylcyclopropyl)phenyl]amino]acetyl]-1-piperazinyl]-1-a zetidinyl]-2-propen-1-one), Salirasib (2-(((2E,6E)-3,7,11-Trimethyldodeca-2,6,10-trien-1-yl)thio)benzoic acid), ML141 (4-(5-(4-methoxyphenyl)-3-phenyl-4,5-dihydropyrazol-1-yl)benzenesulfonamide) and derivatives thereof. These substances may be used alone or in combination. Those skilled in the art are aware that the above compounds have activity as Rac inhibitors (for example, see Cancer research, 2018, 78.12: 3101-3111).
本発明におけるJNKシグナル伝達経路阻害物質の添加時期は、ヒト多能性幹細胞からの下垂体組織の製造の効率改善効果が発揮される限り限定されないが、後述する工程(2)でBMPシグナル伝達経路作用物質を添加する時点ですでに添加されていることが好ましく、分化誘導の開始から72時間以内である。より好ましいJNK阻害剤の添加時期は、分化誘導の開始と同時である。 The timing of addition of the JNK signaling pathway inhibitor in the present invention is not limited as long as the effect of improving the efficiency of production of pituitary tissue from human pluripotent stem cells is exhibited, but it is preferable that it is added already at the time of adding the BMP signaling pathway active substance in step (2) described below, within 72 hours from the start of differentiation induction. The more preferable timing of addition of the JNK inhibitor is simultaneous with the start of differentiation induction.
工程(1’)の培地中には、Wntシグナル伝達経路阻害物質がさらに存在することが好ましい。このような第一工程を工程(1)とする。即ち、工程(1)は、多能性幹細胞をJNKシグナル伝達経路阻害物質及びWntシグナル伝達経路阻害物質の存在下で培養する第一工程である。
Wntシグナル伝達経路とは、Wntファミリー・タンパク質をリガンドとし、主としてFrizzledを受容体とするシグナル伝達経路である。当該シグナル伝達経路としては、古典的Wnt経路(Canonical Wnt pathway)、非古典的Wnt経路(Non-Canonical Wnt pathway)等が挙げられる。古典的Wnt経路は、β -Cateninによって伝達される。非古典的Wnt経路としては、Planar Cell Polarity(PCP)経路、Wnt/JNK経路、Wnt/Calcium経路、Wnt-RAP1経路、Wnt-Ror2経路、Wnt-PKA経路、Wnt-GSK3MT経路、Wnt-aPKC経路、Wnt-RYK経路、Wnt-mTOR経路等が挙げられる。非古典的Wnt経路では、Wnt以外の他のシグナル伝達経路でも活性化される共通のシグナル伝達因子が存在するが、本発明では前記したJNK経路以外のそれらの因子もWntシグナル伝達経路の構成因子とし、それらの因子に対する阻害物質もWntシグナル伝達経路阻害物質に含まれる。
It is preferable that the medium in step (1') further contains a Wnt signaling pathway inhibitor. Such a first step is referred to as step (1). That is, step (1) is a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor.
The Wnt signaling pathway is a signaling pathway that uses Wnt family proteins as ligands and mainly Frizzled as a receptor. Examples of the signaling pathway include the canonical Wnt pathway and the non-canonical Wnt pathway. The canonical Wnt pathway is transmitted by β-Catenin. Examples of the non-canonical Wnt pathway include the Planar Cell Polarity (PCP) pathway, the Wnt/JNK pathway, the Wnt/Calcium pathway, the Wnt-RAP1 pathway, the Wnt-Ror2 pathway, the Wnt-PKA pathway, the Wnt-GSK3MT pathway, the Wnt-aPKC pathway, the Wnt-RYK pathway, and the Wnt-mTOR pathway. In the non-canonical Wnt pathway, there are common signaling factors that are also activated in signaling pathways other than Wnt. In the present invention, these factors other than the above-mentioned JNK pathway are also considered to be constituent factors of the Wnt signaling pathway, and inhibitors of these factors are also included in Wnt signaling pathway inhibitors.
Wntシグナル伝達経路阻害物質は、Wntファミリー・タンパク質により惹起されるシグナル伝達を抑制し得るものである限り限定されない。阻害物質は、核酸、タンパク質、低分子有機化合物のいずれであってもよい。当該物質として例えばWntのプロセシングと細胞外への分泌を阻害する物質、Wntに直接作用する物質(例えば、タンパク質、抗体、アプタマー等)、Wntをコードする遺伝子の発現を抑制する物質(例えば、アンチセンスオリゴヌクレオチド、siRNA、CRISPRi等)、Wnt受容体とWntの結合を阻害する物質、Wnt受容体によるシグナル伝達に起因する生理活性を阻害する物質を挙げることができる。 The Wnt signaling pathway inhibitor is not limited as long as it can suppress signaling induced by Wnt family proteins. The inhibitor may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit Wnt processing and extracellular secretion, substances that act directly on Wnt (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes encoding Wnt (e.g., antisense oligonucleotides, siRNA, CRISPRi, etc.), substances that inhibit the binding between Wnt receptors and Wnt, and substances that inhibit physiological activity resulting from signaling by Wnt receptors.
Wntシグナル伝達経路阻害物質として知られているタンパク質として、secreted Frizzled Related Protein(sFRP)クラスに属するタンパク質(sFRP1~5、Wnt Inhibitory Factor-1(WIF-1)、Cerberus)、Dickkopf(Dkk)クラスに属するタンパク質(Dkk1~4、Kremen)、APCDD1、APCDD1L、Draxinファミリーに属するタンパク質、IGFBP-4、Notum、SOST/Sclerostinファミリーに属するタンパク質等が挙げられる。 Proteins known to inhibit the Wnt signaling pathway include proteins belonging to the secreted frizzled related protein (sFRP) class (sFRP1-5, Wnt inhibitory factor-1 (WIF-1), Cerberus), proteins belonging to the Dickkopf (Dkk) class (Dkk1-4, Kremen), APCDD1, APCDD1L, proteins belonging to the Draxin family, IGFBP-4, Notum, and proteins belonging to the SOST/Sclerostin family.
Wntシグナル伝達経路阻害物質としては、当業者に周知の化合物を使用することができる。古典的Wntシグナル伝達経路の阻害物質として、例えば、Frizzled阻害剤、Dishevelled(Dvl)阻害剤、Tankyrase(TANK)阻害剤、カゼインキナーゼ1阻害剤、カテニン応答性転写阻害剤、p300阻害剤、CREB-binding protein(CBP)阻害剤、BCL-9阻害剤、TCF分解誘導薬(Am J Cancer Res.2015;5(8):2344-2360)等が挙げられる。非古典的Wnt経路の阻害物質として、例えば、Porcupine(PORCN)阻害剤、Calcium/calmodulin-dependent protein kinase II(CaMKII)阻害剤、TGF-β-activated kinase 1(TAK1)阻害剤、Nemo-Like Kinase(NLK)阻害剤、LIM Kinase阻害剤、mammalian target of rapamycin(mTOR)阻害剤、Rac阻害剤、c-Jun NH 2-terminal kinase(JNK)阻害剤、protein kinase C(PKC)阻害剤、Methionine Aminopeptidase 2(MetAP2)阻害剤、Calcineurin阻害剤、nuclear factor of activated T cells(NFAT)阻害剤等が挙げられる。また、作用機序は報告されていないが、Wntシグナル伝達経路阻害物質としてKY02111(N-(6-Chloro-2-benzothiazolyl)-3,4-dimethoxybenzenepropanamide)、KY03-I(2-(4-(3,4-dimethoxyphenyl)butanamide)-6-Iodobenzothiazole)が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。
As the Wnt signaling pathway inhibitor, compounds well known to those skilled in the art can be used. Examples of inhibitors of the classical Wnt signaling pathway include Frizzled inhibitors, Dishevelled (Dvl) inhibitors, Tankyrase (TANK) inhibitors,
PORCN阻害剤として、例えば、IWP-2(N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]acetamide)、IWP-3(2-[[3-(4-fluorophenyl)-3,4,6,7-tetrahydro-4-oxothieno[3,2-d]pyrimidin-2-yl]thio]-N-(6-methyl-2-benzothiazolyl)acetamide)、IWP-4(N-(6-methyl-2-benzothiazolyl)-2-[[3,4,6,7-tetrahydro-3-(2-methoxyphenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl]thio]acetamide)、IWP-L6(N-(5-phenyl-2-pyridinyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]acetamide)、IWP-12(N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-3,6-dimethyl-4-oxothieno[3,2-d]pyrimidin-2-yl)thio]acetamide)、IWP-O1(1H-1,2,3-Triazole-1-acetamide,5-phenyl-N-(5-phenyl-2-pyridinyl)-4-(4-pyridinyl)-)、LGK-974(2-(2’,3-Dimethyl-2,4’-bipyridin-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide)、Wnt-C59(2-[4-(2-Methylpyridin-4-yl)phenyl]-N-[4-(pyridin-3-yl)phenyl]acetamide)、ETC-131、ETC-159(1,2,3,6-Tetrahydro-1,3-dimethyl-2,6-dioxo-N-(6-phenyl-3-pyridazinyl)-7H-purine-7-acetamide)、GNF-1331(N-(6-methoxy-1,3-benzothiazol-2-yl)-2-[(4-propyl-5-pyridin-4-yl-1,2,4-triazol-3-yl)sulfanyl]acetamide)、GNF-6231(N-[5-(4-Acetyl-1-piperazinyl)-2-pyridinyl]-2’-fluoro-3-methyl[2,4’-bipyridine]-5-acetamide)、Porcn-IN-1(N-[[5-fluoro-6-(2-methylpyridin-4-yl)pyridin-3-yl]methyl]-9H-carbazole-2-carboxamide)、RXC004、CGX1321及びこれらの誘導体等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。 PORCN inhibitors include, for example, IWP-2 (N-(6-methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthioeno[3,2-d]pyrimidin-2-yl)thio]acetamide), IWP-3 (2-[[3-(4-fluorophenyl)-3, IWP-4 (N-(6-methyl-2-b) enzothiazolyl)-2-[[3,4,6,7-tetrahydro-3-(2-methoxy phenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl]thio]acetamide), IWP-L6(N-(5-phenyl-2-pyridinyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-p) henylthieno[3,2-d]pyrimidin-2-yl)thio]acetami de), IWP-12(N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-3,6-dimethyl-4-oxothieno[3,2-d]pyrimidin-2-yl)thio]ac etamide), IWP-O1 (1H-1,2,3-Triazole-1-acetamide, 5-ph enyl-N-(5-phenyl-2-pyridinyl)-4-(4-pyridinyl)-), LGK-974(2-(2',3-Dimethyl-2,4'-bipyridin-5-yl)-N-(5-(pyrazin-2-yl)pyridinyl) n-2-yl) acetamide), Wnt-C59 (2-[4-(2-Methylpyridin) -4-yl)phenyl]-N-[4-(pyridin-3-yl)phenyl]acetamide), ETC-131, ETC-159 (1,2,3,6-Tetrahydro-1,3-dimethyl-2,6-dioxo-N-(6-phenyl- 3-pyridazinyl)-7H-purine-7-acetamide), GNF-1331 (N-(6-methoxy-1,3-benzothiazol-2-yl)-2-[(4-propyl-5-pyridin-4-yl-1,2,4-triazol-3-yl)sulfanyl]acetamide), GNF-6231(N-[ 5-(4-Acetyl-1-piperazinyl)-2-pyridinyl]-2'-fluoro- 3-methyl[2,4'-bipyridine]-5-acetamide), Porcn-IN-1 (N-[[5-fluoro-6-(2-methylpyridin-4-yl)pyridin-3-yl]methyl]-9H-carbazole-2-carboxamide), RXC004, CGX1321, and derivatives thereof. These substances may be used alone or in combination.
Wntシグナル伝達経路阻害物質は、好ましくはPORCN阻害剤、KY02111及びKY03-Iからなる群より選ばれる少なくとも1つを含み、より好ましくはPORCN阻害剤を含む。Wntシグナル伝達経路阻害物質は、Wntの非古典的Wnt経路への阻害活性を有する物質を含むこともまた好ましい。Wntシグナル伝達経路阻害物質は、より好ましくはWnt/Planar Cell Polarity(PCP)経路への阻害活性を有する物質を含む。本発明で用いられるPORCN阻害剤は、好ましくはIWP-2、IWP-3、IWP-4、IWP-L6、IWP-12、LGK-974、Wnt-C59、ETC-159及びGNF-6231からなる群より選ばれる少なくとも1つを含み、より好ましくはIWP-2又はWnt-C59を含み、さらに好ましくはIWP-2を含む。 The Wnt signaling pathway inhibitor preferably includes at least one selected from the group consisting of a PORCN inhibitor, KY02111, and KY03-I, and more preferably includes a PORCN inhibitor. It is also preferable that the Wnt signaling pathway inhibitor includes a substance having inhibitory activity against the non-classical Wnt pathway of Wnt. More preferably, the Wnt signaling pathway inhibitor includes a substance having inhibitory activity against the Wnt/Planar Cell Polarity (PCP) pathway. The PORCN inhibitor used in the present invention preferably includes at least one selected from the group consisting of IWP-2, IWP-3, IWP-4, IWP-L6, IWP-12, LGK-974, Wnt-C59, ETC-159, and GNF-6231, more preferably includes IWP-2 or Wnt-C59, and even more preferably includes IWP-2.
培地中のWntシグナル伝達経路阻害物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。下垂体を構成する細胞の製造効率向上の観点からは、例えば、Wntシグナル伝達経路阻害物質としてPORCN阻害剤の1種であるIWP-2を用いる場合は、その濃度は通常約10nM~約50μMであり、好ましくは約10nM~約30μMであり、さらに好ましくは約100nM~約10μMであり、最も好ましくは約0.5μMである。PORCN阻害剤の1種であるWnt-C59を用いる場合は、その濃度は通常約10pM~約1μMであり、好ましくは約100pM~約500nMであり、より好ましくは約50nMである。KY02111を用いる場合は、その濃度は通常約10nM~約50μMであり、好ましくは約10nM~約30μMであり、より好ましくは約100nM~約10μMであり、さらに好ましくは約5μMである。上記以外のWntシグナル伝達経路阻害物質を用いる場合には、上記の濃度と同等のWntシグナル伝達経路阻害活性を示す濃度で用いられることが望ましい。 The concentration of the Wnt signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of improving the production efficiency of cells that constitute the pituitary gland, for example, when IWP-2, a type of PORCN inhibitor, is used as the Wnt signaling pathway inhibitor, its concentration is usually about 10 nM to about 50 μM, preferably about 10 nM to about 30 μM, more preferably about 100 nM to about 10 μM, and most preferably about 0.5 μM. When Wnt-C59, a type of PORCN inhibitor, is used, its concentration is usually about 10 pM to about 1 μM, preferably about 100 pM to about 500 nM, and more preferably about 50 nM. When KY02111 is used, its concentration is usually about 10 nM to about 50 μM, preferably about 10 nM to about 30 μM, more preferably about 100 nM to about 10 μM, and even more preferably about 5 μM. When using a Wnt signaling pathway inhibitor other than the above, it is desirable to use it at a concentration that shows the same Wnt signaling pathway inhibitory activity as the above concentrations.
第一工程(工程(1)又は工程(1’))の培地中には、TGFβシグナル伝達経路阻害物質がさらに存在することが好ましい。第一工程において用いられるTGFβシグナル伝達経路阻害物質としては工程(a)で例示したものと同じものが用いられ得る。工程(a)及び第一工程のTGFβシグナル伝達経路阻害物質は同一であっても異なっていてもよいが、好ましくは同一である。 It is preferable that a TGFβ signaling pathway inhibitor is further present in the medium in the first step (step (1) or step (1')). The TGFβ signaling pathway inhibitor used in the first step may be the same as that exemplified in step (a). The TGFβ signaling pathway inhibitors in step (a) and the first step may be the same or different, but are preferably the same.
培地中のTGFβシグナル伝達経路阻害物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。TGFβ シグナル伝達経路阻害物質としてSB431542を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約100μM、より好ましくは約100nM~約50μM、さらに好ましくは約500nM~約10μ Mの濃度で使用される。また、SB431542以外のTGFβシグナル伝達経路阻害物質を使用する場合、上記濃度のSB431542と同等のTGFβシグナル伝達経路阻害活性を示す濃度で用いられることが望ましい。 The concentration of the TGFβ signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used, within a range that can achieve the above-mentioned effects. When SB431542 is used as the TGFβ signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 100 μM, more preferably about 100 nM to about 50 μM, and even more preferably about 500 nM to about 10 μM. When a TGFβ signaling pathway inhibitor other than SB431542 is used, it is desirable to use it at a concentration that shows the same TGFβ signaling pathway inhibitory activity as SB431542 at the above-mentioned concentration.
第一工程及び以降の工程において、中内胚葉への分化を抑制し、外胚葉・プラコード様組織の製造効率を向上させる観点から、Transforming growth factor-β-activated kinase 1(TAK1)に対する阻害物質を添加することもまた好ましい。TAK1はTGFβ、骨形成タンパク質(BMP)、インターロイキン1(IL-1)、TNF-α等により活性化されるシグナル伝達を媒介する、MAPキナーゼキナーゼキナーゼ(MAPKKK)ファミリーのセリンスレオニンタンパク質キナーゼである。 In the first and subsequent steps, from the viewpoint of suppressing differentiation into mesendoderm and improving the production efficiency of ectoderm/placode-like tissue, it is also preferable to add an inhibitor of transforming growth factor-β-activated kinase 1 (TAK1). TAK1 is a serine-threonine protein kinase of the MAP kinase kinase kinase (MAPKKK) family that mediates signal transduction activated by TGFβ, bone morphogenetic protein (BMP), interleukin 1 (IL-1), TNF-α, etc.
TAK1阻害物質とはTAK1が媒介するシグナル伝達を抑制し得るものである限り限定されない。核酸、タンパク質、低分子有機化合物のいずれであってもよい。当該物質として例えばTAK1と基質の結合を阻害する物質、TAK1のリン酸化を阻害する物質、TAK1の脱リン酸化を促進する物質、TAK1の転写や翻訳を阻害する物質、TAK1の分解を促進する物質等が挙げられる。 The TAK1 inhibitor is not limited as long as it can suppress signal transduction mediated by TAK1. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit the binding of TAK1 to a substrate, substances that inhibit the phosphorylation of TAK1, substances that promote the dephosphorylation of TAK1, substances that inhibit the transcription or translation of TAK1, and substances that promote the degradation of TAK1.
TAK1阻害物質として、例えば、(5Z)-7-Oxozeaenol((3S,5Z,8S,9S,11E)-3,4,9,10-tetrahydro-8,9,16-trihydroxy-14-methoxy-3-methyl-1H-2-benzoxacyclotetradecine-1,7(8H)-dione)、N-Des(aminocarbonyl)AZ-TAK1 inhibitor(3-Amino-5-[4-(4-morpholinylmethyl)phenyl]-2-thiophenecarboxamide)、Takinib(N1-(1-Propyl-1H-benzimidazol-2-yl)-1,3-benzenedicarboxamide)、NG25(N-[4-[(4-Ethyl-1-piperazinyl)methyl]-3-(trifluoromethyl)phenyl]-4-methyl-3-(1H-pyrrolo[2,3-b]pyridin-4-yloxy)-benzamide trihydrochloride)、Sarsasapogenin及びこれらの誘導体、類縁体が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。 As a TAK1 inhibitor, for example, (5Z)-7-Oxozeaenol ((3S, 5Z, 8S, 9S, 11E)-3,4,9,10-tetrahydro-8,9,16-trihydroxy-14-methoxy-3 -methyl-1H-2-benzoxacyclotetradecine-1,7(8H)-dione), N-Des(aminocarbonyl)AZ-TAK1 inhibitor(3-Amino-5-[4-(4-morpholinyl) methyl)phenyl]-2-thiophenecarbox amide), Takinib (N1-(1-Propyl-1H-benzimidazol-2-yl)-1,3-benzenedicarboxamide), NG25 (N-[4-[(4-Ethyl-1-piperazinyl)methyl]-3- (trifluoromethyl)phenyl]-4-methyl-3-(1H-pyrrolo[2,3-b]pyridin-4-yloxy)-benzamide trihydrochloride), Sarsasapo- genin, and their derivatives and analogues. These substances are They may be used alone or in combination.
TAK1阻害物質は、好ましくは(5Z)-7-Oxozeaenolである。第一工程におけるTAK1阻害物質として(5Z)-7-Oxozeaenolを用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約50μM、より好ましくは約100nM~約25μM、さらに好ましくは約500nM~約10μMの濃度で使用される。また、(5Z)-7-Oxozeaenol以外のTAK1阻害物質を使用する場合、上記濃度の(5Z)-7-Oxozeaenolと同等のTAK1阻害活性を示す濃度で用いられることが好ましい。TAK1阻害活性は、例えばCellchemical biology 24.8(2017):1029-1039.に記載のキナーゼアッセイ等の手法により決定することができる。下垂体組織に含有される細胞の割合の制御の観点から、上記TAK1阻害物質は第一工程及び以降の工程の任意の段階で添加しその後に除去することができる。好ましい一態様としては、上記TAK1阻害物質は後述する工程(b)開始時に添加する。 The TAK1 inhibitor is preferably (5Z)-7-Oxozeaenol. When (5Z)-7-Oxozeaenol is used as the TAK1 inhibitor in the first step, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 50 μM, more preferably about 100 nM to about 25 μM, and even more preferably about 500 nM to about 10 μM. When a TAK1 inhibitor other than (5Z)-7-Oxozeaenol is used, it is preferably used at a concentration that exhibits TAK1 inhibitory activity equivalent to that of (5Z)-7-Oxozeaenol at the above concentration. TAK1 inhibitory activity can be determined, for example, by a method such as the kinase assay described in Cell chemical biology 24.8 (2017): 1029-1039. From the viewpoint of controlling the proportion of cells contained in the pituitary tissue, the TAK1 inhibitor can be added at any stage of the first step or any subsequent steps and then removed. In a preferred embodiment, the TAK1 inhibitor is added at the start of step (b) described below.
第一工程において用いられる培地は、上述したようなものである限り特に限定されない。第一工程において用いられる培地は血清培地又は無血清培地であり得る。化学的に未決定な成分の混入を回避する観点から、本明細書においては、無血清培地が好適に用いられる。調製の煩雑さを回避するには、例えば市販のKSR等の血清代替物を適量添加した無血清培地を使用することが好ましい。無血清培地へのKSRの添加量としては、例えばヒトES細胞の場合は、通常約1%から約30%であり、好ましくは約2%から約20%である。無血清培地としては、例えばIMDMとF-12の1:1の混合液に5%KSR、450μM 1-モノチオグリセロール及び1xChemically Defined Lipid Concentrateが添加された培地、又はGMEMに5%~20%KSR、NEAA、ピルビン酸及び2-メルカプトエタノールが添加された培地が挙げられる。 The medium used in the first step is not particularly limited as long as it is as described above. The medium used in the first step may be a serum medium or a serum-free medium. In order to avoid contamination with chemically undefined components, a serum-free medium is preferably used in this specification. To avoid the complexity of the preparation, it is preferable to use a serum-free medium to which an appropriate amount of a serum substitute such as commercially available KSR has been added. The amount of KSR added to the serum-free medium is usually about 1% to about 30%, and preferably about 2% to about 20%, in the case of human ES cells, for example. Examples of serum-free media include a medium in which 5% KSR, 450 μM 1-monothioglycerol, and 1x Chemically Defined Lipid Concentrate are added to a 1:1 mixture of IMDM and F-12, or a medium in which 5% to 20% KSR, NEAA, pyruvic acid, and 2-mercaptoethanol are added to GMEM.
第一工程の開始時において、細胞は接着状態又は浮遊状態のいずれでもよい。好ましい一態様として、多能性幹細胞を単一細胞に分散させた後に再凝集させ、浮遊状態の細胞凝集体を形成させる。このために、第一工程の開始前に、多能性幹細胞、一例として工程(a)で得られた多能性幹細胞を単一細胞に分散する操作を行うことが好ましい。分散させる操作により得られた「分散された細胞」は、好ましくは単一細胞であるが、例えば2以上100以下の少数の細胞からなる細胞の塊を含んでもよく、2以上50以下の細胞からなる細胞の塊を含んでもよい。「分散された細胞」は、例えば単一細胞を7割以上及び細胞の塊を3割以下含んでいてもよく、好ましくは単一細胞を8割以上及び細胞の塊を2割以下含む。 At the start of the first step, the cells may be in either an adherent state or a floating state. In a preferred embodiment, the pluripotent stem cells are dispersed into single cells and then re-aggregated to form floating cell aggregates. For this purpose, it is preferable to carry out an operation of dispersing the pluripotent stem cells, for example the pluripotent stem cells obtained in step (a), into single cells before the start of the first step. The "dispersed cells" obtained by the dispersion operation are preferably single cells, but may also include cell clumps consisting of a small number of cells, for example, 2 to 100 cells, or may include cell clumps consisting of 2 to 50 cells. The "dispersed cells" may, for example, include 70% or more single cells and 30% or less cell clumps, and preferably include 80% or more single cells and 20% or less cell clumps.
多能性幹細胞を分散させる方法としては、機械的分散処理、細胞分散液処理、細胞保護剤添加処理が挙げられ、これらの処理を組み合わせて行ってもよい。細胞を分散させる方法としては、好ましくは細胞保護剤添加処理と同時に細胞分散液処理を行い、次いで機械的分散処理をするとよい。 Methods for dispersing pluripotent stem cells include mechanical dispersion treatment, cell dispersion treatment, and cell protective agent addition treatment, and these treatments may be performed in combination. A preferred method for dispersing cells is to perform cell dispersion treatment simultaneously with cell protective agent addition treatment, followed by mechanical dispersion treatment.
細胞保護剤添加処理に用いられる細胞保護剤としては、FGFシグナル伝達経路作用物質、ヘパリン、Rho-associated protein kinase(ROCK)阻害物質、ミオシン阻害物質、ポリアミン類、統合的ストレス応答(Integrated stress response:ISR)阻害剤、カスパーゼ阻害剤、細胞接着促進物質、血清、又は血清代替物等を挙げることができる。好ましい細胞保護剤としては、ROCK阻害物質が挙げられる。分散により誘導される多能性幹細胞(特に、ヒトの多能性幹細胞)の細胞死を抑制するために、ROCK阻害物質を第一工程の培養開始時から添加することは好ましい。ROCK阻害物質としては、Y-27632((R)-(+)-trans-4-(1-Aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide,dihydrochloride)、Fasudil(HA1077)(1-(5-Isoquinolinylsulfonyl)homopiperazine,hydrochloride)、H-1152(5-[[(2S)-hexahydro-2-methyl-1H-1,4-diazepin-1-yl]sulfonyl]-4-methyl-isoquinoline,dihydrochloride)、HA-1100(Hydroxyfasudil)(1-(1-Hydroxy-5-isoquinolinesulfonyl)homopiperazine,hydrochloride)、Chroman 1((3S)-N-[2-[2-(dimethylamino)ethoxy]-4-(1H-pyrazol-4-yl)phenyl]-6-methoxy-3,4-dihydro-2H-chromene-3-carboxamide)、Belumosudil(KD025、2-[3-[4-[(1H-Indazol-5-yl)amino]quinazolin-2-yl]phenoxy]-N-isopropylacetamide)、HSD1590([2-Methoxy-3-(4,5,10-triazatetracyclo[7.7.0.02,6.012,16]hexadeca-1(9),2(6),3,7,10,12(16)-hexaen-11-yl)phenyl]boronic acid)、CRT0066854((S)-3-phenyl-N1-(2-pyridin-4-yl-5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4-yl)propane-1,2-diamine)、RKI1447(1-(3-hydroxybenzyl)-3-(4-(pyridin-4-yl)thiazol-2-yl)urea)、Ripasudil(4-Fluoro-5-[[(2S)-hexahydro-2-methyl-1H-1,4-diazepin-1-yl]sulfonyl]isoquinoline)、GSK269962A(N-[3-[2-(4-amino-1,2,5-oxadiazol-3-yl)-1-ethylimidazo[4,5-c]pyridin-6-yl]oxyphenyl]-4-(2-morpholin-4-ylethoxy)benzamide)、GSK429286A(N-(6-fluoro-1H-indazol-5-yl)-2-methyl-6-oxo-4-(4-(trifluoromethyl)phenyl)-1,4,5,6-tetrahydropyridine-3-carboxamide)、Y-33075((R)-4-(1-Aminoethyl)-N-1H-pyrrolo[2,3-b]pyridin-4-ylbenzamide)、LX7101(N,N-Dimethylcarbamic acid 3-[[[4-(aminomethyl)-1-(5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-4-piperidinyl]carbonyl]amino]phenyl ester)、AT13148((alphaS)-alpha-(Aminomethyl)-alpha-(4-chlorophenyl)-4-(1H-pyrazol-4-yl)benzenemethanol)、SAR407899(6-(piperidin-4-yloxy)isoquinolin-1(2H)-one hydrochloride)、GSK180736A(4-(4-fluorophenyl)-N-(1H-indazol-5-yl)-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide)、Hydroxyfasudil(1-(1-hydroxy-5-isoquinolinesulfonyl)homopiperazine,HCl)、bdp5290(4-Chloro-1-(4-piperidinyl)-N-[3-(2-pyridinyl)-1H-pyrazol-4-yl]-1H-pyrazole-3-carboxamide)、sr-3677(N-[2-[2-(Dimethylamino)ethoxy]-4-(1H-pyrazol-4-yl)phenyl]-2,3-dihydro-1,4-benzodioxin-2-carboxamidehydrochloride)、CCG-222740(N-(4-Chlorophenyl)-5,5-difluoro-1-(3-(furan-2-yl)benzoyl)piperidine-3-carboxamide)、ROCK inhibitor-2(N-[(1R)-1-(3-methoxyphenyl)ethyl]-4-pyridin-4-ylbenzamide)、Rho-Kinase-IN-1(N-[1-[(4-methylsulfanylphenyl)methyl]piperidin-3-yl]-1H-indazol-5-amine)、ZINC00881524(N-(4,5-dihydronaphtho[1,2-d]thiazol-2-yl)-2-(3,4-dimethoxyphenyl)acetamide)、SB772077B((3S)-1-[[2-(4-Amino-1,2,5-oxadiazol-3-yl)-1-ethyl-1H-imidazo[4,5-c]pyridin-7-yl]carbonyl]-3-pyrrolidinamine dihydrochloride)、Verosudil(N-(1,2-Dihydro-1-oxo-6-isoquinolinyl)-alpha-(dimethylamino)-3-thiopheneacetamide)、GSK-25(4-(4-chloro-2-fluorophenyl)-2-(2-chloropyridin-4-yl)-1-(6-fluoro-1H-indazol-5-yl)-6-methyl-4H-pyrimidine-5-carboxamide)及びこれらの誘導体等を挙げることができる。細胞接着促進物質としては例えばアドへサミン及びアドヘサミン-RGDS誘導体(長瀬産業社製)等が挙げられる。細胞保護剤としては、調製済みの細胞保護剤を用いることもできる。調製済みの細胞保護剤としては、例えば、RevitaCell Supplement(Thermo Fisher Scientific社製)、CloneR、CloneR2(Stemcell Technologies社製)等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。第一工程において、細胞保護剤としてROCK阻害物質であるY-27632を添加する場合は、通常約10nM~約10mM、好ましくは約100nM~約1mM、より好ましくは約1μM~約100μMの濃度となるように培養環境中に添加する。第一工程において、細胞保護剤としてROCK阻害物質であるChroman 1を添加する場合は、通常約10pM~約1mM、好ましくは約100pM~約100μM、より好ましくは約1nM~約10μMの濃度となるように培養環境中に添加する。培養液中のROCK阻害物質の濃度は、多能性幹細胞の細胞死を抑制する必要がある期間(例えば、1~3日間、好ましくは1もしくは2日間)保たれていればよく、培地交換操作を行う際に、ROCK阻害物質を含まない培地で、後述の半量培地交換操作等を行うことで、当該期間経過後徐々に減少させてもよい。また当該期間経過後ROCK阻害物質を含まない培地で全量培地交換操作を行ってもよい。
Cytoprotective agents used in the cytoprotective agent addition treatment include substances acting on the FGF signaling pathway, heparin, Rho-associated protein kinase (ROCK) inhibitors, myosin inhibitors, polyamines, integrated stress response (ISR) inhibitors, caspase inhibitors, cell adhesion promoters, serum, or serum substitutes. A preferred cytoprotective agent is a ROCK inhibitor. In order to suppress cell death of pluripotent stem cells (especially human pluripotent stem cells) induced by dispersion, it is preferable to add a ROCK inhibitor from the start of the culture in the first step. ROCK inhibitors include Y-27632 ((R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide, dihydrochloride), Fasudil (HA1077) (1-(5-isoquinolinylsulfonyl) homopiperazine, hydrochloride), ride), H-1152 (5-[[(2S)-hexahydro-2-methyl-1H-1,4-diazepin-1-yl]sulfonyl]-4-methyl-isoquinoline, dihydrochloride), HA-110 0(Hydroxyfasudil)(1-(1-Hydroxy-5-isoquinolinesulfonyl)h omopiperazine, hydrochloride), Chroman 1 ((3S)-N-[2-[2-(dimethylamino)ethoxy]-4-(1H-pyrazol-4-yl)phenyl]-6-methoxy-3,4-d ihydro-2H-chromene-3-carboxamide), Belumosudil (KD025, 2-[ 3-[4-[(1H-Indazol-5-yl)amino]quinazolin-2-yl]phenoxy]-N-isopropylacetamide), HSD1590([2-Methoxy-3-(4,5,10-triazate tracyclo[7.7.0.02,6.012,16]hexadeca-1(9),2(6),3,7,10,12(16)- hexaen-11-yl) phenyl] boronic acid), CRT0066854 ((S)-3-phenyl-N1-(2-pyridin-4-yl-5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]p yrimidin-4-yl) propane-1,2-diamine), RKI1447 (1-(3-hydro xybenzyl)-3-(4-(pyridin-4-yl)thiazol-2-yl)urea), Ripasudil(4-Fluoro-5-[[(2S)-hexahydro-2-methyl-1H-1,4-diazepin-1-yl ]sulfonyl]isoquinoline), GSK269962A(N-[3-[2-(4-amino-1,2,5- oxadiazol-3-yl)-1-ethylimidazo[4,5-c]pyridin-6-yl]oxyphenyl]-4-(2-morpholin-4-ylethoxy)benzamide), GSK429286A(N-(6-f luoro-1H-indazol-5-yl)-2-methyl-6-oxo-4-(4-(trifluorometh yl) phenyl)-1,4,5,6-tetrahydropyridine-3-carboxamide), Y-33075((R)-4-(1-Aminoethyl)-N-1H-pyrrolo[2,3-b]pyridin-4-ylbenzam ide), LX7101(N,N-Dimethylcarbamic acid 3-[[[4-(aminomet hyl)-1-(5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-4-piperidinyl]carbonyl]amino]phenyl ester), AT13148((alphaS)-alpha-(Amin omethyl)-alpha-(4-chlorophenyl)-4-(1H-pyrazol-4-yl) benzenemethanol), SAR407899 (6-(piperidin-4-yloxy)isoquinolin-1(2H)-one hydrochloride), GSK180736A (4-(4-fluorophenyl)-N- (1H-indazol-5-yl)-6-methyl-2-oxo-1,2,3,4-tetrahydropyri midine-5-carboxamide), Hydroxyfasudil (1-(1-hydroxy-5-isoquinoline sulfonyl)homopiperazine, HCl), bdp5290 (4-Chloro-1-(4-pi peridinyl)-N-[3-(2-pyridinyl)-1H-pyrazol-4-yl]-1H-pyraz ole-3-carboxamide), sr-3677(N-[2-[2-(Dimethylamino)ethoxy]-4-(1H-pyrazol-4-yl)phenyl]-2,3-dihydro-1,4-benzodioxin-2-ca rboxamidehydrochloride), CCG-222740 (N-(4-Chlorophenyl)-5, 5-difluoro-1-(3-(furan-2-yl)benzoyl)piperidine-3-carboxamide), ROCK inhibitor-2(N-[(1R)-1-(3-methoxyphenyl)ethyl]-4-pyri din-4-ylbenzamide), Rho-Kinase-IN-1 (N-[1-[(4-methylsul phanylphenyl)methyl]piperidin-3-yl]-1H-indazol-5-amine), ZINC00881524(N-(4,5-dihydronaphtho[1,2-d]thiazol-2-yl)-2-(3, 4-dimethoxyphenyl) acetamide), SB772077B ((3S)-1-[[2-(4-Amino -1,2,5-oxadiazol-3-yl)-1-ethyl-1H-imidazo[4,5-c]pyridin-7-yl]carbonyl]-3-pyrrolidinamine dihydrochloride), Verosudil (N -(1,2-Dihydro-1-oxo-6-isoquinolinyl)-alpha-(dimethylami Examples of the cell adhesion promoter include adhesamine and adhesamine-RGDS derivatives (manufactured by Nagase & Co., Ltd.). As the cell protective agent, a prepared cell protective agent can also be used. Examples of prepared cell protective agents include RevitaCell Supplement (manufactured by Thermo Fisher Scientific), CloneR, and CloneR2 (manufactured by Stemcell Technologies). These substances may be used alone or in combination. In the first step, when the ROCK inhibitor Y-27632 is added as the cell protective agent, it is usually added to the culture environment at a concentration of about 10 nM to about 10 mM, preferably about 100 nM to about 1 mM, and more preferably about 1 μM to about 100 μM. In the first step, when the
細胞分散液処理に用いられる細胞分散液としては、トリプシン、コラゲナーゼ、ヒアルロニダーゼ、エラスターゼ、プロナーゼ、DNase、パパイン等の酵素及びエチレンジアミン四酢酸等のキレート剤の少なくとも1つを含む溶液を挙げることができる。市販の細胞分散液、例えばTripLE Select(Thermo Fisher Scientific社製)やTripLE Express(Thermo Fisher Scientific社製)、Accumax(Innovative Cell Technologies社製)を用いることもできる。工程(a)の後に得られた多能性幹細胞の処理における好ましい細胞分散液は、TrypLE Selectであるが、これに限定されない。 The cell dispersion liquid used in the cell dispersion treatment may be a solution containing at least one of an enzyme such as trypsin, collagenase, hyaluronidase, elastase, pronase, DNase, papain, etc., and a chelating agent such as ethylenediaminetetraacetic acid. Commercially available cell dispersion liquids, such as TripLE Select (Thermo Fisher Scientific), TripLE Express (Thermo Fisher Scientific), and Accumax (Innovative Cell Technologies), may also be used. A preferred cell dispersion liquid for treating the pluripotent stem cells obtained after step (a) is TrypLE Select, but is not limited to this.
機械的分散処理の方法としては、ピペッティング処理又はスクレーパーでの掻き取り操作が挙げられる。分散された細胞は上記培地中に懸濁される。 Mechanical dispersion methods include pipetting or scraping with a scraper. The dispersed cells are suspended in the above-mentioned medium.
多能性幹細胞を分散させる方法としては、例えば多能性幹細胞のコロニーをROCK阻害物質の存在下で、エチレンジアミン四酢酸又はAccumaxで処理し、さらにピペッティングにより分散させる方法が挙げられる。 One method for dispersing pluripotent stem cells is, for example, treating a colony of pluripotent stem cells with ethylenediaminetetraacetic acid or Accumax in the presence of a ROCK inhibitor, and then dispersing the colony by pipetting.
第一工程において浮遊培養を実施する場合は、分散された多能性幹細胞の懸濁液を非細胞接着性の培養器材中に播種する。培養器材が非接着性である場合、細胞は浮遊培養され、複数の多能性幹細胞が集合して細胞凝集体を形成する。 If suspension culture is performed in the first step, a suspension of dispersed pluripotent stem cells is seeded into a non-adhesive cultureware. If the cultureware is non-adhesive, the cells are cultured in suspension, and multiple pluripotent stem cells gather together to form cell aggregates.
浮遊培養する際、分散された多能性幹細胞を、10cmディッシュのような比較的大きな培養器材に播種することにより、1つの培養器材中に複数の細胞凝集体を同時に形成させてもよいが、細胞凝集体ごとの大きさのばらつきを生じにくくする観点からは、例えば非細胞接着性の96ウェルマイクロプレートのようなマルチウェルプレート(U底、V底)の各ウェルに一定数の分散された多能性幹細胞を播種することが好ましい。これを静置培養すると、細胞が迅速に凝集することにより、各ウェルに1個の細胞凝集体を形成させることができる(Serum-free culture of Embryoid Body-like aggregates with quick reaggregation; SFEBq法)。例えば、培養器材の表面に超親水性のポリマーをコートする等の加工により培養器材を非細胞接着性とすることができる。非細胞接着性のマルチウェルプレートとしては、例えばPrimeSurface96V底プレート(MS-9096V、住友ベークライト社製)等が挙げられる。より迅速に細胞凝集体を形成させるために遠心操作を行ってもよい。各ウェルで形成された細胞凝集体を複数のウェルから回収することにより、均一な細胞凝集体の集団を得ることができる。細胞凝集体が均一であれば、その後の工程において、ウェルごと及び反復実験ごとの製造効率をより安定させることができ、より再現性よく下垂体を構成する細胞を製造することができる。 In suspension culture, dispersed pluripotent stem cells may be seeded in a relatively large culture vessel such as a 10 cm dish to simultaneously form multiple cell aggregates in one culture vessel. However, from the viewpoint of preventing variation in size of each cell aggregate, it is preferable to seed a certain number of dispersed pluripotent stem cells in each well of a multi-well plate (U-bottom, V-bottom) such as a non-cell-adhesive 96-well microplate. When this is cultured statically, the cells rapidly aggregate to form one cell aggregate in each well (Serum-free culture of Embryoid Body-like aggregates with quick reaggregation; SFEBq method). For example, the culture vessel can be made non-cell-adhesive by processing such as coating the surface of the culture vessel with a superhydrophilic polymer. An example of a non-cell-adhesive multi-well plate is the PrimeSurface 96V-bottom plate (MS-9096V, manufactured by Sumitomo Bakelite Co., Ltd.). Centrifugation may be performed to form cell aggregates more quickly. A uniform population of cell aggregates can be obtained by collecting the cell aggregates formed in each well from multiple wells. If the cell aggregates are uniform, the production efficiency for each well and each repeated experiment can be more stable in the subsequent steps, and cells that make up the pituitary gland can be produced with greater reproducibility.
分散された多能性幹細胞から細胞凝集体を形成させるための別の態様としては、一つのウェルが複数のマイクロウェルに分割され、2つ以上の細胞塊が形成される培養器材を用いることができる。言い換えると、本実施形態の一側面において、前記第一工程、第二工程、b工程、及び第三工程のいずれか一つ以上の任意の工程の浮遊培養を、少なくとも1つのウェルが形成されている培養器材中で実施し、前記ウェルは、複数のマイクロウェルに分割されていて、前記マイクロウェルの1つにつき、1つの細胞塊が形成されるように浮遊培養を実施し、1つのウェルにつき分割されたマイクロウェルに相当する数の細胞塊を調製してもよい。上記マイクロウェルとして、底面に細胞が一ヶ所に沈降して凝集体の形成が促進されるすり鉢、下向きの四角錐、凹状等の加工、グリッド、隆起等が複数形成されている培養器材、ないし凝集体を形成しやすいように底面の一部のみに細胞が接着可能な加工を施されている培養器材等を用いることもできる。マイクロウェルを有する培養器材のウェル一つ当たりの培養面積は特に限定されないが、細胞塊を効率よく生産する観点から、好ましくは底面積が1cm2(48ウェルプレート相当)より大きく、より好ましくは2cm2(24ウェルプレート相当)より大きく、さらに好ましくは4cm2(12ウェルプレート相当)より大きい。上記のような培養器材としては、例えば、胚様体形成プレートAggreWell(StemCell Technologies社製)、PAMCELL(ANK社製)、スフェロイドマイクロプレート(Corning社製)、NanoCulture Plate/Dish (Organogenix社製) Cell-able(東洋合成社製)、EZSPHERE(AGCテクノグラス社製)、SPHERICALPLATE 5D(水戸工業社製)、TASCL(シムスバイオ社製)、マイクロウェルバッグ(例、Scientific reports, 2022, 12.1: 1-11.に記載のもの)等が挙げられるが、これに限定されない。 As another aspect for forming cell aggregates from dispersed pluripotent stem cells, a culture vessel in which one well is divided into a plurality of microwells and two or more cell aggregates are formed can be used. In other words, in one aspect of this embodiment, the suspension culture of any one or more of the first step, the second step, the b step, and the third step can be performed in a culture vessel in which at least one well is formed, and the well is divided into a plurality of microwells, and the suspension culture is performed so that one cell aggregate is formed in each of the microwells, and the number of cell aggregates corresponding to the number of divided microwells can be prepared for each well. As the microwell, a culture vessel in which a plurality of mortars, downward pyramids, concaves, grids, protuberances, etc. are formed on the bottom surface, which allows cells to settle in one place and promotes the formation of aggregates, or a culture vessel in which only a part of the bottom surface is processed to allow cells to adhere to the bottom surface so that aggregates can be easily formed can be used. The culture area per well of a culture device having microwells is not particularly limited, but from the viewpoint of efficient production of cell masses, the bottom area is preferably greater than 1 cm2 (equivalent to a 48-well plate), more preferably greater than 2 cm2 (equivalent to a 24-well plate), and even more preferably greater than 4 cm2 (equivalent to a 12-well plate). Examples of the culture equipment include, but are not limited to, the embryoid body formation plate AggreWell (manufactured by StemCell Technologies), PAMCELL (manufactured by ANK), spheroid microplate (manufactured by Corning), NanoCulture Plate/Dish (manufactured by Organogenix), Cell-able (manufactured by Toyo Gosei), EZSPHERE (manufactured by AGC Technoglass), SPHERICALPLATE 5D (manufactured by Mito Kogyo Co., Ltd.), TASCL (manufactured by Sims Bio), and microwell bag (e.g., those described in Scientific reports, 2022, 12.1: 1-11.).
培養器材としては、各ウェル内に細胞凝集体が入ったままの状態でプレート全体の培地を一度に交換することが可能な三次元細胞培養容器を用いることもまた好ましい。このような三次元細胞培養容器としては、例えば、PrimeSurface 96スリットウェルプレート(住友ベークライト社製)等が挙げられる。このプレートには96ウェルのそれぞれの上部に培地が出入りできる細い開口部(スリット)が設けられている。スリットは細胞凝集体が通過しにくい幅に設定されているため、細胞凝集体同士の癒着を防止しながら、プレート全体の培地を一度に交換することができ、操作の効率性及び細胞凝集体の質を向上させることができる。 As the culture equipment, it is also preferable to use a three-dimensional cell culture vessel that allows the culture medium to be replaced for the entire plate at once while the cell aggregates remain in each well. An example of such a three-dimensional cell culture vessel is the PrimeSurface 96 slit well plate (manufactured by Sumitomo Bakelite Co., Ltd.). This plate has narrow openings (slits) at the top of each of the 96 wells through which the culture medium can enter and exit. The slits are set to a width that makes it difficult for cell aggregates to pass through, so the culture medium for the entire plate can be replaced at once while preventing adhesion between the cell aggregates, improving the efficiency of the operation and the quality of the cell aggregates.
第一工程における多能性幹細胞の濃度は、細胞凝集体をより均一に、効率的に形成させるように適宜設定することができる。例えば96ウェルマイクロウェルプレートを用いてヒト多能性幹細胞(例えば、工程(a)から得られたヒトiPS細胞)を浮遊培養する場合、1ウェルあたり通常約1×103から約1×105細胞、好ましくは約3×103から約5×104細胞、より好ましくは約4×103から約2×104細胞、さらに好ましくは約4×103から約1.6×104細胞、特に好ましくは約8×103から約1.2×104細胞となるように調製した液を各ウェルに添加し、プレートを静置して細胞凝集体を形成させる。例えば、1ディッシュあたり約260個のマイクロウェルを有する培養器材であるEZSPHERE SP ディッシュ35mm Type905を用いてヒト多能性幹細胞(例えば、工程(a)から得られたヒトiPS細胞)を浮遊培養する場合、1ディッシュあたり通常約1×104から約1×108細胞、好ましくは約3×104から約5×107細胞、より好ましくは約4×104から約2×107細胞、さらに好ましくは約4×104から約1.6×107細胞、特に好ましくは約8×104から約1.2×107細胞となるように調製した液をディッシュに添加し、ディッシュを静置して細胞凝集体を形成させる。例えば、1ウェルあたり約1800個のマイクロウェルを有する培養器材であるAggreWell 800,6-well plateを用いてヒト多能性幹細胞(例えば、工程(a)から得られたヒトiPS細胞)を浮遊培養する場合、1ウェルあたり通常約1×104から約1×108細胞、好ましくは約3×104から約5×107細胞、より好ましくは約4×104から約2×107細胞、さらに好ましくは約4×104から約1.6×107細胞、特に好ましくは約8×104から約1.2×107細胞となるように調製した液を各ウェルに添加し、プレートを遠心して細胞凝集体を形成させる。細胞数は、血球計算盤で計数することによって求めることができる。 The concentration of pluripotent stem cells in the first step can be appropriately set so as to form cell aggregates more uniformly and efficiently. For example, when human pluripotent stem cells (e.g., human iPS cells obtained from step (a)) are cultured in suspension using a 96-well microwell plate, a liquid prepared so as to obtain about 1 x 10 3 to about 1 x 10 5 cells, preferably about 3 x 10 3 to about 5 x 10 4 cells, more preferably about 4 x 10 3 to about 2 x 10 4 cells, even more preferably about 4 x 10 3 to about 1.6 x 10 4 cells, and particularly preferably about 8 x 10 3 to about 1.2 x 10 4 cells per well is added to each well, and the plate is left to form cell aggregates. For example, when human pluripotent stem cells (e.g., human iPS cells obtained in step (a)) are cultured in suspension using EZSPHERE SP Dish 35 mm Type 905, a cultureware having about 260 microwells per dish, a liquid prepared so that typically about 1 x 104 to about 1 x 108 cells, preferably about 3 x 104 to about 5 x 107 cells, more preferably about 4 x 104 to about 2 x 107 cells, even more preferably about 4 x 104 to about 1.6 x 107 cells, and particularly preferably about 8 x 104 to about 1.2 x 107 cells per dish is added to the dish, and the dish is allowed to stand to form cell aggregates. For example, when human pluripotent stem cells (e.g., human iPS cells obtained from step (a)) are cultured in suspension using AggreWell 800, 6-well plate, a cultureware having about 1800 microwells per well, a solution prepared so that each well contains usually about 1×10 4 to about 1×10 8 cells, preferably about 3×10 4 to about 5×10 7 cells, more preferably about 4×10 4 to about 2×10 7 cells, even more preferably about 4×10 4 to about 1.6×10 7 cells, and particularly preferably about 8×10 4 to about 1.2×10 7 cells is added to each well, and the plate is centrifuged to form cell aggregates. The number of cells can be determined by counting with a hemocytometer.
細胞凝集体を形成させるために必要な浮遊培養の時間は、用いる多能性幹細胞によって適宜決定可能であるが、均一な細胞凝集体を形成するためにはできる限り短時間であることが望ましい。分散された細胞が、細胞凝集体が形成されるに至るまでの工程は、細胞が集合する工程、及び集合した細胞が凝集体を形成する工程にわけられる。分散された細胞を播種する時点(すなわち、浮遊培養開始時)から細胞が集合するまでは、例えば、ヒト多能性幹細胞(ヒトiPS細胞等)の場合には、好ましくは約24時間以内、より好ましくは約12時間以内である。分散された細胞を播種する時点(すなわち浮遊培養開始時)から細胞凝集体が形成されるまでは、例えば、ヒト多能性幹細胞(ヒトiPS細胞等)の場合には、好ましくは約72時間以内、より好ましくは約48時間以内である。細胞凝集体を形成するまでの時間は、細胞を凝集させる用具や、遠心条件等を調整することにより適宜調節することが可能である。 The time required for suspension culture to form cell aggregates can be appropriately determined depending on the pluripotent stem cells used, but it is desirable that the time be as short as possible in order to form uniform cell aggregates. The process from the dispersed cells until they form cell aggregates can be divided into a process in which the cells gather and a process in which the gathered cells form aggregates. For example, in the case of human pluripotent stem cells (human iPS cells, etc.), the time from the time the dispersed cells are seeded (i.e., the time when suspension culture begins) until the cells gather is preferably within about 24 hours, more preferably within about 12 hours. For example, in the case of human pluripotent stem cells (human iPS cells, etc.), the time from the time the dispersed cells are seeded (i.e., the time when suspension culture begins) until the cell aggregates are formed is preferably within about 72 hours, more preferably within about 48 hours. The time until the cell aggregates are formed can be appropriately adjusted by adjusting the tool for aggregating the cells, the centrifugation conditions, etc.
多能性幹細胞を迅速に集合させて細胞凝集体を形成させると、形成された凝集体から分化誘導される細胞において上皮様構造を再現性よく形成させることができる。細胞の凝集体を形成させる実験的な操作としては、例えばウェルの小さなプレート(例えばウェルの底面積が平底換算で0.1~2.0cm2程度のプレート)やマイクロポア等を用いて小さいスペースに細胞を閉じ込める方法、小さな遠心チューブを用いて短時間遠心することで細胞を凝集させる方法が挙げられる。ウェルの小さなプレートとして、例えば24ウェルプレート(面積が平底換算で1.88cm2程度)、48ウェルプレート(面積が平底換算で1.0cm2程度)、96ウェルプレート(面積が平底換算で0.35cm2程度、内径6~8mm程度)、384ウェルプレートが挙げられる。好ましくは96ウェルプレートが挙げられる。ウェルの小さなプレートの形状として、ウェルを上から見たときの底面の形状としては、多角形、長方形、楕円、真円が挙げられ、好ましくは真円が挙げられる。ウェルの小さなプレートの形状として、ウェルを横から見たときの底面の形状としては、外周部が高く内部が低くくぼんだ構造が好ましく、例えば、U底、V底、M底が挙げられ、好ましくはU底又はV底、最も好ましくはV底が挙げられる。ウェルの小さなプレートとして、細胞培養皿(例えば60mm~150mmディッシュ、カルチャーフラスコ)の底面に凹凸又はくぼみがあるものを用いてもよい。ウェルの小さなプレートの底面は、細胞非接着性の底面、好ましくは細胞非接着性コートした底面を用いるのが好ましい。 When pluripotent stem cells are rapidly aggregated to form cell aggregates, epithelial-like structures can be reproducibly formed in cells induced to differentiate from the formed aggregates. Examples of experimental procedures for forming cell aggregates include a method of confining cells in a small space using a small well plate (e.g., a plate with a well bottom area of about 0.1 to 2.0 cm2 calculated as a flat bottom) or a micropore, and a method of aggregating cells by centrifuging for a short time using a small centrifuge tube. Examples of small well plates include 24-well plates (area of about 1.88 cm2 calculated as a flat bottom), 48-well plates (area of about 1.0 cm2 calculated as a flat bottom), 96-well plates (area of about 0.35 cm2 calculated as a flat bottom, inner diameter of about 6 to 8 mm), and 384-well plates. A preferred example is a 96-well plate. Examples of the shape of the small well plate, when viewed from above, include polygonal, rectangular, elliptical, and perfect circle as the shape of the bottom of the well. A preferred example is a perfect circle. As for the shape of the small well plate, the shape of the bottom of the well when viewed from the side is preferably a structure with a high outer periphery and a low inner recess, for example, a U-bottom, a V-bottom, or an M-bottom, preferably a U-bottom or a V-bottom, and most preferably a V-bottom. As the small well plate, a cell culture dish (e.g., a 60 mm to 150 mm dish, culture flask) having an uneven or recessed bottom may be used. The bottom of the small well plate is preferably a non-cell-adhesive bottom, preferably a bottom coated with a non-cell-adhesive material.
細胞凝集体を形成させる別の手法として、立体印刷機、3Dプリンターを用いることもまた好ましい。分散された単一の細胞、ないし複数の細胞から構成されるスフェロイドを、生体適合性を有するインク(バイオインク)に懸濁し、バイオ3Dプリンター(例えば、Cellink社製 BIO X等)で出力する、あるいは細胞集団をニードルに刺して積み上げる(サイフューズ社製 Spike等)といった手法により、所望の形態の細胞集団を調製することができる。 As another method for forming cell aggregates, it is also preferable to use a three-dimensional printing machine or 3D printer. A cell population of the desired shape can be prepared by suspending a dispersed single cell or a spheroid composed of multiple cells in a biocompatible ink (bioink) and printing it out using a bio 3D printer (e.g., Celllink's BIO X, etc.), or by piercing a cell population with a needle and stacking it (Cyfuse's Spike, etc.).
細胞凝集体が形成されたことは、細胞凝集体のサイズ及び細胞数、巨視的形態、組織染色解析による微視的形態及びその均一性、分化及び未分化マーカーの発現及びその均一性、分化マーカーの発現制御及びその同期性、分化効率の凝集体間の再現性等に基づき判断することが可能である。 The formation of cell aggregates can be determined based on the size and cell number of the cell aggregates, the macroscopic morphology, the microscopic morphology and its uniformity determined by tissue staining analysis, the expression and uniformity of differentiated and undifferentiated markers, the control of expression of differentiation markers and their synchronicity, and the reproducibility of differentiation efficiency between aggregates.
第一工程の開始時において、好ましい一態様として、接着培養を実施する。工程(a)後の培養器材上の多能性幹細胞をそのまま第一工程に用いてもよいし、多能性幹細胞を単一細胞に分散させた後に再度接着性の培養器材に播種してもよい。多能性幹細胞の単一細胞への分散後の再播種を実施する際に、適切な細胞外マトリクス又は合成細胞接着分子を足場として用いてもよい。足場により、表面をコーティングした培養器材中で、多能性幹細胞を接着培養できる。細胞外マトリクスは、好ましくはマトリゲル又はラミニンである。合成細胞接着分子としては、ポリ-D-リジン、RGD配列等細胞接着性のドメインを含有する合成ペプチド等が挙げられる。細胞の播種数としては下垂体への分化が生じる限り特に限定されないが、細胞間の接着と相互作用を再現する観点から、培養器材への播種後72時間以内に細胞密度が器材の培養スペースの6割以上に相当するセミコンフルエントに到達するような密度であることもまた好ましい。 At the start of the first step, in a preferred embodiment, adherent culture is performed. The pluripotent stem cells on the culture vessel after step (a) may be used as they are in the first step, or the pluripotent stem cells may be dispersed into single cells and then seeded again on the adherent culture vessel. When reseeding the pluripotent stem cells after dispersing them into single cells, an appropriate extracellular matrix or synthetic cell adhesion molecule may be used as a scaffold. The scaffold allows adherent culture of the pluripotent stem cells in the culture vessel whose surface is coated. The extracellular matrix is preferably matrigel or laminin. Examples of synthetic cell adhesion molecules include synthetic peptides containing a cell adhesive domain such as poly-D-lysine and RGD sequence. The number of cells seeded is not particularly limited as long as differentiation into the pituitary gland occurs, but from the viewpoint of reproducing adhesion and interaction between cells, it is also preferable that the cell density reaches semi-confluence, which corresponds to more than 60% of the culture space of the vessel, within 72 hours after seeding on the culture vessel.
接着性の培養器材として、マイクロパターンが施された培養器材を用いることもまた好ましい。培養器材上のマイクロパターンは、細胞接着性領域と細胞非接着性領域から構成することができ、細胞接着性領域で細胞が接着培養されることが好ましい。細胞接着性領域と細胞非接着性領域の形状は、培養器材上に展開可能である限り限定されない。細胞接着性領域と細胞非接着性領域は、一つの培養器材上に単一の領域が形成されていてもよいし、複数個形成されていてもよい。細胞接着性領域は、接着性を向上させる目的で人工的に処理されていることが好ましい。マイクロパターンが施された培養器材としては、例えばCYTOOchip(CYTOO社製)、ibidi Micropatterning(ibidi社製)等が挙げられる。PDMS製モールドとマトリクス等を用いて培養器材を調製することもできる。あるいは、細胞外マトリクス、細胞接着を促進する基質等でコートされた培養器材を例えば細胞プロセシング装置(Model:CPD-017、片岡製作所社製)等を用いてレーザー等で加工し、細胞接着性領域と細胞非接着性領域を任意の形状に作製しても良い。工程(a)で得られた多能性幹細胞をマイクロパターンが施された培養器材上で培養する場合、例えば既報の方法を参照し実施することができる(Nature protocols,11(11),2223-2232.)。 It is also preferable to use a cultureware with a micropattern as the adhesive cultureware. The micropattern on the cultureware can be composed of a cell adhesive region and a cell non-adhesive region, and it is preferable that cells are cultured in the cell adhesive region. The shape of the cell adhesive region and the cell non-adhesive region is not limited as long as they can be developed on the cultureware. The cell adhesive region and the cell non-adhesive region may be formed as a single region on one cultureware, or multiple regions may be formed. It is preferable that the cell adhesive region is artificially treated for the purpose of improving adhesion. Examples of cultureware with a micropattern include CYTOOChip (manufactured by CYTOO), ibidi Micropatterning (manufactured by ibidi), etc. The cultureware can also be prepared using a PDMS mold and a matrix, etc. Alternatively, a cultureware coated with an extracellular matrix or a substrate that promotes cell adhesion may be processed with a laser or the like using, for example, a cell processing device (Model: CPD-017, Kataoka Seisakusho Co., Ltd.) to create cell adhesive and non-cell adhesive regions of any shape. When culturing the pluripotent stem cells obtained in step (a) on a cultureware with a micropattern, this can be carried out by referring to, for example, a previously reported method (Nature protocols, 11(11), 2223-2232.).
培養器材が培地の灌流を行なうための流路(マイクロ流路)を有することも好ましく、第一工程及びその後の工程において、灌流環境下で細胞を培養してもよい。このような培養器材をマイクロ流体チップともいう。培養器材(例えば、マイクロ流体チップ)は、本発明の製造方法において培養される細胞以外の細胞又は組織を培養する他の培養器材(例えば、マイクロ流体チップ)と流路により接続されていてもよい。これにより、下垂体と他の細胞又は組織との相互作用を再現することができる。下垂体と共培養する他の細胞又は組織としては、下垂体から分泌されるホルモンによる調節を受ける組織、下垂体の成長、分化、成熟、生存を促進する組織、例えば脳、血管、骨、筋肉、脂肪、甲状腺、肝臓、副腎、精巣、卵巣、乳房の細胞又は組織等が挙げられるが、これらに限定されない。培地の灌流の為の方法としては、例えば、マグネティックスターラー、ペリスタルティックポンプ等の使用が挙げられるが、これに限定されない。 It is also preferable that the culture equipment has a flow path (micro-flow path) for perfusing the medium, and cells may be cultured in a perfusion environment in the first step and subsequent steps. Such a culture equipment is also called a microfluidic chip. The culture equipment (e.g., microfluidic chip) may be connected by a flow path to other culture equipment (e.g., microfluidic chip) for culturing cells or tissues other than the cells to be cultured in the manufacturing method of the present invention. This makes it possible to reproduce the interaction between the pituitary gland and other cells or tissues. Examples of other cells or tissues to be co-cultured with the pituitary gland include, but are not limited to, tissues that are regulated by hormones secreted from the pituitary gland, and tissues that promote the growth, differentiation, maturation, and survival of the pituitary gland, such as cells or tissues of the brain, blood vessels, bone, muscle, fat, thyroid gland, liver, adrenal gland, testis, ovary, and breast. Examples of methods for perfusing the medium include, but are not limited to, the use of a magnetic stirrer, a peristaltic pump, etc.
培養器材は、酸素又は培地を透過可能な膜を有してもよい。培養器材は、化合物、増殖因子等の濃度勾配を形成可能であってもよい。膜は、例えば、多孔質膜である。このような膜を有する培養器材においては、膜で隔てた一方で本発明の製造方法によって細胞を培養し、他方でそれ以外の細胞又は組織、フィーダー細胞などを培養することができる。これにより、下垂体を構成する細胞又はその前駆細胞及びこれらを含む細胞集団と、他の細胞又は組織とをコンタミネーションさせることなく培養することができる。 The culture equipment may have a membrane that is permeable to oxygen or culture medium. The culture equipment may be capable of forming a concentration gradient of compounds, growth factors, etc. The membrane is, for example, a porous membrane. In culture equipment having such a membrane, cells can be cultured by the manufacturing method of the present invention on one side separated by the membrane, and other cells or tissues, feeder cells, etc. can be cultured on the other side. This makes it possible to culture cells that constitute the pituitary gland or their precursor cells and cell populations containing these without contamination with other cells or tissues.
第一工程及び以降の工程において、培地交換操作を行う場合、例えば元ある培地を捨てずに新しい培地を加える操作(培地添加操作)、元ある培地を半量程度(元ある培地の体積量の30~90%程度、例えば40~60%程度)捨てて新しい培地を半量程度(元ある培地の体積量の30~90%、例えば40~60%程度)加える操作(半量培地交換操作)、元ある培地を全量程度(元ある培地の体積量の90%以上)捨てて新しい培地を全量程度(元ある培地の体積量の90%以上)加える操作(全量培地交換操作)が挙げられる。 In the first and subsequent steps, when a medium exchange operation is performed, examples include an operation in which new medium is added without discarding the original medium (medium addition operation), an operation in which about half the original medium (about 30-90% of the volume of the original medium, for example about 40-60%) is discarded and about half the new medium (about 30-90% of the volume of the original medium, for example about 40-60%) is added (half medium exchange operation), and an operation in which about the entire amount of the original medium (90% or more of the volume of the original medium) is discarded and about the entire amount of new medium (90% or more of the volume of the original medium) is added (full medium exchange operation).
ある時点で、特定の成分を添加する場合、例えば終濃度を計算した上で、元ある培地を半量程度捨てて、特定の成分を終濃度よりも高い濃度で含む新しい培地を半量程度加える操作(半量培地交換操作)を行ってもよい。ある時点で、元の培地に含まれる成分を希釈して濃度を下げる場合、例えば培地交換操作を、1日に複数回、好ましくは1時間以内に複数回(例えば2~3回)行ってもよい。また、ある時点で、元の培地に含まれる成分を希釈して濃度を下げる場合、細胞又は細胞凝集体を別の培養容器に移してもよい。培地交換操作に用いる道具は特に限定されないが、例えばピペッター、ピペットマン(登録商標)、マルチチャンネルピペット、連続分注器等が挙げられる。例えば、培養器材として96ウェルプレートを用いる場合、マルチチャンネルピペットを使ってもよい。 When adding a specific component at a certain point in time, for example, after calculating the final concentration, an operation may be performed in which about half of the original medium is discarded and about half of a new medium containing the specific component at a concentration higher than the final concentration is added (half medium exchange operation). When diluting a component contained in the original medium to reduce the concentration at a certain point in time, for example, the medium exchange operation may be performed multiple times a day, preferably multiple times within an hour (e.g., 2 to 3 times). Also, when diluting a component contained in the original medium to reduce the concentration at a certain point in time, the cells or cell aggregates may be transferred to another culture vessel. The tools used for the medium exchange operation are not particularly limited, but examples include a pipetter, Pipetman (registered trademark), a multichannel pipette, and a continuous dispenser. For example, when a 96-well plate is used as the culture equipment, a multichannel pipette may be used.
第一工程における培養の時間は、通常8時間~6日間程度、好ましくは12時間~60時間程度である。 The culture time in the first step is usually about 8 hours to 6 days, preferably about 12 hours to 60 hours.
第一工程及び以降の工程において、下垂体の製造効率を向上させる観点から、プラコード領域への分化を促進する化合物を添加することもまた好ましい。上記のような作用を有する化合物として、例えば米国特許US20160326491A1号に記載のBRL-54443、Phenanthroline、Parthenolide等が挙げられる。プラコード領域への分化を促進する化合物としてBRL-54443を用いる場合は通常約10nM~約100μM、Phenanthrolineを用いる場合は通常約10nM~約100μM、Parthenolideを用いる場合は通常約10nM~約100μMの濃度で用いられる。 In the first and subsequent steps, it is also preferable to add a compound that promotes differentiation into the placode region in order to improve the efficiency of pituitary production. Examples of compounds that have the above-mentioned effect include BRL-54443, phenanthrolin, and parthenolide, which are described in US Patent US20160326491A1. When BRL-54443 is used as a compound that promotes differentiation into the placode region, it is usually used at a concentration of about 10 nM to about 100 μM, when phenanthrolin is used, it is usually used at a concentration of about 10 nM to about 100 μM, and when parthenolide is used, it is usually used at a concentration of about 10 nM to about 100 μM.
第一工程において、下垂体への分化誘導効率を改善する観点から、ソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養を実施することもできる。第一工程において用いられるShhシグナル伝達経路作用物質としては工程(a)で例示したものと同様のものが用いられ得る。工程(a)及び第一工程のShhシグナル伝達経路作用物質は同一であっても異なっていてもよいが、好ましくは同一であり、また好ましくはSAGである。
培地中のShhシグナル伝達経路作用物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。第一工程においてShhシグナル伝達経路作用物質としてSAGを用いる場合は、通常、約1nM~約3μM、好ましくは約10nM~約2μM、より好ましくは約30nM~約1μM、更に好ましくは約50nM~約500nMの濃度で使用される。
In the first step, from the viewpoint of improving the efficiency of inducing differentiation into pituitary gland, the culture can be performed in the presence of a substance acting on the Sonic Hedgehog signaling pathway. As the substance acting on the Shh signaling pathway used in the first step, the same substances as those exemplified in step (a) can be used. The substances acting on the Shh signaling pathway in step (a) and the first step may be the same or different, but are preferably the same, and are preferably SAG.
The concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range that can achieve the above-mentioned effects. When SAG is used as the substance acting on the Shh signaling pathway in the first step, it is usually used at a concentration of about 1 nM to about 3 μM, preferably about 10 nM to about 2 μM, more preferably about 30 nM to about 1 μM, and even more preferably about 50 nM to about 500 nM.
<工程(2)>:第二工程
工程(2)は、BMPシグナル伝達経路作用物質及びソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で、第一工程で得られた細胞集団を培養する。第一工程で細胞を浮遊培養している場合は、工程(2)でも引き続き形成された細胞凝集体を浮遊培養すればよい。第一工程で細胞を接着培養している場合は、工程(2)でも引き続き細胞を接着培養すればよい。第一工程で細胞を浮遊培養した後、工程(2)で接着培養してもよい。
<Step (2)>: Second Step In step (2), the cell population obtained in the first step is cultured in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway. If the cells are cultured in suspension in the first step, the formed cell aggregates may be continuously cultured in suspension in step (2). If the cells are cultured in adhesion in the first step, the cells may be continuously cultured in adhesion in step (2). After the cells are cultured in suspension in the first step, they may be cultured in adhesion in step (2).
BMPシグナル伝達経路作用物質とは、BMPにより媒介されるシグナル伝達経路を増強し得る物質である。BMPにより媒介されるシグナル伝達経路を増強し得る物質は、例えば培養環境中のBMPリガンドを安定化し、力価を向上させる物質、I型BMP受容体であるALK-1、ALK-2、ALK-3、ALK-6と結合し、受容体の下流の細胞内シグナル伝達を活性化、惹起する物質、細胞内のBMPシグナル伝達に関与するSmad-1、Smad-5、Smad-8、Smad-9のリン酸化を惹起する物質、Smad-1/5/8/9による遺伝子の転写の活性化や抑制等の機能を誘導・増強する物質等が挙げられる。BMPシグナル伝達経路作用物質としては、例えば、BMP2、BMP4もしくはBMP7等のBMPタンパク質、GDF5、6、7等のGDFタンパク質、抗BMP受容体抗体及びBMP部分ペプチド等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。生物学的活性の見地からのBMPシグナル伝達経路作用物質の定義として、例えば、マウス前駆軟骨細胞株ATDC5、マウス頭蓋冠由来細胞株MC3T3-E1、マウス横紋筋由来細胞株C2C12等の細胞に対する骨芽細胞様細胞への分化誘導能、及びアルカリホスファターゼ産生誘導能を有する物質が挙げられる。上記活性を有する物質としては、例えばBMP2、BMP4、BMP5、BMP6、BMP7、BMP9、BMP10、BMP13/GDF6、BMP14/GDF5、GDF7等が挙げられる。 A substance acting on the BMP signaling pathway is a substance that can enhance the signaling pathway mediated by BMP. Examples of substances that can enhance the signaling pathway mediated by BMP include substances that stabilize BMP ligands in a culture environment and improve their potency, substances that bind to type I BMP receptors ALK-1, ALK-2, ALK-3, and ALK-6 and activate and induce intracellular signaling downstream of the receptor, substances that induce phosphorylation of Smad-1, Smad-5, Smad-8, and Smad-9 involved in intracellular BMP signaling, and substances that induce and enhance functions such as activation and inhibition of gene transcription by Smad-1/5/8/9. Examples of substances acting on the BMP signaling pathway include BMP proteins such as BMP2, BMP4, and BMP7, GDF proteins such as GDF5, 6, and 7, anti-BMP receptor antibodies, and BMP partial peptides. These substances may be used alone or in combination. In terms of biological activity, a substance acting on the BMP signal transduction pathway can be defined as a substance that has the ability to induce differentiation into osteoblast-like cells and the ability to induce alkaline phosphatase production in cells such as mouse precursor chondrocyte cell line ATDC5, mouse calvaria-derived cell line MC3T3-E1, and mouse striated muscle-derived cell line C2C12. Examples of substances having the above activities include BMP2, BMP4, BMP5, BMP6, BMP7, BMP9, BMP10, BMP13/GDF6, BMP14/GDF5, GDF7, etc.
BMP2タンパク質及びBMP4タンパク質は、例えば、R&D Systemsから、BMP7タンパク質は、例えば、Biolegend社から、GDF5タンパク質は、例えば、ペプロテック社から、GDF6タンパク質は、例えば、プライムジーン社から、GDF7タンパク質は、例えば、富士フイルム和光純薬株式会社から入手可能である。BMPシグナル伝達経路作用物質は、好ましくはBMP2、BMP4、BMP7、BMP13及びGDF7からなる群より選ばれる少なくとも1つのタンパク質を含み、より好ましくはBMP4を含む。 BMP2 protein and BMP4 protein are available, for example, from R&D Systems, BMP7 protein is available, for example, from Biolegend, GDF5 protein is available, for example, from Peprotech, GDF6 protein is available, for example, from Primegene, and GDF7 protein is available, for example, from Fujifilm Wako Pure Chemical Industries, Ltd. The BMP signaling pathway active substance preferably includes at least one protein selected from the group consisting of BMP2, BMP4, BMP7, BMP13, and GDF7, and more preferably includes BMP4.
培地中のBMPシグナル伝達経路作用物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。下垂体を構成する細胞の製造効率向上の観点から、BMPシグナル伝達経路作用物質としてBMP4を用いる場合は、通常約1pM~約100nM、好ましくは約10pM~約50nM、より好ましくは約25pM~約25nM、さらに好ましくは約25pM~約5nM、特に好ましくは約100pM~約5nM、最も好ましくは約500pM~約2nMの濃度で使用される。また、BMP4以外のBMPシグナル伝達経路作用物質を使用する場合、上述の濃度のBMP4と同等のBMPシグナル伝達経路促進活性を示す濃度で用いられることが望ましい。当業者であれば、BMPシグナル伝達経路作用物質として、例えば、市販の組み換えBMPタンパク質等を用いる場合、製品添付書類に記載の活性、例えばマウス前駆軟骨細胞株ATDC5に対するアルカリホスファターゼ産生誘導能のED50等の値と上述のBMP4の濃度と活性を比較することにより、添加するBMPシグナル伝達経路作用物質濃度を容易に決定することができる。 The concentration of the substance acting on the BMP signaling pathway in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of improving the production efficiency of cells that constitute the pituitary gland, when BMP4 is used as the substance acting on the BMP signaling pathway, it is usually used at a concentration of about 1 pM to about 100 nM, preferably about 10 pM to about 50 nM, more preferably about 25 pM to about 25 nM, even more preferably about 25 pM to about 5 nM, particularly preferably about 100 pM to about 5 nM, and most preferably about 500 pM to about 2 nM. Furthermore, when a substance acting on the BMP signaling pathway other than BMP4 is used, it is desirably used at a concentration that exhibits the same BMP signaling pathway promoting activity as BMP4 at the above-mentioned concentration. When using, for example, a commercially available recombinant BMP protein as a substance acting on the BMP signaling pathway, a person skilled in the art can easily determine the concentration of the substance acting on the BMP signaling pathway to be added by comparing the activity described in the product insert, such as the ED50 value of the ability to induce alkaline phosphatase production in the mouse precursor chondrocyte cell line ATDC5, with the concentration and activity of the above-mentioned BMP4.
BMPシグナル伝達経路作用物質として、当業者に周知の化合物を使用することもできる。BMPシグナル伝達経路作用物質としては、例えばSmurf1阻害物質、Chk1阻害物質、リン酸化Smad安定化物質等が挙げられる。上記のような活性を有する化合物としては、例えば、A-01([4-[[4-Chloro-3-(trifluoromethyl)phenyl]sulfonyl]-1-piperazinyl][4-(5-methyl-1H-pyrazol-1-yl)phenyl]methanone)、PD 407824(9-Hydroxy-4-phenyl-pyrrolo[3,4-c]carbazole-1,3(2H,6H)-dione)、SB4(2-[[(4-Bromophenyl)methyl]thio]benzoxazole)、SJ000291942(2-(4-Ethylphenoxy)-N-(4-fluoro-3-nitrophenyl)-acetamide)及びこれらの誘導体等が挙げられる。 Compounds well known to those skilled in the art can also be used as BMP signaling pathway active substances. Examples of BMP signaling pathway active substances include Smurf1 inhibitors, Chk1 inhibitors, and phosphorylated Smad stabilizers. Examples of compounds having the above-mentioned activity include A-01 ([4-[[4-Chloro-3-(trifluoromethyl)phenyl]sulfonyl]-1-piperazinyl][4-(5-methyl-1H-pyrazol-1-yl)phenyl]methane), PD 407824 (9-Hydroxy-4-phenyl-pyr Examples include rolo[3,4-c]carbazole-1,3(2H,6H)-dione, SB4 (2-[[(4-bromophenyl)methyl]thio]benzoxazole), SJ000291942 (2-(4-ethylphenoxy)-N-(4-fluoro-3-nitrophenyl)-acetamide) and their derivatives.
工程(2)において用いられるShhシグナル伝達経路作用物質としては工程(a)で例示したものと同様のものが用いられ得る。工程(a)及び工程(2)のShhシグナル伝達経路作用物質、場合によっては工程(1)のShhシグナル伝達経路作用物質は同一であっても異なっていてもよいが、好ましくは同一であり、また好ましくはSAGである。
培地中のShhシグナル伝達経路作用物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。工程(2)においてShhシグナル伝達経路作用物質としてSAGを用いる場合は、通常、約1nM~約5μM、好ましくは約10nM~約4.5μM、より好ましくは約50nM~約4μM、更に好ましくは約100nM~約3μMの濃度で使用される。
The substance acting on the Shh signaling pathway used in step (2) may be the same as those exemplified in step (a). The substances acting on the Shh signaling pathway in steps (a) and (2), and optionally the substance acting on the Shh signaling pathway in step (1), may be the same or different, but are preferably the same, and are preferably SAGs.
The concentration of the substance acting on the Shh signaling pathway in the medium can be appropriately set depending on the substance used within a range that can achieve the above-mentioned effects. When SAG is used as the substance acting on the Shh signaling pathway in step (2), it is usually used at a concentration of about 1 nM to about 5 μM, preferably about 10 nM to about 4.5 μM, more preferably about 50 nM to about 4 μM, and even more preferably about 100 nM to about 3 μM.
工程(2)において用いられる培地は、Shhシグナル伝達経路作用物質及びBMPシグナル伝達経路作用物質を含む限り特に限定されない。工程(2)において用いられる培地としては、第一工程に挙げた培地が挙げられる。 The medium used in step (2) is not particularly limited as long as it contains a substance acting on the Shh signaling pathway and a substance acting on the BMP signaling pathway. Examples of the medium used in step (2) include the medium listed in the first step.
下垂体の製造効率向上の観点から、工程(2)の開始時期は、第一工程における培養開始から好ましくは0.5時間以降6日以内であり、より好ましくは0.5時間以降72時間以内であり、さらに好ましくは24時間以降60時間以内である。浮遊培養を実施する場合、Wntシグナル経路阻害物質存在下で上記期間に工程(2)を開始すると、細胞凝集体の表面に非神経上皮様の組織が形成され、極めて効率よく下垂体が形成される。 From the viewpoint of improving the efficiency of pituitary gland production, the start time of step (2) is preferably 0.5 hours to 6 days after the start of the culture in the first step, more preferably 0.5 hours to 72 hours, and even more preferably 24 hours to 60 hours. When performing suspension culture, if step (2) is started during the above period in the presence of a Wnt signal pathway inhibitor, non-neuroepithelial-like tissue is formed on the surface of the cell aggregates, and pituitary glands are formed extremely efficiently.
第一工程で浮遊培養を行なっている場合、下垂体を構成する細胞の製造効率向上の観点から、工程(2)の開始時期は、好ましくは第一工程において形成された細胞凝集体の表層における1割以上、より好ましくは3割以上、さらに好ましくは5割以上の細胞が互いに密着結合を形成している時期である。細胞凝集体において密着結合が形成しているかは、当業者であれば例えば顕微鏡による観察、抗ZO-1抗体を用いた免疫染色等の手法により容易に判別することができる。 When suspension culture is performed in the first step, from the viewpoint of improving the production efficiency of cells constituting the pituitary gland, the start time of step (2) is preferably the time when at least 10% of the cells on the surface layer of the cell aggregate formed in the first step, more preferably at least 30%, and even more preferably at least 50% of the cells form tight junctions with each other. Those skilled in the art can easily determine whether tight junctions are formed in the cell aggregate by, for example, observation under a microscope or immunostaining using an anti-ZO-1 antibody.
工程(2)におけるBMPシグナル伝達経路作用物質の存在下での培養開始は、第一工程を行った培養容器を用いて、上述の培地交換操作(例えば培地添加操作、半量培地交換操作、全量培地交換操作等)を行ってもよいし、細胞を別の培養容器に移してもよい。 In step (2), the start of culture in the presence of a substance acting on the BMP signaling pathway may be performed by carrying out the above-mentioned medium replacement operation (e.g., medium addition operation, half-medium replacement operation, full-medium replacement operation, etc.) using the culture vessel in which the first step was carried out, or the cells may be transferred to a different culture vessel.
工程(2)におけるBMPシグナル伝達経路作用物質を含む培地での培養の期間は、適宜設定できる。工程(2)における培養の期間は、培養液中のBMPシグナル伝達経路作用物質の濃度が下垂体組織(具体的には、下垂体前駆細胞を含む細胞集団)を誘導するために必要な期間保たれていればよく、通常8時間以上、好ましくは10時間以上、より好ましくは12時間以上、さらに好ましくは14時間以上、最も好ましくは16時間以上である。
当該期間経過後に培地交換操作を行う際に、BMPシグナル伝達経路作用物質を含まない培地で、上述の半量培地交換操作等を行うことで、当該期間経過後、徐々にBMPシグナル伝達経路作用物質の濃度を減少させてもよい。また、当該期間経過後BMPシグナル伝達経路作用物質を含まない培地で全量培地交換操作を行ってもよいが、その場合においても、本明細書においては、Shhシグナル伝達経路作用物質を含む培地で培養が継続されている限り、工程(2)の一部に含まれる。
The period of culture in the medium containing the substance acting on the BMP signaling pathway in step (2) can be appropriately set. The period of culture in step (2) may be any period during which the concentration of the substance acting on the BMP signaling pathway in the culture medium is maintained for a period necessary for inducing pituitary tissue (specifically, a cell population containing pituitary progenitor cells), and is usually 8 hours or more, preferably 10 hours or more, more preferably 12 hours or more, even more preferably 14 hours or more, and most preferably 16 hours or more.
When the medium replacement operation is performed after the lapse of the period, the concentration of the substance acting on the BMP signaling pathway may be gradually reduced after the lapse of the period by performing the above-mentioned half-volume medium replacement operation with a medium not containing a substance acting on the BMP signaling pathway. Also, after the lapse of the period, a full-volume medium replacement operation may be performed with a medium not containing a substance acting on the BMP signaling pathway. Even in this case, as long as the culture is continued in a medium containing a substance acting on the Shh signaling pathway, it is included as part of step (2) in the present specification.
工程(2)におけるShhシグナル伝達経路作用物質を含む培地での培養の期間は、適宜設定できる。なお、工程(1)における浮遊培養がさらにShhシグナル伝達経路作用物質の存在下で実施される場合、下垂体ホルモン(特にACTH)分泌能向上の観点から、工程(1)及び工程(2)におけるShhシグナル伝達経路作用物質の存在下における培養期間は、20日~40日、好ましくは、25日~35日、さらに好ましくは約30日である。 The period of culture in the medium containing the substance acting on the Shh signaling pathway in step (2) can be set appropriately. When the suspension culture in step (1) is further performed in the presence of a substance acting on the Shh signaling pathway, from the viewpoint of improving the ability to secrete pituitary hormones (particularly ACTH), the culture period in the presence of the substance acting on the Shh signaling pathway in steps (1) and (2) is 20 to 40 days, preferably 25 to 35 days, and more preferably about 30 days.
工程(2)及び以降の工程において、下垂体プラコードへの分化を促進する観点から、FGFシグナル伝達経路作用物質を培養環境中に添加することもまた好ましい。FGFシグナル伝達経路作用物質とは、FGF(線維芽細胞増殖因子)により媒介されるシグナル伝達経路を増強し得る物質である限り特に限定はされない。FGFシグナル伝達経路作用物質としては、例えばFGF1、FGF2(bFGFと称することもある。)、FGF3、FGF8、FGF10等のFGFタンパク質、抗FGF受容体抗体、FGF部分ペプチド等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。 In order to promote differentiation into pituitary placodes in step (2) and the subsequent steps, it is also preferable to add an FGF signaling pathway active substance to the culture environment. The FGF signaling pathway active substance is not particularly limited as long as it is a substance that can enhance the signaling pathway mediated by FGF (fibroblast growth factor). Examples of FGF signaling pathway active substances include FGF proteins such as FGF1, FGF2 (also called bFGF), FGF3, FGF8, and FGF10, anti-FGF receptor antibodies, and FGF partial peptides. These substances may be used alone or in combination.
FGF2タンパク質及びFGF8タンパク質は、例えば富士フイルム和光純薬株式会社から入手可能である。FGFシグナル伝達経路作用物質は、好ましくはFGF2、FGF3、FGF8及びFGF10、並びにこれらの改変体からなる群より選ばれる少なくとも1つを含み、より好ましくはFGF2を含み、さらに好ましくは組換えヒトFGF2を含む。 FGF2 protein and FGF8 protein are available, for example, from Fujifilm Wako Pure Chemical Industries, Ltd. The FGF signaling pathway active substance preferably includes at least one selected from the group consisting of FGF2, FGF3, FGF8, FGF10, and modified forms thereof, more preferably includes FGF2, and even more preferably includes recombinant human FGF2.
培地中のFGFシグナル伝達経路作用物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。下垂体を構成する細胞への分化及び細胞の生存と増殖の促進の観点からは、FGFシグナル伝達経路作用物質としてFGF2を用いる場合は、通常約1pg/ml~約100μg/ml、好ましくは約10pg/ml~約50μg/ml、より好ましくは約100pg/ml~約10μg/ml、さらに好ましくは約500pg/ml~約1μg/ml、最も好ましくは約1ng/ml~約200ng/mlの濃度で使用される。また、FGF2以外のFGFシグナル伝達経路作用物質を使用する場合、上記濃度のFGF2と同等のFGFシグナル伝達経路促進活性を示す濃度で用いられることが望ましい。添加する物質のFGFシグナル伝達経路促進活性については、例えば、3T3細胞を用いた細胞増殖試験等の手法により測定することができる。 The concentration of the FGF signaling pathway active substance in the medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of differentiation into cells that constitute the pituitary gland and promotion of cell survival and proliferation, when FGF2 is used as the FGF signaling pathway active substance, it is usually used at a concentration of about 1 pg/ml to about 100 μg/ml, preferably about 10 pg/ml to about 50 μg/ml, more preferably about 100 pg/ml to about 10 μg/ml, even more preferably about 500 pg/ml to about 1 μg/ml, and most preferably about 1 ng/ml to about 200 ng/ml. When using an FGF signaling pathway active substance other than FGF2, it is desirable to use it at a concentration that shows the same FGF signaling pathway promoting activity as FGF2 at the above concentration. The FGF signaling pathway promoting activity of the added substance can be measured, for example, by a method such as a cell proliferation test using 3T3 cells.
培地中でのFGFタンパク質の活性を保持する目的から、FGFタンパク質を含む培地中にヘパリン、ヘパラン硫酸を添加することも好ましい。ヘパリンはナトリウム塩として、例えば、富士フイルム和光純薬株式会社から入手可能である。培地中のヘパリン又はヘパラン硫酸の濃度は、上述の効果を達成可能な範囲で適宜設定することが可能である。培地中のヘパリンナトリウムの濃度は、通常約1ng/ml~約100mg/ml、好ましくは約10ng/ml~約50mg/ml、より好ましくは約100ng/ml~約10mg/ml、さらに好ましくは約500ng/ml~約1mg/ml、最も好ましくは約1μg/ml~約200μg/mlである。ヘパラン硫酸を用いる場合、上記濃度のヘパリンと同様のFGFタンパク質保護の活性をもつ濃度であることが好ましい。37℃等での細胞培養環境下でFGFタンパク質の活性を保持する目的から、例えば米国特許US8772460B2号に記載のThermostable FGF2等のFGFの改変体や生分解性ポリマーにFGF2を結合させたStemBeads FGF2等のFGF2徐放性ビーズを用いることもまた好ましい。Thermostable FGF2は、例えば、HumanZyme社から入手可能である。StemBeads FGF2は例えばStemCulture社から入手可能である。 In order to maintain the activity of the FGF protein in the medium, it is also preferable to add heparin or heparan sulfate to the medium containing the FGF protein. Heparin is available as a sodium salt, for example from Fujifilm Wako Pure Chemical Industries, Ltd. The concentration of heparin or heparan sulfate in the medium can be appropriately set within a range that can achieve the above-mentioned effects. The concentration of heparin sodium in the medium is usually about 1 ng/ml to about 100 mg/ml, preferably about 10 ng/ml to about 50 mg/ml, more preferably about 100 ng/ml to about 10 mg/ml, even more preferably about 500 ng/ml to about 1 mg/ml, and most preferably about 1 μg/ml to about 200 μg/ml. When heparan sulfate is used, it is preferable that the concentration has the same FGF protein protection activity as the above-mentioned concentration of heparin. For the purpose of maintaining the activity of FGF protein in a cell culture environment at 37°C or the like, it is also preferable to use modified FGF such as Thermostable FGF2 described in US Patent No. US8772460B2, or FGF2 sustained-release beads such as StemBeads FGF2 in which FGF2 is bound to a biodegradable polymer. Thermostable FGF2 is available, for example, from HumanZyme. StemBeads FGF2 is available, for example, from StemCulture.
工程(2)及び以降の工程におけるFGFシグナル伝達経路作用物質の添加時期は適宜設定できる。好ましい一態様としては、工程(2)のBMPシグナル伝達経路作用物質の添加より6時間以降、より好ましくは12時間以降、さらにこのましくは18時間以降にFGFシグナル伝達経路作用物質を添加する。 The timing of addition of the substance acting on the FGF signaling pathway in step (2) and the subsequent steps can be set as appropriate. In a preferred embodiment, the substance acting on the FGF signaling pathway is added 6 hours or more, more preferably 12 hours or more, and even more preferably 18 hours or more after the addition of the substance acting on the BMP signaling pathway in step (2).
工程(2)において、工程(a)又は第一工程において用いた添加物、例えば、JNKシグナル伝達経路阻害物質、Wntシグナル伝達経路阻害物質、TGFβシグナル伝達経路阻害物質、TAK1阻害物質等を引き続き添加することもまた好ましい。工程(2)で添加するJNKシグナル伝達経路阻害物質、Wntシグナル伝達経路阻害物質又はTGFβシグナル伝達経路阻害物質は、それ以前の工程に用いられた物質と異なっていてもよいが、好ましくは同一である。添加物の濃度及び種類については、適宜調整することができる。これらの物質の添加時期は、工程(2)の開始と同時であってもよいし、異なっていてもよい。 In step (2), it is also preferable to continue adding the additives used in step (a) or the first step, such as a JNK signaling pathway inhibitor, a Wnt signaling pathway inhibitor, a TGFβ signaling pathway inhibitor, a TAK1 inhibitor, etc. The JNK signaling pathway inhibitor, Wnt signaling pathway inhibitor, or TGFβ signaling pathway inhibitor added in step (2) may be different from the substance used in the previous step, but is preferably the same. The concentration and type of additives can be adjusted as appropriate. The timing of addition of these substances may be simultaneous with the start of step (2) or may be different.
<工程(b)>:b工程
工程(b)は、BMPシグナル伝達経路阻害物質の添加条件下で、工程(2)で得られた細胞集団を培養する。工程(2)で細胞を浮遊培養している場合は、工程(b)でも引き続き形成された細胞凝集体を浮遊培養すればよい。工程(2)で細胞を接着培養している場合は、工程(b)でも引き続き細胞を接着培養すればよい。
<Step (b)>: Step b In step (b), the cell population obtained in step (2) is cultured under conditions in which a substance inhibiting the BMP signaling pathway is added. If the cells are cultured in suspension in step (2), the cell aggregates formed may be cultured in suspension in step (b). If the cells are cultured in adhesion in step (2), the cells may be cultured in adhesion in step (b).
BMPシグナル伝達経路阻害物質とは、BMPファミリー・タンパク質により惹起されるシグナル伝達を抑制し得るものである限り限定されない。核酸、タンパク質、低分子有機化合物のいずれであってもよい。当該物質として、例えば、BMPのプロセシングと細胞外への分泌を阻害する物質、BMPに直接作用する物質(例えば、タンパク質、抗体、アプタマー等)、BMPをコードする遺伝子の発現を抑制する物質(例えば、アンチセンスオリゴヌクレオチド、siRNA等)、BMP受容体とBMPの結合を阻害する物質、BMP受容体によるシグナル伝達に起因する生理活性を阻害する物質を挙げることができる。BMP受容体にはI型BMP受容体とII型BMP受容体が存在し、I型BMP受容体としてはBMPR1A、BMPR1B、ACVR、II型BMP受容体としてはTGF-beta R-II、ActR-II、ActR-IIB、BMPR2、MISR-IIが知られている。 The BMP signaling pathway inhibitor is not limited as long as it can suppress signaling induced by BMP family proteins. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include substances that inhibit BMP processing and extracellular secretion, substances that act directly on BMP (e.g., proteins, antibodies, aptamers, etc.), substances that suppress the expression of genes encoding BMP (e.g., antisense oligonucleotides, siRNA, etc.), substances that inhibit the binding of BMP receptors to BMP, and substances that inhibit physiological activity resulting from signaling by BMP receptors. There are type I BMP receptors and type II BMP receptors, and BMPR1A, BMPR1B, and ACVR are known as type I BMP receptors, while TGF-beta R-II, ActR-II, ActR-IIB, BMPR2, and MISR-II are known as type II BMP receptors.
BMPシグナル伝達経路阻害物質として知られているタンパク質として、例えば、Noggin、Chordin、Follistatin、Gremlin、Inhibin、Twisted Gastrulation、Coco、DANファミリーに属する分泌タンパク質等が挙げられる。上記工程(2)において培養液中にBMPシグナル伝達経路作用物質を添加していることから、以降のBMPシグナル伝達経路をより効果的に阻害するという観点から、工程(b)におけるBMPシグナル伝達経路阻害物質は、細胞外へのBMPの分泌よりも後のシグナル伝達経路を阻害する物質、例えばBMP受容体とBMPの結合を阻害する物質、BMP受容体によるシグナル伝達に起因する生理活性を阻害する物質等を含むことが好ましく、より好ましくはI型BMP受容体の阻害剤を含む。 Proteins known as BMP signaling pathway inhibitors include, for example, Noggin, Chordin, Follistatin, Gremlin, Inhibin, Twisted Gastrulation, Coco, and secreted proteins belonging to the DAN family. Since a substance acting on the BMP signaling pathway is added to the culture medium in step (2) above, from the viewpoint of more effectively inhibiting the subsequent BMP signaling pathway, it is preferable that the BMP signaling pathway inhibitor in step (b) includes a substance that inhibits the signaling pathway subsequent to the secretion of BMP outside the cell, such as a substance that inhibits the binding between BMP receptors and BMP, or a substance that inhibits physiological activity resulting from signaling by BMP receptors, and more preferably includes an inhibitor of type I BMP receptor.
BMPシグナル伝達経路阻害物質として、当業者に周知の化合物を使用することもできる。BMPシグナル伝達経路阻害物質としては、例えばI型BMP受容体の阻害物質が挙げられる。上記のような活性を有する化合物としては、例えば、K02288(3-[(6-Amino-5-(3,4,5-trimethoxyphenyl)-3-pyridinyl]phenol)、Dorsomorphin(6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)pyrazolo[1,5-a]pyrimidine)、LDN-193189(4-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride)、LDN-212854(5-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline)、LDN-214117(1-(4-(6-methyl-5-(3,4,5-trimethoxyphenyl)pyridin-3-yl)phenyl)piperazine)、ML347(5-[6-(4-Methoxyphenyl)pyrazolo[1,5-a]pyrimidin-3-yl]quinoline)、DMH1(4-(6-(4-Isopropoxyphenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline)、DMH2(4-[6-[4-[2-(4-Morpholinyl)ethoxy]phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]-quinoline)、Compound 1(3-(1,2,3-benzothiadiazol-6-yl)-1-[2-(cyclohex-1-en-1-yl)ethyl]urea)、VU0465350(7-(4-isopropoxyphenyl)-3-(1H-pyrazol-4-yl)imidazo[1,2-a]pyridine)、VU0469381(5-(6-(4-methoxyphenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinolone)、OD36(4-chloro-7,10-dioxa-13,17,18,21-tetrazatetracyclo[12.5.2.12,6.017,20]docosa-1(20),2(22),3,5,14(21),15,18-heptaene)、OD52、E6201((3S,4R,5Z,8S,9S,11E)-14-(ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9,10-tetrahydro-1H-benzo[c][1]oxacyclotetradecine-1,7(8H)-dione)、Saracatinib(N-(5-chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(oxan-4-yloxy)quinazolin-4-amine)、BYL719((2S)-1-N-[4-methyl-5-[2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl]-1,3-thiazol-2-yl]pyrrolidine-1,2-dicarboxamide)等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。 Compounds well known to those skilled in the art can also be used as BMP signaling pathway inhibitors. Examples of BMP signaling pathway inhibitors include inhibitors of type I BMP receptors. Compounds having the above-mentioned activity include, for example, K02288 (3-[(6-Amino-5-(3,4,5-trimethoxyphenyl)-3-pyridinyl]phenol), Dorsomorphin (6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)pyrazolo[1,5-a]pyrimid), and ine), LDN-193189 (4-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride), LDN-212854 (5 -[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quin oline), LDN-214117 (1-(4-(6-methyl-5-(3,4,5-trimethoxyphenyl)pyridin-3-yl)phenyl)piperazine), ML347(5-[6-(4-Methoxyphenyl) pyrazolo[1,5-a]pyrimidin-3-yl]quinoline), DMH1(4-(6-(4-Isopr opoxyphenyl) pyrazolo[1,5-a]pyrimidin-3-yl)quinoline), DMH2(4-[6-[4-[2-(4-Morpholinyl)ethoxy]phenyl]pyrazolo[1,5-a]py rimidin-3-yl]-quinoline), Compound 1(3-(1,2,3-benzothiadiazol-6-y l)-1-[2-(cyclohex-1-en-1-yl)ethyl]urea), VU0465350(7-(4-isopropoxyphenyl)-3-(1H-pyrazol-4-yl)imidazo[1,2-a]pyridine), V U0469381 (5-(6-(4-methoxyphenyl)pyrazolo[1,5-a]pyrimidin-3-yl) quinolone), OD36(4-chloro-7,10-dioxa-13,17,18,21-tetrazatetracyclo[12.5.2.12,6.017,20]docosa-1(20),2(22),3,5,14(21),15 , 18-heptaene), OD52, E6201 ((3S, 4R, 5Z, 8S, 9S, 11E)-14-(ethylamino) -8,9,16-trihydroxy-3,4-dimethyl-3,4,9,10-tetrahydro-1H-benzo[c][1]oxacyclotetradecine-1,7(8H)-dione), Saracatinib (N-(5-ch loro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)et hoxy]-5-(oxan-4-yloxy)quinazolin-4-amine, BYL719 ((2S)-1-N-[4-methyl-5-[2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl]-1,3-thiazol-2-yl]pyrrolidine-1,2-dicarboxamide), etc. These substances may be used alone or in combination.
BMPシグナル伝達経路阻害物質は、好ましくはI型BMP受容体阻害剤であり、より好ましくはK02288、Dorsomorphin、LDN-193189、LDN-212854、LDN-214117、ML347、DMH1及びDMH2からなる群より選ばれる少なくとも1つを含み、さらに好ましくはK02288乃至LDN-193189を含む。 The BMP signaling pathway inhibitor is preferably a type I BMP receptor inhibitor, more preferably at least one selected from the group consisting of K02288, Dorsomorphin, LDN-193189, LDN-212854, LDN-214117, ML347, DMH1 and DMH2, and even more preferably K02288 to LDN-193189.
培地中のBMPシグナル伝達経路阻害物質の濃度は、上述の効果を達成可能な範囲で用いる物質に応じて適宜設定することが可能である。下垂体組織の形成効率の観点から、工程(b)におけるBMPシグナル伝達経路阻害物質としてK02288を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約50μM、より好ましくは約100nM~約50μM、さらに好ましくは約500nM~約25μMの濃度で使用される。BMPシグナル伝達経路阻害物質としてLDN-193189を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約10μM、より好ましくは約25nM~約1μM、さらに好ましくは約100nM~約500nMの濃度で使用される。BMPシグナル伝達経路阻害物質としてLDN-212854を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約10μM、より好ましくは約25nM~約5μM、さらに好ましくは約250nM~約3μMの濃度で使用される。BMPシグナル伝達経路阻害物質としてML347を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約50μM、より好ましくは約100nM~約50μM、さらに好ましくは約1μM~約25μMの濃度で使用される。BMPシグナル伝達経路阻害物質としてDMH2を用いる場合は、通常約1nM~約100μM、好ましくは約10nM~約10μM、より好ましくは約25nM~約5μM、さらに好ましくは約250nM~約3μMの濃度で使用される。また、K02288以外のBMPシグナル伝達経路阻害物質を使用する場合、上記濃度のK02288と同等のBMPシグナル伝達経路阻害活性を示す濃度で用いられることが望ましい。 The concentration of the BMP signaling pathway inhibitor in the culture medium can be set appropriately depending on the substance used within a range that can achieve the above-mentioned effects. From the viewpoint of the efficiency of pituitary tissue formation, when K02288 is used as the BMP signaling pathway inhibitor in step (b), it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 50 μM, more preferably about 100 nM to about 50 μM, and even more preferably about 500 nM to about 25 μM. When LDN-193189 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 10 μM, more preferably about 25 nM to about 1 μM, and even more preferably about 100 nM to about 500 nM. When LDN-212854 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 10 μM, more preferably about 25 nM to about 5 μM, and even more preferably about 250 nM to about 3 μM. When ML347 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 50 μM, more preferably about 100 nM to about 50 μM, and even more preferably about 1 μM to about 25 μM. When DMH2 is used as the BMP signaling pathway inhibitor, it is usually used at a concentration of about 1 nM to about 100 μM, preferably about 10 nM to about 10 μM, more preferably about 25 nM to about 5 μM, and even more preferably about 250 nM to about 3 μM. In addition, when using a BMP signaling pathway inhibitor other than K02288, it is desirable to use it at a concentration that exhibits the same BMP signaling pathway inhibitory activity as K02288 at the above concentration.
工程(2)を実施した後に工程(b)を開始する時期に関しては、適宜設定できる。工程(b)を開始する時期は、通常工程(2)を開始してから8時間以上、15日間以内、好ましくは10時間以上、12日間以内、より好ましくは12時間以上、9日間以内、さらに好ましくは14時間以上、8日間以内、最も好ましくは16時間以上、7日間以内である。 The timing for starting step (b) after carrying out step (2) can be set appropriately. The timing for starting step (b) is usually at least 8 hours and within 15 days, preferably at least 10 hours and within 12 days, more preferably at least 12 hours and within 9 days, even more preferably at least 14 hours and within 8 days, and most preferably at least 16 hours and within 7 days after starting step (2).
工程(2)及び以降の工程において、培地に副腎皮質ホルモン類を添加することにより、細胞集団を副腎皮質ホルモン類により処理してもよい。副腎皮質ホルモン類処理により、下垂体プラコード及び/又はラトケ嚢からACTH産生細胞以外の下垂体ホルモン産生細胞(即ち、GH産生細胞、PRL産生細胞、TSH産生細胞、LH産生細胞、FSH産生細胞等)への分化が促進される。副腎皮質ホルモン類としては、ハイドロコルチゾン、酢酸コルチゾン、酢酸フルドロコルチゾン等の天然糖質コルチコイド;デキサメサゾン、ベタメタゾン、プレドニゾロン、メチルプレドニゾロン、トリアムシノロン等の人工的に合成された糖質コルチコイド等を挙げることができるが、これらに限定されない。 In step (2) and the subsequent steps, the cell population may be treated with corticosteroids by adding corticosteroids to the medium. Treatment with corticosteroids promotes differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells other than ACTH-producing cells (i.e., GH-producing cells, PRL-producing cells, TSH-producing cells, LH-producing cells, FSH-producing cells, etc.). Examples of corticosteroids include, but are not limited to, natural glucocorticoids such as hydrocortisone, cortisone acetate, and fludrocortisone acetate; and artificially synthesized glucocorticoids such as dexamethasone, betamethasone, prednisolone, methylprednisolone, and triamcinolone.
培地中における、副腎皮質ホルモン類の濃度は、下垂体プラコード及び/又はラトケ嚢から、下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化を促進し得る限り特に限定されず、また、副腎皮質ホルモン類の種類により適宜設定することができる、例えば、ハイドロコルチゾンの場合、通常100ng/ml以上、好ましくは、1μg/ml以上である。下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化に悪影響がない限りハイドロコルチゾン濃度の上限値は特にないが、培養コストの観点から、通常1000μg/ml以下、好ましくは100μg/ml以下である。一態様において、培地中のハイドロコルチゾン濃度は、通常約100ng/ml~約1000μg/ml、好ましくは約1~約100μg/mlである。副腎皮質ホルモン類として、デキサメサゾンを使用する場合、その培地中の濃度は、ハイドロコルチゾンの1/25程度とすることが出来る。 The concentration of the adrenal cortical hormones in the medium is not particularly limited as long as it can promote differentiation from the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells), and can be appropriately set depending on the type of adrenal cortical hormone. For example, in the case of hydrocortisone, it is usually 100 ng/ml or more, preferably 1 μg/ml or more. As long as there is no adverse effect on differentiation into pituitary hormone-producing cells (excluding ACTH-producing cells), there is no particular upper limit for the hydrocortisone concentration, but from the viewpoint of culture costs, it is usually 1000 μg/ml or less, preferably 100 μg/ml or less. In one embodiment, the hydrocortisone concentration in the medium is usually about 100 ng/ml to about 1000 μg/ml, preferably about 1 to about 100 μg/ml. When dexamethasone is used as the adrenal cortical hormone, its concentration in the medium can be about 1/25 of that of hydrocortisone.
工程(2)及び以降の工程において、培地に副腎皮質ホルモン類を添加する時期は、下垂体プラコード及び/又はラトケ嚢から、下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化を促進し得る限り特に限定されず、第二工程開始時から培地に副腎皮質ホルモン類を添加してもよいし、第二工程開始後、副腎皮質ホルモン類を添加しない培地中で一定期間培養後、培地に副腎皮質ホルモン類を添加してもよい。好適には、第二工程を開始後、細胞集団中に、ACTH産生細胞の出現が確認された段階で、培地に副腎皮質ホルモン類を添加する。即ち、細胞凝集塊中に、ACTH産生細胞の出現が確認されるまでは、細胞凝集塊を、副腎皮質ホルモン類を添加しない培地中で培養し、ACTH産生細胞の出現が確認された後に、副腎皮質ホルモン類を含む培地中で第二工程乃至以降の工程を継続する。ACTH産生細胞の出現は、ACTHに対する抗体を用いて免疫組織学的染色により確認することが出来る。ヒト多能性幹細胞を用いた場合、一般的に、第一工程開始から30日以降であれば、ACTH産生細胞の出現が期待できるので、一態様において第一工程開始から30日以降に、培地に副腎皮質ホルモン類を添加する。 In step (2) and the subsequent steps, the timing of adding the corticosteroids to the medium is not particularly limited as long as it can promote differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells). The corticosteroids may be added to the medium from the start of the second step, or the corticosteroids may be added to the medium after a certain period of culture in a medium without corticosteroids after the start of the second step. Preferably, the corticosteroids are added to the medium when the appearance of ACTH-producing cells is confirmed in the cell population after the start of the second step. That is, until the appearance of ACTH-producing cells is confirmed in the cell aggregate, the cell aggregate is cultured in a medium without corticosteroids, and after the appearance of ACTH-producing cells is confirmed, the second step and the subsequent steps are continued in a medium containing corticosteroids. The appearance of ACTH-producing cells can be confirmed by immunohistological staining using an antibody against ACTH. When human pluripotent stem cells are used, the appearance of ACTH-producing cells can generally be expected 30 days or more after the start of the first step, so in one embodiment, corticosteroids are added to the medium 30 days or more after the start of the first step.
細胞凝集塊を副腎皮質ホルモン類で処理する期間は、下垂体プラコード及び/又はラトケ嚢から、下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化を促進し得る限り特に限定されないが、通常、副腎皮質ホルモン類非処理群と比較して、副腎皮質ホルモン類処理群において、下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化の促進が確認されるまで、細胞凝集塊を副腎皮質ホルモン類で処理する。処理期間は、通常、7日以上、好ましくは12日以上である。処理期間の上限値は、特に限定されないが、副腎皮質ホルモン類非処理群と比較して、副腎皮質ホルモン類処理群において、下垂体ホルモン産生細胞(但し、ACTH産生細胞を除く)への分化の促進が確認された段階で、培地から副腎皮質ホルモン類を除去してもよい。 The period for treating the cell aggregate with the corticosteroids is not particularly limited as long as it is possible to promote differentiation of the pituitary placode and/or Rathke's pouch into pituitary hormone-producing cells (excluding ACTH-producing cells), but the cell aggregate is usually treated with the corticosteroids until promotion of differentiation into pituitary hormone-producing cells (excluding ACTH-producing cells) is confirmed in the corticosteroid-treated group compared to the corticosteroid-untreated group. The treatment period is usually 7 days or more, preferably 12 days or more. The upper limit of the treatment period is not particularly limited, but the corticosteroids may be removed from the medium at the stage when promotion of differentiation into pituitary hormone-producing cells (excluding ACTH-producing cells) is confirmed in the corticosteroid-treated group compared to the corticosteroid-untreated group.
工程(2)及び以降の工程において、下垂体への分化及び成長ホルモン産生細胞への分化を促進する観点から、レチノイン酸シグナル伝達経路作用物質の存在下で行うこともまた好ましい。レチノイン酸伝達経路作用物質としては、例えばレチノイン酸受容体(RAR)又はレチノイドX受容体(RXR)に結合し、下流の転写を活性化させる物質等が挙げられる。上記のような作用を有する化合物としては、例えばオールトランスレチノイン酸、イソトレチノイン、9-cisレチノイン酸、TTNPB(4-[(E)-2-[(5,5,8,8-Tetramethyl-5,6,7,8-tetrahydronaphthalene)-2-yl]-1-propenyl]benzoic acid)、Ch55(4-[(E)-3-(3,5-di-tert-butylphenyl)-3-oxo-1-propenyl]benzoic acid)、EC19(3-[2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)ethynyl]benzoic acid)、EC23(4-[2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)ethynyl]-benzoicacid)、Fenretinide(4-hydroxyphenylretinamide)、Acitretin((all-e)-9-(4-methoxy-2,3,6-trimethylphenyl)-3,7-dimethyl-2,4,6,8-nonatetraen)、Trifarotene、Adapalene、AC 261066(4-[4-(2-Butoxyethoxy-)-5-methyl-2-thiazolyl]-2-fluorobenzoicacid)、AC 55649(4-N-Octylbiphenyl-4-carboxylic acid)、AM 580(4-[(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carboxamido]benzoic acid)、AM80(4-[(5,5,8,8-Tetramethyl-6,7-dihydronaphthalen-2-yl)carbamoyl]benzoic acid)、BMS 753(4-[[(2,3-Dihydro-1,1,3,3-tetramethyl-2-oxo-1H-inden-5-yl)carbonyl]amino]benzoicacid)、BMS 961(3-Fluoro-4-[(r)-2-hydroxy-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-acetylamino]-benzoic acid)、CD1530(4-(6-Hydroxy-7-tricyclo[3.3.1.13,7]dec-1-yl-2-naphthalenyl)benzoicacid)、CD2314(5-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-3-thiophenecarboxylic acid)、CD437(2-naphthalenecarboxylicacid,6-(4-hydroxy-3-tricyclo(3.3.1.1(3,7))dec-1-ylphen))、CD271(6-[3-(1-Adamantyl)-4-methoxyphenyl]-2-naphthalene carboxylic acid)及びこれらの誘導体等が挙げられる。これらの物質は単独又は組み合わせて用いてもよい。 From the viewpoint of promoting differentiation into pituitary gland and growth hormone-producing cells, it is also preferable to carry out step (2) and the subsequent steps in the presence of a substance that acts on the retinoic acid signaling pathway. Examples of substances that act on the retinoic acid signaling pathway include substances that bind to retinoic acid receptors (RAR) or retinoid X receptors (RXR) and activate downstream transcription. Examples of compounds that have the above-mentioned effects include all-trans retinoic acid, isotretinoin, 9-cis retinoic acid, TTNPB (4-[(E)-2-[(5,5,8,8-Tetramethyl-5,6,7,8-tetrahydronaphthalene)-2-yl]-1-propenyl]benzoic acid), Ch55 (4-[(E)-3-(3,5 -di-tert-butylphenyl)-3-oxo-1-propenyl]benzoic acid), EC19(3-[2-(5,6,7,8-Tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)e thynyl]benzoic acid), EC23(4-[2-(5,6,7,8-Tetrahydro-5,5,8 , 8-tetramethyl-2-naphthalenyl)ethynyl]-benzoicacid), Fenretinide (4-hydroxyphenylretinamide), Acitretin ((all-e)-9-(4-metho xy-2,3,6-trimethylphenyl)-3,7-dimethyl-2,4,6,8-nonatetra en), Trifarotene, Adapalene, AC 261066 (4-[4-(2-Butoxyethoxy-)-5-methyl-2-thiazolyl]-2-fluorobenzoic acid), AC 55649 (4-N-Oc tylbiphenyl-4-carboxylic acid), AM 580(4-[(5,6,7,8-Tetrahydr o-5,5,8,8-tetramethyl-2-naphthalenyl)carboxamide]benzoic acid), AM80(4-[(5,5,8,8-Tetramethyl-6,7-dihydronaphthalenyl-2-yl) carbamoyl]benzoic acid), BMS 753 (4-[[(2,3-Dihydro-1,1,3,3 -tetramethyl-2-oxo-1H-inden-5-yl)carbonyl]amino]benzoicacid), BMS 961 (3-Fluoro-4-[(r)-2-hydroxy-2-(5,5,8,8-tetramethyl-5 ,6,7,8-tetrahydro-naphthalen-2-yl)-acetylamino]-benzoic a cid), CD1530 (4-(6-Hydroxy-7-tricyclo[3.3.1.13,7]dec-1-yl-2-naphthalenyl)benzoicacid), CD2314 (5-(5,6,7,8-Tetrahydro-5,5,8, 8-tetramethyl-2-anthracenyl)-3-thiophenecarboxylic acid) , CD437 (2-naphthalenecarboxylic acid, 6-(4-hydroxy-3-triclo(3.3.1.1(3,7))dec-1-ylphen)), CD271 (6-[3-(1-adamantyl)-4-methoxyphenyl]-2-naphthalene carboxylic acid) and derivatives thereof. These substances may be used alone or in combination.
工程(2)及び以降の工程におけるレチノイン酸伝達経路作用物質は、好ましくはオールトランスレチノイン酸乃至EC23を含む。培地中のレチノイン酸伝達経路作用物質の濃度は、上述の効果を達成可能な範囲であれば特に限定されないが、レチノイン酸伝達経路作用物質としてEC23を用いる場合は、例えばEC23の濃度が約10pM~約30μMであり、好ましくは約100pM~約20μMであり、より好ましくは約10nM~約10μ Mであり、さらに好ましくは約100nM~約5μMである。EC23以外のレチノイン酸伝達経路作用物質を使用する場合、上記濃度のEC23と同等のレチノイン酸伝達経路作用活性を示す濃度で用いられることが望ましい。 The retinoic acid pathway acting substance in step (2) and the subsequent steps preferably includes all-trans retinoic acid or EC23. The concentration of the retinoic acid pathway acting substance in the medium is not particularly limited as long as it is within a range in which the above-mentioned effects can be achieved, but when EC23 is used as the retinoic acid pathway acting substance, the concentration of EC23 is, for example, about 10 pM to about 30 μM, preferably about 100 pM to about 20 μM, more preferably about 10 nM to about 10 μM, and even more preferably about 100 nM to about 5 μM. When a retinoic acid pathway acting substance other than EC23 is used, it is desirable to use it at a concentration that exhibits retinoic acid pathway acting activity equivalent to that of EC23 at the above-mentioned concentration.
分化傾向の調節の観点からは、工程(2)及び以降の工程をNotchシグナル伝達経路阻害物質の存在下で行うこともまた好ましい。本明細書において、Notchシグナル伝達経路とは、細胞膜上に発現する受容体であるNotchタンパク質と隣接細胞の膜上に発現するNotchリガンド(Delta、Jagged等)との直接相互作用により活性化されるシグナル伝達経路を表す。Notchシグナルが伝達された細胞においては、Notchタンパク質が段階的にプロセシングを受け膜上で切り出された細胞内ドメインが核内へと運ばれて下流遺伝子の発現を制御する。 From the viewpoint of regulating differentiation tendency, it is also preferable to carry out step (2) and the subsequent steps in the presence of a Notch signaling pathway inhibitor. In this specification, the Notch signaling pathway refers to a signaling pathway that is activated by direct interaction between the Notch protein, which is a receptor expressed on the cell membrane, and the Notch ligand (Delta, Jagged, etc.) expressed on the membrane of an adjacent cell. In cells to which the Notch signal is transmitted, the Notch protein is processed in stages, and the intracellular domain cut out on the membrane is transported into the nucleus to control the expression of downstream genes.
Notchシグナル伝達経路阻害物質は、Notchにより媒介されるシグナル伝達を抑制し得るものである限り特に限定されない。核酸、タンパク質、低分子有機化合物のいずれであってもよい。当該物質として例えば、機能欠失型のNotch受容体及びリガンド、Notchのプロセシング(S1切断)を阻害する物質、Notch及びNotchリガンドの糖鎖修飾を阻害する物質、細胞膜移行を阻害する物質、Notchの細胞内ドメイン(NICD)のプロセシング(S2切断、S3切断)を阻害する物質(γセクレターゼ阻害剤)、NICDを分解する物質、NICD依存的な転写を阻害する物質等を挙げることができる。 The Notch signaling pathway inhibitor is not particularly limited as long as it can suppress signaling mediated by Notch. It may be any of nucleic acids, proteins, and low molecular weight organic compounds. Examples of such substances include functionally defective Notch receptors and ligands, substances that inhibit Notch processing (S1 cleavage), substances that inhibit glycosylation of Notch and Notch ligands, substances that inhibit cell membrane translocation, substances that inhibit processing (S2 cleavage, S3 cleavage) of the intracellular domain of Notch (NICD) (gamma secretase inhibitors), substances that degrade NICD, and substances that inhibit NICD-dependent transcription.
Notchシグナル伝達経路阻害物質として、当業者に周知の化合物を使用することもできる。Notchシグナル伝達経路阻害物質としての活性を有する化合物として、例えば、DAPT(N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester)、DBZ((2S)-2-[[2-(3,5-difluorophenyl)acetyl]amino]-N-[(7S)-5-methyl-6-oxo-7H-benzo[d][1]benzazepin-7-yl]propanamide)、MDL28170(benzyl N-[(2S)-3-methyl-1-oxo-1-[[(2S)-1-oxo-3-phenylpropan-2-yl]amino]butan-2-yl]carbamate)、FLI-06(cyclohexyl 2,7,7-trimethyl-4-(4-nitrophenyl)-5-oxo-1,4,6,8-tetrahydroquinoline-3-carboxylate)、L-685,458(tert-butyl N-[6-[[1-[(1-amino-1-oxo-3-phenylpropan-2-yl)amino]-4-methyl-1-oxopentan-2-yl]amino]-5-benzyl-3-hydroxy-6-oxo-1-phenylhexan-2-yl]carbamate)、CB-103(6-(4-tert-butylphenoxy)pyridin-3-amine)及びこれらの誘導体等、並びにOnco Targets Ther.2013;6:943-955.に記載の物質等が挙げられる。Notchシグナル伝達経路阻害物質は、好ましくはDAPTを含む。 Compounds well known to those skilled in the art can also be used as Notch signaling pathway inhibitors. Examples of compounds having activity as Notch signaling pathway inhibitors include DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester), DBZ ((2S)-2-[[2-(3,5-difluorophenyl)acetyl]amino]-N-[( 7S)-5-methyl-6-oxo-7H-benzo[d][1]benzazepin-7-yl]propanamide), MDL28170 (benzyl N-[(2S)-3-methyl-1-oxo-1-[[(2S)-1-o xo-3-phenylpropan-2-yl]amino]butan-2-yl]carbamate), FLI- 06 (cyclohexyl 2,7,7-trimethyl-4-(4-nitrophenyl)-5-oxo-1,4,6,8-tetrahydroquinoline-3-carboxylate), L-685,458 (tert-butyl N -[6-[[1-[(1-amino-1-oxo-3-phenylpropan-2-yl)amino ]-4-methyl-1-oxopentan-2-yl]amino]-5-benzyl-3-hydroxy-6-oxo-1-phenylhexan-2-yl]carbamate), CB-103 (6-(4-tert-butylphenoxy)pyridin-3-amine) and derivatives thereof, as well as substances described in Onco Targets Ther. 2013; 6: 943-955. The Notch signaling pathway inhibitor preferably includes DAPT.
培地中におけるNotchシグナル伝達経路阻害物質の濃度は、上述の効果を達成可能な範囲であれば特に限定されないが、例えば、Notchシグナル伝達経路阻害物質としてDAPTを用いる場合は、例えば、DAPTの濃度が約100pM~約50μMであり、好ましくは約1nM~約30μMであり、より好ましくは約100nM~約20μMであり、さらに好ましくは約1μM~約10μMである。DAPT以外のNotchシグナル伝達経路阻害物質を使用する場合、上記濃度のDAPTと同等のNotchシグナル伝達経路阻害活性を示す濃度で用いられることが望ましい。 The concentration of the Notch signaling pathway inhibitor in the medium is not particularly limited as long as it is within a range in which the above-mentioned effects can be achieved, but for example, when DAPT is used as the Notch signaling pathway inhibitor, the concentration of DAPT is, for example, about 100 pM to about 50 μM, preferably about 1 nM to about 30 μM, more preferably about 100 nM to about 20 μM, and even more preferably about 1 μM to about 10 μM. When using a Notch signaling pathway inhibitor other than DAPT, it is desirable to use it at a concentration that exhibits Notch signaling pathway inhibitory activity equivalent to that of DAPT at the above-mentioned concentration.
<工程(3)>、<工程(3’)>:第三工程
第三工程は、ソニック・ヘッジホッグシグナル(Shh)伝達経路作用物質の非添加条件下で、工程(2)又は工程(b)で培養した細胞集団を培養する。第二工程で得られた細胞集団をShhシグナル伝達経路作用物質の非存在下で培養し、下垂体組織を含む細胞集団を得る第三工程を工程(3)、b工程で得られた細胞集団をShhシグナル伝達経路作用物質の非存在下で培養し、下垂体組織を含む細胞集団を得る第三工程を工程(3’)とする。工程(2)又は工程(b)で細胞を浮遊培養している場合は、第三工程でも引き続き形成された細胞凝集体を浮遊培養すればよい。工程(2)又は工程(b)で細胞を接着培養している場合は、第三工程でも引き続き細胞を接着培養すればよい。工程(2)又は工程(b)で細胞を浮遊培養した後、第三工程で接着培養してもよい。
<Step (3)>, <Step (3')>: Third Step In the third step, the cell population cultured in step (2) or step (b) is cultured under conditions in which a substance acting on the Sonic Hedgehog signaling (Shh) pathway is not added. The third step of culturing the cell population obtained in the second step in the absence of a substance acting on the Shh signaling pathway to obtain a cell population containing pituitary tissue is called step (3), and the third step of culturing the cell population obtained in step b in the absence of a substance acting on the Shh signaling pathway to obtain a cell population containing pituitary tissue is called step (3'). When the cells are cultured in suspension in step (2) or step (b), the formed cell aggregates may be continuously cultured in suspension in the third step. When the cells are cultured in adhesion in step (2) or step (b), the cells may be continuously cultured in adhesion in the third step. After the cells are cultured in suspension in step (2) or step (b), they may be cultured in adhesion in the third step.
第三工程において用いられる培地は、Shhシグナル伝達経路作用物質を含有しない限り特に限定されない。本工程におけるShhシグナル伝達経路作用物質の非添加条件とは、意図的にShhシグナル伝達経路作用物質を細胞集団の培養環境に添加しない条件を表し、細胞集団による自己分泌等で意図せずShhシグナル伝達経路作用物質が培養環境中に含まれる場合も、Shhシグナル伝達経路作用物質の非添加条件に含まれる。第三工程において用いられる培地としては、第一工程に挙げた培地や10%乃至20%のKSRを含有するgfCDM培地等が挙げられる。 The medium used in the third step is not particularly limited as long as it does not contain a substance acting on the Shh signaling pathway. The condition of not adding a substance acting on the Shh signaling pathway in this step refers to a condition in which a substance acting on the Shh signaling pathway is not intentionally added to the culture environment of the cell population, and also includes a case in which a substance acting on the Shh signaling pathway is unintentionally included in the culture environment due to autocretion by the cell population, etc. Examples of the medium used in the third step include the medium listed in the first step and gfCDM medium containing 10% to 20% KSR.
第三工程及び以降の工程において、下垂体を構成する細胞の生存及び分化成熟を促進する観点から、細胞凝集体をゲル中に包埋して培養する工程を含んでもよい。ゲルとしては、例えばアガロース、メチルセルロース、コラーゲン、マトリゲル等を用いたゲルが挙げられ、マトリゲルを用いることが好ましい。 The third and subsequent steps may include a step of embedding the cell aggregates in a gel and culturing them in order to promote the survival and differentiation/maturation of the cells that make up the pituitary gland. Examples of gels include gels made of agarose, methylcellulose, collagen, matrigel, etc., and it is preferable to use matrigel.
第三工程及び以降の工程のゲル中に包埋して細胞を培養する工程において、細胞凝集体をそのまま包埋してもよいし、分散及び単離した後の細胞をゲル中に播種してもよい。セルソーター等を用いて基底細胞等特定の細胞種を分取した後に播種してもよい。ゲル中に包埋する培養法のさらなる一態様として、線維芽細胞、間葉系細胞、血管系の細胞等の下垂体以外の細胞との共培養を実施することもできる。上記のようなゲル包埋培養は、例えば、Nature 501,373-379(2013)、Nature,499,481-484(2013)、Nat Protoc 14,518-540(2019)、Genes 2020,11,603等を参照して実施することができる。
In the third and subsequent steps of culturing cells by embedding them in a gel, the cell aggregates may be embedded as they are, or the cells may be seeded in the gel after being dispersed and isolated. Specific cell types such as basal cells may be separated using a cell sorter or the like and then seeded. As a further embodiment of the culture method of embedding cells in a gel, co-culture with cells other than the pituitary gland, such as fibroblasts, mesenchymal cells, and vascular cells, may also be performed. The gel-embedded culture described above may be performed by referring to, for example,
第三工程及び以降の工程において、細胞への栄養及び酸素供給を改善し、物質交換を改善する目的から、細胞が物理的に揺動される培養方法を実施することも好ましい。このような培養方法としては、振盪培養、回転培養、攪拌培養等の静置培養以外の方法が挙げられる。振盪培養、回転培養、攪拌培養等を実施するための手段は特に限定されないが、例えば、細胞を培養している培養器材をローテーター、シェーカー等に設置する、又は細胞をスターラー等が回転している環境下に置くことにより実施することができる。振盪培養、回転培養、攪拌培養の速度等のパラメーターは当業者であれば細胞への傷害を生じない範囲で適宜設定可能である。例えば波動形揺動の3Dシェーカー(例えば、Mini-Shaker 3D、Biosan社製)を用いて振盪培養を実施する場合は、例えば5~60rpm、好ましくは5~40rpm、より好ましくは5~20rpmの範囲で振盪速度範囲を設定可能である。往復式のシェーカー(例えばNS-LR、アズワン社製)を用いて振盪培養を実施する場合は、例えば15~60rpm、好ましくは15~50rpm、より好ましくは15~45rpmの範囲で振盪速度範囲を設定可能である。シーソー式のシェーカー(例えばNS-S、アズワン社製)を用いて振盪培養を実施する場合は、例えば、5~50rpm、好ましくは5~40rpm、より好ましくは5~30rpmの範囲で振盪速度範囲を設定可能である。細胞凝集体を攪拌培養、回転培養で培養する場合は、例えばスピナーフラスコ(例えば、3152、コーニング社製)をマグネティックスターラー上に設置し、細胞凝集体が目視で沈降しない程度の回転数で培養を実施することもできる。三次元回転浮遊培養装置(例えば、CellPet CUBE、ジェイテック社製;Clinostar、Celvivo社製)を用いて培養を実施することもできる。細胞への摩擦等の物理的な傷害を抑制する観点から、前記のゲルに包埋した細胞凝集体を振盪培養、回転培養又は攪拌培養することもまた好ましい。 In the third and subsequent steps, it is also preferable to carry out a culture method in which the cells are physically shaken in order to improve the supply of nutrients and oxygen to the cells and to improve material exchange. Such culture methods include methods other than stationary culture, such as shaking culture, rotary culture, and agitation culture. The means for carrying out shaking culture, rotary culture, agitation culture, and the like are not particularly limited, but for example, shaking culture, rotary culture, agitation culture, and the like can be carried out by placing the culture equipment in which the cells are cultured on a rotator, shaker, or the like, or placing the cells in an environment in which a stirrer or the like is rotating. Those skilled in the art can appropriately set parameters such as the speed of shaking culture, rotary culture, and agitation culture within a range that does not cause damage to the cells. For example, when shaking culture is carried out using a 3D shaker with a wave-type agitation (e.g., Mini-Shaker 3D, manufactured by Biosan), the shaking speed range can be set, for example, to 5 to 60 rpm, preferably 5 to 40 rpm, and more preferably 5 to 20 rpm. When shaking culture is performed using a reciprocating shaker (e.g., NS-LR, manufactured by AS ONE), the shaking speed range can be set, for example, in the range of 15 to 60 rpm, preferably 15 to 50 rpm, and more preferably 15 to 45 rpm. When shaking culture is performed using a seesaw shaker (e.g., NS-S, manufactured by AS ONE), the shaking speed range can be set, for example, in the range of 5 to 50 rpm, preferably 5 to 40 rpm, and more preferably 5 to 30 rpm. When culturing cell aggregates by stirring culture or rotary culture, for example, a spinner flask (e.g., 3152, manufactured by Corning) can be placed on a magnetic stirrer and culture can be performed at a rotation speed at which the cell aggregates do not settle with the naked eye. Culture can also be performed using a three-dimensional rotary suspension culture device (e.g., CellPet CUBE, manufactured by J-Tech; Clinostar, manufactured by Celvivo). From the viewpoint of suppressing physical damage to the cells, such as friction, it is also preferable to subject the cell aggregates embedded in the gel to shaking culture, rotation culture, or stirring culture.
第三工程及び以降の工程において、細胞死を抑制し、細胞の増殖を促進する観点から、高酸素の雰囲気下で培養することもまた好ましい。培養過程における高酸素条件は、例えば細胞を培養するインキュベーターに酸素ボンベを接続し、人工的に酸素を供給することにより実現できる。かかる目的での酸素濃度は、通常25%~80%であり、より好ましくは30%~60%である。 In the third and subsequent steps, it is also preferable to culture in a high-oxygen atmosphere from the viewpoint of suppressing cell death and promoting cell proliferation. High-oxygen conditions during the culture process can be achieved, for example, by connecting an oxygen tank to the incubator in which the cells are cultured and artificially supplying oxygen. The oxygen concentration for this purpose is usually 25% to 80%, and more preferably 30% to 60%.
第三工程及び以降の工程において、細胞凝集体を培養する培地中への酸素供給量を増やす観点から、ガス交換効率の高い培養器材を用いることもできる。このような培養器材の例として、細胞培養ディッシュ、プレートの底面をガス透過性のフイルムとしたLumoxディッシュ(ザルスタット株式会社製)、VECELL 96well plate(株式会社ベセル製)等が挙げられる。前述した高酸素濃度条件下での培養と組み合わせて用いることもまた好ましい。 In the third and subsequent steps, culture equipment with high gas exchange efficiency can be used to increase the amount of oxygen supplied to the medium in which the cell aggregates are cultured. Examples of such culture equipment include cell culture dishes, Lumox dishes (manufactured by Sarstedt Co., Ltd.) with a gas-permeable film on the bottom of the plate, and VECELL 96-well plates (manufactured by Vecel Co., Ltd.). It is also preferable to use this in combination with the culture under high oxygen concentration conditions described above.
第三工程及び以降の工程において、細胞凝集体中の非神経上皮組織の構造を維持する観点から、細胞保護剤を培地に添加することもできる。第三工程及び以降の工程において用いられる細胞保護剤としては、上述したFGFシグナル伝達経路作用物質、ヘパリン、ROCK阻害物質、基底膜標品、ミオシン阻害物質、ポリアミン類、ISR阻害剤、カスパーゼ阻害剤、血清、又は血清代替物等が挙げられる。ミオシン阻害物質としては例えば非筋型ミオシンII ATPアーゼの阻害物質であるBlebbistatin、ミオシン軽鎖キナーゼ(MLCK)の阻害物質であるML-7、ML-9、W-7、MLCK inhibitor peptide 18及びこれらの誘導体等が挙げられる。添加する細胞保護剤は第一工程で添加したものと異なっていてもよいが、好ましくは同一である。好ましい細胞保護剤としては、ROCK阻害物質が挙げられる。第三工程及び以降の工程において、細胞保護剤としてROCK阻害物質であるY-27632を添加する場合は、通常約10nM~約10mM、好ましくは約100nM~約1mM、より好ましくは約1μM~約100μMの濃度となるように培養環境中に添加する。ROCK阻害物質であるChroman 1を添加する場合は通常約10pM~約1mM、好ましくは約100pM~約100μM、より好ましくは約1nM~約10μMの濃度となるように培養環境中に添加する。細胞保護剤として非筋型ミオシンII ATPアーゼの阻害物質であるBlebbistatinを添加する場合は、通常約10nM~約10mM、好ましくは約100nM~約1mM、より好ましくは約1μM~約100μMの濃度となるように培養環境中に添加する。
In the third and subsequent steps, a cell protective agent may be added to the medium from the viewpoint of maintaining the structure of the non-neuroepithelial tissue in the cell aggregate. Examples of cell protective agents used in the third and subsequent steps include the above-mentioned FGF signaling pathway active substances, heparin, ROCK inhibitors, basement membrane preparations, myosin inhibitors, polyamines, ISR inhibitors, caspase inhibitors, serum, or serum substitutes. Examples of myosin inhibitors include blebbistatin, which is an inhibitor of nonmuscle myosin II ATPase, and ML-7, ML-9, W-7, MLCK inhibitor peptide 18, and derivatives thereof, which are inhibitors of myosin light chain kinase (MLCK). The cell protective agent added may be different from that added in the first step, but is preferably the same. A preferred cell protective agent is a ROCK inhibitor. In the third and subsequent steps, when the ROCK inhibitor Y-27632 is added as a cell protective agent, it is usually added to the culture environment at a concentration of about 10 nM to about 10 mM, preferably about 100 nM to about 1 mM, and more preferably about 1 μM to about 100 μM. When the
第三工程及び以降の工程において、細胞保護剤以外の非神経上皮組織の構造を維持する作用を有する物質を添加することもできる。上記のような物質として例えば細胞接着を促進する物質、基底膜成分の合成を促進する物質、基底膜成分の分解を阻害する物質等が挙げられる。細胞接着を促進する物質は細胞-細胞間の接着、細胞-基底膜間の接着、細胞-培養器材間の接着等いずれを促進するものであってもよいし、細胞接着に関与する因子の産生を促すものであってもよい。細胞接着を促進する物質として例えばアドヘサミン、アドヘサミン-RGDS誘導体、Pyrintegrin、Biotin tripeptide-1、Acetyl Tetrapeptide-3、RGDS Peptide及びこれらの誘導体等が挙げられる。基底膜成分の合成を促進する物質として例えばアスコルビン酸誘導体等が挙げられる。アスコルビン酸誘導体としては、例えばアスコルビン酸リン酸ナトリウム、アスコルビン酸リン酸マグネシウム、アスコルビン酸2-グルコシド、3-O-エチルアスコルビン酸、テトラヘキシルデカン酸アスコルビル、パルミチン酸アスコルビル、ステアリン酸アスコルビル、アスコルビン酸-2リン酸-6パルミチン酸、グリセリルオクチルアスコルビン酸等が挙げられる。基底膜成分の分解を阻害する物質として、例えば、マトリックスメタロプロテアーゼ及びセリンプロテアーゼの阻害剤等が挙げられる。基底膜成分の合成を促進する物質であるアスコルビン酸誘導体の一種であるアスコルビン酸2-リン酸を添加する場合は、通常10μg/ml以上、1000μg/ml以下、好ましくは30μg/ml以上500μg/ml以下、さらに好ましくは50μg/ml以上300μg/ml以下の濃度となるように培養環境中に添加する。他のアスコルビン酸及びアスコルビン酸の誘導体等を添加する際は、上記の濃度とモル当量が同程度となるように添加すればよい。 In the third step and subsequent steps, a substance other than a cytoprotectant that has the effect of maintaining the structure of non-neuroepithelial tissue can also be added. Examples of such substances include substances that promote cell adhesion, substances that promote the synthesis of basement membrane components, and substances that inhibit the decomposition of basement membrane components. The substance that promotes cell adhesion may promote cell-cell adhesion, cell-basement membrane adhesion, or cell-culture equipment adhesion, or may promote the production of factors involved in cell adhesion. Examples of substances that promote cell adhesion include adhesamine, adhesamine-RGDS derivatives, pyrintegrin, biotin tripeptide-1, acetyl tetrapeptide-3, RGDS peptide, and derivatives thereof. Examples of substances that promote the synthesis of basement membrane components include ascorbic acid derivatives. Examples of ascorbic acid derivatives include sodium ascorbyl phosphate, magnesium ascorbyl phosphate, ascorbyl 2-glucoside, 3-O-ethyl ascorbic acid, ascorbyl tetrahexyldecanoate, ascorbyl palmitate, ascorbyl stearate, ascorbyl 2-phosphate-6 palmitate, and glyceryl octyl ascorbic acid. Examples of substances that inhibit the decomposition of basement membrane components include inhibitors of matrix metalloproteases and serine proteases. When ascorbic acid 2-phosphate, which is a type of ascorbic acid derivative that promotes the synthesis of basement membrane components, is added, it is usually added to the culture environment at a concentration of 10 μg/ml or more and 1000 μg/ml or less, preferably 30 μg/ml or more and 500 μg/ml or less, and more preferably 50 μg/ml or more and 300 μg/ml or less. When adding other ascorbic acid or ascorbic acid derivatives, etc., add them so that the molar equivalents are approximately the same as the above concentrations.
第三工程及び以降の工程において、下垂体細胞の生存を促進する観点から、酸化ストレスを軽減する作用を有する物質を添加することも好ましい。上記のような活性を有する物質として例えば抗酸化物質、フリーラジカルスカベンジャー作用を有する物質、NADPHオキシダーゼ阻害物質、シクロオキシゲナーゼ阻害物質、リポキシゲナーゼ(LOX)阻害物質、スーパーオキシドジスムターゼ(SOD)様物質、Nrf2活性化剤等が挙げられる。上記のような活性を有する物質として、例えば、アスコルビン酸、N-アセチル-L-システイン、酢酸(±)-α-トコフェロール、Apocynin(4’-Hydroxy-3’-methoxyacetophenone)、ニコチンアミド、タウリン(2-アミノエタンスルホン酸)、IM-93(1-Isopropyl-3-(1-methyl-1H-Indole-3-yl)-4-(N,N-dimethyl-1,3-propanediamine)-1H-Pyrrole-2、5H-dione)、Caffeic Acid(3,4-Dihydroxycinnamic Acid)、Celastrol(3-Hydroxy-24-nor-2-oxo-1(10),3,5,7-friedelatetraen-29-oic Acid;Tripterin)、Ebselen(2-Phenyl-1,2-benzisoselenazol-3(2H)-one)、(-)-Epigallocatechin Gallate((2R,3R)-2-(3,4,5-Trihydroxyphenyl)-3,4-dihydro-1[2H]-benzopyran-3,5,7-triol-3-(3,4,5-trihydroxybenzoate))、EUK-8(N,N’-Bis(salicylideneamino)ethane-manganese(II))、Edaravone(3-Methyl-1-phenyl-2-pyrazolin-5-one)、MnTBAP(Mn(III)tetrakis(4-benzoicacid)porphyrin Chloride)、Nordihydroguaiaretic Acid、Resveratrol(trans-3,4,5-Trihydroxystilbene)及びこれらの誘導体等が挙げられるが、これらに限定はされない。細胞培養用に調製済の試薬(例えば、抗酸化サプリメント、Sigma Aldrich社製、A1345)を用いることもできる。本発明で用いる酸化ストレスを軽減する作用を有する物質は、好ましくはアスコルビン酸、N-アセチル-L-システイン並びにこれらの誘導体からなる群より選ばれる少なくとも1つを含む。アスコルビン酸は、例えば、その誘導体であるアスコルビン酸2リン酸として、約1nM~約1M、好ましくは約10nM~約100mM、より好ましくは約100nM~約10mM、さらに好ましくは約1μM~約3mMの濃度で、N-アセチル-L-システインは、例えば、約1nM~約1M、好ましくは約10nM~約100mM、より好ましくは約100nM~約10mM、さらに好ましくは約1μM~約5mMの濃度で培地中に添加することができる。 In the third and subsequent steps, from the viewpoint of promoting the survival of pituitary cells, it is also preferable to add a substance that has the effect of reducing oxidative stress. Examples of substances having such activity include antioxidants, substances with free radical scavenging action, NADPH oxidase inhibitors, cyclooxygenase inhibitors, lipoxygenase (LOX) inhibitors, superoxide dismutase (SOD)-like substances, Nrf2 activators, etc. Examples of substances having the above-mentioned activity include ascorbic acid, N-acetyl-L-cysteine, (±)-α-tocopherol acetate, apocynin (4'-hydroxy-3'-methoxyacetophenone), nicotinamide, taurine (2-aminoethanesulfonic acid), IM-93 (1-isopropyl-3-(1-methyl-1H-indole-3-yl)-4-(N,N-dimethyl-1,3-pr Celastrol (3-Hydroxy-24-nor-2-oxo-1 (10), 3,5, 7-friedelatetraen-29-oic Acid; Tripterin), Ebselen (2-Phenyl-1, 2-benzisoselenazol-3 (2H)-one), (-)-Epigallocatechin Gallate ((2R,3R)-2-(3,4,5-Trihydroxyphenyl)-3,4-dihydro-1[2H]-benzopyran-3,5,7-triol-3-(3, 4,5-trihydroxybenzoate)), EUK-8(N,N'-Bis(salicylideneamino)ethane-manganese(II)), E Examples of such antioxidants include, but are not limited to, daravonone (3-methyl-1-phenyl-2-pyrazolin-5-one), MnTBAP (Mn(III) tetrakis(4-benzoic acid) porphyrin chloride), Nordihydroguaiaretic Acid, Resveratrol (trans-3,4,5-Trihydroxystilbene) and their derivatives. Reagents already prepared for cell culture (e.g., antioxidant supplements, Sigma Aldrich, A1345) can also be used. The substance having the effect of reducing oxidative stress used in the present invention preferably includes at least one selected from the group consisting of ascorbic acid, N-acetyl-L-cysteine and their derivatives. Ascorbic acid can be added to the medium, for example, in the form of its derivative ascorbic acid 2-phosphate, at a concentration of about 1 nM to about 1 M, preferably about 10 nM to about 100 mM, more preferably about 100 nM to about 10 mM, and even more preferably about 1 μM to about 3 mM, and N-acetyl-L-cysteine can be added to the medium at a concentration of, for example, about 1 nM to about 1 M, preferably about 10 nM to about 100 mM, more preferably about 100 nM to about 10 mM, and even more preferably about 1 μM to about 5 mM.
第三工程及び以降の工程において、下垂体細胞の生存を促進する観点から、ストレス応答シグナル伝達経路に対する阻害物質(ストレスに対する細胞内シグナル伝達機構を阻害する物質)を添加することもまた好ましい。ストレス応答性MAPキナーゼ経路(stress-activated protein kinase:SAPK)は、ストレスに対する細胞内シグナル伝達機構の主要なものの一つである。ストレス応答性MAPキナーゼ経路の阻害剤としては、例えば、MAP3K阻害剤、MAP2K阻害剤、ASK阻害剤、MEK阻害剤、Akt阻害剤、Rhoファミリーキナーゼ阻害剤、JNK阻害剤、p38阻害剤、MSK阻害剤、STAT阻害剤、NF-κB阻害剤、CAMK阻害剤等が挙げられる。 In the third and subsequent steps, from the viewpoint of promoting survival of pituitary cells, it is also preferable to add an inhibitor of the stress-responsive signaling pathway (a substance that inhibits the intracellular signaling mechanism in response to stress). The stress-responsive MAP kinase pathway (stress-activated protein kinase: SAPK) is one of the main intracellular signaling mechanisms in response to stress. Examples of inhibitors of the stress-responsive MAP kinase pathway include MAP3K inhibitors, MAP2K inhibitors, ASK inhibitors, MEK inhibitors, Akt inhibitors, Rho family kinase inhibitors, JNK inhibitors, p38 inhibitors, MSK inhibitors, STAT inhibitors, NF-κB inhibitors, and CAMK inhibitors.
MEK阻害剤としては、例えば、Selumetinib(AZD6244,6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide)、Mirdametinib(PD0325901,N-[(2R)-2,3-dihydroxypropoxy]-3,4-difluoro-2-(2-fluoro-4-iodoanilino)benzamide)、Trametinib(GSK1120212,N-[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide)、U0126(1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene)、PD184352(CI-1040,2-(2-chloro-4-iodoanilino)-N-(cyclopropylmethoxy)-3,4-difluorobenzamide)、PD98059(2-(2-amino-3-methoxyphenyl)chromen-4-one)、BIX 02189(3-[N-[3-[(dimethylamino)methyl]phenyl]-C-phenylcarbonimidoyl]-2-hydroxy-N,N-dimethyl-1H-indole-6-carboxamide)、Pimasertib(AS-703026,N-[(2S)-2,3-dihydroxypropyl]-3-(2-fluoro-4-iodoanilino)pyridine-4-carboxamide)、Pelitinib(EKB-569,(E)-N-[4-(3-chloro-4-fluoroanilino)-3-cyano-7-ethoxyquinolin-6-yl]-4-(dimethylamino)but-2-enamide)、BIX 02188(3-[N-[3-[(dimethylamino)methyl]phenyl]-C-phenylcarbonimidoyl]-2-hydroxy-1H-indole-6-carboxamide)、TAK-733(3-[(2R)-2,3-dihydroxypropyl]-6-fluoro-5-(2-fluoro-4-iodoanilino)-8-methylpyrido[2,3-d]pyrimidine-4,7-dione)、AZD8330(2-(2-fluoro-4-iodoanilino)-N-(2-hydroxyethoxy)-1,5-dimethyl-6-oxopyridine-3-carboxamide)、Binimetinib(MEK162,6-(4-bromo-2-fluoroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide)、SL-327((Z)-3-amino-3-(4-aminophenyl)sulfanyl-2-[2-(trifluoromethyl)phenyl]prop-2-enenitrile)、Refametinib(RDEA119,N-[3,4-difluoro-2-(2-fluoro-4-iodoanilino)-6-methoxyphenyl]-1-[(2S)-2,3-dihydroxypropyl]cyclopropane-1-sulfonamide)、GDC-0623(5-(2-fluoro-4-iodoanilino)-N-(2-hydroxyethoxy)imidazo[1,5-a]pyridine-6-carboxamide)、BI-847325(3-[3-[N-[4-[(dimethylamino)methyl]phenyl]-C-phenylcarbonimidoyl]-2-hydroxy-1H-indol-6-yl]-N-ethylprop-2-ynamide)、RO5126766(CH5126766,3-[[3-fluoro-2-(methylsulfamoylamino)pyridin-4-yl]methyl]-4-methyl-7-pyrimidin-2-yloxychromen-2-one)、Cobimetinib(GDC-0973,[3,4-difluoro-2-(2-fluoro-4-iodoanilino)phenyl]-[3-hydroxy-3-[(2S)-piperidin-2-yl]azetidin-1-yl]methanone)及びこれらの誘導体等が挙げられる。 Examples of MEK inhibitors include Selumetinib (AZD6244, 6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide), Mirdametinib (PD0325901, N-[(2R)-2,3-dihydroxypropoxy]-3,4-difluoro-2-(2-fluoro-4-iodoanilino)benzamide), Trametinib (GSK1120212, N -[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide), U012 6(1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene), PD184352(CI-1040,2-(2-chloro-4-iodoanilino)-N-(cyclopropyl methoxy)-3,4-dif luorobenzamide), PD98059 (2-(2-amino-3-methoxyphenyl)chromen-4-one), BIX 02189 (3-[N-[3-[(dimethylamino)methyl]phenyl]-C-ph enylcarbonimidoyl]-2-hydroxy-N,N-dimethyl-1H-indole-6-carboxamide), Pimasertib (AS-703026, N-[(2S)-2,3-dihydroxypropyl]-3-(2 -fluoro-4-iodoanil ino)pyridine-4-carboxamide), Pelitinib (EKB-569, (E)-N-[4-(3-chloro-4-fluoroanilino)-3-cyano-7-ethoxyquinolin-6-yl]-4-(dim) but-2-enamide), BIX 02188 (3-[N-[3-[(dimethylamino)methyl]phenyl]-C-phenylcarbonimidoyl]-2-hydroxy-1H-indole-6-c arboxamide), TAK-73 3(3-[(2R)-2,3-dihydroxypropyl]-6-fluoro-5-(2-fluoro-4-iodoanilino)-8-methylpyrido[2,3-d]pyrimidine-4,7-dione), AZD8330(2 -(2-fluoro-4-iodoanilino)-N-(2-hydroxyethoxy)-1,5-dimethyl-6-oxopyridine-3-carboxamide), Binimetinib (MEK162,6-(4-bromo-2-f luoroanilino)-7-f luoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide), SL-327((Z)-3-amino-3-(4-aminophenyl)sulfanyl-2-[2-(t rifluoromethyl) phenyl]prop-2-enenitrile), Refametinib (RDEA119,N-[3,4-difluoro-2-(2-fluoro-4-iodoanilino)-6-methoxyphenyl]- 1-[(2S)-2,3-dihydroxyp [ropyl]cyclopropane-1-sulfonamide), GDC-0623 (5-(2-fluoro-4-iodoanilino)-N-(2-hydroxyethoxy)imidazo[1,5-a]pyridine-6-carbo xamide), BI-847325 (3-[3-[N-[4-[(dimethylamino)methyl]phenyl]-C-phenylcarbonimidoyl]-2-hydroxy-1H-indol-6-yl]-N-ethylprop-2 -ynamide), RO512676 6 (CH5126766, 3-[[3-fluoro-2-(methylsulfamoylamino)pyridin-4-yl]methyl]-4-methyl-7-pyrimidin-2-yloxychromen-2-one), cobimetinib (GDC-0973, [3,4-difluoro-2-(2-fluoro-4-iodoanilino)phenyl]-[3-hydroxy-3-[(2S)-piperidin-2-yl]azetidin-1-yl]methaneone) and their derivatives.
p38阻害剤としては、例えば、SB203580(4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine)、Doramapimod(BIRB 796,1-[5-tert-butyl-2-(4-methylphenyl)pyrazol-3-yl]-3-[4-(2-morpholin-4-ylethoxy)naphthalen-1-yl]urea)、SB202190(FHPI,4-[4-(4-fluorophenyl)-5-pyridin-4-yl-1H-imidazol-2-yl]phenol)、Ralimetinib dimesylate(5-[2-tert-butyl-4-(4-fluorophenyl)-1H-imidazol-5-yl]-3-(2,2-dimethylpropyl)imidazo[4,5-b]pyridin-2-amine;methanesulfonic acid)、VX-702(6-(N-carbamoyl-2,6-difluoroanilino)-2-(2,4-difluorophenyl)pyridine-3-carboxamide)、PH-797804(3-[3-bromo-4-[(2,4-difluorophenyl)methoxy]-6-methyl-2-oxopyridin-1-yl]-N,4-dimethylbenzamide)、Neflamapimod(VX-745,5-(2,6-dichlorophenyl)-2-(2,4-difluorophenyl)sulfanylpyrimido[1,6-b]pyridazin-6-one)、TAK-715(N-[4-[2-ethyl-4-(3-methylphenyl)-1,3-thiazol-5-l]pyridin-2-yl]benzamide)、PD 169316(4-[4-(4-fluorophenyl)-2-(4-nitrophenyl)-1H-imidazol-5-yl]pyridine)、TA-02(4-[2-(2-fluorophenyl)-4-(4-fluorophenyl)-1H-imidazol-5-yl]pyridine)、SD 0006(1-[4-[3-(4-chlorophenyl)-4-pyrimidin-4-yl-1H-pyrazol-5-yl]piperidin-1-yl]-2-hydroxyethanone)、Pamapimod(6-(2,4-difluorophenoxy)-2-(1,5-dihydroxypentan-3-ylamino)-8-methylpyrido[2,3-d]pyrimidin-7-one)、BMS-582949(4-[5-(cyclopropylcarbamoyl)-2-methylanilino]-5-methyl-N-propylpyrrolo[2,1-f][1,2,4]triazine-6-carboxamide)、SB239063(4-[4-(4-fluorophenyl)-5-(2-methoxypyrimidin-4-yl)imidazol-1-yl]cyclohexan-1-ol)、Skepinone-L(13-(2,4-difluoroanilino)-5-[(2R)-2,3-dihydroxypropoxy]tricyclo[9.4.0.03,8]pentadeca-1(11),3(8),4,6,12,14-hexaen-2-one)、DBM 1285(N-cyclopropyl-4-[4-(4-fluorophenyl)-2-piperidin-4-yl-1,3-thiazol-5-yl]pyrimidin-2-amine;dihydrochloride)、SB 706504(1-cyano-2-[2-[[8-(2,6-difluorophenyl)-4-(4-fluoro-2-methylphenyl)-7-oxopyrido[2,3-d]pyrimidin-2-yl]amino]ethyl]guanidine)、SCIO 469(2-[6-chloro-5-[(2R,5S)-4-[(4-fluorophenyl)methyl]-2,5-dimethylpiperazine-1-carbonyl]-1-methylindol-3-yl]-N,N-dimethyl-2-oxoacetamide)、Pexmetinib(1-[5-tert-butyl-2-(4-methylphenyl)pyrazol-3-yl]-3-[[5-fluoro-2-[1-(2-hydroxyethyl)indazol-5-yl]oxyphenyl]methyl]urea)、UM-164(2-[[6-[4-(2-hydroxyethyl)piperazin-1-yl]-2-methylpyrimidin-4-yl]amino]-N-[2-methyl-5-[[3-(trifluoromethyl)benzoyl]amino]phenyl]-1,3-thiazole-5-carboxamide)、p38 MAPK Inhibitor(4-(2,4-difluorophenyl)-8-(2-methylphenyl)-7-oxido-1,7-naphthyridin-7-ium)、p38 MAP Kinase Inhibitor III(4-[5-(4-fluorophenyl)-2-methylsulfanyl-1H-imidazol-4-yl]-N-(1-phenylethyl)pyridin-2-amine)、p38 MAP Kinase Inhibitor IV(3,4,6-trichloro-2-(2,3,5-trichloro-6-hydroxyphenyl)sulfonylphenol)、CAY105571(4-[5-(4-fluorophenyl)-2-[4-(methylsulfonyl)phenyl]-1H-imidazol-4-yl]-pyridine)及びこれらの誘導体等が挙げられる。 Examples of p38 inhibitors include SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine), Doramapimod (BIRB 796, 1-[5-tert-butyl-2-(4-methylphenyl)pyrazo l-3-yl]-3-[4-(2-morpholin-4-ylethoxy)naphthalen-1-yl]urea), SB202190 (FHPI,4-[4-(4-fluorophenyl)-5-pyridin-4-yl-1H-imidaz ol-2-yl]phenol), Ralimetinib dimesylate (5-[2-t ert-butyl-4-(4-fluorophenyl)-1H-imidazol-5-yl]-3-(2,2-dimethylpropyl)imidazo[4,5-b]pyridin-2-amine; methanesulfonic ac id), VX-702 (6-(N-carbamoyl-2,6-difluoroanilino) -2-(2,4-difluorophenyl)pyridine-3-carboxamide), PH-797804 (3-[3-bromo-4-[(2,4-difluorophenyl)methoxy]-6-methyl-2-oxopyridin -1-yl]-N,4-dimethylbenzamide), Neflamapimod ( VX-745,5-(2,6-dichlorophenyl)-2-(2,4-difluorophenyl)sulfanylpyrimido[1,6-b]pyridazin-6-one), TAK-715(N-[4-[2-ethyl-4-(3- methylphenyl)-1,3-thiazol-5-l]pyridin-2-yl]b TA-02 (4-[2-(2-fluorophenyl)-4- (4-fluorophenyl)-1H-imidazol-5-yl]pyridine), SD 0006 (1-[4-[3-(4-chlorophenyl)-4-pyrimidin-4-yl-1H-pyrazol-5-yl]piperidin-1-yl]-2-hydroxyethanone), Pamapimod (6-(2,4-difl) urophenoxy)-2-(1,5-dihydroxypentan-3-ylamino )-8-methylpyrido[2,3-d]pyrimidin-7-one), BMS-582949 (4-[5-(cyclopropylcarbamoyl)-2-methylanilino]-5-methyl-N-propylpyrrolo[ 2,1-f][1,2,4]triazine-6-carboxamide), SB2390 63 (4-[4-(4-fluorophenyl)-5-(2-methoxypyrimidin-4-yl)imidazol-1-yl]cyclohexan-1-ol), Skepinone-L (13-(2,4-difluoroanilino) -5-[(2R)-2,3-dihydroxypropoxy]tricyclo[9.4.0 .. 03,8] pentadeca-1(11),3(8),4,6,12,14-hexaen-2-one), DBM 1285(N-cyclopropyl-4-[4-(4-fluorophenyl)-2-piperidin-4-yl-1,3-thia zol-5-yl]pyrimidin-2-amine; dihydrochloride) , SB 706504 (1-cyano-2-[2-[[8-(2,6-difluorophenyl)-4-(4-fluoro-2-methylphenyl)-7-oxopyrido[2,3-d]pyrimidin-2-yl]amino]ethyl ]guanidine), SCIO 469 (2-[6-chloro-5-[(2R,5S )-4-[(4-fluorophenyl)methyl]-2,5-dimethylpiperazine-1-carbonyl]-1-methylindol-3-yl]-N,N-dimethyl-2-oxoacetamide), Pexmet inib(1-[5-tert-butyl-2-(4-methylphenyl)pyrazo l-3-yl]-3-[[5-fluoro-2-[1-(2-hydroxyethyl)indazol-5-yl]oxyphenyl]methyl]urea), UM-164(2-[[6-[4-(2-hydroxyethyl)piperaz in-1-yl]-2-methylpyrimidin-4-yl]amino]-N-[2-me thyl-5-[[3-(trifluoromethyl)benzoyl]amino]phenyl]-1,3-thiazole-5-carboxamide), p38 MAPK Inhibitor (4-(2,4-difluorophenyl) l)-8-(2-methylphenyl)-7-oxido-1,7-naphthyridin- 7-ium), p38 MAP Kinase Inhibitor III (4-[5-(4-fluorophenyl)-2-methylsulfanyl-1H-imidazol-4-yl]-N-(1-phenylethyl)pyridin-2 -amine), p38 MAP Kinase Inhibitor IV (3,4,6-tri Examples include chloro-2-(2,3,5-trichloro-6-hydroxyphenyl)sulfonylphenol, CAY105571 (4-[5-(4-fluorophenyl)-2-[4-(methylsulfonyl)phenyl]-1H-imidazol-4-yl]-pyridine) and their derivatives.
JNK阻害剤としては、例えば、第一工程で記載したものと同様の物が挙げられる。本発明で用いるストレスに対する細胞内シグナル伝達機構を阻害する物質は、好ましくはMEK阻害剤、p38阻害剤、JNK阻害剤からなる群から選ばれる一つ以上である。p38阻害剤としてSB203580を用いる場合は、通常約1nM~約1mM、好ましくは約10nM~約100μM、より好ましくは約100nM~約10μM、さらに好ましくは約500nM~約5μMの濃度で、培地中に添加することができる。MEK阻害剤としてPD0325901を用いる場合は、通常約1nM~約1mM、好ましくは約10nM~約100μM、より好ましくは約100nM~約10μM、さらに好ましくは約500nM~約5μMの濃度で、培地中に添加することができる。JNK阻害剤としてJNK-IN-8を用いる場合は、第一工程に記載の濃度と同様の濃度で、培地中に添加することができる。他のMEK阻害剤、p38阻害剤、JNK阻害剤を用いる場合は上記阻害剤の添加濃度と同等の阻害活性を有する濃度で添加することが好ましい。 Examples of JNK inhibitors include those described in the first step. The substance used in the present invention that inhibits the intracellular signal transduction mechanism in response to stress is preferably one or more selected from the group consisting of MEK inhibitors, p38 inhibitors, and JNK inhibitors. When SB203580 is used as the p38 inhibitor, it can be added to the medium at a concentration of usually about 1 nM to about 1 mM, preferably about 10 nM to about 100 μM, more preferably about 100 nM to about 10 μM, and even more preferably about 500 nM to about 5 μM. When PD0325901 is used as the MEK inhibitor, it can be added to the medium at a concentration of usually about 1 nM to about 1 mM, preferably about 10 nM to about 100 μM, more preferably about 100 nM to about 10 μM, and even more preferably about 500 nM to about 5 μM. When JNK-IN-8 is used as the JNK inhibitor, it can be added to the medium at a concentration similar to that described in the first step. When using other MEK inhibitors, p38 inhibitors, or JNK inhibitors, it is preferable to add them at a concentration that has the same inhibitory activity as the above inhibitors.
5.下垂体組織を含む細胞集団
本発明における下垂体ホルモン産生細胞を含む細胞集団は、下垂体ホルモン産生細胞を含む細胞凝集体であってよい。
当該細胞凝集体は、下垂体ホルモン産生細胞を全体に含む、一態様においては均等に含む細胞凝集体であってもよい。下垂体ホルモン産生細胞を多く含む部分構造(本明細書において下垂体組織ともいう)と下垂体ホルモン産生細胞の量が少ないもしくは下垂体ホルモン産生細胞が存在しない部分構造をともに含む細胞凝集体(下垂体組織と、下垂体以外の組織を含む細胞集団ともいう)であってもよい。
本発明の治療剤は、上述の細胞凝集体のいずれを含有していてもよい。
本発明の治療剤に含有される下垂体組織を含む細胞集団は、一態様では、下垂体組織を含み、下垂体以外の組織を含まない細胞凝集体であってよい。当該細胞凝集体に含まれる細胞については、上記1に記載の通りである。
本発明の治療剤に含有される下垂体組織を含む細胞集団は、一態様では、下垂体組織と、下垂体以外の組織をともに含む細胞集団であってよく、該細胞集団は、1)神経系細胞又は神経組織及び2)下垂体組織を含む細胞集団であってもよい。
該細胞集団は、上記した製造方法1により得ることができる。
5. Cell Populations Comprising Pituitary Tissue The cell populations comprising pituitary hormone-producing cells of the present invention may be cell aggregates comprising pituitary hormone-producing cells.
The cell aggregate may be a cell aggregate containing pituitary hormone-producing cells throughout, and in one embodiment, containing them evenly.The cell aggregate may be a cell aggregate containing both a partial structure containing many pituitary hormone-producing cells (also referred to as pituitary tissue in this specification) and a partial structure containing a small amount of pituitary hormone-producing cells or no pituitary hormone-producing cells (also referred to as a cell population containing pituitary tissue and tissue other than the pituitary gland).
The therapeutic agent of the present invention may contain any of the cell aggregates described above.
In one embodiment, the cell population containing pituitary tissue contained in the therapeutic agent of the present invention may be a cell aggregate containing pituitary tissue and not containing any tissue other than the pituitary gland. The cells contained in the cell aggregate are as described in 1 above.
In one embodiment, the cell population comprising pituitary tissue contained in the therapeutic agent of the present invention may be a cell population comprising both pituitary tissue and tissue other than the pituitary gland, and the cell population may be a cell population comprising 1) nervous cells or nervous tissue and 2) pituitary tissue.
The cell population can be obtained by the
上記の製造方法1により得られる本発明の細胞集団における1)神経系細胞又は神経組織は、一態様において中枢神経系の細胞若しくは組織、又はその前駆組織であり、中枢神経系の細胞又は組織としては、間脳(具体的には視床下部)及びそれらの組織に含まれる細胞(前駆細胞を含む)が挙げられ、より好ましくは視床下部もしくはその前駆組織、又はこれらに含まれる細胞であり、さらに好ましくは組織中に脳室様の構造を有している視床下部もしくはその前駆組織又はこれらに含まれる細胞である。1)神経系細胞又は神経組織は、例えばN-Cadherin陽性の神経上皮組織である。
In one embodiment, the 1) nervous system cells or nervous tissue in the cell population of the present invention obtained by the above-mentioned
上記の製造方法1により得られる本発明の細胞集団における2)下垂体組織は、一態様において非神経上皮組織と連続して形成されており、非神経上皮組織及び下垂体組織が1)神経系細胞又は神経組織を被覆していることがさらに好ましい。
上記した製造方法1により得られる前記非神経上皮組織は、一態様において口腔上皮又はその前駆組織である。下垂体組織は下垂体ホルモン産生細胞やその前駆細胞である下垂体前駆細胞を含むことが好ましく、下垂体幹細胞を含んでいても良く、濾胞星状細胞を含んでいてもよく、下垂体ホルモン産生細胞、下垂体前駆細胞、下垂体幹細胞、濾胞星状細胞全てを含んでいてもよい。
下垂体組織中に下垂体ニッチが形成されていることもまた好ましく、下垂体ニッチが下垂体前葉と中葉の間に残る遺残腔周辺のMCLニッチ様の構造であることもまた好ましく、下垂体ニッチが実質層ニッチ様の構造であることもまた好ましく、MCLニッチ様の構造と実質層ニッチ様の構造をともに含むことがさらに好ましい。
In one embodiment, 2) pituitary tissue in the cell population of the present invention obtained by the above-mentioned
In one embodiment, the non-neural epithelial tissue obtained by the above-mentioned
It is also preferable that a pituitary niche is formed in the pituitary tissue, and it is also preferable that the pituitary niche is an MCL niche-like structure around the residual cavity remaining between the anterior and intermediate lobes of the pituitary gland, and it is also preferable that the pituitary niche is a parenchymal layer niche-like structure, and it is even more preferable that it contains both an MCL niche-like structure and a parenchymal layer niche-like structure.
上記の製造方法1により得られる本発明の細胞集団に含まれる3)間葉系細胞は、例えば、Nestin、Vimentin、Cadherin-11、Laminin、CD44、CD90及びCD105からなる群から選ばれる少なくとも1種の間葉系細胞マーカーを発現する。
本発明に含まれうる非神経上皮組織は、例えば、サイトケラチン、E-Cadherin及びEpCAMからなる群から選ばれる少なくとも1種の非神経上皮組織マーカーを発現する。
本発明の治療薬に含有される細胞集団に含まれ得る下垂体幹細胞は、例えばSox2、Sox9、E-Cadherin、Nestin、S100β、GFRα2、Prop1、CD133、β-Catenin、Klf4、Oct4、Pax6、コクサッキーウイルス・アデノウイルス共通受容体(CXADR)、PRRX1/2、Ephrin-B2及びACEからなる群から選ばれる少なくとも1種の下垂体幹細胞マーカーを発現する。本発明の細胞集団の好ましい態様は、下垂体幹細胞マーカー(例、CXADR)陽性の下垂体幹細胞を含む。該細胞集団における下垂体幹細胞数の割合(下垂体幹細胞数の存在割合)が1%以上、好ましくは3%以上又は5%以上であってもよい
3) Mesenchymal cells contained in the cell population of the present invention obtained by the above-mentioned
Non-neuroepithelial tissues that can be included in the present invention express at least one non-neuroepithelial tissue marker selected from the group consisting of, for example, cytokeratin, E-Cadherin, and EpCAM.
Pituitary stem cells that may be contained in the cell population contained in the therapeutic agent of the present invention express at least one pituitary stem cell marker selected from the group consisting of, for example, Sox2, Sox9, E-Cadherin, Nestin, S100β, GFRα2, Prop1, CD133, β-Catenin, Klf4, Oct4, Pax6, coxsackievirus-adenovirus common receptor (CXADR), PRRX1/2, Ephrin-B2, and ACE. A preferred embodiment of the cell population of the present invention contains pituitary stem cells that are positive for a pituitary stem cell marker (e.g., CXADR). The proportion of pituitary stem cells in the cell population (proportion of pituitary stem cells present) may be 1% or more, preferably 3% or more or 5% or more.
6.下垂体組織の製造方法
本発明の治療薬に含有される下垂体組織の製造方法について、上記、「2.下垂体組織を含む細胞集団の製造方法」により得られた下垂体組織を含む細胞集団から下垂体組織を回収してもよい。一実施態様は、下記工程(1)、(2)及び(4)を含む。
(1)多能性幹細胞を、JNKシグナル伝達経路阻害物質とWntシグナル伝達経路阻害物質の存在下で培養し、細胞集団を得る第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質及びソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養(好ましくは浮遊培養)し、下垂体組織を含む細胞集団を得る第二工程、
(4)第二工程で得られた細胞集団から下垂体組織を回収する第四工程。
第一工程及び第二工程は、上記、「2.下垂体組織を含む細胞集団の製造方法」の第一工程及び第二工程と同様にして実施することができる。また、所望により第一工程の前にa工程を実施してもよい。また、所望により第二工程と第四工程の間に、第三工程を実施してもよい。
下垂体組織を含む細胞集団から下垂体組織を回収する第四工程は、形成された細胞集団が接着培養等で得られた平面的な組織である場合は、例えば顕微鏡観察下でニードル等を用い、下垂体組織を物理的に剥離する等の手法で回収することができる。形成された細胞集団が細胞塊等の立体的な組織である場合は、顕微鏡観察下でピンセット等を用い、細胞塊の外側(ラトケ嚢部分)に形成される下垂体組織を剥離・回収することによっておこなわれる。下垂体組織は、例えばNature communications,2016,7.に記載されているように、得られた細胞塊の表層にある半透明な薄い上皮として判別することができる。第四工程における細胞集団(細胞塊)からの下垂体組織の回収方法として、凍結融解、好ましくは緩慢凍結法を用いることもできる。当該方法は、外側に下垂体組織及び内側に間葉系神経や神経上皮組織を有する細胞塊を凍結融解することで物理的処理を施すことなく外側の下垂体組織が細胞塊から剥離されるというものである。
6. Method for Producing Pituitary Tissue In the method for producing the pituitary tissue contained in the therapeutic agent of the present invention, the pituitary tissue may be collected from the cell population containing pituitary tissue obtained by the above-mentioned "2. Method for Producing a Cell Population Containing Pituitary Tissue". One embodiment includes the following steps (1), (2) and (4).
(1) a first step of culturing pluripotent stem cells in the presence of a JNK signaling pathway inhibitor and a Wnt signaling pathway inhibitor to obtain a cell population;
(2) a second step of culturing (preferably in suspension) the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary tissue;
(4) A fourth step of recovering pituitary tissue from the cell population obtained in the second step.
The first and second steps can be carried out in the same manner as the first and second steps in "2. Method for producing a cell population containing pituitary tissue" above. If desired, step a may be carried out before
In the fourth step of recovering pituitary tissue from a cell population containing pituitary tissue, when the formed cell population is a planar tissue obtained by adhesion culture or the like, the pituitary tissue can be recovered by a method such as physically peeling off the pituitary tissue using a needle under a microscope. When the formed cell population is a three-dimensional tissue such as a cell mass, the pituitary tissue formed on the outside of the cell mass (Rathke's pouch portion) is peeled off and recovered using tweezers under a microscope. The pituitary tissue can be identified as a semi-transparent thin epithelium on the surface of the obtained cell mass, as described in Nature communications, 2016, 7. In the fourth step, freezing and thawing, preferably slow freezing, can also be used as a method for recovering pituitary tissue from a cell population (cell mass). In this method, the outer pituitary tissue is peeled off from the cell mass without physical treatment by freezing and thawing a cell mass having pituitary tissue on the outside and mesenchymal nerve or neuroepithelial tissue on the inside.
一態様において、本発明の治療薬に含有される下垂体ホルモン産生細胞、下垂体ホルモン産生細胞を含む細胞集団、又は下垂体ホルモン産生細胞を含む細胞凝集体、下垂体組織を含む細胞集団、あるいは下垂体組織を含む細胞集団から回収される下垂体組織は、上述の製造方法により製造し得る。 In one embodiment, the pituitary hormone-producing cells, cell populations containing pituitary hormone-producing cells, or cell aggregates containing pituitary hormone-producing cells, cell populations containing pituitary tissue, or pituitary tissue recovered from cell populations containing pituitary tissue, contained in the therapeutic agent of the present invention, can be produced by the production method described above.
7.精製体の製造方法
上述の「1.本発明の治療薬及び細胞移植」にある、目的外細胞(下垂体ホルモン産生細胞以外の細胞)が含まれない、又は目的外細胞が含まれる割合を低下させた細胞集団(精製体)を得るために、以下の工程を行ってもよい。具体的には、上述の「4.下垂体組織を含む細胞集団の製造方法1」に記載の第二工程(工程(2))の後にさらに、以下:第二工程で得られた下垂体ホルモン産生細胞を含む細胞集団から下垂体ホルモン産生細胞を選別し、回収する工程を行ってもよい。該工程は、自体公知の方法(例:国際公開第2023/054395号)を参考に行ってもよい。
7. Method for producing purified product In order to obtain a cell population (purified product) that does not contain unintended cells (cells other than pituitary hormone-producing cells) or has a reduced proportion of unintended cells, as described in the above "1. Therapeutic drug and cell transplantation of the present invention", the following steps may be performed. Specifically, after the second step (step (2)) described in the above "4. Method for producing a cell population containing
該工程では、第二工程で得られた下垂体組織を含む細胞集団からEpCAMを発現する細胞(EpCAM陽性細胞集団)を分離する。該分離工程を行う前に、第二工程で得られた下垂体組織を含む細胞集団を分散させる工程を行ってもよい。 In this step, cells expressing EpCAM (EpCAM-positive cell population) are separated from the cell population containing pituitary tissue obtained in the second step. Prior to carrying out this separation step, a step of dispersing the cell population containing pituitary tissue obtained in the second step may be carried out.
該分散させる工程では、まず得られた下垂体組織を含む細胞集団のROCK阻害剤による前処理を行い得る。当該前処理は、以降の下垂体組織を含む細胞集団の分散により誘導される細胞死を抑制するために、ROCK阻害剤を、当該前処理開始前から添加することが好ましい。ROCK阻害剤は、例えば、当該処理開始の少なくとも24時間前、少なくとも12時間前、少なくとも6時間前、少なくとも3時間前、少なくとも2時間前、少なくとも1時間前に添加する。ROCK阻害剤としては、Y-27632((R)-(+)-trans-4-(1-Aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide,dihydrochloride)等を挙げることができる。当該処理に際して用いられるROCK阻害剤の濃度は、以降の下垂体組織を含む細胞集団の分散により誘導される細胞死を抑制し得る濃度である。例えば、Y-27632について、このような濃度は、通常、約0.1~200μM、好ましくは約2~50μMである。ROCK阻害剤の濃度を添加する期間内で変動させてもよく、例えば期間の後半で濃度を半減させることができる。当該前処理を行うための培地は、工程(2)において用いる培地を用いることができる。なお、上記培地中に既に所望する濃度でROCK阻害剤が添加されている場合は、当該前処理工程を行う必要はない。 In the dispersion step, the obtained cell population containing pituitary tissue may first be pretreated with a ROCK inhibitor. In order to suppress cell death induced by the subsequent dispersion of the cell population containing pituitary tissue, it is preferable to add the ROCK inhibitor before the start of the pretreatment. The ROCK inhibitor is added, for example, at least 24 hours, at least 12 hours, at least 6 hours, at least 3 hours, at least 2 hours, or at least 1 hour before the start of the treatment. Examples of ROCK inhibitors include Y-27632 ((R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide, dihydrochloride). The concentration of the ROCK inhibitor used in this treatment is a concentration that can suppress cell death induced by the subsequent dispersion of the cell population containing the pituitary tissue. For example, for Y-27632, such a concentration is usually about 0.1 to 200 μM, preferably about 2 to 50 μM. The concentration of the ROCK inhibitor may be varied during the period of addition, for example, the concentration may be reduced by half in the latter half of the period. The medium used in step (2) can be used as the medium for carrying out this pretreatment. Note that if the ROCK inhibitor has already been added to the medium at the desired concentration, there is no need to carry out this pretreatment step.
次に、上記前処理を行った下垂体組織を含む細胞集団を、酵素処理により分散させる。具体的には、まず、上記前処理を行った該細胞集団を、PBSを含む培養器に移し、同培地で洗浄する。分散のための酵素としては、細胞を分散し得る限り特に限定されないが、例えば、パパイン、EDTA;トリプシン、コラゲナーゼ(コラゲナーゼ タイプI~VII)、メタロプロテアーゼ、ヒアルロニダーゼ、エラスターゼ、ディスパーゼ、デオキシリボヌクレアーゼ等の酵素やこれらの混合物が挙げられる。好ましい酵素としては、パパインが挙げられる。酵素処理の条件(温度、時間等)は、用いる酵素等により適宜設定し得る。また、当該酵素処理を促進させるために、当該処理前に、下垂体組織を含む細胞集団を物理的(例:メス、ハサミ等)に細断するか、或いは該細胞集団に物理的(例:メス、ハサミ等)に切れ込みを入れる工程を行ってもよい。 Next, the cell population containing the pituitary tissue that has been pretreated as described above is dispersed by enzyme treatment. Specifically, the cell population that has been pretreated as described above is first transferred to an incubator containing PBS and washed with the same medium. The enzyme for dispersion is not particularly limited as long as it can disperse the cells, but examples include enzymes such as papain, EDTA, trypsin, collagenase (collagenase types I to VII), metalloprotease, hyaluronidase, elastase, dispase, deoxyribonuclease, and mixtures thereof. A preferred enzyme is papain. The conditions for the enzyme treatment (temperature, time, etc.) can be appropriately set depending on the enzyme used, etc. In addition, in order to promote the enzyme treatment, a step of physically cutting the cell population containing the pituitary tissue (e.g., scalpel, scissors, etc.) into small pieces or physically cutting the cell population (e.g., scalpel, scissors, etc.) may be performed before the treatment.
上記酵素処理後、浮遊細胞を回収し、再度、上記酵素処理を行ってもよい。複数回繰り返してもよい。当該酵素処理も、上述の酵素等で行うことができ、好ましい酵素としては、EDTA;トリプシン、コラゲナーゼ(コラゲナーゼ タイプI~VIII)、メタロプロテアーゼ、ヒアルロニダーゼ、エラスターゼ、ディスパーゼ、デオキシリボヌクレアーゼ等の酵素やこれらの混合物が挙げられる。好ましい酵素としては、コラゲナーゼが挙げられ、より好ましくはコラゲナーゼ タイプIである。酵素処理の条件(温度、時間等)は、用いる酵素等により適宜設定し得る。 After the enzyme treatment, the floating cells may be collected and the enzyme treatment may be carried out again. This may be repeated multiple times. The enzyme treatment may be carried out using the enzymes described above, and preferred enzymes include EDTA; trypsin, collagenase (collagenase types I to VIII), metalloprotease, hyaluronidase, elastase, dispase, deoxyribonuclease, and mixtures thereof. Preferred enzymes include collagenase, more preferably collagenase type I. The conditions for the enzyme treatment (temperature, time, etc.) may be set appropriately depending on the enzyme used.
上記酵素処理後、浮遊細胞を回収し、再度、上記酵素処理を行う。当該酵素処理も、上述の酵素等で行うことができ、好ましい酵素としては、EDTA;トリプシンが挙げられ、より好ましくは、EDTA;トリプシンとデオキシリボヌクレアーゼである。また、EDTA;トリプシンの代わりに、TrypLE (Invitrogen)等の市販品を用いてもよい。酵素処理の条件(温度、時間等)は、用いる酵素等により適宜設定し得る。上記一連の酵素処理により単細胞懸濁液を調製し得る。また、単細胞懸濁液を調製する際に、自体公知の方法により、死細胞を除去してもよい。 After the enzyme treatment, the floating cells are collected and the enzyme treatment is carried out again. The enzyme treatment can be carried out with the enzymes mentioned above, and preferred enzymes include EDTA; trypsin, and more preferably EDTA; trypsin and deoxyribonuclease. Commercially available products such as TrypLE (Invitrogen) may be used in place of EDTA; trypsin. The enzyme treatment conditions (temperature, time, etc.) can be appropriately set depending on the enzymes used. A single-cell suspension can be prepared by the above series of enzyme treatments. When preparing a single-cell suspension, dead cells may be removed by a method known per se.
該工程では、以上の通りにして得られる分散された細胞集団から、EpCAMを発現する細胞が分離される。分散された細胞集団から、EpCAMを発現する所望の細胞を分離する方法としては、フローサイトメトリーやマスサイトメトリーを用いた方法、磁気細胞分離法などが挙げられ、これらの方法は、自体公知の方法を用いて行うことができる。例えば、EpCAMを発現する細胞は、当該細胞とEpCAM分子に特異的に結合する物質(例:抗体等)とを接触させる工程を含む方法により分離することができる。上記物質には、それ自体に検出可能な標識(例:GFP、PE)が付加されているもの、及びそれ自体には標識が付加されていないものも含まれる。上記物質が、それ自体に標識が付加されていないものである場合、当該物質を直接又は間接的に認識する検出可能な標識が付加された物質をさらに使用することで前記分離が可能となる。例えば、前記物質が抗体である場合には、蛍光色素、金属同位体又はビーズ(例:磁気ビーズ)を当該抗体に直接的又は間接的に担持させ、それにより細胞表面のマーカーを標識することが可能であり、当該標識に基づいて細胞を分離することが可能である。この際用いる抗体は、1種類のみ、又は2種類以上の抗体であってもよい。該工程を行った後、上述の工程(3)を行ってもよい。
該工程を行うタイミングは、第二工程(工程(2))の後に限定されない。一態様として、工程(2)の後さらに工程(b)を行ってから、該工程を行うこともでき、また別の態様では、工程(2)の後に、工程(b)を行い、その後に工程(3)を行い、さらにその後に該工程を行うこともできる。これらの態様において、該工程を行った後、上述の工程(3)を行ってもよい。
In this step, cells expressing EpCAM are separated from the dispersed cell population obtained as described above. Methods for separating desired cells expressing EpCAM from dispersed cell populations include methods using flow cytometry or mass cytometry, magnetic cell separation methods, etc., and these methods can be performed using methods known per se. For example, cells expressing EpCAM can be separated by a method including a step of contacting the cells with a substance (e.g., antibody, etc.) that specifically binds to EpCAM molecules. The above-mentioned substances include those that have a detectable label (e.g., GFP, PE) attached thereto, and those that have no label attached thereto. When the above-mentioned substances have no label attached thereto, the separation can be achieved by further using a substance that has a detectable label attached thereto that directly or indirectly recognizes the substance. For example, when the substance is an antibody, a fluorescent dye, a metal isotope, or beads (e.g., magnetic beads) can be directly or indirectly supported on the antibody, thereby labeling a marker on the cell surface, and the cells can be separated based on the label. In this case, only one type of antibody may be used, or two or more types of antibodies may be used. After carrying out this step, the above-mentioned step (3) may be carried out.
The timing of carrying out this step is not limited to after the second step (step (2)). In one embodiment, step (b) may be carried out after step (2) and then this step may be carried out. In another embodiment, step (b) may be carried out after step (2), and then step (3) may be carried out, and then this step may be carried out. In these embodiments, the above-mentioned step (3) may be carried out after carrying out this step.
8.下垂体ホルモン産生細胞を含む組成物及び治療方法
本発明の治療薬に含有される下垂体ホルモン産生細胞は、細胞移植療法を所望する下垂体の障害に基づく疾患に関して、治療上有効量を投与すればよく、移植対象の、年齢、体重、疾患の重篤度等の要因により変化し得、特に限定されないが、例えば、10×103細胞以上もしくは10×103細胞~10×1012細胞程度、好ましくは10×104細胞以上もしくは10×104細胞~10×1011細胞程度、さらに好ましくは10×105細胞以上もしくは10×105細胞~10×1010細胞程度とすることができる。また、本発明の治療薬に、下垂体ホルモン産生細胞を含む細胞集団、下垂体ホルモン産生細胞を含む細胞凝集体、下垂体組織を含む細胞集団、あるいは下垂体組織を含む細胞集団から回収される下垂体組織として下垂体ホルモン産生細胞を含有させる場合も、細胞移植療法を所望する下垂体の障害に基づく疾患に関して、治療上有効量を投与すればよく、移植対象の、年齢、体重、疾患の重篤度等の要因により適宜変化し得る。
8. Compositions Comprising Pituitary Hormone-Producing Cells and Treatment Methods The pituitary hormone-producing cells contained in the therapeutic agent of the present invention may be administered in a therapeutically effective amount for a disease based on a pituitary disorder for which cell transplantation therapy is desired, and may vary depending on factors such as the age, weight, and severity of the disease of the transplantation subject, and are not particularly limited, and may be, for example, 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells, preferably 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells, and more preferably 10 x 10 cells or more, or about 10 x 10 cells to 10 x 10 cells. Furthermore, when the therapeutic agent of the present invention contains pituitary hormone-producing cells as a cell population containing pituitary hormone-producing cells, a cell aggregate containing pituitary tissue, or pituitary tissue recovered from a cell population containing pituitary tissue, it is sufficient to administer a therapeutically effective amount for a disease due to a pituitary disorder for which cell transplantation therapy is desired, and this can be varied appropriately depending on factors such as the age, weight, and severity of the disease of the transplantation subject.
本発明の治療薬に含有される下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を、細胞移植療法に用いる場合、拒絶反応が起こらないという観点から、移植先の個体のHLA遺伝子型が同一若しくは実質的に同一である体細胞から樹立したiPS細胞に由来する細胞を用いることが望ましい。ここで、「実質的に同一」とは、移植した細胞に対して免疫抑制剤により免疫反応が抑制できる程度にHLA遺伝子型が一致していることであり、例えば、HLA-A、HLA-B及びHLA-DRの3遺伝子座あるいはHLA-Cを加えた4遺伝子座、又は更にHLA-DP及びHLA-DQを加えた6遺伝子座が一致するHLA型を有する体細胞である。年齢や体質などの理由から充分な細胞が得られない場合には、ポリエチレングリコールやシリコーンのようなカプセル、多孔性の容器などに包埋して拒絶反応を回避した状態で移植することも可能である。 When the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention are used in cell transplantation therapy, it is desirable to use cells derived from iPS cells established from somatic cells with the same or substantially the same HLA genotype of the recipient individual, from the viewpoint of preventing rejection reactions. Here, "substantially the same" means that the HLA genotype matches the transplanted cells to such an extent that the immune response can be suppressed by an immunosuppressant, for example, somatic cells with HLA types matching the three loci of HLA-A, HLA-B, and HLA-DR, or four loci including HLA-C, or six loci including HLA-DP and HLA-DQ. If sufficient cells cannot be obtained due to age, constitution, etc., they can also be transplanted in a state where they are embedded in a capsule or porous container made of polyethylene glycol or silicone to avoid rejection reactions.
本発明の治療薬は、医薬上許容される水性液体と混合するなどして、調製することができる。従って、一態様において、本発明の下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を製剤化する工程を含む、下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を含有する下垂体の障害に基づく疾患の治療薬の製法も提供される。かかる製法は、本発明の下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を準備する工程を含んでいてもよい。さらに、本発明の下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を保存する工程を含むこともできる。 The therapeutic agent of the present invention can be prepared by mixing with a medicamentously acceptable aqueous liquid, for example. Thus, in one embodiment, there is also provided a method for producing a therapeutic agent for a disease due to a disorder of the pituitary gland, which contains pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, and includes a step of formulating the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention. Such a production method may include a step of preparing the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention. It may also include a step of preserving the pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells, of the present invention.
本発明の治療薬に含まれ得る前記医薬上許容される水性液体には、例えば、緩衝剤、等張化剤、pH調整剤、抗酸化剤、キレート剤等を適宜選択して、移植される下垂体ホルモン産生細胞の生存率及び生理活性に影響を与えない範囲で含有させることができる。
緩衝剤としては、例えば、リン酸緩衝剤、ホウ酸緩衝剤、クエン酸緩衝剤、酒石酸緩衝剤、酢酸緩衝剤、アミノ酸、イプシロン-アミノカプロン酸などが挙げられる。等張化剤としては、D-ソルビトール、D-グルコース、D-マンニトールなどの糖類、グリセリン、プロピレングリコールなどの多価アルコール類、塩化ナトリウムなどの塩類、ホウ酸などが挙げられる。
キレート剤としては、エデト酸ナトリウム、クエン酸などが挙げられる。
pH調整剤としては、例えば、水酸化ナトリウム、水酸化カリウム、炭酸ナトリウム、炭酸水素ナトリウム、ホウ酸又はその塩(ホウ砂)、塩酸、クエン酸又はその塩(クエン酸ナトリウム、クエン酸二水素ナトリウム等)、リン酸又はその塩(リン酸水素二ナトリウム、リン酸二水素カリウム等)、酢酸又はその塩(酢酸ナトリウム、酢酸アンモニウム等)、酒石酸又はその塩(酒石酸ナトリウム等)等が挙げられる。
抗酸化剤としては、例えば、アスコルビン酸、グルタチオン、亜硫酸水素ナトリウム、乾燥亜硫酸ナトリウム、ピロ亜硫酸ナトリウム、トコフェロール等が挙げられる。
「製薬学上許容される水性液体」としては、具体的には、生理食塩水、ブドウ糖やその他の補助薬を含む等張液(例えば、D-ソルビトール、D-マンニトール、塩化ナトリウムなど)などの水性液を挙げることができる。
本発明の治療薬は、例えば、無痛化剤(例えば、塩化ベンザルコニウム、塩酸プロカインなど)、安定剤(例えば、ヒト血清アルブミン、ポリエチレングリコールなど)、保存剤、酸化防止剤などと配合してもよい。
The pharma- ceutically acceptable aqueous liquid that may be contained in the therapeutic agent of the present invention may contain, for example, an appropriate selection of a buffer, an isotonicity agent, a pH adjuster, an antioxidant, a chelating agent, etc., within a range that does not affect the viability and physiological activity of the transplanted pituitary hormone-producing cells.
Examples of the buffer include phosphate buffer, borate buffer, citrate buffer, tartrate buffer, acetate buffer, amino acid, epsilon-aminocaproic acid, etc. Examples of the isotonicity agent include sugars such as D-sorbitol, D-glucose, D-mannitol, etc., polyhydric alcohols such as glycerin, propylene glycol, etc., salts such as sodium chloride, boric acid, etc.
Chelating agents include sodium edetate and citric acid.
Examples of pH adjusters include sodium hydroxide, potassium hydroxide, sodium carbonate, sodium bicarbonate, boric acid or a salt thereof (borax), hydrochloric acid, citric acid or a salt thereof (sodium citrate, sodium dihydrogen citrate, etc.), phosphoric acid or a salt thereof (disodium hydrogen phosphate, potassium dihydrogen phosphate, etc.), acetic acid or a salt thereof (sodium acetate, ammonium acetate, etc.), tartaric acid or a salt thereof (sodium tartrate, etc.), and the like.
Examples of antioxidants include ascorbic acid, glutathione, sodium hydrogen sulfite, dry sodium sulfite, sodium pyrosulfite, and tocopherol.
Specific examples of the "pharmaceutical acceptable aqueous liquid" include aqueous liquids such as physiological saline, isotonic solutions containing glucose and other auxiliary drugs (eg, D-sorbitol, D-mannitol, sodium chloride, etc.).
The therapeutic agent of the present invention may be compounded with, for example, a soothing agent (eg, benzalkonium chloride, procaine hydrochloride, etc.), a stabilizer (eg, human serum albumin, polyethylene glycol, etc.), a preservative, an antioxidant, and the like.
また、本発明の治療剤は、血管新生促進剤と組み合わせて併用剤(組み合わせ剤)としてもよい。血管新生促進剤は、医薬的に許容されるものであれば特に限定されないが、例えば、血管内皮成長因子(VEGF)、線維芽細胞増殖因子:酸性(aFGF)及び塩基性(bFGF)、上皮成長因子(EGF)、顆粒球マクロファージ-コロニー刺激因子(GM-CSF)、肝細胞増殖因子(HGF)、スフィンゴシン-1-リン酸(S1P)などが挙げれる。併用剤とする場合、有効成分をそれぞれ別個に製剤化してもよいし、それぞれの有効成分を単剤として製剤化してもよい。また、単剤として製剤化する場合、本発明の併用剤は、各製剤をそれぞれ1又は2以上含んでもよい。
別個に製剤化する場合には特に限定はないが、血管新生促進剤の持続放出等を企図して、生体適合性を有するゲル(特に、ヒドロゲル等)やフイルム等に該剤を含有させてもよい。生体適合性を有するゲルやフイルム等は、生体吸収性であってもよい。上述のゲルやフイルム等は、特に限定されないが、例えば、羊膜、ポリフッ化ビニリデン(PVDF)、ポリテトラフルオロエチレン(PTFE)、ポリウレタン、ポリプロピレン、ポリエステル、塩化ビニル、ポリカーボネート、アクリル、シリコーン、MPC(2-メタクリロイルオキシエチルホスホリルコリン)、ポリ乳酸、ポリグリコール酸、乳酸・グリコール酸コポリマー(PLGA)、フィブリン、ゼラチン、コラーゲン、コラーゲンアルギン酸ナトリウム、ポリ(N-イソプロピルアクリルアミド)(PIPAAm)をポリエチレングリコール(PEG)で架橋した温度応答性ゲル、ヒアルロン酸、グリコサミノグリカン、プロテオグリカン、コンドロイチン、セルロース、アガロース、カルボキシメチルセルロース、キチン、キトサン、ゼラチン、アテロコラーゲン、エラスチン、フィブロネクチン、プロネクチン、ラミニン、テネイシン、フィブロイン、エンタクチン、トロンボスポンジン、レトロネクチン、デキストリン、トレハロース等を用いて作製されたものであってもよい。市販の生体適合性を有するゲルやフイルム等としては、例えばDuragen(Integra Japan社製)、Surgicel(ジョンソン・エンド・ジョンソン社製)、スポンゼル(LTLファーマ社製)等が挙げられる。
The therapeutic agent of the present invention may be combined with an angiogenesis promoter to form a combination drug (combination drug). The angiogenesis promoter is not particularly limited as long as it is medicamentically acceptable, and examples thereof include vascular endothelial growth factor (VEGF), fibroblast growth factor: acidic (aFGF) and basic (bFGF), epidermal growth factor (EGF), granulocyte macrophage-colony stimulating factor (GM-CSF), hepatocyte growth factor (HGF), and sphingosine-1-phosphate (S1P). When used as a combination drug, the active ingredients may be formulated separately, or each active ingredient may be formulated as a single agent. When formulated as a single agent, the combination drug of the present invention may contain one or more of each formulation.
When the agent is formulated separately, there is no particular limitation, but the agent may be contained in a biocompatible gel (particularly, a hydrogel, etc.), film, etc., with the intention of sustained release of the angiogenesis promoter, etc. The biocompatible gel, film, etc. may be bioabsorbable. The above-mentioned gel, film, etc., are not particularly limited, but examples thereof include amniotic membrane, polyvinylidene fluoride (PVDF), polytetrafluoroethylene (PTFE), polyurethane, polypropylene, polyester, vinyl chloride, polycarbonate, acrylic, silicone, MPC (2-methacryloyloxyethyl phosphorylcholine), polylactic acid, polyglycolic acid, lactic acid-glycolic acid copolymer (PLGA), fibrin, gelatin, collagen, collagen sodium alginate, poly(N-isopropyl acrylate), ... The gel may be a temperature-responsive gel in which polyacrylamide (PIPAAm) is crosslinked with polyethylene glycol (PEG), hyaluronic acid, glycosaminoglycan, proteoglycan, chondroitin, cellulose, agarose, carboxymethylcellulose, chitin, chitosan, gelatin, atelocollagen, elastin, fibronectin, pronectin, laminin, tenascin, fibroin, entactin, thrombospondin, retronectin, dextrin, trehalose, etc. Examples of commercially available biocompatible gels and films include Duragen (Integra Japan), Surgicel (Johnson & Johnson), and Spongel (LTL Pharma).
本発明の併用剤において、各有効成分がそれぞれ別個に製剤化されている場合、各製剤は、同時に又は時間差をおいて、同一経路又は別経路で対象に投与(移植)することができる。また各製剤の投与(移植)量や投与(移植)回数は、血管新生促進剤の種類、投与対象の症状、齢、体重、薬物受容性等の種々の条件により、適宜設定することができる。 When each active ingredient in the combination drug of the present invention is formulated separately, each formulation can be administered (transplanted) to a subject simultaneously or at different times, via the same route or via different routes. The administration (transplantation) amount and number of administrations (transplants) of each formulation can be appropriately determined depending on various conditions such as the type of angiogenesis promoter, the symptoms, age, weight, and drug tolerance of the subject to be administered.
一例としては、ゼラチンヒドロゲル持続放出デバイスに塩基性線維芽細胞増殖因子(FGF)を含有させたものを移植部位に予め処置しておき、一定期間経過後に本発明の治療剤を移植する方法が挙げられる。 One example is a method in which a gelatin hydrogel sustained release device containing basic fibroblast growth factor (FGF) is applied to the transplant site in advance, and the therapeutic agent of the present invention is transplanted after a certain period of time has passed.
本発明の治療薬は、細胞の凍結保存に通常使用される条件で凍結保存された状態で提供され、用時融解して用いることもできる。その場合、血清若しくはその代替物、有機溶剤(例、DMSO)等をさらに含んでいてもよい。この場合、血清若しくはその代替物の濃度は、特に限定されるものではないが約1~約30%(v/v)、好ましくは約5~約20%(v/v)であり得る。有機溶剤の濃度は、特に限定されるものではないが0~約50%(v/v)、好ましくは約5~約20%(v/v)であり得る。 The therapeutic agent of the present invention is provided in a frozen state under conditions normally used for cryopreservation of cells, and can be thawed when needed. In this case, it may further contain serum or a substitute thereof, an organic solvent (e.g., DMSO), etc. In this case, the concentration of serum or a substitute thereof is not particularly limited, but may be about 1 to about 30% (v/v), preferably about 5 to about 20% (v/v). The concentration of the organic solvent is not particularly limited, but may be 0 to about 50% (v/v), preferably about 5 to about 20% (v/v).
さらに本発明の治療薬に含有される下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等の細胞集団は、上述の通り、対象の皮下組織及び/又は筋肉組織に移植することで下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患を治療し得る。従って、本発明は、皮下組織及び/又は筋肉組織へ、治療有効量の下垂体ホルモン産生細胞あるいは下垂体ホルモン産生細胞を含む細胞凝集体等を投与することにより、下垂体の障害に基づく疾患を治療することを特徴とする治療方法も提供する。 Furthermore, as described above, cell populations such as pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells contained in the therapeutic agent of the present invention can be transplanted into the subcutaneous tissue and/or muscle tissue of a subject to treat diseases caused by disorders of the pituitary gland that contain pituitary hormone-producing cells. Therefore, the present invention also provides a treatment method characterized by treating diseases caused by disorders of the pituitary gland by administering a therapeutically effective amount of pituitary hormone-producing cells or cell aggregates containing pituitary hormone-producing cells to the subcutaneous tissue and/or muscle tissue.
本発明の治療薬に含有される下垂体ホルモン産生細胞、下垂体ホルモン産生細胞を含む細胞集団、下垂体ホルモン産生細胞を含む細胞凝集体、下垂体組織を含む細胞集団、あるいは下垂体組織を含む細胞集団から回収される下垂体組織は、ゲノム編集により遺伝子を操作された多能性幹細胞を原料として用いて製造されたものであってもよい。ゲノム編集の対象となる遺伝子は、上述の本発明の下垂体ホルモン産生細胞等の製造方法による下垂体組織の分化に関与する遺伝子、該製造方法によって副生する下垂体以外の目的外細胞への分化に関わる遺伝子、下垂体から分泌されるホルモン関連遺伝子、疾患の感染に関わる遺伝子等であるが、これに限定されない。 The pituitary hormone-producing cells, cell populations containing pituitary hormone-producing cells, cell aggregates containing pituitary hormone-producing cells, cell populations containing pituitary tissue, or pituitary tissue recovered from cell populations containing pituitary tissue contained in the therapeutic agent of the present invention may be produced using pluripotent stem cells whose genes have been manipulated by genome editing as a raw material. Genes that are the subject of genome editing include, but are not limited to, genes involved in the differentiation of pituitary tissue by the above-mentioned method for producing pituitary hormone-producing cells, etc. of the present invention, genes involved in the differentiation into unintended cells other than the pituitary gland that are by-produced by the production method, hormone-related genes secreted from the pituitary gland, genes involved in the infection of diseases, etc.
9.モデル動物
本発明の態様として、非ヒト動物の皮下組織及び/又は筋肉組織へヒト下垂体ホルモン産生細胞を投与することを含む、担ヒト下垂体組織モデル非ヒト動物の作製方法が挙げられる。
一態様において、当該非ヒト動物は、ヒト以外の哺乳動物であれば特に限定はなく、具体的には、物質の薬効や安全性を評価する目的で利用される哺乳動物が挙げられる。当該哺乳動物としては、マウス、ラット、ハムスターもしくはモルモット、イヌ、ウサギ、又はキツネザル、ロリス、ツバイ又はサルが挙げられる。
ヒト下垂体ホルモン産生細胞としては、上記の任意のヒト下垂体ホルモン産生細胞を含む細胞集団が挙げられる。当該ヒト下垂体ホルモン産生細胞を移植する皮下組織と筋肉組織としては、上記1のとおりである。当該ヒト下垂体ホルモン産生細胞の非ヒト動物への移植は、本明細書実施例に準じて行えば良い。
一態様において、本発明のモデル非ヒト動物は、外科的に下垂体の一部又は全てが除去された動物であってよい。
9. Model Animals An embodiment of the present invention includes a method for producing a non-human animal model bearing human pituitary tissue, which comprises administering human pituitary hormone-producing cells into the subcutaneous tissue and/or muscle tissue of the non-human animal.
In one embodiment, the non-human animal is not particularly limited as long as it is a mammal other than a human, and specifically includes a mammal used for evaluating the efficacy or safety of a substance, such as a mouse, a rat, a hamster or a guinea pig, a dog, a rabbit, or a lemur, a loris, a tree shrew, or a monkey.
The human pituitary hormone-producing cells include a cell population containing any of the above-mentioned human pituitary hormone-producing cells. The subcutaneous tissue and muscle tissue into which the human pituitary hormone-producing cells are transplanted are as described above in 1. The human pituitary hormone-producing cells may be transplanted into a non-human animal in accordance with the Examples of the present specification.
In one embodiment, the model non-human animal of the present invention may be an animal in which part or all of the pituitary gland has been surgically removed.
本発明は、上記作製方法により得られる担ヒト下垂体組織モデル非ヒト動物を包含する。
本発明は、前記担ヒト下垂体組織モデル非ヒト動物に被験物質を投与することを含む、当該被験物質の薬効又は安全性を評価する方法を包含する。
一態様において、当該非ヒト動物への被験物質の投与方法には特に限定はなく、経口又は非経口(静脈内投与、皮下投与、皮内投与等を含むがこれらに限定されない)で投与することができる。
一態様において、被験物質を当該非ヒト動物へ投与し、当該非ヒト動物体内の下垂体ホルモンの濃度(例えば血中濃度)を測定することにより、当該被験物質が、下垂体ホルモンの分泌又はインビボ作用に及ぼす影響を調べることができる。
すなわち、一態様において、本発明の評価方法は、以下の工程を含んでいてもよい:
(i)前記担ヒト下垂体組織モデル非ヒト動物に、被験物質を投与する工程、
(ii)被験物質を投与された非ヒト動物の、下垂体ホルモンの量又は下垂体ホルモンの活性を調べる工程、及び
(iii)(ii)の結果に基づき、当該被験物質が下垂体ホルモンの分泌又は活性に及ぼす影響を評価する工程。
The present invention includes a non-human animal model bearing human pituitary tissue obtained by the above-mentioned production method.
The present invention encompasses a method for evaluating the efficacy or safety of a test substance, which comprises administering the test substance to the non-human animal model bearing human pituitary tissue.
In one embodiment, the method of administering the test substance to the non-human animal is not particularly limited, and the substance can be administered orally or parenterally (including, but not limited to, intravenous administration, subcutaneous administration, intradermal administration, etc.).
In one aspect, the effect of the test substance on the secretion or in vivo action of a pituitary hormone can be examined by administering the test substance to the non-human animal and measuring the concentration of the pituitary hormone in the non-human animal (e.g., blood concentration).
That is, in one embodiment, the evaluation method of the present invention may comprise the following steps:
(i) administering a test substance to the non-human animal model bearing human pituitary gland tissue;
(ii) examining the amount of pituitary hormone or activity of pituitary hormone in a non-human animal administered the test substance; and (iii) evaluating the effect of the test substance on the secretion or activity of the pituitary hormone based on the results of (ii).
また、一態様において、本発明の評価方法は、下垂体の障害に基づく疾患の予防又は治療薬のスクリーニング方法であり、以下の工程を含んでいてもよい:
(i)前記担ヒト下垂体組織モデル非ヒト動物に、被験物質を投与する工程、
(ii)被験物質を投与された非ヒト動物の、下垂体ホルモンの量又は下垂体ホルモンの活性を調べる工程、及び
(iii)下垂体ホルモンの量又は下垂体ホルモンの活性が増大した/低減した被験物質を下垂体の障害に基づく疾患の予防又は治療薬の候補として選択する工程。
In one embodiment, the evaluation method of the present invention is a method for screening for a preventive or therapeutic agent for a disease caused by a pituitary gland disorder, and may comprise the following steps:
(i) administering a test substance to the non-human animal model bearing human pituitary gland tissue;
(ii) examining the amount or activity of the pituitary hormone in the non-human animal administered the test substance; and (iii) selecting the test substance in which the amount or activity of the pituitary hormone is increased/decreased as a candidate for a preventive or therapeutic drug for a disease based on a disorder of the pituitary gland.
本発明のスクリーニング方法で用いる被験物質としては、例えば、細胞抽出物、細胞培養上清、微生物発酵産物、海洋生物由来の抽出物、植物抽出物、精製タンパク質又は粗タンパク質、ペプチド、非ペプチド化合物、合成低分子化合物、及び天然化合物が挙げられる。前記被験物質はまた、(1)生物学的ライブラリー、(2)デコンヴォルーションを用いる合成ライブラリー法、(3)「1ビーズ1化合物(one-bead one-compound)」ライブラリー法、及び(4)アフィニティクロマトグラフィー選別を使用する合成ライブラリー法を含む当技術分野で公知のコンビナトリアルライブラリー法における多くのアプローチのいずれかを使用して得ることができる。アフィニティクロマトグラフィー選別を使用する生物学的ライブラリー法はペプチドライブラリーに限定されるが、その他の4つのアプローチはペプチド、非ペプチドオリゴマー、又は化合物の低分子化合物ライブラリーに適用できる(Lam(1997)Anticancer Drug Des. 12:145-67)。分子ライブラリーの合成方法の例は、当技術分野において見出され得る(DeWitt et al.(1993)Proc. Natl. Acad. Sci. USA 90:6909-13; Erb et al.(1994)Proc. Natl. Acad. Sci. USA 91:11422-6; Zuckermann et al.(1994)J. Med. Chem. 37:2678-85; Cho et al.(1993)Science 261:1303-5; Carell et al.(1994)Angew. Chem. Int. Ed. Engl. 33:2059; Carell et al.(1994)Angew. Chem. Int. Ed. Engl. 33:2061; Gallop et al.(1994)J. Med. Chem. 37:1233-51)。化合物ライブラリーは、溶液(Houghten(1992)Bio/Techniques 13:412-21を参照のこと)又はビーズ(Lam(1991)Nature 354:82-4)、チップ(Fodor(1993)Nature 364:555-6)、細菌(米国特許第5,223,409号)、胞子(米国特許第5,571,698号、同第5,403,484号、及び同第5,223,409号)、プラスミド(Cull et al.(1992)Proc. Natl. Acad. Sci. USA 89:1865-9)若しくはファージ(Scott and Smith(1990)Science 249:386-90; Devlin(1990)Science 249:404-6; Cwirla et al.(1990)Proc. Natl. Acad. Sci. USA 87:6378-82; Felici(1991)J. Mol. Biol. 222:301-10; 米国特許出願第2002103360号)として作製され得る。 The test substances used in the screening method of the present invention include, for example, cell extracts, cell culture supernatants, microbial fermentation products, extracts from marine organisms, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic small molecule compounds, and natural compounds. The test substances can also be obtained using any of the many approaches in combinatorial library methods known in the art, including (1) biological libraries, (2) synthetic library methods using deconvolution, (3) "one-bead one-compound" library methods, and (4) synthetic library methods using affinity chromatography selection. While the biological library method using affinity chromatography selection is limited to peptide libraries, the other four approaches can be applied to small molecule compound libraries of peptides, non-peptide oligomers, or compounds (Lam (1997) Anticancer Drug Des. 12:145-67). Examples of methods for the synthesis of molecular libraries can be found in the art (DeWitt et al. (1993) Proc. Natl. Acad. Sci. USA 90:6909-13; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91:11422-6; Zuckermann et al. (1994) J. Med. Chem. 37:2678-85; Cho et al. (1993) Science 261:1303-5; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2061; Gallop et al. al. (1994) J. Med. Chem. 37:1233-51). Chemical libraries have been developed in solution (see Houghten (1992) Bio/Techniques 13:412-21) or on beads (Lam (1991) Nature 354:82-4), on chips (Fodor (1993) Nature 364:555-6), on bacteria (U.S. Pat. No. 5,223,409), spores (U.S. Pat. Nos. 5,571,698, 5,403,484, and 5,223,409), plasmids (Cull et al. (1992) Proc. Natl. Ac. 1999, 10:131-132), and on spores (U.S. Pat. Nos. 5,571,698, 5,403,484, and 5,223,409). ad. Sci. USA 89:1865-9) or as phages (Scott and Smith (1990) Science 249:386-90; Devlin (1990) Science 249:404-6; Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87:6378-82; Felici (1991) J. Mol. Biol. 222:301-10; U.S. Patent Application No. 2002103360).
以下に実施例を挙げて本発明をより具体的に説明するが、本発明はこれらに何ら限定されるものではない。 The present invention will be explained in more detail below with reference to examples, but the present invention is not limited to these in any way.
実施例1:下垂体ホルモン産生細胞の移植部位の検討
<材料及び方法>
(1)ヒト胚性幹細胞(hESC)の維持及び分化培養(下垂体オルガノイド)
hESC(KhES-1)を、理化学研究所バイオリソース研究センターから入手し、日本政府のhESC研究ガイドラインに従って使用した。全ての実験プロトコールと手順は、名古屋大学大学院医学研究科の倫理委員会によって承認された(承認ES-001)。hESCの維持及び分化培養を、Scientific Reports,4,3594(2014)に記載の方法に準じてフィーダーフリー条件で培養した。フィーダーフリー培地としてはStemFit培地(AK03、味の素社製)、フィーダーフリー足場にはLaminin511-E8(ニッピ社製)を用いた。
具体的な維持培養操作としては、まずサブコンフレントになったhESC(KhES-1株)を、PBSにて洗浄後、TrypLE Select(Life Technologies社製)を用いて単一細胞へ分散した。その後、前記単一細胞へ分散されたヒトES細胞を、Laminin511-E8にてコートしたプラスチック培養ディッシュに播種し、Y27632(ROCK阻害物質、10μM)存在下、StemFit培地にてフィーダーフリー培養した。前記プラスチック培養ディッシュとして、6ウェルプレート(イワキ社製、細胞培養用、培養面積9.4cm2)を用いた場合、前記単一細胞へ分散されたヒトES細胞の播種細胞数は2.4×104とした。播種した1日後に、Y27632を含まないStemFit培地に全量培地交換した。以降、1日~2日に一回Y27632を含まないStemFit培地にて全量培地交換した。その後、播種した7日後にサブコンフレント(培養面積の6割が細胞に覆われる程度)になるまで培養した。
Example 1: Examination of transplantation sites of pituitary hormone-producing cells <Materials and methods>
(1) Maintenance and differentiation culture of human embryonic stem cells (hESC) (pituitary organoids)
hESCs (KhES-1) were obtained from the RIKEN BioResource Research Center and used in accordance with the Japanese government's hESC research guidelines. All experimental protocols and procedures were approved by the Ethics Committee of the Nagoya University Graduate School of Medicine (approval ES-001). hESC maintenance and differentiation cultures were cultured under feeder-free conditions according to the method described in Scientific Reports, 4, 3594 (2014). StemFit medium (AK03, Ajinomoto Co., Inc.) was used as the feeder-free medium, and Laminin511-E8 (Nippi Co., Ltd.) was used as the feeder-free scaffold.
As a specific maintenance culture operation, first, subconfluent hESC (KhES-1 strain) was washed with PBS and dispersed into single cells using TrypLE Select (Life Technologies). Then, the human ES cells dispersed into the single cells were seeded on a plastic culture dish coated with Laminin511-E8 and cultured feeder-free in StemFit medium in the presence of Y27632 (ROCK inhibitor, 10 μM). When a 6-well plate (Iwaki, for cell culture, culture area 9.4 cm 2 ) was used as the plastic culture dish, the seeded cell number of the human ES cells dispersed into the single cells was 2.4×10 4. One day after seeding, the entire medium was replaced with StemFit medium not containing Y27632. Thereafter, the entire medium was replaced with StemFit medium not containing Y27632 once every 1 to 2 days, and then the cells were cultured until they became subconfluent (about 60% of the culture area was covered with cells) 7 days after seeding.
分化誘導操作としては、ヒトES細胞(KhES-1株)を、StemFit培地を用いて、播種した6日後にStemFit培地の培地交換と同時にSB431542(5μM)とSAG(300nM)を添加して24時間培養した。調製したサブコンフレントのヒトES細胞を、TrypLE Select(Life Technologies社製)を用いて細胞分散液処理し、さらにピペッティング操作により単一細胞に分散した。その後、上記単一細胞に分散されたヒトES細胞を非細胞接着性の96穴培養プレート(PrimeSurface 96V底プレート、MS-9096V、住友ベークライト社製)の1ウェルあたり1.0×104細胞になるように100μLの無血清培地に浮遊させ、37℃、5%CO2の条件下で浮遊培養した。その際の無血清培地(gfCDM+KSR)には、F-12培地とIMDM培地の1:1混合液に5%KSR、450μM 1-モノチオグリセロール、1xChemically defined lipid concentrateを添加した無血清培地を用いた。浮遊培養開始時(分化誘導開始後0日目)に、前記無血清培地にY27632(終濃度20μM)、IWP-2(0.5μM)、SB431542(1μM)、JNK-IN-8(1μM)、SAG(100nM)を添加した。
As a differentiation induction operation, human ES cells (KhES-1 strain) were cultured for 24 hours using StemFit medium, 6 days after seeding, with the addition of SB431542 (5 μM) and SAG (300 nM) at the same time as changing the medium to StemFit medium. The prepared subconfluent human ES cells were treated with cell dispersion liquid using TrypLE Select (Life Technologies), and further dispersed into single cells by pipetting. Thereafter, the human ES cells dispersed into the single cells were suspended in 100 μL of serum-free medium so that the number of cells was 1.0 × 10 4 per well of a non-cell-adhesive 96-well culture plate (PrimeSurface 96V bottom plate, MS-9096V, Sumitomo Bakelite Co., Ltd.), and the suspension culture was performed under the conditions of 37 ° C. and 5% CO 2 . The serum-free medium (gfCDM+KSR) used was a 1:1 mixture of F-12 medium and IMDM medium, to which 5% KSR, 450 μM 1-monothioglycerol, and 1x chemically defined lipid concentrate had been added. At the start of suspension culture (
分化誘導開始後2日目にY27632を含まず、IWP-2(0.5μM)、SB431542(1μM)、JNK-IN-8(1μM)、BMP4(終濃度0.5nM)、SAG(終濃度700nM)を含む無血清培地を1ウェルあたり100μL加えた。その後、分化誘導開始後6、9、12、15、19、22、26日目にY27632とBMP4を含まず、IWP-2(0.5μM)、SB431542(1μM)、JNK-IN-8(1μM)とSAGを含む無血清培地を用いて半量培地交換を行った。19日目以降は、培養時の酸素分圧を40%とした。
分化誘導開始後29日目に細胞凝集体を10cm浮遊培養用ディッシュへとディッシュ1枚あたり60個の細胞凝集体を移し、無血清培地を用いて40%O2、5%CO2の条件下で浮遊培養を継続した。分化誘導開始後29日目から50日目は10%KSRを添加した無血清培地を、分化誘導開始後50日目以降は20%KSRを添加した無血清培地を用いて、培養した。
On the second day after the start of differentiation induction, 100 μL of serum-free medium containing IWP-2 (0.5 μM), SB431542 (1 μM), JNK-IN-8 (1 μM), BMP4 (final concentration 0.5 nM), and SAG (final concentration 700 nM) was added per well. Thereafter, on the 6th, 9th, 12th, 15th, 19th, 22nd, and 26th days after the start of differentiation induction, half of the medium was replaced using serum-free medium containing IWP-2 (0.5 μM), SB431542 (1 μM), JNK-IN-8 (1 μM), and SAG, without Y27632 and BMP4. From the 19th day onwards, the oxygen partial pressure during culture was set to 40%.
On the 29th day after the start of differentiation induction, the cell aggregates were transferred to 10 cm suspension culture dishes at 60 cell aggregates per dish, and suspension culture was continued using serum-free medium under conditions of 40% O2 and 5% CO2. From the 29th to 50th day after the start of differentiation induction, serum-free medium supplemented with 10% KSR was used for culture, and from the 50th day after the start of differentiation induction, serum-free medium supplemented with 20% KSR was used for culture.
前記、分化誘導開始後100日目の細胞凝集体を、それぞれ4%パラホルムアルデヒドで固定し、凍結切片を作製した。これらの凍結切片に関し、下垂体ホルモン産生細胞マーカーである副腎皮質刺激ホルモン(ACTH)(抗ACTH抗体、Fitzgerald社製、マウス)、上皮細胞マーカーであるE-cadherin(抗E-cadherin抗体、TAKARA社製、ラット)、下垂体の前駆マーカーであるLhx3(抗Lhx3抗体、自家製(非特許文献1及び2)、ウサギ)を用いて免疫染色を行った。DAPIで細胞核を染色した。これらの染色された切片を、共焦点レーザー走査型顕微鏡(オリンパス社製)を用いて観察し、免疫染色像を取得した。その結果、上記分化誘導法で誘導された分化誘導開始後100日目の細胞凝集体には、E-cadherin陽性の上皮組織の一部にACTH陽性の下垂体ホルモン産生細胞が存在していることを確認した(図1)。右下のスケールバーは100μmを示す。マウスへの移植には、分化後100~200日で採取した下垂体オルガノイド(PO)を使用した。
The cell aggregates on the 100th day after the start of differentiation induction were fixed with 4% paraformaldehyde and frozen sections were prepared. These frozen sections were immunostained using adrenocorticotropic hormone (ACTH) (anti-ACTH antibody, Fitzgerald, mouse), which is a pituitary hormone-producing cell marker, E-cadherin (anti-E-cadherin antibody, TAKARA, rat), which is an epithelial cell marker, and Lhx3 (anti-Lhx3 antibody, homemade (
(2)マウス下垂体切除術
全ての動物実験は、名古屋大学大学院医学研究科の動物実験委員会によって承認され、動物のケアと使用に関する施設のガイドラインに従って行った。8~9週齢の重症複合免疫不全症(SCID)雄マウス(C.B-17/Icr-Hsd-Prkdcscid、日本エスエルシー株式会社、静岡、日本)を、経耳下垂体切除に供した。マウスを、3つの薬剤(メデトミジン0.75mg/kg、ミダゾラム4mg/kg、ブトルファノール5mg/kg)の混合物の腹腔内(i.p.)注射で麻酔し(Kawai S, et al. Effect of three types of mixed anesthetic agents alternate to ketamine in mice. Exp Anim. 2011; 60: 481-487)、そして0.2ml生理食塩水を含む1ml注射器に取り付けた針(KN-390、株式会社夏目製作所、東京、日本)を用いて、下垂体組織を、耳道を介してトルコ鞍から吸引した。該手順の後、マウスにメデトミジン拮抗薬アチパメゾール(0.75mg/kg、i.p.)を注射した。
(2) Mouse hypophysectomy All animal experiments were approved by the Animal Care and Use Committee of the Nagoya University Graduate School of Medicine and were performed in accordance with the institutional guidelines for the care and use of animals. Male severe combined immunodeficiency (SCID) mice (CB-17/Icr-Hsd-Prkdc scid , Japan SLC Co., Ltd., Shizuoka, Japan) aged 8-9 weeks were subjected to transhypopituitary glandectomy. Mice were anesthetized with an intraperitoneal (i.p.) injection of a mixture of three drugs (medetomidine 0.75 mg/kg,
(3)採血とACTH測定
ACTHレベルを、下垂体機能のバイオマーカーとして評価した。血液サンプルを、尾の切断によって採取し、ヒトCRH(2μg/kg、i.p.、タナベ、ニプロESファーマ株式会社、大阪、日本)の投与の前と1時間後にサンプリングした。血漿を、1000×gで15分間、4℃で遠心分離することで、血液サンプルから分離した。血漿ACTHアッセイはACTH ELISAキット(MD bioproducts、Oakdale、MN)を使用し、吸光度(ACTH濃度)を、Cytation 5(Biotek、Winooski、VT)を用いて読み取った。CRH負荷テストを、下垂体切除術の1週間後、及びPO移植治療の1/~4週間後に、偽手術マウスを含めて6か月後まで実施した(図2A)。CRH刺激後の血漿ACTHレベルが10pg/ml未満のマウスを下垂体機能低下症として分類し、被験者として使用した。
(3) Blood sampling and ACTH measurement ACTH levels were evaluated as a biomarker of pituitary function. Blood samples were collected by tail snip and sampled before and 1 h after administration of human CRH (2 μg/kg, i.p., Tanabe, Nipro ES Pharma Co., Ltd., Osaka, Japan). Plasma was separated from the blood samples by centrifugation at 1000×g for 15 min at 4°C. Plasma ACTH assay was performed using an ACTH ELISA kit (MD bioproducts, Oakdale, MN), and absorbance (ACTH concentration) was read using a Cytation 5 (Biotek, Winooski, VT). CRH loading tests were performed 1 week after hypophysectomy and 1/4 weeks after PO transplantation treatment, including sham-operated mice, up to 6 months later (Figure 2A). Mice with plasma ACTH levels below 10 pg/ml after CRH stimulation were classified as hypopituitaric and used as subjects.
(4)In vitroでのPOの自発的ACTH分泌の測定
5つのPOを2.5mlの細胞培養培地(Iscove改変ダルベッコ培地(Sigma-Aldrich、St.Louis、MO);Ham’s F12(Thermo Fisher Scientific、Waltham、MA)(1:1);1% GlutaMAX(Thermo Fisher Scientific);1% Chemically Defined Lipid Concentrate(Thermo Fisher Scientific);450μM 1-thioglycerol(Sigma-Aldrich);及び20% KnockOut Serum Replacement(Thermo Fisher Scientific)にて、37℃で72時間インキュベートした。培養上清を回収した。上清中のACTH濃度を、日本で臨床的に使用されている電気化学発光免疫測定法(ECLIA)キット(株式会社エスアールシー、東京、日本)を使用して決定した。
(4) Measurement of spontaneous ACTH secretion in PO in vitro Five POs were cultured in 2.5 ml of cell culture medium (Iscove's modified Dulbecco's medium (Sigma-Aldrich, St. Louis, MO); Ham's F12 (Thermo Fisher Scientific, Waltham, MA) (1:1); 1% GlutaMAX (Thermo Fisher Scientific); 1% Chemically Defined Lipid Concentrate (Thermo Fisher Scientific); 450 μM 1-thioglycerol (Sigma-Aldrich); and 20% The cells were incubated in KnockOut Serum Replacement (Thermo Fisher Scientific) at 37°C for 72 hours. The culture supernatant was collected. The ACTH concentration in the supernatant was determined using an electrochemiluminescence immunoassay (ECLIA) kit (SRC Co., Ltd., Tokyo, Japan) that is used clinically in Japan.
(5)移植方法
マウスをイソフルランで麻酔し、仰臥位に置いた。鼠蹊部の皮下白色脂肪組織(ISWAT)移植では、左鼠径部を剃毛した。4mmの垂直皮膚切開を行い、皮下脂肪組織にポケットを作製し、顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った(図2B~G)。偽手術群では、左鼠蹊部に垂直皮膚切開を行い、皮下脂肪に小さなポケットを作製し、PO移植なしで外科的創閉鎖を行った。無血管領域(AR)移植群では、左背部皮膚に垂直皮膚切開を行い、5つのPOを配置した。全ての下垂体機能低下マウスは、副腎クリーゼを予防するために、0.2mg/0.61ml/マウスのデキサメタゾンの筋肉内注射に供した。
(5) Transplantation Method Mice were anesthetized with isoflurane and placed in a supine position. For subcutaneous white adipose tissue (ISWAT) transplantation in the groin, the left groin was shaved. A 4 mm vertical skin incision was made, a pocket was created in the subcutaneous adipose tissue, and five POs harvested from cell culture were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed following nylon suture closure on the PO (Fig. 2B-G). In the sham operation group, a vertical skin incision was made in the left groin, a small pocket was created in the subcutaneous fat, and surgical wound closure was performed without PO transplantation. In the avascular area (AR) transplantation group, a vertical skin incision was made in the left dorsal skin, and five POs were placed. All hypopituitarized mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
(6)POを移植したマウスの評価
体重を追跡し、体重減少率を評価した。ランニングホイールアクティビティテストを、ランニングホイール デバイス(ENV-044; Med Associates、Georgia、VT)を用いて行った。
(6) Evaluation of PO-implanted mice Body weight was monitored and the rate of weight loss was evaluated. Running wheel activity test was performed using a running wheel device (ENV-044; Med Associates, Georgia, VT).
(7)組織学的評価
SCIDマウスから移植した細胞凝集体、皮膚、脂肪を10%ホルマリンで固定し、パラフィン浸透のために脱水し、スライドミクロトームで切片にした。5μmの切片をヘマトキシリンエオシン(H&E)で染色するか、核の4’,6-ジアミジノ-2-フェニルインドール対比染色による様々な抗原の免疫蛍光顕微鏡法に供した。抗原標的には、ACTH(マウス、1:200、10C-CR1096M1;Fitzgerald)、LHX3(LIMホメオボックスタンパク質3、ウサギ、1:3000、自家製(非特許文献1及び2))、ヒト核(マウス、1:1000、MAB4383;Millipore)、E-カドヘリン(ラット、1:50、M108;タカラバイオ株式会社)及びSMA(平滑筋アクチン、マウス、1:200、M0851;DAKO)を含む。
(7) Histological Evaluation: Cell aggregates, skin, and fat from SCID mice were fixed in 10% formalin, dehydrated for paraffin infiltration, and sectioned on a slide microtome. Five-μm sections were stained with hematoxylin and eosin (H&E) or subjected to immunofluorescence microscopy for various antigens with nuclear 4',6-diamidino-2-phenylindole counterstaining. Antigen targets include ACTH (mouse, 1:200, 10C-CR1096M1; Fitzgerald), LHX3 (
(8)PO及び未分化hESCからのRNA抽出及びcDNA合成
製造元の指示に従って、RNeasy Mini Kit(Qiagen、Hilden、Germany)を用いて、PO及び未分化hESCからRNAを抽出した。RNAの品質を、TapeStation 4150(Agilent Technologies、Santa Clara、CA)を用いて評価した。ReverTra Ace qPCR RT Master Mix with gDNA Remover(東洋紡、大阪、日本)を用いてcDNAを合成した。
(8) RNA extraction and cDNA synthesis from PO and undifferentiated hESC RNA was extracted from PO and undifferentiated hESC using RNeasy Mini Kit (Qiagen, Hilden, Germany) according to the manufacturer's instructions. RNA quality was assessed using TapeStation 4150 (Agilent Technologies, Santa Clara, CA). cDNA was synthesized using ReverTra Ace qPCR RT Master Mix with gDNA Remover (Toyobo, Osaka, Japan).
(9)定量PCR
LightCycler 480 システム(Roche Diagnostics、Rotkreuz、Switzerland)を用いて、5つのPO及び5つの移植したPO(10,000 ESC/サンプルから区別)の定量PCR(qPCR)を行った。データを、内因性コントロールとしてGAPDHのデータに正規化し、標準曲線に基づく相対定量を使用して決定した。用いたプライマーを、以下の表1に示す。
(9) Quantitative PCR
Quantitative PCR (qPCR) was performed on five POs and five transplanted POs (separated from 10,000 ESCs/sample) using a LightCycler 480 system (Roche Diagnostics, Rotkreuz, Switzerland). Data were normalized to that of GAPDH as an endogenous control and relative quantification based on a standard curve was determined. The primers used are shown in Table 1 below.
(10)統計解析
全てのデータを、SPSS統計ソフトウェア(バージョン 28.0.0.0、IBM、Armonk、NY)を用いて解析した。データは、平均±標準誤差として示す。2群間の比較は、スチューデントのt検定を用いて行った。多群間の比較は、事後テューキー検定を用いた一元配置分散分析(one-way ANOVA)によって行った。<0.05(*)、<0.01(**)、及び<0.001(***)のP値を有意とみなした。
(10) Statistical Analysis All data were analyzed using SPSS statistical software (version 28.0.0.0, IBM, Armonk, NY). Data are presented as mean ± standard error. Comparison between two groups was performed using Student's t-test. Comparison between multiple groups was performed by one-way analysis of variance (ANOVA) with post-hoc Tukey's test. P values of <0.05 ( * ), <0.01 ( ** ), and <0.001 ( *** ) were considered significant.
<結果>
(1)皮下移植法の評価
以下の4つの方法:
(i)ヘパリンを含む塩基性線維芽細胞増殖因子(FGF)を含有するゼラチンヒドロゲル持続放出デバイス(Uematsu SS, et al. The optimization of the prevascularization procedures for improving subcutaneous islet engraftment. Transplantation. 2018; 102: 387-395)(図3A、B:GEL)又は(ii)医療上承認された血管アクセスカテーテル(Pepper AR, et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat Biotechnol. 2015; 33: 518-523)(図3C、デバイスレス、DL);
(iii)ISWATでの移植片配置(Yasunami Y, et al. A novel subcutaneous site of islet transplantation superior to the liver. Transplantation. 2018; 102: 945-952)(図3D、E);及び
(iv)背中の皮膚の直下の比較的ARでの移植片配置(図3F、no treatment:NT)を行った。皮下移植において、血液供給が移植片の運命を決定すると仮定した。ARでの配置は、他の3つのオプションの対照としての役割を果たす。脂肪組織は本質的に血管に富むが、GEL及びDL血管新生誘導(pre-vascularization)方法は移植部位への血液供給を増加させる。しかしながら、これらの方法は、移植の2週間前又は1か月前にそれぞれ侵襲的な操作を必要とし、下垂体機能低下マウスにストレスを与える可能性がある。hESC由来POの移植後1か月の血漿ACTHレベルは、ISWATコホートマウスで最も高かった(PO培養培地のACTHレベル30100pg/ml;全て同じロットから移植されたPO)。従って、ISWAT移植が最もよいと考えられた(図3G、H)。すなわち、下垂体の投与方法として、鼠径部移植及び/又は皮下白色脂肪移植が良いことが分かった。
<Results>
(1) Evaluation of subcutaneous implantation methods The following four methods:
(i) a gelatin hydrogel sustained release device containing basic fibroblast growth factor (FGF) with heparin (Uematsu SS, et al. The optimization of the prevascularization procedures for improving subcutaneous islet engraftment. Transplantation. 2018; 102: 387-395) (Fig. 3A,B: GEL) or (ii) a medically approved vascular access catheter (Pepper AR, et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat Biotechnol. 2015; 33: 518-523) (Fig. 3C, device-less, DL);
(iii) graft placement in ISWAT (Yasunami Y, et al. A novel subcutaneous site of islet transplantation superior to the liver. Transplantation. 2018; 102: 945-952) (Fig. 3D, E); and (iv) graft placement in the relative AR just under the skin on the back (Fig. 3F, no treatment: NT). In subcutaneous transplantation, we hypothesized that blood supply determines the fate of the graft. Placement in the AR serves as a control for the other three options. Although adipose tissue is inherently highly vascularized, the GEL and DL pre-vascularization methods increase blood supply to the transplantation site. However, these methods require
(2)ISWAT、偽手術、及びAR群間の比較
ISWAT群(n=6)、偽手術群(n=6)、及びAR群(n=5)の血漿ACTHレベルを6か月後まで追跡調査した。AR群は、上記のように、血管分布の進んだISWATの研究の対照として機能する。移植前の基礎値血漿ACTHレベルとCRH刺激した血漿ACTHレベルは、3群間で有意差はなかった(ISWAT対偽対AR、基礎;4.6±1.8pg/ml対2.3±1.6pg/ml対2.5±1.1pg/ml、p=0.27-0.92、刺激;1.7±0.8pg/ml対1.7±2.3pg/ml対7.2±2.1pg/ml、p=0.06-0.72)。
(2) Comparison between ISWAT, sham, and AR groups Plasma ACTH levels were followed up for 6 months in the ISWAT group (n=6), sham group (n=6), and AR group (n=5). The AR group served as a control for the study of the highly vascularized ISWAT, as described above. Pre-implant basal and CRH-stimulated plasma ACTH levels were not significantly different among the three groups (ISWAT vs. sham vs. AR, basal; 4.6±1.8pg/ml vs. 2.3±1.6pg/ml vs. 2.5±1.1pg/ml, p=0.27-0.92; stimulated; 1.7±0.8pg/ml vs. 1.7±2.3pg/ml vs. 7.2±2.1pg/ml, p=0.06-0.72).
移植後、基礎値血漿ACTHレベル(「basal」、図4A)は、偽手術群よりもISWAT群の方が一貫して高かった。移植後2、4、8、及び17週で、ACTH値は群間で有意に異なった(p<0.001-0.05、図4A)。CRH刺激した血漿ACTHレベル(「stimulated」、図4A)も、偽手術群よりもISWAT群の方が高く、2、4、8、21、及び26週で群間に統計的に有意な差があった(p<0.001-0.005、図4A)。 After transplantation, basal plasma ACTH levels ("basal", Figure 4A) were consistently higher in the ISWAT group than in the sham group. At 2, 4, 8, and 17 weeks after transplantation, ACTH values were significantly different between groups (p<0.001-0.05, Figure 4A). CRH-stimulated plasma ACTH levels ("stimulated", Figure 4A) were also higher in the ISWAT group than in the sham group, with statistically significant differences between groups at 2, 4, 8, 21, and 26 weeks (p<0.001-0.005, Figure 4A).
PO培養液中のACTHレベルによって評価したin vitroでの移植前ACTH分泌は、ISWAT群とAR群の間で有意差はなかった(ISWAT対AR、44416±8435pg/ml対45800±17291pg/ml、p=0.876)。移植後、基礎値血漿ACTHレベル及びCRH刺激した血漿ACTHレベルは、AR群で偽手術群より高く、またISWAT群より低く、2週間でのISWAT群の基礎値血漿ACTHレベルは、AR群より有意に高く(p=0.009)、そして2、8、及び21週間でのISWAT群のCRH刺激した血漿ACTHレベルは、AR群よりも有意に高かった(p<0.001-0.013)(図4A)。これらの結果は、脂肪組織などの血管が発達した部位に移植された場合、血管が発達していない部位よりも、皮下組織においてPOがより効率的にACTHを放出することを示した。 In vitro pre-transplant ACTH secretion, assessed by ACTH levels in PO culture medium, was not significantly different between the ISWAT and AR groups (ISWAT vs. AR, 44,416 ± 8,435 pg/ml vs. 45,800 ± 17,291 pg/ml, p = 0.876). After transplantation, basal and CRH-stimulated plasma ACTH levels were higher in the AR group than in the sham-operated group and lower than in the ISWAT group, basal plasma ACTH levels in the ISWAT group were significantly higher than those in the AR group at 2 weeks (p = 0.009), and CRH-stimulated plasma ACTH levels in the ISWAT group were significantly higher than those in the AR group at 2, 8, and 21 weeks (p < 0.001-0.013) (Figure 4A). These results showed that when PO was implanted in a vascularized site such as adipose tissue, it released ACTH more efficiently in the subcutaneous tissue than in a site with poor blood flow.
ランニングホイールアクティビティテストでは、偽手術群よりもISWAT群でより大きな活動が見られた(図4B)。ISWAT群の体重減少率は、偽手術群と比較して少量であった(図4C)。 In the running wheel activity test, the ISWAT group showed greater activity than the sham group (Figure 4B). The weight loss rate in the ISWAT group was smaller than that in the sham group (Figure 4C).
(3)皮下移植後の肉眼的及び組織学的所見
ISWAT移植後4週間の肉眼検査では、移植片血管新生が明らかであった(図5A、移植した細胞凝集体:図中矢印、移植片に関連する血管:図中矢じり)。移植後21週目に、肉眼的に脂肪組織は観察されなかったが、移植片は損傷を受けていないように見えた(図5B、図中矢印)。移植後21週目に纏めて採取した皮膚とその下の移植片の顕微鏡検査では、移植した細胞凝集体は皮下組織内にあった(図5C、I、図中黒ボックス:図5D-N)。蛍光免疫顕微鏡法により、移植片がACTH、E-カドヘリン(口腔外胚葉のマーカー)、及びLHX3(下垂体前駆細胞マーカー)を発現していることが明らかになった(図5D、E、F、J、K、L)。ヒト核の同時発現は、当該細胞が移植したhESC由来下垂体細胞であることを示した(図5F、G、L、M)。さらに、血管壁マーカーであるSMAは、移植片の周囲及び内部にクラスターとして発現し(図5H、N)、血管新生を示しす。これらの観察結果は、hESC由来のPOがin vivoで生着して機能することを示す。対照的に、AR移植の後継物の免疫染色では生着が見られたが、ISWAT移植よりも生存可能な領域が少なく、そしてSMAの発現が少なかった(図6、図6A右上ボックス:図6B-F、図6A左下ボックス:図6G-K)。図5および図6の右下のスケールバーは100μmを示す。
(3) Macroscopic and histological findings after subcutaneous transplantation
(4)hESC由来POの血管新生の促進
hESC由来のPOが皮下移植後に機能することを認め、血管新生が皮下移植されたPOの生着にどのように影響するかという疑問が生じた。血管内皮増殖因子(VEGF)、塩基性線維芽細胞増殖因子(FGF2)、及びアンジオポエチン2(ANGPT2)は、初期段階の血管新生に関連している(Carmeliet P, et al. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011; 473: 298-307、Ortega S, et al. Neuronal defects and delayed wound healing in mice lacking fibroblast growth factor 2. Proc Natl Acad Sci U S A. 1998; 95: 5672-5677、及びNag S, et al. Angiopoietins are expressed in the normal rat pituitary gland. Endocr Pathol. 2005; 16: 67-73)。これらの遺伝子が、POに発現しているか否かをqPCRで評価した。移植前のPO、移植12時間後に採取したPO、及び対照として未分化hESCを分析した。POのVEGFA、VEGFB、VEGFC、及びANGPT2の発現レベルは、未分化hESCよりも有意に高かった。特にVEGFCとANGPT2の発現レベルは、移植後に上昇した(図7A~E)。これらの結果は、PO自体が血管新生因子を発現し、皮下組織への生着を促進する可能性があることを示唆していた。
(4) Promotion of angiogenesis of hESC-derived PO Having observed that hESC-derived PO functions after subcutaneous transplantation, the question arose as to how angiogenesis affects the engraftment of subcutaneously transplanted PO. Vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF2), and angiopoietin 2 (ANGPT2) are involved in the early stages of angiogenesis (Carmeliet P, et al. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011; 473: 298-307, Ortega S, et al. Neuronal defects and delayed wound healing in mice lacking
最後に、皮下PO移植片の生存における血管新生の重要性をさらに評価するために、VEGF阻害剤であるベバシズマブ(2mg/kg、腹腔内、2x/週、2週間(Lin Y, et al. A strategy of vascular-targeted therapy for liver fibrosis. Hepatology. 2022; 76: 660-675);Selleck、Houston、TX)を投与することにより、in vivoでPOの機能を試験した。ISWAT移植後の下垂体機能低下症SCIDマウスをベバシズマブ投与群(n=9)とビヒクル投与群(n=9)に分けた。移植に用いたPOの培地中のACTHレベルは、群間で有意差はなかったが(ベバシズマブ対ビヒクル、19445±6774pg/ml対20295±6610pg/ml、p=0.93)、CRH刺激後の血漿ACTHレベルは、ビヒクル群よりもベバシズマブ群で有意に低かった(4.6±3.0pg/ml対34.5±11.7pg/ml、p=0.035)。上記結果から、血管新生がISWATの生着PO機能にとって重要であることが示唆される(図7F)。 Finally, to further evaluate the importance of angiogenesis in the survival of subcutaneous PO grafts, we tested PO function in vivo by administering the VEGF inhibitor bevacizumab (2 mg/kg, i.p., 2x/week for 2 weeks (Lin Y, et al. A strategy of vascular-targeted therapy for liver fibrosis. Hepatology. 2022; 76: 660-675); Selleck, Houston, TX). Hypopituitaric SCID mice after ISWAT transplantation were divided into bevacizumab-treated (n=9) and vehicle-treated (n=9) groups. Although ACTH levels in the culture medium of POs used for transplantation were not significantly different between groups (bevacizumab vs. vehicle, 19445±6774 pg/ml vs. 20295±6610 pg/ml, p=0.93), plasma ACTH levels after CRH stimulation were significantly lower in the bevacizumab group than in the vehicle group (4.6±3.0 pg/ml vs. 34.5±11.7 pg/ml, p=0.035). These results suggest that angiogenesis is important for ISWAT engraftment PO function (Figure 7F).
本実施例では、hESC由来のPOを下垂体機能低下症SCIDマウスの皮下組織に移植し、そして機能し得ることを実証した。移植したマウスは循環へのACTHの放出でCRHに応答した。これは、注入したCRHが移植したACTH産生細胞を刺激したことを示す。ISWAT移植とAR移植との比較では、血管が豊富な皮下脂肪組織は、背中の皮膚の下の比較的血管の少ない位置のスペースよりも優れており、より持続的なACTH分泌が得られることを示した。PSC由来のPO移植は、身体活動レベルと体重を改善する(Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells. Nat Commun. 2016; 7: 10351、及びSuga H, et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature. 2011; 480: 57-62)。本実施例ではこれを確認した。さらに、hESC由来のPOは、VEGFやANGPT2などの血管新生因子を発現し、POが自律的に血管新生と生着を促進することを示唆していた。VEGFC及びANGPT2発現のレベルは、移植後のPOで上昇した。これは、おそらく、脂肪細胞から放出されるサイトカインであるアディポネクチンがこれに寄与している(Sakata N, et al. Mechanism of transplanted islet engraftment in visceral white adipose tissue. Transplantation. 2020; 104: 2516-2527)。最後に、ベバシズマブ投与によってACTH分泌が減少したことは、少なくとも皮下移植において血管新生が重要であることを示している。これらの結果は、皮下部位が選択されている場合、hESC由来のPOの脂肪組織内移植を達成し得ることをサポートしている。 In this example, we demonstrate that hESC-derived POs can be transplanted into the subcutaneous tissue of hypopituitaric SCID mice and are functional. Transplanted mice responded to CRH with the release of ACTH into the circulation, indicating that the injected CRH stimulated the transplanted ACTH-producing cells. Comparison of ISWAT and AR transplants demonstrated that the vascularized subcutaneous adipose tissue was superior to the relatively vascularized space under the skin on the back, resulting in more sustained ACTH secretion. PSC-derived PO transplantation improves physical activity levels and body weight (Ozone C, et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells. Nat Commun. 2016; 7: 10351; and Suga H, et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature. 2011; 480: 57-62). This was confirmed in the present study. Furthermore, hESC-derived PO expressed angiogenic factors such as VEGF and ANGPT2, suggesting that PO autonomously promotes angiogenesis and engraftment. The levels of VEGFC and ANGPT2 expression were increased in PO after transplantation. This is likely due to the contribution of adiponectin, a cytokine released by adipocytes (Sakata N, et al. Mechanism of transplanted islet engraftment in visceral white adipose tissue. Transplantation. 2020; 104: 2516-2527). Finally, bevacizumab administration reduced ACTH secretion, indicating that angiogenesis is important, at least in subcutaneous transplantation. These results support that intraadipose tissue engraftment of hESC-derived POs may be achievable if the subcutaneous site is selected.
本実施例における皮下移植したPSC由来のPOにおける機能の実証は、再生医療技術における重要なステップである。腎被膜下移植と皮下移植は大きく異なる。POのレシピエント患者は下垂体機能低下症に苦しんでいるため、侵襲的処置などのストレスに敏感である。腎被膜下移植と皮下移植の両方に全身麻酔が必要であるが、ヒトでは、皮下移植は局所麻酔下で行うことができ、別の方法よりも侵襲性が大幅に低くなる。腎被膜下移植は、正常な腎臓に損傷を与える可能性がある。対照的に、皮下移植中の付随的損傷のリスクは低い。手術自体は簡単で、マウスでもヒトでも迅速に行うことができるため、おそらく外来での作業が可能である。PSC由来細胞の移植で重要なのは、腫瘍が発生した場合の移植片の除去である。本発明において採用した皮下移植は、比較的単純で非侵襲的な除去を可能にする。本実施例で実証したように、無血管部位(AR)への移植よりも脂肪組織への移植の方が効果的であることは、臨床応用にとって重要である。 The demonstration of functionality in subcutaneously transplanted PSC-derived POs in this example is an important step in regenerative medicine technology. Subrenal capsule transplantation and subcutaneous transplantation are significantly different. PO recipient patients suffer from hypopituitarism and are therefore sensitive to stresses such as invasive procedures. Both subrenal capsule transplantation and subcutaneous transplantation require general anesthesia, but in humans, subcutaneous transplantation can be performed under local anesthesia, making it significantly less invasive than the alternative methods. Subrenal capsule transplantation can cause damage to normal kidneys. In contrast, the risk of collateral damage during subcutaneous transplantation is low. The surgery itself is simple and can be performed quickly in both mice and humans, likely allowing for outpatient work. A key aspect of transplantation of PSC-derived cells is the removal of the graft in the event of tumor development. Subcutaneous transplantation as employed in this invention allows for relatively simple and non-invasive removal. As demonstrated in this example, transplantation into adipose tissue is more effective than transplantation into an avascular site (AR), which is important for clinical application.
実施例2:下垂体ホルモン産生細胞の移植部位のさらなる検討(鼠径部皮下、腋窩皮下、筋肉)
<材料及び方法>
実施例1と同様の分化誘導法を用いてhESC由来POを作製した。マウスへの移植には、分化後90-100日で採取したPOを使用した。
Example 2: Further investigation of transplantation sites of pituitary hormone-producing cells (inguinal subcutaneous, axillary subcutaneous, muscle)
Materials and Methods
hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
(1)移植方法
実施例1と同様にマウスに下垂体切除術を施し、採血およびACTH測定を行った。実施例1と同様に、マウスに鼠径部のISWAT移植(Inguinal)を行った。腋窩の皮下白色脂肪組織移植(Axilla)では、イソフルラン吸入麻酔下に、マウスを仰臥位に置き、左腋窩部を剃毛した。4mmの垂直皮膚切開を行い、皮下脂肪組織にポケットを作製し、顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。臀部筋肉内移植(Muscle)では、イソフルラン吸入麻酔下に、マウスを腹臥位に置き、左臀部を剃毛した。4mmの垂直皮膚切開を行い、臀部の筋肉にポケットを作製し、顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。全ての下垂体機能低下マウスは、副腎クリーゼを予防するために、0.2mg/0.61ml/マウスのデキサメタゾンの筋肉内注射に供した。
(1) Transplantation Method As in Example 1, mice were hypophysectomized, and blood samples were taken and ACTH was measured. As in Example 1, mice were subjected to inguinal ISWAT transplantation (Inguinal). For axillary subcutaneous white adipose tissue transplantation (Axilla), mice were placed in a supine position and the left axillary region was shaved under isoflurane inhalation anesthesia. A 4 mm vertical skin incision was made, a pocket was made in the subcutaneous adipose tissue, and five POs taken from cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Nylon suture closure on the PO was followed by skin closure. For gluteal intramuscular transplantation (Muscle), mice were placed in a prone position and the left gluteal region was shaved under isoflurane inhalation anesthesia. A 4 mm vertical skin incision was made, a pocket was made in the gluteal muscle, and five POs taken from cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed followed by nylon suture closure over the PO. All hypopituitaric mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
(2)統計解析
全てのデータを、R(Ver 4.3.0)を用いて解析した。データは、平均±標準誤差として示す。多群間の比較は、クリスカルウォリス検定によって行った。<0.05(*)のP値を有意とみなした。
(2) Statistical Analysis All data were analyzed using R (Version 4.3.0). Data are presented as mean ± standard error. Comparisons between multiple groups were performed using the Criskall-Wallis test. A P value of <0.05 ( * ) was considered significant.
<結果>
鼠径部のISWATに加えて適している移植部位を評価するために、Inguinal群(n=2)、Axilla群(n=3)、及びMuscle群(n=4)の血漿ACTHレベルを6か月後まで追跡調査した(図8)。移植前の基礎値血漿ACTHレベルとCRH刺激した血漿ACTHレベルは、3群間で有意差はなかった。(鼠径部白色脂肪組織対腋窩部白色脂肪組織対臀部筋肉内、基礎;27.81±13.20pg/ml対14.58±4.52pg/ml対23.00±8.93pg/ml、p=0.717、刺激;61.33±23.91pg/ml対44.40±14.35pg/ml対53.58±14.85pg/ml、p=0.76)。腋窩皮下白色脂肪組織移植群、筋肉内移植群は、ISWAT群と比較しても同等であった。すなわち、ISWATに加えて腋窩皮下白色脂肪組織移植、筋肉内移植も移植部位として適していると考えられる。
<Results>
To evaluate suitable transplantation sites in addition to the ISWAT in the groin, plasma ACTH levels were followed up for 6 months in the Inguinal (n=2), Axilla (n=3), and Muscle (n=4) groups (Figure 8). Pre-transplant basal and CRH-stimulated plasma ACTH levels were not significantly different among the three groups (inguinal white adipose tissue vs. axillary white adipose tissue vs. gluteal intramuscular, basal: 27.81±13.20 pg/ml vs. 14.58±4.52 pg/ml vs. 23.00±8.93 pg/ml, p=0.717; stimulated: 61.33±23.91 pg/ml vs. 44.40±14.35 pg/ml vs. 53.58±14.85 pg/ml, p=0.76). The axillary subcutaneous white adipose tissue transplantation group and intramuscular transplantation group were comparable when compared with the ISWAT group. In other words, in addition to ISWAT, axillary subcutaneous white adipose tissue transplantation and intramuscular transplantation are also suitable transplantation sites.
その後、n数を増やし再現性を確認した。Axilla群は2匹、Inguinal群は1匹、Muscle群は2匹追加した。Axilla群(n=5)、Inguinal群(n=3)、及びMuscle群(n=6)の血漿ACTHレベルを6か月後まで追跡調査した。移植前の基礎値血漿ACTHレベルとCRH刺激した血漿ACTHレベルは、n数を増やしても3群間で有意差はなかった。(腋窩部白色脂肪組織対鼠径部白色脂肪組織対臀部筋肉内、基礎;15.17±3.58pg/ml対19.05±9.39pg/ml対32.23±9.41pg/ml、p=0.6771、刺激;38.83±9.11pg/ml対48.87±17.97pg/ml対83.50±21.93pg/ml、p=0.6269)。 Then, the n number was increased to confirm reproducibility. Two animals were added to the Axilla group, one to the Inguinal group, and two to the Muscle group. Plasma ACTH levels in the Axilla group (n=5), Inguinal group (n=3), and Muscle group (n=6) were followed up for 6 months. There were no significant differences between the basal plasma ACTH levels before transplantation and CRH-stimulated plasma ACTH levels among the three groups, even when the n number was increased. (Axillary white adipose tissue vs. inguinal white adipose tissue vs. gluteal intramuscular tissue, basal: 15.17±3.58 pg/ml vs. 19.05±9.39 pg/ml vs. 32.23±9.41 pg/ml, p=0.6771, stimulated: 38.83±9.11 pg/ml vs. 48.87±17.97 pg/ml vs. 83.50±21.93 pg/ml, p=0.6269).
実施例3:精製後の細胞凝集体(精製体)の移植検討(鼠径部皮下)
<材料及び方法>
(1)hESC由来POから下垂体ホルモン産生細胞(精製体)への精製
実施例1と同様の分化誘導法で得られた、分化誘導開始後64-76日目のhESC由来POで、抗EpCAM抗体を用いたセルソーティング(精製)を実施した。セルソーティングとしては、1.前処理、2.細胞凝集体の分散、3.EpCAM陽性細胞の分離の3つの工程にわけて実施した。
Example 3: Examination of transplantation of purified cell aggregates (purified bodies) (subcutaneously in the groin)
Materials and Methods
(1) Purification of pituitary hormone-producing cells (purified bodies) from hESC-derived PO Cell sorting (purification) using anti-EpCAM antibody was carried out on hESC-derived PO on days 64-76 after the start of differentiation induction, which was obtained by the same differentiation induction method as in Example 1. The cell sorting was carried out in three steps: 1. pretreatment, 2. dispersion of cell aggregates, and 3. separation of EpCAM-positive cells.
1.Y27632による前処理
分散処理による細胞死を抑制するために、作業日の前日にY27632(20μM)を培地に添加し、細胞凝集体を前処理した。セルソーティング前までの以降の操作では、細胞が曝されるすべての溶液に20μM Y27632を添加した。
1. Pretreatment with Y27632 To suppress cell death due to dispersion treatment, Y27632 (20 μM) was added to the medium on the day before the operation to pretreat the cell aggregates. In the subsequent operations before cell sorting, 20 μM Y27632 was added to all solutions to which the cells were exposed.
2.細胞凝集体の分散
酵素処理による分散を促進するために、細胞凝集体にメスで細断または切込みを入れた。次に細胞凝集体を、PBSにて洗浄後、神経細胞分散キットの分散液(Wako社製)を加えて、37℃で30分酵素処理を行い、さらにピペッティング操作により細胞凝集体をほぐした。反応終了後、DMEM/F12(Wako社製)にて洗浄し、コラゲナーゼ溶液を加えて、37℃で30分旋回振盪(140~150rpm)した。コラゲナーゼ溶液の組成は、上記のDMEM/F12に0.2%コラゲナーゼtype I(Wako社製)および0.1%BSA(Themofisher社製)を添加したものである。コラゲナーゼ処理終了後、PBSにて洗浄し、10×TrypLE Select溶液(Themofisher社製)+0.2mg/ml DNaseI(Roche社製)を加えて、37℃で10分酵素処理後、さらにピペッティング操作により単一細胞に分散した。20%KSRを添加した無血清培地にて懸濁・中和し、遠心後に10μg/ml DnaseI、20% KSRを添加した無血清培地で再懸濁した単一細胞を、70μmセルストレイナーに通して凝集塊を取り除いた。
2. Dispersion of cell aggregates To promote dispersion by enzyme treatment, the cell aggregates were chopped or cut with a scalpel. Next, the cell aggregates were washed with PBS, and then the dispersion liquid of the nerve cell dispersion kit (manufactured by Wako) was added, and the enzyme treatment was performed at 37°C for 30 minutes, and the cell aggregates were further loosened by pipetting. After the reaction was completed, the cells were washed with DMEM/F12 (manufactured by Wako), and a collagenase solution was added and the cells were swirled and shaken (140-150 rpm) at 37°C for 30 minutes. The composition of the collagenase solution was the above-mentioned DMEM/F12 to which 0.2% collagenase type I (manufactured by Wako) and 0.1% BSA (manufactured by Thermofisher) were added. After collagenase treatment, the cells were washed with PBS, and 10x TrypLE Select solution (Thermofisher) + 0.2 mg/ml DNase I (Roche) was added and treated with the enzyme at 37°C for 10 minutes, after which the cells were dispersed into single cells by pipetting. The cells were suspended and neutralized in serum-free medium containing 20% KSR, centrifuged, and then resuspended in serum-free medium containing 10 μg/ml DNase I and 20% KSR. The single cells were passed through a 70 μm cell strainer to remove aggregates.
3.FACSによるEpCAM陽性細胞の分離
上記で得られた単一細胞を、遠心後に1mM EDTA、1%FBSを添加したDMEM/F-12溶液に懸濁させた。PE標識抗EpCAM抗体(Miltenyi社製)を添加し、4℃で10分インキュベートし、蛍光活性化セルソーティング(FACS)を行い、EpCAM陽性細胞を分離・回収した。FACSのためのセルソーターとして、Stream-In-Air方式の機種であるBD社のFACS Aria Fusionを用いた。
精製したEpCAM陽性細胞を、非細胞接着性細胞非接着性96ウェル培養プレートの1ウェルあたり10×104細胞になるように200μLの20%KSR、20μM Y27632を添加した無血清培地で再凝集させ、37℃、40%O2、5%CO2の条件下で浮遊培養した。浮遊培養開始後3日目以降は、Y27632を含まず、20%KSRを添加した無血清培地を用いて、3日~4日に一度の半量交換を行い、長期培養した。
3. Separation of EpCAM-positive cells by FACS The single cells obtained above were suspended in a DMEM/F-12 solution containing 1 mM EDTA and 1% FBS after centrifugation. PE-labeled anti-EpCAM antibody (Miltenyi) was added, incubated at 4°C for 10 minutes, and fluorescence-activated cell sorting (FACS) was performed to separate and collect EpCAM-positive cells. A BD FACS Aria Fusion, a Stream-In-Air type cell sorter, was used as the cell sorter for FACS.
The purified EpCAM-positive cells were reaggregated in 200 μL of serum-free medium supplemented with 20% KSR and 20 μM Y27632 to give 10×10 4 cells per well of a non-adhesive 96-well culture plate, and cultured in suspension at 37° C., 40% O 2 , and 5% CO 2. From the third day after the start of suspension culture, serum-free medium supplemented with 20% KSR without Y27632 was used, and half of the medium was replaced every 3 to 4 days for long-term culture.
(2)移植方法
マウスへの移植には、分化後137~144日で採取した精製した下垂体ホルモン産生細胞を使用した。実施例1と同様にマウスに下垂体切除術を施し、採血およびACTH測定を行った。鼠径部のISWAT移植は、実施例1と同様に行った。全ての下垂体機能低下マウスは、副腎クリーゼを予防するために、0.2mg/0.61ml/マウスのデキサメタゾンの筋肉内注射に供した。
(2) Transplantation Method Purified pituitary hormone-producing cells harvested 137-144 days after differentiation were used for transplantation into mice. Mice were hypophysectomized as in Example 1, and blood samples were collected and ACTH was measured. ISWAT transplantation into the groin was performed as in Example 1. All hypopituitarism mice were given an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
<結果>
精製した下垂体ホルモン産生細胞をISWATに移植し(n=2)、血漿ACTHレベルを3か月後まで追跡調査した(図9)。移植後、移植術前よりも基礎値血漿ACTH濃度の改善を認めた。また、基礎値と同様にCRH刺激した血漿ACTHレベル(「stimulated」)についても改善が認められた。
<Results>
Purified pituitary hormone-producing cells were transplanted into ISWAT (n=2), and plasma ACTH levels were followed up for 3 months (Figure 9). After transplantation, the basal plasma ACTH concentration was improved compared to before transplantation. In addition, CRH-stimulated plasma ACTH levels ("stimulated") were also improved, as were the basal levels.
実施例4:下垂体ホルモン産生細胞の移植部位のさらなる検討(鼠径部皮下、腋窩皮下、筋肉)
<材料及び方法>
実施例1と同様の分化誘導法を用いてhESC由来POを作製した。マウスへの移植には、分化後90-100日で採取したPOを使用した。
Example 4: Further investigation of transplantation sites of pituitary hormone-producing cells (inguinal subcutaneous, axillary subcutaneous, muscle)
Materials and Methods
hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
(1)移植方法
実施例2と同様にマウスに下垂体切除術を施し、マウスに鼠径部のISWAT移植(Inguinal)、腋窩の皮下白色脂肪組織移植(Axilla)、臀部筋肉内移植(Muscle)を行った。
(1) Transplantation Method Mice were hypophysectomized in the same manner as in Example 2, and then transplanted with ISWAT in the groin (Inguinal), subcutaneous white adipose tissue in the axilla (Axilla), or intramuscular transplantation in the gluteal region (Muscle).
(2)統計解析
生存時間解析では、ログランク検定にボンフェローニ補正を行った。<0.05(*)のP値を有意とみなした。
(2) Statistical Analysis For survival time analysis, the log-rank test was subjected to Bonferroni correction. A P value of <0.05 (*) was considered significant.
<結果>
生存率を評価するために、非移植群(sham、n=3)、Axilla群(n=5)、Inguinal群(n=3)、及びMuscle群(n=6)を336日後まで追跡調査した(図10)。腋窩部白色脂肪組織/腋窩白色脂肪組織/臀部筋肉内移植群は、偽手術群と比較して生存率が改善した(p=0.0002367)。
Results
To evaluate survival rates, the non-transplant group (sham, n = 3), the Axilla group (n = 5), the Inguinal group (n = 3), and the Muscle group (n = 6) were followed up for 336 days ( FIG. 10 ). The axillary white adipose tissue/axillary white adipose tissue/gluteal intramuscular transplant group showed improved survival rates compared to the sham surgery group (p = 0.0002367).
実施例5:血管新生誘導による移植効果の検討
<材料及び方法>
実施例1と同様の分化誘導法を用いてhESC由来POを作製した。マウスへの移植には、分化後90-100日で採取したPOを使用した。
Example 5: Examination of the transplantation effect by angiogenesis induction <Materials and methods>
hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 90-100 days after differentiation was used for transplantation into mice.
(1)移植方法
アルツディスポ関節注25mgとヘパリンを含む塩基性線維芽細胞増殖因子(FGF)を混合して作製したMixtureをそれぞれDuragen 人工硬膜(Integra Japan 株式会社)/Surgicel SNOW(Johnson & Johnson株式会社)に浸しシートを作製した。マウスをイソフルランで麻酔し、腹臥位に置いた。細胞移植をする1週間前に背部正中の皮下脂肪組織に4mmの線状皮膚切開を行い、皮下脂肪組織にポケットを作製し、Mixtureを含有するDuragenシート(Duragen群)、Mixtureを含有するSurgicelシート(Surgicel群)をそれぞれ皮下ポケットに1週間留置した。またMixtureをシリンジ使用下に1週間連続で局所注射を行った(bFGF群)。Duragen群に関しては、前処置時の皮膚切開を利用し細胞移植時に留置したシートを取り除き、顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。Surgicel群に関しては、前処置時の皮膚切開を利用し、Surgicelは生体吸収性であり取り除く必要がないため、そのまま顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。bFGF群では、前処置時の皮膚切開を利用し顕微鏡下でワイドボアチップを使用して細胞培養液から採取した5つのPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。無血管領域(AR)移植群では、背部正中皮膚に線状皮膚切開を行い、5つのPOを配置した。全ての下垂体機能低下マウスは、副腎クリーゼを予防するために、0.2mg/0.61ml/マウスのデキサメタゾンの筋肉内注射に供した。
(1) Transplantation Method Mixtures prepared by mixing 25 mg of Artz Dispo joint injection and basic fibroblast growth factor (FGF) containing heparin were soaked in Duragen artificial dura mater (Integra Japan Co., Ltd.)/Surgicel SNOW (Johnson & Johnson Co., Ltd.) to prepare sheets. Mice were anesthetized with isoflurane and placed in a prone position. One week before cell transplantation, a 4 mm linear skin incision was made in the subcutaneous adipose tissue in the midline of the back, a pocket was made in the subcutaneous adipose tissue, and a Duragen sheet containing the mixture (Duragen group) and a Surgicel sheet containing the mixture (Surgicel group) were placed in the subcutaneous pocket for one week. The mixture was also locally injected using a syringe for one week (bFGF group). For the Duragen group, the sheet placed during cell transplantation was removed using the skin incision during pretreatment, and five POs taken from the cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed following nylon suture closure on the PO. For the Surgicel group, the skin incision during pretreatment was used, and five POs taken from the cell culture medium were placed in the pocket using a wide-bore tip under a microscope, since Surgicel is bioabsorbable and does not need to be removed. Skin closure was performed following nylon suture closure on the PO. For the bFGF group, the skin incision during pretreatment was used, and five POs taken from the cell culture medium were placed in the pocket using a wide-bore tip under a microscope. Skin closure was performed following nylon suture closure on the PO. For the avascular area (AR) transplantation group, a linear skin incision was made in the midline skin of the back, and five POs were placed. All hypopituitaric mice were subjected to an intramuscular injection of dexamethasone at 0.2 mg/0.61 ml/mouse to prevent adrenal crisis.
<結果>
血流の乏しい皮下移植部位への細胞移植に備え、移植腔周囲の血流を促進し細胞の生着を高めるために、AR群(n=2)、bFGF群(n=3)、Duragen群(n=3)、及びSurgicel群(n=3)の血漿ACTHレベルを1か月後まで追跡調査した(図11)。移植後、基礎値血漿ACTHレベル(「basal」)は、Duragen群において最も改善する傾向を認めた。また、CRH刺激した血漿ACTHレベル(「stimulated」)も、基礎値血漿ACTHレベルと同様にDuragen群において最も改善する傾向を認めた。(AR群対bFGF群対Duragen群対Surgicel群、基礎;14.7±20.05pg/ml対17.12±16.37pg/ml対37.17±34.52pg/ml対2.39±2.66pg/ml、刺激;42.54±20.05pg/ml対60.77±16.37pg/ml対109.22±34.52pg/ml対6.29±2.66pg/ml)。
Results
In preparation for cell transplantation into a subcutaneous transplantation site with poor blood flow, in order to promote blood flow around the transplant cavity and increase cell engraftment, plasma ACTH levels were followed up for one month in the AR group (n=2), bFGF group (n=3), Duragen group (n=3), and Surgicel group (n=3) ( FIG. 11 ). After transplantation, the basal plasma ACTH level ("basal") tended to improve the most in the Duragen group. In addition, the CRH-stimulated plasma ACTH level ("stimulated") also tended to improve the most in the Duragen group, similar to the basal plasma ACTH level. (AR vs. bFGF vs. Duragen vs. Surgicel, Basal; 14.7±20.05 pg/ml vs. 17.12±16.37 pg/ml vs. 37.17±34.52 pg/ml vs. 2.39±2.66 pg/ml, Stimulated; 42.54±20.05 pg/ml vs. 60.77±16.37 pg/ml vs. 109.22±34.52 pg/ml vs. 6.29±2.66 pg/ml).
実施例6:霊長類に対する免疫抑制剤下でのヒトES細胞由来下垂体オルガノイドの皮下移植検討
<材料及び方法>
実施例1と同様の分化誘導法を用いてhESC由来POを作製した。サルへの移植には、分化後103~117日で採取したPOを使用した。
Example 6: Subcutaneous transplantation of human ES cell-derived pituitary organoids into primates under immunosuppressive drug conditions
Materials and Methods
hESC-derived PO was prepared using the same differentiation induction method as in Example 1. PO harvested 103 to 117 days after differentiation was used for transplantation into monkeys.
(2)サルと下垂体切除術
全ての動物実験は、滋賀医科大学の動物実験委員会によって承認され、動物のケアと使用に関する施設のガイドラインに従って行った。本研究では、体重6.22kgの健康な成体オスのカニクイザルを用いた。
術前の血液検査と術中ナビゲーションのためのCT検査は、サルに鎮静剤を投与した状態で術前に行った。下垂体前葉機能評価のため、ACTH、コルチゾール、成長ホルモン(GH)、IGF-1の血清濃度を術前に採取した(株式会社エスアールエル、東京、日本)。同様に、下垂体後葉機能を評価するためにアルギニン・バソプレシン(AVP)値を測定した。
手術前投薬としてアトロピン(50μg/kg)を筋肉内投与し、麻酔を行った。ケタミン(5mg/kg)とキシラジン(1mg/kg)を筋肉内投与した。気管チューブを挿入し、サルの口の左側に固定した。デキサメタゾン(120μg)、サイロキシン(10μg/body)、デスモプレシン(0.5μg)およびセフトリアキソン(20mg/kg)は、それぞれステロイド補充および抗生物質療法として手術直前に静脈内投与した。手術中の麻酔にはイソフルランを1~3%の濃度で吸入投与した。
蝶形骨下垂体摘出術には内視鏡的経口腔経蝶形骨アプローチを行った。下垂体の正確な方向は術中ナビゲーションシステムで確認した。うつ伏せになったサルの頭を術者側に少し回転させた。上顎の犬歯を糸で上方に引っ張り、サルの口をできるだけ大きく開けた。軟口蓋を正中線で直線的に切開した後、糸で軟口蓋を側方に後退させた。蝶形骨の前の粘膜を正中線で直線的に切開し、蝶形骨を露出させた。ダイヤモンドバー付きの高速ドリルで蝶形骨に穴をあけた。錐体型蝶形骨洞の内部には手術の目印となるようなものがないため、この手術中にナビゲーションシステムで何度も手術の方向を確認した。蝶形骨と鞍底を除去した後、硬膜の鞍をはさみで切断した。下垂体と茎を摘出し、病理組織標本を凍結保存した。角度付き内視鏡で精査し、下垂体が残存していないことを確認した。鞍は腹部皮下脂肪層から採取した脂肪移植片で再建し、術後の脳脊髄液(CSF)漏出を防ぐためにフィブリン接着剤を用いた。再建後の出血や髄液漏れは確認されなかった。軟口蓋を縫合して手術終了とした。点滴はサルが意識を取り戻した直後に中止した。サルは覚醒後、餌を食べることができ、自由に水を飲むことができた。セフトリアキソン(20mg/kg)を術後7日間筋肉内投与した。術後に疼痛が出現した場合には、ブプレノルフィン(4μg/kg)の筋肉内投与を考慮した。
下垂体機能低下症には、ホルモン補充を毎日筋肉内投与した。デキサメタゾン、サイロキシン、バソプレシンの用量は、ヒトの下垂体機能低下症の臨床用量に基づいて決定した。デキサメタゾンは120μg/日から40μg/日に漸減し術後66日からコートリル1.5mg経口内服へ変更した。サイロキシン(10μg/日)は術直後より筋肉内注射をした。尿崩症(DI)はバソプレシン(0.5μg/日)と自由飲水で治療した。サルの水分バランスは、水分摂取量と尿量を正確に測定することが困難であったため、体重と血清ナトリウム(Na)値で評価した。水分と電解質の不均衡は血液検査で評価した。感染は手術創、体温、摂餌量から評価した。術後17日目に鎮静下でホルモン刺激試験を行った。コルチコトロピン放出ホルモン(CRH)(1.5μg/kg)および成長ホルモン放出因子(GRF)(1μg/kg)を静脈内投与した後、下垂体前葉機能を測定した。注射60分後に採血を行い、DIの有無および血中AVP濃度に基づいて下垂体後葉機能を評価した。
(2) Monkeys and hypophysectomy All animal experiments were approved by the Animal Care and Use Committee of the Shiga University of Medical Science and were performed in accordance with the institutional guidelines for the care and use of animals. Healthy adult male cynomolgus monkeys weighing 6.22 kg were used in this study.
Preoperative blood tests and CT scans for intraoperative navigation were performed before surgery while the monkeys were sedated. Serum concentrations of ACTH, cortisol, growth hormone (GH), and IGF-1 were collected before surgery to evaluate anterior pituitary function (SRL Co., Ltd., Tokyo, Japan). Similarly, arginine vasopressin (AVP) levels were measured to evaluate posterior pituitary function.
Anesthesia was induced by intramuscular administration of atropine (50 μg/kg) as preoperative medication. Ketamine (5 mg/kg) and xylazine (1 mg/kg) were administered intramuscularly. A tracheal tube was inserted and fixed to the left side of the monkey's mouth. Dexamethasone (120 μg), thyroxine (10 μg/body), desmopressin (0.5 μg), and ceftriaxone (20 mg/kg) were administered intravenously immediately before surgery as steroid replacement and antibiotic therapy, respectively. Anesthesia during surgery was administered by inhalation of isoflurane at a concentration of 1-3%.
An endoscopic transoral transsphenoidal approach was used for sphenoidal hypophysectomy. The exact direction of the pituitary gland was confirmed by an intraoperative navigation system. The monkey was placed prone and the head was rotated slightly toward the surgeon. The maxillary canines were pulled upward with a thread to open the monkey's mouth as wide as possible. The soft palate was incised linearly in the midline, and then the soft palate was retracted laterally with a thread. The mucosa in front of the sphenoid bone was incised linearly in the midline to expose the sphenoid bone. A hole was drilled in the sphenoid bone with a high-speed drill with a diamond burr. Because there are no surgical landmarks inside the petrous sphenoid sinus, the surgical direction was confirmed many times during this operation by the navigation system. After removing the sphenoid bone and the floor of the sella, the dural sella was cut with scissors. The pituitary gland and stalk were removed, and the pathological specimens were frozen and preserved. Close examination with an angled endoscope confirmed that no pituitary gland remained. The saddle was reconstructed with a fat graft taken from the abdominal subcutaneous fat layer, and fibrin glue was used to prevent postoperative cerebrospinal fluid (CSF) leakage. No bleeding or CSF leakage was observed after reconstruction. The soft palate was sutured to complete the operation. The infusion was discontinued immediately after the monkey regained consciousness. After awakening, the monkey was able to eat food and drink water freely. Ceftriaxone (20 mg/kg) was administered intramuscularly for 7 days after surgery. In the event of postoperative pain, intramuscular administration of buprenorphine (4 μg/kg) was considered.
For hypopituitarism, hormone replacement was administered intramuscularly daily. The doses of dexamethasone, thyroxine, and vasopressin were determined based on the clinical doses for hypopituitarism in humans. Dexamethasone was gradually tapered from 120 μg/day to 40 μg/day, and then changed to oral administration of 1.5 mg of cortryl from 66 days after surgery. Thyroxine (10 μg/day) was administered intramuscularly immediately after surgery. Diabetes insipidus (DI) was treated with vasopressin (0.5 μg/day) and free drinking of water. The monkeys' water balance was evaluated by body weight and serum sodium (Na) value, because it was difficult to accurately measure water intake and urine volume. Water and electrolyte imbalance was evaluated by blood tests. Infection was evaluated by the surgical wound, body temperature, and food intake. A hormone stimulation test was performed under sedation on the 17th day after surgery. Anterior pituitary function was measured after intravenous administration of corticotropin-releasing hormone (CRH) (1.5 μg/kg) and growth hormone-releasing factor (GRF) (1 μg/kg). Blood was collected 60 minutes after injection and posterior pituitary function was evaluated based on the presence or absence of DI and blood AVP concentration.
(3)免疫抑制療法
サルへの免疫抑制剤長期投与による副作用がないことを確認するため半年間の経過観察期間を置いた。Tacの場合は0.3mg/dayで開始し、目標トラフを15~20ng/mLとする。MMFは100 mg/dayで開始し、副作用をみながら100mg~400mgで調整した。またetanerceptは移植1時間前、移植後3、7、10日目に2.5mgを皮下注射する。rATGは移植12時間前から初回投与量として1.5mg/kgを12時間かけて投与し、さらに移植開始時から12時間かけて1.5mg/kgを投与する。その後はそれぞれ12時間の休薬ののちに2回投与する。つまり1.5mg/kgを1回、1.5mg/kgを3回投与することとなり、総投与量は6mg/kgとなる。
(3) Immunosuppressant therapy A six-month follow-up period was set to confirm that there were no side effects from long-term administration of immunosuppressants to monkeys. In the case of Tac, the dose was started at 0.3 mg/day, with a target trough of 15-20 ng/mL. MMF was started at 100 mg/day, and adjusted to 100-400 mg while monitoring side effects. Etanercept was subcutaneously injected at 2.5 mg one hour before transplantation and on the 3rd, 7th, and 10th days after transplantation. rATG was administered at an initial dose of 1.5 mg/kg over 12 hours starting 12 hours before transplantation, and then at 1.5 mg/kg over 12 hours from the start of transplantation. After that, it was administered twice after a 12-hour break. In other words, 1.5 mg/kg was administered once and 1.5 mg/kg three times, for a total dose of 6 mg/kg.
(4)採血とACTH測定
ACTHレベル、Cortisolレベルを、下垂体機能のバイオマーカーとして評価した。血液サンプルを、手背の静脈によって採取し、ヒトCRH(2μg/kg、腹腔内、タナベ、ニプロESファーマ株式会社、大阪、日本)の投与の前と1時間後にサンプリングした。採血後、ヘパリン化し、遠心にて血漿を採取した。1mLずつ分注してマイナス80℃で保存後、血上清中のACTH濃度/Cortisol濃度を、日本で臨床的に使用されている電気化学発光免疫測定法(ECLIA)キット(株式会社エスアールエル、東京、日本)を使用して決定した。移植治療後1か月目までは毎週ACTH濃度/Cortisol濃度を測定した。その後は2週間ごとにACTH濃度/Cortisol濃度を測定した。
CRH負荷テストを、下垂体切除術の17日後、及びPO移植治療の4週間後に実施した。
移植片のコルチゾールに対するネガティブフィードバックを評価した。血液サンプルを、手背の静脈によって採取し、デキサメタゾン1mgの投与の前と2時間後にサンプリングした。採血後、ヘパリン化し、遠心にて血漿を採取した。1mLずつ分注してマイナス80℃で保存後、血上清中のACTH濃度を、日本で臨床的に使用されている電気化学発光免疫測定法(ECLIA)キット(株式会社エスアールエル、東京、日本)を使用して決定した。デキサメタゾン抑制試験をPO移植治療の6週後に実施した。
(4) Blood collection and ACTH measurement ACTH and Cortisol levels were evaluated as biomarkers of pituitary function. Blood samples were collected via the dorsal vein of the hand and sampled before and 1 hour after administration of human CRH (2 μg/kg, intraperitoneally, Tanabe, Nipro ES Pharma Co., Ltd., Osaka, Japan). After blood collection, the blood was heparinized and centrifuged to collect plasma. After storing at −80° C. in 1 mL aliquots, the ACTH/Cortisol concentrations in the blood supernatant were determined using an electrochemiluminescence immunoassay (ECLIA) kit (SRL Co., Ltd., Tokyo, Japan) that is used clinically in Japan. ACTH/Cortisol concentrations were measured weekly until one month after transplantation treatment. Thereafter, ACTH/Cortisol concentrations were measured every two weeks.
CRH loading tests were performed 17 days after hypophysectomy and 4 weeks after PO implantation treatment.
The negative feedback of the graft on cortisol was evaluated. Blood samples were collected via the dorsal vein of the hand before and 2 hours after administration of 1 mg dexamethasone. After blood collection, the blood was heparinized and centrifuged to obtain plasma. After storage at -80°C in 1 mL aliquots, ACTH concentrations in the blood supernatant were determined using an electrochemiluminescence immunoassay (ECLIA) kit (SRL Co., Ltd., Tokyo, Japan), which is used clinically in Japan. A dexamethasone suppression test was performed 6 weeks after PO transplantation treatment.
(5)In vitroでのPOの自発的ACTH分泌の測定
実施例1と同様の方法を用いてIn vitroでのPOの自発的ACTH分泌POを測定した。
(5) Measurement of Spontaneous ACTH Secretion from PO in Vitro Using the same method as in Example 1, spontaneous ACTH secretion from PO in vitro was measured.
(6)移植方法
アルツディスポ関節注25mgとヘパリンを含む塩基性線維芽細胞増殖因子(FGF)を混合して作製したMixtureをそれぞれDuragen 人工硬膜(Integra Japan株式会社)に浸しシートを作製した。サルを下垂体切除術時と同様の方法で麻酔し、腹臥位に置いた。細胞移植をする1週間前に背部正中の皮下脂肪組織に4mmの線状皮膚切開を行い、皮下脂肪組織にポケットを作製し、Mixtureを含有するDuragenシートを皮下ポケットに1週間留置した。細胞移植時に前処置時の皮膚切開を利用し細胞移植時に留置したシートを取り除き、目視下にHBSSでwashした1440個(5個/20g)のPOをポケットに配置した。PO上のナイロン縫合閉鎖に続いて、皮膚閉鎖を行った。術後副腎クリーゼを予防するために、4.5mgハイドロコートンの静脈内注射に供した。
(6) Transplantation Method A mixture of 25 mg of Artz Dispo joint injection and basic fibroblast growth factor (FGF) containing heparin was soaked in Duragen artificial dura mater (Integra Japan Co., Ltd.) to prepare a sheet. Monkeys were anesthetized in the same manner as in hypophysectomy and placed in a prone position. One week before cell transplantation, a 4 mm linear skin incision was made in the subcutaneous adipose tissue in the midline of the back, a pocket was made in the subcutaneous adipose tissue, and a Duragen sheet containing the mixture was placed in the subcutaneous pocket for one week. During cell transplantation, the sheet placed during cell transplantation was removed using the skin incision made during pretreatment, and 1440 POs (5 pieces/20 g) washed with HBSS under visual observation were placed in the pocket. Following nylon suture closure on the PO, skin closure was performed. To prevent postoperative adrenal crisis, the patient was given an intravenous injection of 4.5 mg hydrocortone.
<結果>
PO培養液中のACTHレベルによって評価したin vitroでの移植前ACTH分泌は、平均ACTH濃度は13123pg/mlであった。移植後、基礎値血漿ACTHレベル及びCRH刺激した血漿ACTHレベルは、移植前より高い値を推移していた(図12A、13A)。また基礎値Cortisolレベルに関しても術前内服コートリル内服時と同等の値を示した(図12B)。移植6週目に行ったデキサメタゾン抑制試験では、血漿ACTHレベルはデキサメタゾン投与後2時間値で投与前より低い値を示した(図13B)。
<Results>
Pre-transplant ACTH secretion in vitro was evaluated by ACTH levels in PO culture medium, with a mean ACTH concentration of 13123 pg/ml. After transplantation, basal plasma ACTH levels and CRH-stimulated plasma ACTH levels remained higher than before transplantation (Figures 12A and 13A). Basal Cortisol levels were also comparable to those observed when oral Cortisol was administered preoperatively (Figure 12B). In a dexamethasone suppression test performed 6 weeks after transplantation,
本発明によれば、侵襲性が低く、移植した細胞が腫瘍化した際に切除することが容易であるような移植部位(皮下組織(特に皮下脂肪組織)及び/又は筋肉組織)に移植されるように用いられることを特徴とする、下垂体ホルモン産生細胞を含有する下垂体の障害に基づく疾患の治療薬を提供することが可能となるため有用である。また、本発明の治療薬により、下垂体ホルモン産生細胞の臨床応用の実現が可能となるため有用である。また、本発明の方法により、化合物の薬効及び安全性評価に有用な、ホルモン分泌能が長期間維持されるヒト下垂体組織を有するモデル動物の作出が可能となるため有用である。 The present invention is useful because it makes it possible to provide a therapeutic drug for diseases caused by disorders of the pituitary gland, which contains pituitary hormone-producing cells and is characterized by being used to transplant into a transplantation site (subcutaneous tissue (particularly subcutaneous adipose tissue) and/or muscle tissue) that is less invasive and easy to resect when the transplanted cells become tumorous. The therapeutic drug of the present invention is also useful because it makes it possible to realize clinical applications of pituitary hormone-producing cells. The method of the present invention is also useful because it makes it possible to create a model animal having human pituitary tissue that maintains hormone secretion ability for a long period of time, which is useful for evaluating the efficacy and safety of compounds.
本出願は、日本で出願された特願2023-021207(出願日:2023年2月14日)及び特願2023-176873(出願日:2023年10月12日)を基礎としており、その内容は本明細書に全て包含されるものである。 This application is based on patent application No. 2023-021207 (filing date: February 14, 2023) and patent application No. 2023-176873 (filing date: October 12, 2023) filed in Japan, the contents of which are incorporated in their entirety into this specification.
Claims (22)
(1)多能性幹細胞をc-jun N末キナーゼ(JNK)シグナル伝達経路阻害物質及びWntシグナル伝達経路阻害物質の存在下で培養し、細胞集団を得る第一工程、
(2)第一工程で得られた細胞集団を、BMPシグナル伝達経路作用物質及びソニック・ヘッジホッグシグナル伝達経路作用物質の存在下で培養し、下垂体ホルモン産生細胞を含む細胞集団を得る第二工程。 The therapeutic agent according to any one of claims 1 to 11, wherein the pituitary hormone-producing cells are pituitary hormone-producing cells produced by a method comprising the steps of:
(1) a first step of culturing pluripotent stem cells in the presence of a c-jun N-terminal kinase (JNK) signaling pathway inhibitor and a Wnt signaling pathway inhibitor to obtain a cell population;
(2) A second step of culturing the cell population obtained in the first step in the presence of a substance acting on the BMP signaling pathway and a substance acting on the Sonic Hedgehog signaling pathway to obtain a cell population containing pituitary hormone-producing cells.
(a)多能性幹細胞を、フィーダー細胞非存在下で、1)TGFβファミリーシグナル伝達経路阻害物質及び/又はソニック・ヘッジホッグシグナル伝達経路作用物質、並びに2)未分化維持因子を含む培地で培養するa工程。 The therapeutic agent according to claim 12, characterized in that the process further comprises the following step a before the first step:
(a) A step a of culturing pluripotent stem cells in a medium containing 1) a TGFβ family signaling pathway inhibitor and/or a Sonic hedgehog signaling pathway agonist, and 2) an undifferentiated state maintenance factor, in the absence of feeder cells.
第二工程で得られた下垂体ホルモン産生細胞を含む細胞集団から下垂体ホルモン産生細胞を選別し、回収する工程。 The therapeutic agent according to claim 12 or 13, characterized in that the process further comprises the following step after the second step:
A step of selecting and recovering pituitary hormone-producing cells from the cell population containing pituitary hormone-producing cells obtained in the second step.
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2025501112A JPWO2024171957A1 (en) | 2023-02-14 | 2024-02-09 |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2023021207 | 2023-02-14 | ||
| JP2023-021207 | 2023-02-14 | ||
| JP2023-176873 | 2023-10-12 | ||
| JP2023176873 | 2023-10-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024171957A1 true WO2024171957A1 (en) | 2024-08-22 |
Family
ID=92421780
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2024/004478 Ceased WO2024171957A1 (en) | 2023-02-14 | 2024-02-09 | Transplantation of pituitary hormone-producing cells |
Country Status (2)
| Country | Link |
|---|---|
| JP (1) | JPWO2024171957A1 (en) |
| WO (1) | WO2024171957A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019103129A1 (en) * | 2017-11-24 | 2019-05-31 | 住友化学株式会社 | Method for producing cell mass including pituitary tissue, and cell mass thereof |
| WO2023277135A1 (en) * | 2021-06-30 | 2023-01-05 | 住友化学株式会社 | Method for producing cells constituting nasal epithelium, and cell population including cells constituting nasal epithelium or progenitor cells thereof |
| WO2023054396A1 (en) * | 2021-09-30 | 2023-04-06 | 住友化学株式会社 | Method for producing cell mass including pituitary tissue and cell mass |
-
2024
- 2024-02-09 JP JP2025501112A patent/JPWO2024171957A1/ja active Pending
- 2024-02-09 WO PCT/JP2024/004478 patent/WO2024171957A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019103129A1 (en) * | 2017-11-24 | 2019-05-31 | 住友化学株式会社 | Method for producing cell mass including pituitary tissue, and cell mass thereof |
| WO2023277135A1 (en) * | 2021-06-30 | 2023-01-05 | 住友化学株式会社 | Method for producing cells constituting nasal epithelium, and cell population including cells constituting nasal epithelium or progenitor cells thereof |
| WO2023054396A1 (en) * | 2021-09-30 | 2023-04-06 | 住友化学株式会社 | Method for producing cell mass including pituitary tissue and cell mass |
Non-Patent Citations (9)
| Title |
|---|
| HIROYU SASAKI, HIDETAKA SUGA, KAZUTO TAKEUCHI, YUICHI MIZUTA, HIDEYUKI HAMADE: "O2-6-1 Efficacy of subcutaneous transplantation of human ES cell-derived pituitary cells in mice", NIHON NAIBUNPI GAKKAI ZASSHI - FOLIA ENDOCRINOLOGICA JAPONICA, NIHON NAIBUNPI GAKKAI, JP, vol. 99, no. 1, 1 May 2023 (2023-05-01), JP , pages 308, XP009556728, ISSN: 0029-0661 * |
| NAOAKI SAKATA; TAKESHI AOKI; GUMPEI YOSHIMATSU; HARUYUKI TSUCHIYA; TATSUO HATA; YU KATAYOSE; SHINICHI EGAWA; MICHIAKI UNNO: "Strategy for clinical setting in intramuscular and subcutaneous islet transplantation", DIABETES/METABOLISM RESEARCH AND REVIEWS, WILEY, LONDON,, GB, vol. 30, no. 1, 14 January 2014 (2014-01-14), GB , pages 1 - 10, XP072260659, ISSN: 1520-7552, DOI: 10.1002/dmrr.2463 * |
| SASAKI HIROO, SUGA HIDETAKA, TAKEUCHI KAZUHITO, NAGATA YUICHI, HARADA HIDEYUKI, KONDO TATSUMA, ITO EIJI, MAEDA SACHI, SAKAKIBARA M: "Subcutaneous transplantation of human embryonic stem cells-derived pituitary organoids", FRONTIERS IN ENDOCRINOLOGY, FRONTIERS RESEARCH FOUNDATION, CH, vol. 14, CH , XP093200445, ISSN: 1664-2392, DOI: 10.3389/fendo.2023.1130465 * |
| SASAKI, HIROO ET AL.: "Subcutaneous transplantation of human pluripotent stem cell-derived pituitary cells for the realization of regenerative medicine", NIHON NAIBUNPI GAKKAI ZASSHI - FOLIA ENDOCRINOLOGICA JAPONICA, NIHON NAIBUNPI GAKKAI, JP, vol. 99, no. S.HPT, 1 August 2023 (2023-08-01), JP , pages 4 - 6, XP009556721, ISSN: 0029-0661, DOI: 10.1507/endocrine.99.S.HPT_4 * |
| SUGA, HIDETAKA, ARIMA, HIROSHI: "My medicine using human induced pluripotent stem cell-derived pituitary organoid", JOURNAL OF CLINICAL AND EXPERIMENTAL MEDICINE, BIOMEDICAL DRUGS PUBLICATION, JP, vol. 276, no. 6, 6 February 2021 (2021-02-06), JP , pages 654 - 661, XP009557441, ISSN: 0039-2359 * |
| SUMI, SHOICHIRO: "New Methods for Islet Transplantation", TAN TO SUI - BILIARY TRACT AND PANCREAS, IGAKU TOSHO SHUPPAN, TOKYO, JP, vol. 38, no. 9, 1 September 2017 (2017-09-01), JP , pages 863 - 867, XP009557442, ISSN: 0388-9408 * |
| TAGA SHIORI, SUGA HIDETAKA, NAKANO TOKUSHIGE, KUWAHARA ATSUSHI, INOSHITA NAOKO, KODANI YU, NAGASAKI HIROSHI, SATO YOSHITAKA, TSUMU: "Generation and purification of ACTH-secreting hPSC-derived pituitary cells for effective transplantation", STEM CELL REPORTS, CELL PRESS, UNITED STATES, vol. 18, no. 8, 1 August 2023 (2023-08-01), United States , pages 1657 - 1671, XP093200447, ISSN: 2213-6711, DOI: 10.1016/j.stemcr.2023.05.002 * |
| YASUNAMI, Y. et al., A Novel Subcutaneous Site of Islet Transplantation Superior to the Liver, Transplantation, June 2018, vol. 102, no. 6, pp. 945-952, doi: 10.1097/TP.0000000000002162, ISSN 0041-1337 * |
| ZIMMER BASTIAN, PIAO JINGHUA, RAMNARINE KIRAN, TOMISHIMA MARK J., TABAR VIVIANE, STUDER LORENZ: "Derivation of Diverse Hormone-Releasing Pituitary Cells from Human Pluripotent Stem Cells", STEM CELL REPORTS, CELL PRESS, UNITED STATES, vol. 6, no. 6, 14 June 2016 (2016-06-14), United States , pages 858 - 872, XP055824600, ISSN: 2213-6711, DOI: 10.1016/j.stemcr.2016.05.005 * |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2024171957A1 (en) | 2024-08-22 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP3047017B1 (en) | Methods of mammalian retinal stem cell production and applications | |
| ES2741969T3 (en) | Method for stem cell culture | |
| JP2023011944A (en) | Differentiation of cortical neurons from human pluripotent stem cells | |
| EP3231863B1 (en) | Method for producing therapeutic corneal endothelial substitute cell sphere | |
| CN110684734A (en) | Midbrain dopamine (DA) neurons for implantation | |
| EP3105320B1 (en) | Synthetic retina | |
| ES2902859T3 (en) | Method for producing adenohypophysis or precursor tissue thereof | |
| JPWO2013051722A1 (en) | Method for producing corneal endothelial cell | |
| EP3178925B1 (en) | Method for producing retinal ganglion cells | |
| WO2015061568A1 (en) | Reprogramming cardiomyocytes with one transcription factor | |
| WO2023277135A1 (en) | Method for producing cells constituting nasal epithelium, and cell population including cells constituting nasal epithelium or progenitor cells thereof | |
| CN116323677A (en) | Retinal pigment epithelium and photoreceptor bilayer and uses thereof | |
| US20240392241A1 (en) | Method for producing cell mass including pituitary tissue and cell mass | |
| US20240400983A1 (en) | Cell aggregate including pituitary hormone-producing cells and method for producing same | |
| WO2024171957A1 (en) | Transplantation of pituitary hormone-producing cells | |
| HK40109168A (en) | Method for producing cell mass including pituitary tissue and cell mass | |
| CN104968298A (en) | Methods for using autologous fibroblasts to alter skin identiy | |
| WO2022172960A1 (en) | Maturation agent | |
| HK40109173A (en) | Cell aggregate including pituitary hormone-producing cells and method for producing same | |
| CN118043447A (en) | Preparation method of cell population containing pituitary tissue and cell population | |
| CN118318033A (en) | Cell aggregates containing pituitary hormone-producing cells and preparation method thereof | |
| JP2024094292A (en) | Sheet-shaped cell structure containing upper respiratory tract cells, and method for producing and using same | |
| JP2025177429A (en) | Method for producing primitive nodule 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: 24756802 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2025501112 Country of ref document: JP Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025501112 Country of ref document: JP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |