WO2024014497A1 - 細胞移植用のフィブリンゲルシート - Google Patents
細胞移植用のフィブリンゲルシート Download PDFInfo
- Publication number
- WO2024014497A1 WO2024014497A1 PCT/JP2023/025809 JP2023025809W WO2024014497A1 WO 2024014497 A1 WO2024014497 A1 WO 2024014497A1 JP 2023025809 W JP2023025809 W JP 2023025809W WO 2024014497 A1 WO2024014497 A1 WO 2024014497A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- fibrin gel
- gel sheet
- sheet
- fibrin
- 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
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L27/00—Materials for grafts or prostheses or for coating grafts or prostheses
- A61L27/36—Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
- A61L27/38—Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0676—Pancreatic 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
- A61K35/37—Digestive system
- A61K35/39—Pancreas; Islets of Langerhans
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L27/00—Materials for grafts or prostheses or for coating grafts or prostheses
- A61L27/14—Macromolecular materials
- A61L27/22—Polypeptides or derivatives thereof, e.g. degradation products
- A61L27/225—Fibrin; Fibrinogen
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L27/00—Materials for grafts or prostheses or for coating grafts or prostheses
- A61L27/36—Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
- A61L27/38—Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
- A61L27/3804—Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
- A61L27/3834—Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L27/00—Materials for grafts or prostheses or for coating grafts or prostheses
- A61L27/50—Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
- A61L27/52—Hydrogels or hydrocolloids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L27/00—Materials for grafts or prostheses or for coating grafts or prostheses
- A61L27/50—Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
- A61L27/58—Materials at least partially resorbable by the body
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/45—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/50—Proteins
- C12N2533/56—Fibrin; Thrombin
Definitions
- the present invention relates to a fibrin gel sheet for cell transplantation, in which cells are uniformly dispersed and encapsulated in a fibrin gel sheet, and which has an unprecedentedly large size, and a method for manufacturing the same.
- Functional differentiated cells derived from pluripotent stem cells such as induced pluripotent cells and embryonic stem cells (ES cells) are expected to serve as a cell source for transplantation and regenerative medicine.
- pluripotent stem cells such as induced pluripotent cells and embryonic stem cells (ES cells)
- ES cells embryonic stem cells
- Non-Patent Document 1 discloses a sheet-like structure formed by sandwiching pancreatic islet cells encapsulated in fibrin gel between Vycryl (registered trademark) meshes of 7 mm x 7 mm. It is described that it was transplanted into a site of a recipient diabetic model rat that had undergone angiogenesis treatment in advance.
- Vycryl registered trademark
- Non-Patent Document 2 discloses a sheet-like structure formed by sandwiching pancreatic islet cells encapsulated in a hydrogel containing polyvinyl alcohol (PVA) between polyethylene terephthalate (PET) meshes with a size of 20 mm x 15 mm. It is described that the construct was transplanted into a recipient diabetic model rat.
- PVA polyvinyl alcohol
- PET polyethylene terephthalate
- Patent Document 1 discloses a sheet-like structure in which pancreatic islet cells encapsulated in a hydrogel containing PVA are sandwiched between circular PET meshes with a diameter of 15 mm, and the structure is used in a diabetic model as a recipient. It has been reported that it was transplanted into mice.
- the previously reported sheet-like structure in which functional differentiated cells are encapsulated in a biodegradable gel is intended for transplantation into diabetic model animals (rats and mice), and its size is When targeting large mammals such as humans, the size may not be sufficient, and the number of cells loaded may not be sufficient. Therefore, in this field, there has been a strong desire to develop the above-mentioned structure that has a larger size and contains more cells, which can also be used in humans.
- the present inventors tried to create the above-mentioned structure having a larger size using fibrin gel, and found that the larger amount of fibrin gel prepared to create the larger size did not reach the desired size. It was discovered that when trying to spread the material to a size of 1 and mold it into a sheet, it gelled (solidified) before it was sufficiently spread, making it difficult to mold it to the desired size. Furthermore, when trying to mold a large amount of fibrin gel into a sheet of the desired size while continuously injecting it, the cells in the solution will precipitate, and the cell content will decrease from the beginning to the end of the continuous injection. It has become clear that only the structure described above can be produced in which the cell density gradually decreases and, as a result, the cell density is not uniform. Heterogeneity in cell density is undesirable because it may reduce therapeutic efficiency and affect long-term engraftment of transplanted cells.
- the present invention aims to solve these problems discovered by the present inventors, and has a larger size than ever before, and at the same time, a larger number of cells are uniformly dispersed and encapsulated.
- the object is to provide a sheet-like fibrin gel structure (ie, a fibrin gel sheet).
- the present inventors have found that, in the formation of fibrin gel, by adjusting the blending amount of fibrinogen and thrombin and/or the temperature during gelation (solidification), it is possible to It has been discovered that the rate of gelling (solidification) of the gel can be adjusted and that it can be spread and shaped into fibrin gel sheets having a desired larger size.
- a fibrin gel in forming a fibrin gel, by adding and suspending biodegradable particles to a fibrinogen solution in which cells are suspended, or by adding and dissolving a biodegradable gelling agent to the fibrinogen solution.
- a biodegradable gelling agent By suppressing cell precipitation, cells can be maintained in a uniformly dispersed and suspended state, and by gelling (solidifying) this, even in molding by continuous injection, It has been found that a fibrin gel sheet with uniform cell density can be obtained.
- a fibrin gel sheet for cell transplantation in which cells are uniformly dispersed and encapsulated in a fibrin gel sheet, has a surface area of 2.25 cm 2 or more, and a thickness of 1 mm or less.
- the fibrin gel sheet of [1] in which cells are encapsulated in an amount exceeding 1.5 ⁇ 10 6 cells/cm 2 .
- the fibrin gel sheet of [1] or [2], wherein the cells are in the form of spheroids.
- the fibrin gel sheet of [5] which contains a biodegradable gelling agent in an amount of 0.2 to 2 w/v%.
- the fibrin gel sheet of [8] which has a size that allows the biodegradable particles to pass through an opening of 100 ⁇ m to 1000 ⁇ m.
- a laminate of fibrin gel sheets for cell transplantation comprising a plurality of laminated fibrin gel sheets according to any one of [1] to [12].
- a fibrin gel sheet used in a cell transplant treatment method in which cells are uniformly dispersed and encapsulated in a fibrin gel sheet, has a surface area of 2.25 cm 2 or more, and a thickness of 1 mm or less .
- [8a] The fibrin gel sheet according to any one of [1a] to [4a], further encapsulating biodegradable particles.
- the fibrin gel sheet of [8a] which has a size that allows the biodegradable particles to pass through an opening of 100 ⁇ m to 1000 ⁇ m.
- the fibrin gel sheet according to any one of [8a] to [10a] wherein the biodegradable particles are gelatin gel particles.
- the fibrin gel sheet according to any one of [1a] to [10a] further comprising a support.
- [33a] A laminate of fibrin gel sheets used in a cell transplant treatment method, which is formed by laminating a plurality of fibrin gel sheets according to any one of [1a] to [12a].
- This specification includes the content described in the specification and/or drawings of Japanese Patent Application No. 2022-113546 filed on July 14, 2022, which is the basis of the priority of this application. All publications, patents, and patent applications cited herein are incorporated by reference in their entirety.
- the present invention it is possible to provide a fibrin gel sheet that has a larger size than hitherto available and in which more cells are uniformly dispersed and encapsulated, as well as a method for producing the same.
- FIG. 1 is a photographic diagram showing the appearance of (A) a fibrin gel mass containing iPIC and (B) a fibrin gel sheet containing iPIC. Scale bar: 10mm.
- Figure 2 is a graph showing the analysis results of blood human c-peptide concentration measured over time by ELISA method in nude rats in which fibrin gel masses containing iPIC or fibrin gel sheets containing iPIC were subcutaneously implanted. be.
- Figure 3 shows the results of histological analysis of the implantation site 10 days and 6 weeks after implantation in nude rats in which (A) fibrin gel mass containing iPIC or (B) fibrin gel sheet containing iPIC was subcutaneously implanted.
- FIG. Scale bar 1mm.
- FIG. 4 is a graph showing the results of analyzing the relationship between thrombin concentration and fibrin gel gelation time.
- Figure 5 shows the appearance (A) of a fibrin gel sheet containing large animal/human-sized iPICs manufactured by a method using a cooled metal mold, and the positions 1 to 4 of the sheet observed using a phase contrast microscope. It is a photographic figure showing the result (B) observed by.
- FIG. 6 is a photographic diagram showing the state of iPIC suspended in (A) fibrinogen solution and (B) fibrinogen+gelatin microsphere solution and left to stand for a predetermined period of time. Arrowheads in (A) indicate precipitation of iPIC.
- Figure 7 shows the analysis results of blood human c-peptide concentration measured over time by ELISA method in nude rats subcutaneously implanted with fibrin gel sheets containing iPIC or fibrin + gelatin microsphere gel sheets containing iPIC. It is a graph diagram.
- Figure 8 is a photographic diagram (A) showing the appearance and cross section of a fibrin + gelatin microsphere gel sheet containing large animal/human-sized iPICs produced manually by pipette injection and using a 3D printer, and the brightness distribution of each sheet.
- FIG. 9 is a graph showing the measurement results of the number of cells (concentration) in the fibrinogen+collagen solution discharged from the syringe.
- FIG. 10 is a graph showing the analysis results of blood human c-peptide concentration measured by ELISA method in nude rats in which a fibrin + gelatin microsphere gel sheet containing iPIC or a fibrin + collagen gel sheet containing iPIC was subcutaneously implanted. It is a diagram.
- FIG. 11 is a graph showing the measurement results of the number of cells contained in six arbitrary locations (1 to 6) of a fibrin + collagen gel sheet containing large animal/human-sized iPICs produced using a 3D printer. .
- “about” means up to plus or minus 25%, 20%, 10%, 8%, 6%, 5%, 4%, 3%, 2% or 1%, respectively, with respect to the reference value. Indicates a fluctuating value. Preferably, the term “about” indicates a range of plus or minus 15%, 10%, 5%, or 1%, respectively, relative to the reference value.
- not using feeder cells basically means not containing feeder cells, and not using a medium preconditioned by culturing feeder cells. Therefore, the medium does not contain substances such as growth factors and cytokines secreted from feeder cells.
- feeder cell refers to a cell that is co-cultured with another type of cell to provide an environment that supports the cell and allows it to grow.
- Feeder cells may be from the same species as the cells they support or from a different species.
- human dermal fibroblasts or human embryonic stem cells may be used as feeders for human cells, primary cultures of mouse embryonic fibroblasts, and immortalized mouse embryonic fibroblasts may be used.
- Feeder cells can be inactivated, such as by radiation or mitomycin C treatment.
- adheresion refers to the attachment of cells to a container, e.g., attachment of cells to a sterile plastic (or coated plastic) cell culture dish or flask in the presence of an appropriate medium. It refers to the fact that Some cells cannot be maintained or grow in culture unless they are attached to a cell culture vessel. In contrast, non-adherent cells can be maintained and expanded in culture without being attached to a container.
- culture refers to maintaining, growing, and/or differentiating cells in an in vitro environment.
- “Culture” means sustaining, proliferating, and/or differentiating cells in a tissue or outside the body, eg, in a cell culture dish or flask.
- Culture includes two-dimensional culture (plane culture) and three-dimensional culture (suspension culture).
- purify and purification refer to removing impurities in a composition, such as a composition of cells, to make it pure with respect to specific components.
- “Purified” when used to describe the composition of a cell is one in which the amount of impurity is relative to the proportion of such components in the composition of the cell before it is purified. Refers to a composition of cells that has a comparatively reduced purity and increased purity of certain components.
- a composition of cells can be purified with respect to a target cell type, such that the proportion of target cell types is increased compared to the proportion of target cells present in the composition of cells prior to purification.
- Compositions of cells can also be purified for target cell types by cell selection and sorting methods known in the art.
- compositions of cells can also be purified by certain sorting or selection processes as described herein.
- the method of purifying the target cells results in the target cells being at least 70%, 80%, 85%, 90%, 95%, 97%, 98% or 99% pure. , impurities (including contaminating cells) may be undetectable.
- growth factor is an endogenous protein that promotes differentiation and/or proliferation of specific cells.
- growth factors include epidermal growth factor (EGF), acidic fibroblast growth factor (aFGF), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), and insulin-like growth factor 1.
- IGF-1 insulin-like growth factor 2
- IGF-2 insulin-like growth factor 2
- KGF keratinocyte growth factor
- NGF nerve growth factor
- PDGF platelet-derived growth factor
- TGF- ⁇ transforming growth factor ⁇
- VEGF Vascular endothelial growth factor
- transferrin various interleukins (e.g., IL-1 to IL-18), various colony-stimulating factors (e.g., granulocyte/macrophage-colony stimulating factor (GM-CSF)), Included are various interferons (such as IFN- ⁇ ) and other cytokines that have effects on stem cells, such as stem cell factor (SCF) and erythropoietin (Epo).
- SCF stem cell factor
- Epo erythropoietin
- ROCK inhibitor refers to a substance that inhibits Rho kinase (ROCK: Rho-associated, coiled-coil containing protein kinase), and refers to a substance that inhibits either ROCK I or ROCK II. You can.
- the ROCK inhibitor is not particularly limited as long as it has the above function, and for example, N-(4-pyridinyl)-4 ⁇ -[(R)-1-aminoethyl]cyclohexane-1 ⁇ -carboxamide (herein, Y27632 ), Fasudil (HA1077), (2S)-2-methyl-1-[(4-methyl-5-isoquinolinyl)sulfonyl]hexahydro-1H-1,4-diazepine (H-1152), 4 ⁇ -[(1R)-1-aminoethyl]-N-(4-pyridyl)benzene-1 ⁇ -carboxamide (Wf-536), N-(1H-pyrrolo[2,3-b]pyridin-4-yl)- 4PER[(R)-1-aminoethyl]cyclohexane-1 ⁇ -carboxamide (Y-30141), N-(3- ⁇ [2-(4-amino-1,2,5
- ROCK inhibitors are not limited to these, and antisense oligonucleotides and siRNAs against ROCK mRNA, antibodies that bind to ROCK, dominant negative ROCK mutants, etc. can also be used as ROCK inhibitors, and commercially available are available or can be synthesized according to known methods.
- a “GSK3 ⁇ inhibitor” is a substance that has inhibitory activity against GSK3 ⁇ (glycogen synthase kinase 3 ⁇ ).
- GSK3 (glycogen synthase kinase 3) is a type of serine/threonine protein kinase and is involved in many signal pathways involved in glycogen production, apoptosis, stem cell maintenance, and the like. GSK3 exists in two isoforms, ⁇ and ⁇ .
- the "GSK3 ⁇ inhibitor” used in the present invention is not particularly limited as long as it has GSK3 ⁇ inhibitory activity, and may be a substance that has both GSK3 ⁇ inhibitory activity and GSK3 ⁇ inhibitory activity.
- CHIR98014 (2-[[2-[(5-nitro-6-aminopyridin-2-yl)amino]ethyl]amino]-4-(2,4-dichlorophenyl)-5-(1H -imidazol-1-yl)pyrimidine), CHIR99021 (6-[[2-[[4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl)-2-pyrimidinyl] TDZD-8 (4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione), SB216763 (3-(2,4-dichlorophenyl) )-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione), TWS-119(3-[6-(3-aminophenyl)-7H-pyrrolo[2 , 3-d]pyrimidin-4-
- GSK3 ⁇ inhibitors are not limited to these, and antisense oligonucleotides and siRNAs against GSK3 ⁇ mRNA, antibodies that bind to GSK3 ⁇ , dominant negative GSK3 ⁇ mutants, etc. can also be used as GSK3 ⁇ inhibitors, and commercially available are available or can be synthesized according to known methods.
- serum replacement refers to, for example, KnockOutTM Serum Replacement (KSR: Thermo Fisher Scientific), StemSure (registered trademark) Serum Replacement (Wako), B-27 supplement. , N2-supplements, albumin (e.g. lipids) rich albumin), insulin, transferrin, fatty acids, collagen precursors, trace elements (e.g. zinc, selenium (e.g. sodium selenite)), 2-mercaptoethanol, 3' thiol glycerol or mixtures thereof (e.g. ITS-G). ).
- Preferred serum replacements include B-27 supplement, KSR, StemSure® Serum Replacement, and ITS-G.
- concentration in the medium is 0.01 to 10% by weight, preferably 0.1 to 2% by weight. In the present invention, it is preferable to use a "serum substitute" in place of serum.
- factor having CDK8/19 inhibitory activity means any substance that has CDK8/19 inhibitory activity. In contrast to other proteins of the same CDK family, CDK8 is not required for cell proliferation and inhibition of CDK8 has no significant effect under normal conditions. CDK19 and CDK8 are similar, and inhibition of CDK8 is usually accompanied by inhibition of CDK19. Conventionally known "factors having CDK8/19 inhibitory activity" can be used, and such CDK8/19 inhibitors can be found in patent documents or non-patent documents.
- Factors with CDK8/19 inhibitory activity are not limited to these, and antisense oligonucleotides and siRNAs against CDK8/19 mRNA, antibodies that bind to CDK8/19, dominant negative CDK8/19 mutants, etc. It can be used as a factor having 19-inhibitory activity, and can be commercially available or synthesized according to known methods.
- the present invention relates to a fibrin gel sheet for cell transplantation (hereinafter sometimes simply referred to as "fibrin gel sheet of the present invention"), in which cells are uniformly dispersed and encapsulated in a fibrin gel sheet. It is.
- the fibrin gel sheet of the present invention is characterized by having a size that could not be achieved with conventional fibrin gel sheets for cell transplantation.
- the surface area of the sheet (also referred to as one side of the sheet) is 2.25 cm 2 or more, preferably 4 cm 2 or more, more preferably 9 cm 2 or more, even more preferably 16 cm 2 or more, even more preferably 25 cm 2 or more, particularly preferably 36 cm 2 In particular, it is more preferably 49 cm 2 or more, especially more preferably 64 cm 2 or more, even more preferably 81 cm 2 or more, particularly preferably 100 cm 2 or more, and the upper limit is not particularly limited, and the size and shape of the transplant site Although it is possible to select the size as appropriate depending on the size, for example, the size may be 400 cm 2 or less.
- the range of the surface area of the fibrin gel sheet of the present invention can be expressed using two numerical values respectively selected from the lower limit and upper limit.
- the surface area of the fibrin gel sheet of the present invention is 2.25 cm 2 or more. ⁇ 400cm 2 or less, preferably 4cm 2 or more and 400cm 2 or less, more preferably 9cm 2 or more and 400cm 2 or less, even more preferably 16cm 2 or more and 400cm 2 or less, even more preferably 25cm 2 or more and 400cm 2 or less, especially Preferably 36 cm 2 or more and 400 cm 2 or less, especially more preferably 49 cm 2 or more and 400 cm 2 or less, even more preferably 64 cm 2 or more and 400 cm 2 or less, even more preferably 81 cm 2 or more and 400 cm 2 or less, particularly preferably
- the size can be appropriately selected from the range of 100 cm 2 or more and 400 cm 2 or less.
- the shape of the above-mentioned one side of the fibrin gel sheet of the present invention is not particularly limited, and can be appropriately selected depending on the size and shape of the transplant site. , octagon, etc.), rounded polygons, circles, ellipses, etc., but are not limited to these.
- the fibrin gel sheet of the present invention has a rectangular shape
- its vertical length is 15 mm or more, preferably 20 mm or more, more preferably 30 mm or more, still more preferably 40 mm or more, even more preferably 50 mm or more, Particularly preferably 60 mm or more, particularly more preferably 70 mm or more, particularly still more preferably 80 mm or more, even more preferably 90 mm or more, particularly preferably 100 mm or more
- the upper limit is not particularly limited, but for example, 200 mm or less. I can do it.
- the fibrin gel sheet of the present invention has a horizontal length of 15 mm or more, preferably 20 mm or more, more preferably 30 mm or more, even more preferably 40 mm or more, even more preferably 50 mm or more, particularly preferably 60 mm or more, especially more preferably is 70 mm or more, particularly preferably 80 mm or more, even more preferably 90 mm or more, particularly preferably 100 mm or more, and the upper limit thereof is not particularly limited, but may be, for example, 200 mm or less.
- the size of the fibrin gel sheet of the present invention is (vertical length) width x width length (horizontal length x length may be acceptable)), 15 mm or more to 200 mm or less x 15 mm or more to 200 mm or less, preferably 20 mm or more to 200 mm or less x 20 mm or more to 200 mm or less, More preferably 30 mm or more and 200 mm or less x 30 mm or more and 200 mm or less, still more preferably 40 mm or more and 200 mm or less x 40 mm or more and 200 mm or less, even more preferably 50 mm or more and 200 mm or less x 50 mm or more and 200 mm or less, particularly preferably 60 mm.
- the size can be appropriately selected from the range of ⁇ 200 mm or less x 90 mm or more and 200 mm or less, particularly preferably 100 mm or more - 200 mm or less x 100 mm or more and 200 mm or less.
- the size of the fibrin gel sheet of the present invention is 15 mm or more x 15 mm or more, preferably 20 mm x 20 mm. , 20 mm x 30 mm, or 30 mm x 30 mm, more preferably 40 mm x 40 mm, 40 mm x 50 mm, 50 mm x 50 mm, 40 mm x 60 mm, or 60 mm x 60 mm, even more preferably 70 mm x 70 mm, 80 mm x 80 mm, 90 mm x 90 mm, 100 mm. x 100 mm, or 200 mm x 200 mm, etc., but are not limited to these.
- the thickness of the fibrin gel sheet of the present invention can be appropriately set depending on factors such as the size and amount of encapsulated cells and the size of the transplant site, but is preferably 1 mm or less. Since the thickness of the fibrin gel sheet of the present invention is 1 mm or less, nutrients and oxygen can be efficiently supplied to the cells contained in the fibrin gel sheet, and the fibrin gel can be quickly decomposed after transplantation. achievable and desirable.
- the thickness of the fibrin gel sheet of the present invention can be, for example, 1 mm or less, 900 ⁇ m or less, 800 ⁇ m or less, 700 ⁇ m or less, 600 ⁇ m or less, 500 ⁇ m or less, or 400 ⁇ m or less, and the lower limit is not particularly limited, but for example, 100 ⁇ m or more, 200 ⁇ m or more, or 300 ⁇ m or more.
- the range of the thickness of the fibrin gel sheet of the present invention can be expressed using two numerical values respectively selected from the upper and lower limits.
- the thickness of the fibrin gel sheet of the present invention is 100 ⁇ m or more and 1 mm or less.
- the thickness of the fibrin gel sheet of the present invention is about 400 ⁇ m.
- the thickness of the fibrin gel sheet of the present invention does not need to be uniform throughout the sheet, and even the thickest part may be within the above range.
- the expression that cells are "uniformly dispersed and encapsulated" means that the seeded cells are not confined to any part of the fibrin gel sheet of the present invention, but are widely distributed within the fibrin gel sheet. means to be All cells do not need to be completely embedded in the fibrin gel sheet, but in a cell culture seeded on the surface of the fibrin gel and cultured adherently, the cells are localized only on the surface of the fibrin gel sheet.
- cells are "dispersed and encapsulated.” More specifically, in this specification, cells are "uniformly dispersed and encapsulated” at any plurality of locations (for example, 2, 4, or 6 locations, etc.) on the fibrin gel sheet of the present invention. ) means that the deviation of each cell number (cell density) included in a predetermined range is small, and preferably the value of each cell number is within ⁇ 20% of their average value, preferably ⁇ 15%. It means within a range of ⁇ 10%, more preferably within ⁇ 10%.
- the expression that cells are "uniformly dispersed and encapsulated” refers to the positional relationship between the seeded cells, for example, cells seeded on the surface or inside of the fibrin gel. is cultured and proliferated, and if the seeded cells come into contact with/combine with each other, it cannot be said that the cells are at least dispersed among the seeded cells, so the cells are It does not fall under the condition of being "uniformly dispersed and contained.”
- Examples of such a state in which the seeded cells are in contact/bond are a cell culture (e.g., cell sheet, etc.) formed by seeding and culturing on the surface/inside of a fibrin gel, Such aspects are not intended to be included in the present invention.
- the morphology of the seeded cells may be, for example, a state of a cell mass or cell aggregate in which cells aggregate and aggregate, i.e., a spheroid. This is not intended to exclude the possibility that the cells may be in the form of , or that the seeded cells will proliferate.
- cells are cells to be transplanted to a patient who requires them for cell therapy, and can be appropriately selected depending on the patient's symptoms, such as epidermis, nerves, brain, spinal cord, etc.
- Cells that constitute organs and tissues such as (but not limited to) the esophagus, stomach, small intestine, large intestine, bladder, urethra, lung, thyroid, pancreas, liver, muscle, skeleton, heart, blood vessels, spleen, kidney, and blood. Examples include cells having functions equivalent to or similar to those of the progenitor cells.
- Cells may be used alone or in combination with different cells.
- the form of cells in the fibrin gel sheet of the present invention is not particularly limited, and may be cells separated from each other (single cell state), or cell clusters or cell aggregates in which cells aggregate and aggregate.
- the cells may exist in the form of spheroids, or may be cells cultured in suspension. Examples of cells in the form of spheroids include, but are not limited to, pancreatic islet cells, hepatocytes, cardiomyocytes, and the like.
- cells may be cells collected from a living body, cultured cells, or cells obtained by freezing and thawing these cells.
- “cells” are cells derived from pluripotent stem cells obtained by inducing differentiation from pluripotent stem cells.
- pluripotency refers to the ability to differentiate into tissues and cells with various different forms and functions, and to differentiate into cells of any of the three germ layers. “Pluripotency” means that it cannot differentiate into a scutellum and therefore has no ability to form an individual, whereas “totipotency” means that it can differentiate into any tissue in the body, including the scutellum. ” is distinguished from.
- multipotency refers to the ability to differentiate into a limited number of cell lines.
- mesenchymal stem cells, hematopoietic stem cells, and neural stem cells are multipotent but not pluripotent.
- pluripotent stem cells refer to embryonic stem cells (ES cells) and similar pluripotent cells, that is, to various tissues of the body (endoderm, mesoderm, ectoderm). refers to cells that potentially have the ability to differentiate into all germ layers). Examples of cells having pluripotency similar to ES cells include "induced pluripotent stem cells” (sometimes referred to herein as “iPS cells”). Preferably, in the present invention, the pluripotent stem cells are human pluripotent stem cells.
- ES cells various mouse ES cell lines established by inGenius, RIKEN, etc. can be used for mouse ES cells, and for human ES cells, available from the National Institutes of Health.
- Various human ES cell lines established by NIH, RIKEN, Kyoto University, and Cellartis are available.
- ES cell lines include NIH's CHB-1 to CHB-12 lines, RUES1 line, RUES2 line, HUES1 to HUES28 line, etc., WiCell Research Institute's H1 line and H9 line, RIKEN's KhES-1 line, KhES-2 line, etc. strains, KhES-3 strain, KhES-4 strain, KhES-5 strain, SSES1 strain, SSES2 strain, SSES3 strain, etc. can be used.
- “Induced pluripotent stem cells” refer to cells obtained by reprogramming mammalian somatic cells or undifferentiated stem cells by introducing specific factors (nuclear reprogramming factors).
- specific factors nuclear reprogramming factors
- Yamanaka et al. established iPS cells by introducing four factors, Oct3/4, Sox2, Klf4, and c-Myc, into mouse fibroblasts.
- Nanog-iPS cells were established using Nanog expression as an indicator (Okita, K., Ichisaka, T. , and Yamanaka, S. (2007). Nature 448, 313-317.), iPS cells produced by a c-Myc-free method (Nakagawa M, Yamanaka S., et al.
- induced pluripotent stem cells Yu J., Thomson JA. et al., Science (2007) 318: 1917-1920.
- induced pluripotent stem cells created by Daley et al. Park IH, Daley GQ. et al., Nature (2007) 451:141-146
- induced pluripotent stem cells created by Sakurada et al. Japanese Unexamined Patent Publication No. 2008-307007, etc.
- human iPS cell lines include RIKEN's HiPS-RIKEN-1A strain, HiPS-RIKEN-2A strain, HiPS-RIKEN-12A strain, Nips-B2 strain, Kyoto University's Ff-WJ-18 strain, and Ff- I01s01 strain, Ff-I01s02 strain, Ff-I01s04 strain, Ff-I01s06 strain, Ff-I14s03 strain, Ff-I14s04 strain, QHJI01s01 strain, QHJI01s04 strain, QHJI14s03 strain, QHJI14s04 strain, 253G1 strain, 201B7 strain, 40 9B2 strain, 454E2 606A1 stock, 610B1 stock, 648A1 stock, CDI's MyCell iPS Cells (21525.102.10A) stock, MyCell
- Pluripotent stem cell-derived cells that can be used in the present invention include, but are not limited to, iPS cell-derived pancreatic islet cells (hereinafter sometimes referred to as "iPIC").
- iPICs include insulin producing cells and/or pancreatic ⁇ cells.
- Insulin-producing cells are cells characterized by the expression of at least one marker of insulin and NK6 homeobox 1 (NKX6.1).
- Pancreatic ⁇ cells are more mature cells than insulin-producing cells and are characterized by the expression of at least one marker of MAFA, UCN3, and IAPP.
- the term “marker” refers to a cell antigen or its gene that is specifically expressed by a predetermined cell type, such as a "marker protein” or “marker gene.”
- the marker is a cell surface marker, in which case enrichment, isolation, and/or detection of viable cells can be performed.
- the marker can be a positive selection marker or a negative selection marker.
- Detection of a marker protein can be performed using an immunological assay (ELISA, immunostaining, flow cytometry, etc.) using an antibody specific to the marker protein.
- Detection of marker genes can be performed using nucleic acid amplification methods and/or nucleic acid detection methods (RT-PCR, microarrays, biochips, etc.) known in the art.
- RT-PCR nucleic acid detection methods
- “positive” for a marker protein means that it is detected as positive by flow cytometry
- negative means that it is below the detection limit by flow cytometry.
- positive” for a marker gene means that it is detected by RT-PCR, and “negative” means that it is below the detection limit by RT-PCR.
- “Expression” is defined as the transcription and/or translation of a specific nucleotide sequence driven by a promoter within a cell.
- iPIC can be obtained from pluripotent stem cells by inducing differentiation according to known methods (WO2009/012428, WO2016/021734, Stem Cell Research (2015) 14, 185-197). That is, iPIC can be obtained using the following differentiation induction step: Step 1) Inducing differentiation from pluripotent stem cells to definitive endoderm cells; Step 2) Inducing differentiation from definitive endoderm cells to gastrula cells; Step 3) Inducing differentiation from gastrula cells to posterior foregut cells; Step 4) Inducing differentiation from posterior foregut cells to pancreatic progenitor cells; Step 5) Inducing differentiation from pancreatic progenitor cells to endocrine progenitor cells; Step 6) Differentiation of endocrine progenitor cells into iPICs is induced. Each step will be explained below, but the induction of differentiation into each cell is not limited to these methods.
- Step 1) Differentiation into definitive endoderm cells Pluripotent stem cells are first differentiated into definitive endoderm cells. Methods for inducing definitive endoderm from pluripotent stem cells are already known, and any of these methods may be used.
- the pluripotent stem cells are differentiated into definitive endoderm cells by being cultured in a medium containing activin A, more preferably in a medium containing activin A, a ROCK inhibitor, or a GSK3 ⁇ inhibitor.
- the number of cells at the start of culture is not particularly limited, and is 22,000 to 150,000 cells/cm 2 , preferably 22,000 to 100,000 cells/cm 2 , and more preferably 22,000 to 80,000 cells/cm 2 .
- the culture period is 1 to 4 days, preferably 1 to 3 days, particularly preferably 3 days.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%. Culture may be performed by either two-dimensional culture or three-dimensional culture.
- the media used in this step include RPMI 1640 medium, MEM medium, iMEM medium, DMEM/F12 medium, Improved MEM Zinc Option medium, Improved MEM/1% B-27/Penisilin Streptomycin medium, MCDB131/20mM Glucose/ NaHCO3 / Basal media used for culturing mammalian cells can be used, such as FAF-BSA/ITS-X/GlutaMAXTM/Ascorbic Acid/Penisilin Streptomycin media.
- the concentration of activin A in the medium is usually 30 to 200 ng/mL, preferably 50 to 150 ng/mL, more preferably 70 to 120 ng/mL, particularly preferably about 100 ng/mL.
- activin A can be included in the medium at low doses, for example in an amount of 5-100 ng/mL, preferably 5-50 ng/mL, more preferably 5-10 ng/mL.
- the concentration of activin A in the medium is about 0.1-100 ng/mL, preferably about 1-50 ng/mL, more preferably about 3-10 ng/mL.
- the concentration of the GSK3 ⁇ inhibitor in the medium is appropriately set depending on the type of GSK3 ⁇ inhibitor used.
- the concentration is usually 2 to 5 ⁇ M, preferably 2 to 4 ⁇ M, particularly preferably about It is 3 ⁇ M.
- the concentration of the ROCK inhibitor in the medium is appropriately set depending on the type of ROCK inhibitor used.
- the concentration is usually 5 to 20 ⁇ M, preferably 5 to 15 ⁇ M, particularly preferably about It is 10 ⁇ M.
- Insulin can further be added to the medium.
- Insulin can be included in the medium in an amount of 0.01-20 ⁇ M, preferably 0.1-10 ⁇ M, more preferably 0.5-5 ⁇ M.
- the concentration of insulin in the medium may be, but is not limited to, the concentration of insulin contained in the added B-27 supplement.
- the cells are cultured for one day in a medium containing activin A, a ROCK inhibitor, and a GSK3 ⁇ inhibitor, and then cultured for two more days in a medium containing only activin A, while changing the medium every day.
- pluripotent stem cells are cultured in the presence of low doses of activin A in a medium containing 0.01-20 ⁇ M insulin, followed by a second culture in medium without insulin. It can be manufactured by performing the following steps.
- Step 2) Differentiation into gastrula cells
- the definitive endoderm cells obtained in step 1) are further cultured in a medium containing growth factors to induce differentiation into gastrula cells.
- the culture period is 2 to 8 days, preferably about 4 days.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%. Culture may be performed by either two-dimensional culture or three-dimensional culture.
- a basic medium used for culturing mammalian cells can be used.
- growth factors serum substitutes, vitamins, antibiotics, etc. may be added to the medium as appropriate.
- EGF EGF, KGF, and FGF10 are preferred, EGF and/or KGF are more preferred, and KGF is even more preferred.
- the concentration of the growth factor in the medium is appropriately set depending on the type of growth factor used, but is usually about 0.1 nM to 1000 ⁇ M, preferably about 0.1 nM to 100 ⁇ M.
- the concentration is about 5-2000 ng/mL (i.e., about 0.8-320 nM), preferably about 5-1000 ng/mL (i.e., about 0.8-160 nM), more preferably about 10-1000 ng/mL (i.e., about 0.8-160 nM). 1000 ng/mL (ie, approximately 1.6-160 nM).
- the concentration is about 5-2000 ng/mL (ie, about 0.3-116 nM), preferably about 10-1000 ng/mL (ie, about 0.6-58 nM).
- the concentration is usually 5 to 150 ng/mL, preferably 30 to 100 ng/mL, particularly preferably about 50 ng/mL.
- Step 3) Differentiation into posterior foregut cells
- the gastrula cells obtained in step 2) are further cultured in a medium containing growth factors, cyclopamine, noggin, etc., and induced to differentiate into posterior foregut cells.
- the culture period is 1 to 5 days, preferably about 2 days. Culture may be performed by either two-dimensional culture or three-dimensional culture.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%.
- a basic medium used for culturing mammalian cells can be used.
- growth factors serum substitutes, vitamins, antibiotics, etc. may be added to the medium as appropriate.
- EGF EGF, KGF, and FGF10 are preferred, EGF and/or KGF are more preferred, and KGF is even more preferred.
- the concentration of the growth factor in the medium is appropriately set depending on the type of growth factor used, but is usually about 0.1 nM to 1000 ⁇ M, preferably about 0.1 nM to 100 ⁇ M.
- the concentration is about 5-2000 ng/mL (i.e., about 0.8-320 nM), preferably about 5-1000 ng/mL (i.e., about 0.8-160 nM), more preferably about 10-1000 ng/mL (i.e., about 0.8-160 nM). 1000 ng/mL (ie, approximately 1.6-160 nM).
- the concentration is about 5-2000 ng/mL (ie, about 0.3-116 nM), preferably about 10-1000 ng/mL (ie, about 0.6-58 nM).
- the concentration is usually 5 to 150 ng/mL, preferably 30 to 100 ng/mL, particularly preferably about 50 ng/mL.
- the concentration of cyclopamine in the medium is not particularly limited, but is usually 0.5 to 1.5 ⁇ M, preferably 0.3 to 1.0 ⁇ M, particularly preferably about 0.5 ⁇ M.
- the concentration of Noggin in the medium is not particularly limited, but is usually 10 to 200 ng/mL, preferably 50 to 150 ng/mL, particularly preferably about 100 ng/mL.
- Step 4) Differentiation into pancreatic progenitor cells
- the posterior foregut cells obtained in step 3) are further treated in a medium containing a factor having CDK8/19 inhibitory activity, preferably a factor having CDK8/19 inhibitory activity and a growth factor.
- the culture period is 2 to 10 days, preferably about 5 days. Culture may be performed by either two-dimensional culture or three-dimensional culture.
- the posterior foregut cells obtained in step 3 were incubated with 0.25% trypsin-EDTA solution.
- the cells are treated and dispersed in the liquid by pipetting to obtain a cell dispersion, the resulting dispersion is centrifuged, the collected cells are resuspended in a small amount of new medium, and the cell suspension is Re-seeded in the new medium in step 4).
- a basic medium used for culturing mammalian cells can be used.
- growth factors serum substitutes, vitamins, antibiotics, etc. may be added to the medium as appropriate.
- the various compounds or salts thereof mentioned above can be used, and the amount added to the medium is determined as appropriate depending on the compound or salt thereof used, but it is usually about 0.00001 ⁇ M. ⁇ 5 ⁇ M, preferably 0.00001 ⁇ M to 1 ⁇ M.
- the concentration of the factor having CDK8/19 inhibitory activity in the medium is preferably a concentration that achieves 50% or more inhibitory activity against CDK8/19.
- EGF EGF, KGF, and FGF10 are preferred, KGF and/or EGF are more preferred, and KGF and EGF are even more preferred.
- the concentration of the growth factor in the medium is appropriately set depending on the type of growth factor used, but is usually about 0.1 nM to 1000 ⁇ M, preferably about 0.1 nM to 100 ⁇ M.
- the concentration is about 5-2000 ng/mL (i.e., about 0.8-320 nM), preferably about 5-1000 ng/mL (i.e., about 0.8-160 nM), more preferably about 10-1000 ng/mL (i.e., about 0.8-160 nM). 1000 ng/mL (ie, approximately 1.6-160 nM).
- the concentration is about 5-2000 ng/mL (ie, about 0.3-116 nM), preferably about 10-1000 ng/mL (ie, about 0.6-58 nM).
- the concentrations are usually 5 to 150 ng/mL for EGF, preferably 30 to 100 ng/mL, particularly preferably about 50 ng/mL, and 10 to 200 ng/mL for KGF.
- it is 50 to 150 ng/mL, particularly preferably about 100 ng/mL.
- the first day of culturing in step 4) may be carried out in the presence of a ROCK inhibitor, and thereafter culturing may be carried out in a medium containing no ROCK inhibitor.
- the medium may contain a protein kinase C (PKC) activator.
- PKC protein kinase C
- PDBu PKC activator II
- TPB PKC activator V
- the PKC activator is added at a concentration of about 0.1 to 100 ng/mL, preferably about 1 to 50 ng/mL, more preferably about 3 to 10 ng/mL.
- dimethyl sulfoxide and/or activin (1 to 50 ng/mL) may be added to the medium.
- a serum substitute eg, B-27 supplement, ITS-G
- amino acids, L-glutamine, GlutaMAX (product name) may be added to the medium.
- antibiotics e.g. Antibiotic-Antimycotic, penicillin, streptomycin, or mixtures thereof
- antibacterial agents For example, amphotericin B
- antioxidants pyruvate, buffers, inorganic salts, etc.
- concentration in the medium is usually 0.01 to 20% by weight, preferably 0.1 to 10% by weight.
- Culture may be performed by either two-dimensional culture or three-dimensional culture.
- cell culture is performed by adhesive culture without using feeder cells.
- culture containers such as dishes, flasks, microplates, and cell culture sheets such as OptiCell (product name) (Nunc) are used.
- Culture vessels may be surface treated to improve adhesion (hydrophilicity) with cells, collagen, gelatin, poly-L-lysine, poly-D-lysine, laminin, fibronectin, Matrigel (e.g. BD Matrigel (Japan)). It is preferably coated with a cell adhesion substrate such as Becton Dickinson, Inc.) or vitronectin.
- a culture vessel coated with type I collagen, Matrigel, fibronectin, vitronectin, poly-D-lysine, etc. is preferable, and a culture vessel coated with Matrigel or poly-D-lysine is more preferable.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%.
- the pancreatic progenitor cells obtained in step 4) can be further purified using a known surface marker such as glycoprotein 2 (GP2).
- GP2 glycoprotein 2
- the above purification can be performed by a method known per se, for example, using beads on which anti-GP2 antibodies are immobilized.
- Step 5) Differentiation into endocrine progenitor cells
- the pancreatic progenitor cells obtained in step 4) are further cultured in a medium containing growth factors to induce differentiation into endocrine progenitor cells.
- Culture may be performed by either two-dimensional culture or three-dimensional culture.
- the pancreatic progenitor cells obtained in step 4) are treated with a 0.25% trypsin-EDTA solution and dispersed in the solution by pipetting to obtain a cell dispersion, The resulting dispersion is centrifuged, the collected cells are resuspended in a small amount of new medium, and the cell suspension is re-inoculated into the new medium in step 5).
- the culture period is 2 to 3 days, preferably about 2 days.
- a basic medium used for culturing mammalian cells can be used. According to a previous report (Nature Biotechnology 2014; 32:1121-1133), SANT1, retinoic acid, ALK5 inhibitor II, T3, and LDN were added to the medium, and further Wnt inhibitor, ROCK inhibitor, and FGF (preferably FGF2) were added. , serum substitutes, vitamins, antibiotics, etc. may be added as appropriate.
- the culture is performed in a non-adhesive manner without using feeder cells. During culture, dishes, flasks, microplates, porous plates (Nunc), etc., or bioreactors are used.
- the culture container is preferably surface-treated to reduce adhesion to cells.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%.
- the endocrine progenitor cells obtained in step 5) can be further purified using a known surface marker such as glycoprotein 2 (GP2).
- GP2 glycoprotein 2
- the above purification can be performed by a method known per se, for example, using beads on which anti-GP2 antibodies are immobilized.
- Step 6) Differentiation into iPIC
- the endocrine progenitor cells obtained in step 5) are further cultured in a medium containing growth factors to induce differentiation into iPIC.
- the culture period is 10 to 30 days, preferably about 10 to 20 days.
- a basic medium used for culturing mammalian cells can be used.
- the medium contained ALK5 inhibitor II, T3, LDN, ⁇ -secretase inhibitor XX, ⁇ -secretase inhibitor RO, N-cysteine, AXL inhibitor, and ascorbic acid according to a previous report (Nature Biotechnology 2014; 32:1121-1133).
- Wnt inhibitors, ROCK inhibitors, FGF (preferably FGF2), serum substitutes, vitamins, antibiotics, etc. may be added as appropriate.
- the medium may be supplemented with ALK5 inhibitor II, T3, LDN, the ⁇ -secretase inhibitor RO, and ascorbic acid, or T3, ALK5 inhibitor II, ZnSO 4 , heparin, N-acetylcysteine, Trolox, and R428 may also be added.
- the culture may be performed by either two-dimensional culture or three-dimensional culture. Culture does not use feeder cells. In the case of three-dimensional culture, non-adhesive culture is used. During culture, dishes, flasks, microplates, porous plates (Nunc), etc., or bioreactors are used.
- the culture container is preferably surface-treated to reduce adhesion to cells.
- the culture temperature is not particularly limited, but the culture is carried out at 30 to 40°C (for example, 37°C). Further, the carbon dioxide concentration in the culture container is, for example, about 5%.
- the fibrin gel sheet of the present invention is characterized by having a higher number of cells (higher density of cells) that could not be achieved with conventional fibrin gel sheets for cell transplantation . More than .5 ⁇ 10 6 , preferably 2 ⁇ 10 6 or more, more preferably 2.5 ⁇ 10 6 or more, even more preferably 3 ⁇ 10 6 or more, even more preferably 4.5 ⁇ 10 6 As mentioned above, it is particularly preferably contained in an amount of 5 x 10 6 or more, and the upper limit is not particularly limited, but it can be, for example, 10 x 10 6 or less, preferably 7 x 10 6 or less.
- the fibrin gel sheet of the present invention includes cells 1. More than 5 ⁇ 10 6 to 10 ⁇ 10 6 or less/cm 2 , preferably 2 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , more preferably 2.5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , more preferably 3 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , even more preferably 4.5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , particularly preferably in an amount appropriately selected from the range of 5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 .
- the above-mentioned iPIC usually exists in the form of cell clusters having a size of about 100 ⁇ m to 200 ⁇ m, but the fibrin gel sheet of the present invention has more than 3,000 iPIC cells per cm 2 , preferably 4,000 iPICs per cm 2 . 20,000 or more, more preferably 5,000 or more, even more preferably 6,000, even more preferably 9,000, particularly preferably 10,000 or more, and the upper limit is not particularly limited, but for example, 20,000 or more, Preferably, the number can be 14,000 or less.
- the range of the number of iPICs (cell clusters) contained in the fibrin gel sheet of the present invention can be expressed using two numerical values respectively selected from the lower limit and upper limit values.
- the fibrin gel sheet of the present invention iPIC (cell clusters), more than 3,000 to 20,000 cells/cm 2 , preferably 4,000 to 20,000 cells/cm 2 , more preferably 5,000 to 20,000 cells/cm 2 , even more preferably 6,000 cells/cm 2 . It may be included in an amount appropriately selected from the range of from 9,000 to 20,000 pieces/cm 2 , more preferably from 9,000 to 20,000 pieces/cm 2 , particularly preferably from 10,000 to 20,000 pieces/cm 2 . can.
- the fibrin gel is formed by allowing fibrinogen and thrombin to interact in a suitable solvent (eg, water, physiological saline, etc.).
- a suitable solvent eg, water, physiological saline, etc.
- the amounts of fibrinogen and thrombin used to form the fibrin gel and the method for mixing the two can be appropriately determined based on the method for producing a fibrin gel sheet of the present invention, which will be described in detail below.
- the fibrin gel sheet of the present invention further includes a biodegradable material having a particulate form (hereinafter sometimes referred to as "biodegradable particles").
- biodegradable particles a biodegradable material having a particulate form
- biodegradable particles are added and suspended in a fibrinogen solution together with cells in the method for producing a fibrin gel sheet of the present invention, which is detailed below, and are dispersed in the solution to suppress cell precipitation. This allows cells to be dispersed and encapsulated in the fibrin gel sheet that is finally obtained.
- the biodegradable material constituting the "biodegradable particles” may be any material that can be decomposed in vivo by hydrolysis, etc., and is not particularly limited, but examples include polylactic acid (PLA), Caprolactone (PCL), polyurethane (PU), polyethylene glycol (PEG), polyhydroxyethyl methacrylate, polyester, polyglycolic acid (PGA), polylactic acid-co-glycolic acid (PLGA), poly(3-hydroxybutyrate-co) -hydroxyvalerate) (PHBV), poly(ethylene-co-vinyl acetate) (PEVA) polyacrylamide, polyethylene oxide, polyethylene amine, polyhydroxybutyric acid, poly(N-vinylpyrrolidone), polyvinyl alcohol, polypropylene fumarate, poly Acrylic acid, poly-e-caprolactone, polymethacrylic acid, polyvinylidene difluoride (PVDF), pectic acid, hyaluronic acid, heparin sul
- the biodegradable particles are made from a gel-like biodegradable material selected from the group consisting of gelatin, collagen, hyaluronic acid, carboxymethylcellulose, PLA, PGA, PLGA, etc., modified versions thereof, and combinations thereof. configured.
- the form of the "biodegradable particles” may be any form that can suppress cell precipitation and maintain dispersion in the solution, such as block, bead, pellet, sheet, gel, etc. It can have any morphology and can be solid or porous.
- the form of the "biodegradable particles” is preferably a three-dimensional shape, such as a sphere, a polyhedron such as a tetrahedron, a hexahedron, a cylinder, a prism, a cone, a truncated cone, a pyramid, a truncated pyramid, a torus, a disc, Examples include ellipsoids, modified solids thereof, etc., but are not limited thereto.
- the size of the "biodegradable particles” may be any size that can suppress cell precipitation and maintain dispersion in the solution, and that does not hinder the formation of the fibrin gel sheet of the present invention, with an opening of 100 ⁇ m or more.
- the size may be such that it can pass through 1000 ⁇ m, preferably 100 ⁇ m to 800 ⁇ m, more preferably 150 ⁇ m to 400 ⁇ m, and still more preferably 200 ⁇ m.
- the biodegradable particles are gelatin gel and have a spherical shape that can pass through an opening of 200 ⁇ m.
- the amount of biodegradable particles contained in the fibrin gel sheet of the present invention may be any amount that can suppress cell precipitation and maintain dispersion in the fibrinogen solution and does not hinder the formation of the fibrin gel sheet of the present invention.
- the amount of biodegradable particles in the fibrinogen solution is 10 to 30 w/v%, preferably 15 to 25 w/v%, more preferably 18 to 22 w/v%, and even more preferably 20 w/v%. can be included.
- the biodegradable particles may maintain their form and shape, or may be partially or entirely integrated with the fibrin gel.
- the fibrin gel sheet of the present invention further includes a biodegradable gelling agent.
- the "biodegradable gelling agent” is added to and dissolved in a fibrinogen solution together with cells, imparts viscosity to the solution, and This suppresses precipitation of cells in solution and enables maintenance of dispersion, thereby allowing cells to be dispersed and encapsulated in the fibrin gel sheet finally obtained.
- the "biodegradable gelling agent” is a substance that can be decomposed in vivo by hydrolysis, etc., and is also capable of imparting a viscosity to the solution that can suppress cell precipitation and maintain dispersion.
- the material is not particularly limited as long as it does not interfere with the formation of the fibrin gel sheet of the present invention, and for example, the above-mentioned biodegradable materials can be used (excluding fibrin).
- the biodegradable gelling agent includes collagen, hyaluronic acid, heparin sulfate, chondroitin sulfate, heparan sulfate proteoglycan, carboxymethyl cellulose, alginic acid, cellulose, silk fibroin, keratin, gelatin, dextran, PEG, etc., and modified products thereof, Biodegradable materials selected from the group consisting of: and combinations thereof can be used.
- the amount of the biodegradable gelling agent contained in the fibrin gel sheet of the present invention is an amount that can suppress cell precipitation and maintain dispersion in the fibrinogen solution, and that does not interfere with the formation of the fibrin gel sheet of the present invention.
- the amount of the biodegradable gelling agent in the fibrinogen solution is 0.2 to 2 w/v%, preferably 0.4 to 1.5 w/v%, more preferably 0.5 to 1 w/v%. v %, more preferably 0.75 w/v %.
- the fibrin gel sheet of the present invention can further include a support.
- the support supports the fibrin gel sheet of the present invention during production and/or use, and can contribute to improving the operability of the fibrin gel sheet.
- the support may be made of the aforementioned biodegradable materials or non-biodegradable materials, such as polyester (PEs), polypropylene (PP), polyethylene (PE), polytetrafluoroethylene (PTFE), polyethylene terephthalate (PET), nylon, It is made of silk (not limited thereto) and has any shape (eg, sheet, mesh, plate, woven fabric, nonwoven fabric, etc.) capable of supporting the fibrin gel sheet of the present invention.
- the support may be adhered to the surface of the fibrin gel sheet of the present invention, or a part or the entire support may be fixed in the fibrin gel sheet of the present invention.
- the fibrin gel sheet of the present invention includes a mesh-like support made of polyester or polylactic acid-co-glycolic acid.
- the fibrin gel sheet of the present invention may be provided in the form of a laminate in which a plurality of fibrin gel sheets are stacked.
- the laminate may be one in which each fibrin gel sheet contains the same cells, or may be a combination of fibrin gel sheets in which some or all of the sheets contain different cells.
- the present invention also relates to a method for producing a fibrin gel sheet of the present invention (hereinafter sometimes referred to as the "method of the present invention"), and the method is a method in which cells are suspended.
- the fibrinogen solution is reacted with thrombin to form and gel (solidify) it into a sheet having a surface area of 2.25 cm 2 or more and a thickness of 1 mm or less.
- Fibrin gel can be obtained by combining fibrinogen and thrombin in an appropriate solvent (eg, water, physiological saline, etc.) and gelling (solidifying) the mixture.
- Cells are uniformly dispersed and suspended in the fibrinogen solution, and the amount of cells can be determined based on the fibrin gel sheet after being molded, that is, 1.5 x 10 6 per cm 2 of fibrin gel sheet. More than 2 x 10 6 pieces, preferably 2 x 10 6 or more, more preferably 2.5 x 10 6 or more, even more preferably 3 x 10 6 or more, even more preferably 4.5 x 10 6 or more, particularly preferably Although the upper limit is not particularly limited, it can be suspended in an amount of 5 x 10 6 or more, for example, 10 x 10 6 or less, preferably 7 x 10 6 or less.
- the range of the number of cells suspended in the fibrinogen solution can be expressed using two numbers selected from the lower limit and upper limit, respectively.
- More than ⁇ 10 6 to 10 ⁇ 10 6 or less/cm 2 preferably 2 ⁇ 10 6 or more to 10 ⁇ 10 6 or less /cm 2 , more preferably 2.5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , more preferably 3 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , even more preferably 4.5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 , Particularly preferably, they can be suspended in an amount appropriately selected from the range of 5 ⁇ 10 6 or more to 10 ⁇ 10 6 or less/cm 2 .
- the fibrinogen solution when the cells are iPICs, contains iPICs in a cell mass of more than 3,000, preferably 4,000 or more, more preferably more than 4,000 per cm 2 of fibrin gel sheet.
- the amount is 5,000 or more, more preferably 6,000 or more, even more preferably 9,000 or more, particularly preferably 10,000 or more, and the upper limit is not particularly limited, but for example, 20,000 or less, preferably 14,000 or less. It can be suspended in an amount that becomes .
- the range of the number of iPIC (cell clusters) suspended in the fibrinogen solution can be expressed using two numbers selected from the lower limit and upper limit, respectively.
- more than 3000 to less than 20000 pieces/cm 2 preferably more than 4000 to less than 20000 pieces/cm 2 , more preferably more than 5000 to less than 20000 pieces/cm 2 , even more preferably more than 6000 pieces/cm 2 Suspended in an amount appropriately selected from the range of ⁇ 20,000 cells/cm 2 , more preferably 9,000 to 20,000 cells/cm 2 , particularly preferably 10,000 to 20,000 cells/cm 2 . be able to.
- the surface area of one side of the sheet is 2.25 cm 2 or more, preferably 4 cm 2 or more, more preferably 9 cm 2 or more, more preferably 16 cm 2 or more, even more preferably 25 cm 2 or more, especially preferably 36 cm 2 or more, especially more preferably 49 cm 2 or more, even more preferably 64 cm 2 or more, even more preferably 81 cm 2 or more,
- the size is 100 cm 2 or more, and the upper limit is not particularly limited and can be appropriately selected depending on the size and shape of the transplant site, but the size can be, for example, 400 cm 2 or less.
- the range of the surface area of the fibrin gel sheet obtained by molding can be expressed using two values selected from the lower limit and upper limit, for example, the surface area of the fibrin gel sheet obtained by molding is 2.25 cm. 2 or more and 400 cm 2 or less, preferably 4 cm 2 or more and 400 cm 2 or less, more preferably 9 cm 2 or more and 400 cm 2 or less, even more preferably 16 cm 2 or more and 400 cm 2 or less , even more preferably 25 cm 2 or more and 400 cm 2 or less.
- the size can be appropriately selected from the range of 100 cm 2 or more to 400 cm 2 or less.
- the shape of the above-mentioned one side of the fibrin gel sheet obtained by molding is not particularly limited, and can be appropriately selected depending on the size and shape of the implantation site. Examples include, but are not limited to, polygons with rounded corners, circles, ellipses, etc. (squares, octagons, etc.).
- the vertical length is 15 mm or more, preferably 20 mm or more, more preferably 30 mm or more, even more preferably 40 mm or more, even more preferably 50 mm or more, particularly preferably 60 mm or more.
- the upper limit is not particularly limited, but can be, for example, 200 mm or less
- the horizontal length is 15 mm or more, preferably 20 mm or more, more preferably 30 mm or more, even more preferably 40 mm or more, even more preferably 50 mm or more, especially preferably 60 mm or more, especially more preferably 70 mm or more, even more preferably
- the length is 80 mm or more, particularly preferably 90 mm or more, particularly preferably 100 mm or more, and the upper limit thereof is not particularly limited, but may be, for example, 200 mm or less.
- the range of the vertical length and horizontal length can be expressed using two numerical values respectively selected from the lower limit and upper limit.
- the size of the fibrin gel sheet obtained by molding is (vertical length).
- Length x horizontal length horizontal length x vertical length may be used
- 15 mm or more and 200 mm or less x 15 mm or more and 200 mm or less preferably 20 mm or more and 200 mm or less x 20 mm or more and 200 mm or less , more preferably 30 mm or more and 200 mm or less x 30 mm or more and 200 mm or less, still more preferably 40 mm or more and 200 mm or less x 40 mm or more and 200 mm or less, even more preferably 50 mm or more and 200 mm or less x 50 mm or more and 200 mm or less, particularly preferably 60 mm or more and 200 mm or less x 60 mm or more and 200 mm or less, especially more preferably 70 mm or more and 200 mm or less x 70 mm or more and
- the size of the fibrin gel sheet obtained by molding is 15 mm or more x 15 mm or more, preferably 20 mm x 20 mm, 20 mm x 30 mm, or 30 mm x 30 mm, more preferably 40 mm x 40 mm, 40 mm x 50 mm, 50 mm x 50 mm, 40 mm x 60 mm, or 60 mm x 60 mm, even more preferably 70 mm x 70 mm, 80 mm x 80 mm, 90 mm x 90 mm, Examples include 100 mm x 100 mm, 200 mm x 200 mm, etc., but the size is not limited to these.
- the thickness of the fibrin gel sheet obtained by molding can be appropriately set depending on factors such as the size and amount of the encapsulated cells and the size of the transplant site, but is preferably 1 mm or less. Since the thickness of the fibrin gel sheet obtained by molding is 1 mm or less, nutrients and oxygen can be efficiently supplied to the cells contained in the fibrin gel sheet, and the fibrin gel can be rapidly decomposed after transplantation. can be achieved and is preferable.
- the thickness of the fibrin gel sheet obtained by molding can be, for example, 1 mm or less, 900 ⁇ m or less, 800 ⁇ m or less, 700 ⁇ m or less, 600 ⁇ m or less, 500 ⁇ m or less, or 400 ⁇ m or less, and the lower limit is not particularly limited, but for example, 100 ⁇ m or more, It can be 200 ⁇ m or more, or 300 ⁇ m or more. Further, the range of the thickness of the fibrin gel sheet obtained by molding can be expressed using two values selected from the upper and lower limits, for example, the thickness of the fibrin gel sheet obtained by molding is 100 ⁇ m or more.
- the thickness can be 1 mm or less, 100 ⁇ m to 800 ⁇ m or less, 200 ⁇ m to 600 ⁇ m or less, or 300 ⁇ m or more to 400 ⁇ m or less, and the thickness can be appropriately selected from these ranges.
- the thickness of the fibrin gel sheet obtained by molding can be about 400 ⁇ m. Note that the thickness of the fibrin gel sheet obtained by molding does not need to be uniform throughout the sheet, and even the thickest part may be within the above range.
- the gelation (solidification) rate of the fibrin gel to be formed is adjusted by adjusting the blending amount of fibrinogen and thrombin and/or the temperature during molding, and the fibrin gel having a desired size is adjusted. This allows it to be spread and molded into a gel sheet.
- the fibrinogen solution contains fibrinogen in an amount of 5 to 150 mg/mL, preferably 10 to 80 mg/mL, more preferably 20 mg/mL, and thrombin is 0.4 U or less, preferably 0.0 U per 1 mg of fibrinogen. It is 3U or less or 0.25U or less, more preferably 0.2U or less or 0.15U or less, even more preferably 0.1U or less or 0.08U or less, and the lower limit is not particularly limited, but the blending ratio is 0.04U or more. Act in an amount that becomes .
- thrombin can act on fibrinogen in any form, and may be added directly to the fibrinogen solution, or after being dissolved in an appropriate solvent (e.g., water, physiological saline, etc.) to form a thrombin solution, May be mixed with fibrinogen solution.
- an appropriate solvent e.g., water, physiological saline, etc.
- any means can be used to mold the fibrinogen solution reacted with thrombin into a sheet having a desired size and thickness.
- any molding means conventionally known in the field of manufacturing polymer sheets (e.g. , molding, extrusion molding, calendar molding, inflation molding, etc.), and is preferably carried out by molding.
- the fibrinogen solution can be easily injected into a sheet mold and spread and molded using a spatula, trowel, etc. as necessary.
- the fibrinogen solution reacted with thrombin is shaped under cooling.
- the cooling temperature may be any temperature that allows the fibrin gel solution to be molded into a sheet having a desired size and thickness, and is not particularly limited, for example, 2 to 8°C, preferably 2 to 6°C. °C, more preferably 4°C. Cooling may be performed as long as the fibrin gel solution can be cooled, and the fibrin gel solution may be cooled, or the molding means (for example, a mold, etc.) in contact with the fibrin gel solution may be cooled.
- the above-mentioned support may be integrated with the fibrin gel sheet before the molded fibrin gel solution gels (solidifies).
- the support may be adhered to the surface of the fibrin gel solution, or part or all of it may be immersed in the fibrin gel solution. By gelling (solidifying) this, it is possible to obtain a fibrin gel sheet that is integrally provided with the support.
- biodegradable particles are further added and suspended in the fibrinogen solution in which cells are suspended, or a “biodegradable gelling agent” is added and dissolved.
- a biodegradable gelling agent is added and dissolved.
- biodegradable particles can be used, and can suppress cell precipitation and maintain dispersion in a fibrinogen solution in which cells are suspended, and can be used in the fibrin gel sheet of the present invention.
- Biodegradable particles can be added and suspended in an amount that does not interfere with cell formation; for example, biodegradable particles can be added to a fibrinogen solution in which cells are suspended in an amount of 10 to 30 w/v%, preferably 15 to 25 w/v%, more preferably It is added and suspended in an amount of 18 to 22 w/v%, more preferably 20 w/v%.
- the biodegradable particles are gelatin gel particles, have a spherical shape that can pass through an opening of 200 ⁇ m, and are added in an amount of 20 w/v % to a fibrinogen solution in which cells are suspended; suspend
- the "biodegradable gelling agent” can be one defined above, which can suppress cell precipitation and maintain dispersion in the fibrinogen solution in which the cells are suspended, and which can be used for the fibrin of the present invention.
- the biodegradable gelling agent can be added and dissolved in an amount that does not interfere with the formation of a gel sheet, for example, 0.2 to 2 w/v%, preferably 0.4 to 1%, of a biodegradable gelling agent is added to a fibrinogen solution in which cells are suspended. It is added and dissolved in an amount of .5 w/v%, more preferably 0.5 to 1 w/v%, even more preferably 0.75 w/v%.
- the biodegradable gelling agent is collagen, which is added and dissolved in an amount of 0.75 w/v % to the fibrinogen solution in which the cells are suspended.
- the fibrinogen solution contains fibrinogen in an amount of 5 to 150 mg/mL, preferably 20 to 100 mg/mL, more preferably 80 mg/mL.
- Thrombin may be used in an amount sufficient to gel (solidify) the fibrinogen solution, for example, 0.5 to 5 U, preferably 1 to 4 U, more preferably 1.5 to 3 U per mg of fibrinogen. It is applied in an amount of .5U.
- Thrombin can be made to act on fibrinogen in any form, and can be added directly to a fibrinogen solution by dissolving it in an appropriate solvent (e.g., water, physiological saline, etc.) to make a thrombin solution, and then adding it to the fibrinogen solution.
- an appropriate solvent e.g., water, physiological saline, etc.
- the effect can be exerted by adding thrombin, by including thrombin in the "substrate” described in detail below, or by a combination of a plurality of them. If the substrate contains thrombin, the fibrinogen and thrombin can be combined by applying a fibrinogen solution to the substrate.
- any means can be used to form the fibrinogen solution in which cells are suspended into a sheet having a desired size and thickness, such as coating the fibrin gel solution on an appropriate base material, It can be carried out by one or a combination of any forming means conventionally known in the sheet manufacturing field (for example, mold forming, extrusion molding, calendar molding, inflation molding, etc.), and preferably by molding onto a suitable base material. It is done by coating.
- any forming means conventionally known in the sheet manufacturing field (for example, mold forming, extrusion molding, calendar molding, inflation molding, etc.), and preferably by molding onto a suitable base material. It is done by coating.
- it is possible to maintain uniform dispersion of suspended cells, and the fibrinogen solution in which cells are suspended can be injected not only at once but also continuously ( Even when the injection process takes a long time from the start to the end of the injection, it is possible to easily form the cell into a sheet in which the cells are uniformly dispersed.
- the "base material” may be anything that has a flat surface that allows a sheet to be formed on its surface, and is not particularly limited, but it is preferable to use the above-mentioned support as the base material. can.
- the fibrinogen solution in which cells are suspended can be applied to the substrate by any means, for example, by injection, by using one or more of a spatula, a trowel, a spray, a bioprinter (2D or 3D printer), etc. can be performed manually and/or mechanically, preferably mechanically using a bioprinter.
- the fibrin gel sheet of the present invention produced by the method of the present invention can be used in a cell culture medium such as, but not limited to, Dulbecco's modified Eagle's medium (DMEM), RPMI1640 medium, MEM medium, CMRL medium, etc. or a buffer, such as Stored in an isotonic solution such as, but not limited to, phosphate buffered saline (PBS), Hank's Balanced Salt Solution (HBSS), etc., such as, but not limited to, physiological saline until use. I can do it.
- DMEM Dulbecco's modified Eagle's medium
- RPMI1640 medium MEM medium
- CMRL medium CMRL medium
- a buffer such as Stored in an isotonic solution such as, but not limited to, phosphate buffered saline (PBS), Hank's Balanced Salt Solution (HBSS), etc., such as, but not limited to, physiological saline until use. I can do it.
- the fibrin gel sheet of the present invention can be used depending on the above-mentioned cells to be dispersed and encapsulated, such as epidermis, nerves, brain, spinal cord, esophagus, stomach, small intestine, large intestine, bladder, urethra, lung, etc.
- Cells that have the same or similar functions as cells constituting organs and tissues such as (but not limited to) the thyroid, pancreas, liver, muscle, skeleton, heart, blood vessels, spleen, kidney, blood, etc., and their progenitor cells. It can be used in cell therapy methods to enhance or maintain the functions of organs and tissues, or to supplement or supplement the functions of those organs and tissues that have been reduced or lost due to diseases, disorders, etc. used for transplantation into patients in need.
- the fibrin gel sheet of the present invention when the fibrin gel sheet of the present invention contains iPIC derived from pluripotent stem cells, the fibrin gel sheet of the present invention can reduce the blood sugar level (glucose) in the transplanted patient by the action of insulin and glucagon secreted by the iPIC. ) levels can be improved and/or maintained, and blood sugar levels can be controlled to normal values. Accordingly, the fibrin gel sheets of the present invention containing iPICs can be used to treat or prevent diseases, disorders, or conditions in which it is necessary to improve and/or maintain blood sugar levels.
- the fibrin gel sheets of the present invention containing iPICs can be used to treat or prevent diseases, disorders, or conditions in which it is necessary to improve and/or maintain blood sugar levels.
- Such diseases, disorders, or conditions include diabetes (type 1 diabetes, type 2 diabetes), altered fasting and postprandial glucose levels, and hypoglycemia (e.g., hypoglycemia due to insulin administration in diabetic patients). These include, but are not limited to.
- treatment means treatment, cure, prevention, or improvement of remission of a disease, disorder, or symptom, or reduction of the rate of progression of a disease, disorder, or symptom.
- prevention means reducing the possibility or risk of developing a disease, disorder, or symptom, or delaying the onset of a disease, disorder, or symptom.
- the fibrin gel sheet of the present invention is transplanted to patients who require it for cell therapy, such as mammals such as mice, rats, hamsters, rabbits, cats, dogs, cows, sheep, monkeys, and humans. (but not limited to), preferably large mammals, and particularly preferably humans.
- the fibrin gel sheet of the present invention When the fibrin gel sheet of the present invention is implanted, the fibrin gel is rapidly decomposed, and the encapsulated cells can quickly come into contact with the host tissue and receive oxygen, nutrients, etc. Therefore, the fibrin gel sheet of the present invention can be transplanted into tissues with poor blood vessels (for example, subcutaneously), and prior angiogenesis treatment may not be necessary or necessary upon transplantation.
- the fibrin gel sheet of the present invention allows one or more sheets to be transplanted depending on the patient's symptoms and the size of the transplant site, and since the fibrin gel sheet of the present invention is thin, Depending on the situation, a plurality of sheets may be stacked and transplanted.
- iPIC iPIC is produced using human iPS cells (Ff-I14-s04) according to the above steps 1)-6) and previously reported (Stem Cell Research (2015) 14, 185-197; Nature Biotechnology 2014; 32:1121-1133). It was prepared by inducing differentiation from a strain.
- pancreatic progenitor cells prepared by inducing differentiation from human iPS cells were mixed with differentiation factors (SANT1, retinoic acid, T3, LDN, Wnt inhibitor, ROCK inhibitor, FGF2) in a differentiation induction medium containing ALK5iII (10 ⁇ M) ( The cells were cultured for 2 days in Improved MEM/1% B-27/Penisilin Streptomycin medium) to induce differentiation into endocrine progenitor cells.
- differentiation factors SANT1, retinoic acid, T3, LDN, Wnt inhibitor, ROCK inhibitor, FGF2
- a differentiation induction medium Improved MEM/1% B-27/Penisilin Streptomycin medium
- ALK5iII (10 ⁇ M) was used together with differentiation factors (T3, LDN, ⁇ -secretase inhibitor RO, FGF receptor 1 inhibitor PD-166866).
- differentiation induction containing ALK5iII (10 ⁇ M) along with T3, LDN, ⁇ -secretase inhibitor RO, N-acetylcysteine, AXL inhibitor R428, ascorbic acid, ROCK inhibitor, ZnSO 4 , heparin, and Trolox was performed.
- the cells were cultured for 4 days in a medium (MCDB131/20mM Glucose/NaHCO 3 /FAF-BSA/ITS-X/Glutamax/Penisilin Streptomycin medium) to induce differentiation into iPIC.
- a medium MCDB131/20mM Glucose/NaHCO 3 /FAF-BSA/ITS-X/Glutamax/Penisilin Streptomycin medium
- the obtained iPIC had the form of an aggregate (spheroid) (about 150 ⁇ m in diameter) in which about 500 cells aggregated.
- a fibrin gel mass containing iPIC and a fibrin gel sheet containing iPIC were subcutaneously implanted into nude rats under anesthesia. After transplantation, blood was collected from the tail vein, plasma was collected, and human c-peptide concentration was measured by ELISA.
- Figure 1 shows the appearance of the fibrin gel mass and fibrin gel sheet. In all cases, iPIC was retained within the gel, and no cell leakage was observed.
- Figure 2 shows the results of measuring the blood human c-peptide concentration over time in nude rats in which a fibrin gel mass containing iPIC or a fibrin gel sheet containing iPIC was subcutaneously implanted
- Figure 3 shows the results 10 days after implantation. The results of histological analysis of each transplanted site after 6 weeks are shown.
- Example 2 Production of large animal/human size fibrin gel sheet (manufacturing method 1_method using a cooled metal mold) ⁇ Method> ⁇ Evaluation of gelation time
- fibrinogen solution 40 mg/mL
- thrombin solution 31 U/mL, 15.6 U/mL, 10.4 U/mL, 5.2 U/mL, 3.1 U/mL
- Example 3 Production of large animal/human size fibrin gel sheet (manufacturing method 2-1_fibrin + gelatin microsphere gel sheet) ⁇ Method> ⁇ Confirmation of maintenance of cell dispersion Dissolve gelatin to 2w/v% using physiological saline, cool in a refrigerator at 4°C to gel, and then pass the gelatin gel through a mesh with an opening of 200 ⁇ m. As a result, gelatin gel microspheres (hereinafter referred to as "gelatin microspheres”) of about 200 ⁇ m were produced. Gelatin microspheres were suspended in a fibrinogen solution (40 mg/mL) to a concentration of 20 w/v% to prepare a fibrinogen+gelatin microsphere solution.
- a fibrinogen solution 40 mg/mL
- the fibrin gel sheet containing the iPIC described in Example 1 and the fibrin+gelatin microsphere gel sheet containing the above iPIC were implanted subcutaneously into nude rats under anesthesia. After transplantation, blood was collected from the tail vein, plasma was collected, and human c-peptide concentration was measured by ELISA.
- Polyester mesh (69% open area, size 40x60 mm) impregnated with 300 ⁇ L of thrombin solution (62.5 U/mL) and glycolic acid/lactic acid polyester (90/10, poly(glycolide-co-L-lactide), size
- 300 ⁇ L of thrombin solution (62.5 U/mL) and glycolic acid/lactic acid polyester (90/10, poly(glycolide-co-L-lactide), size
- 600 ⁇ L of thrombin solution was applied to the entire surface and left to stand for 5 minutes to gel.
- a thin layer of encapsulated fibrin + gelatin microsphere gel sheet was prepared.
- FIG. 6 shows the state of iPIC suspended in a fibrinogen solution and a fibrinogen+gelatin microsphere solution and left standing.
- fibrinogen solution FIG. 6(A)
- precipitation of iPIC was observed after 10 minutes of standing (arrowhead).
- fibrinogen + gelatin microsphere solution FIG. 6(B)
- iPIC remained dispersed even after 30 minutes of standing.
- the iPIC was retained within the gel sheet, and no cell leakage was observed.
- FIG. 7 shows the results of measuring the blood human c-peptide concentration over time in nude rats in which a fibrin gel sheet containing iPIC or a fibrin + gelatin microsphere gel sheet containing iPIC was subcutaneously implanted. There was no significant difference in the amount of human c-peptide in the blood between the fibrin gel sheet and the fibrin + gelatin microsphere gel sheet, and even the use of gelatin microspheres in the sheet had a negative effect on the secretion of human c-peptide by iPIC. It was shown that there is no.
- Figure 8 shows a fibrin + gelatin microsphere gel sheet containing large animal/human-sized iPICs, which was produced by manual pipette injection and using a 3D printer. With either method, it was possible to produce the fibrin + gelatin microsphere gel sheet of large animal/human size, iPIC was retained within the gel, and no cell leakage was observed (FIG. 8(A)).
- Example 4 Preparation of large animal/human size fibrin gel sheet (manufacturing method 2-2_fibrin + collagen gel sheet) ⁇ Method> - Confirmation of maintenance of cell dispersibility Fibrinogen solution (80 mg/mL) and collagen solution (3 w/v %) were mixed at a ratio of 3:1 to prepare a fibrinogen + collagen solution.
- iPIC was suspended in a fibrinogen+collagen solution and then filled into a syringe. The syringe is held vertically with the tip facing down, and after 0, 10, 20, and 30 minutes of standing, a certain amount of the ejected liquid is collected and the number of cells (concentration) is measured. The dispersibility retention function was evaluated.
- Fibrinogen solution 80 mg/mL
- collagen solution 3 w/v%
- iPIC cells were collected into a 1.5 mL tube and centrifuged, the medium supernatant was removed, and 25 ⁇ L of fibrinogen+collagen solution was added and suspended.
- 25 ⁇ L of thrombin solution was applied to the entire surface and allowed to stand for 5 minutes to gel, thereby producing a thin layer of fibrin+collagen gel sheet containing iPIC.
- a fibrin + gelatin microsphere gel sheet containing iPIC as described in Example 3 "in vivo test" and a fibrin + collagen gel sheet containing iPIC described above were implanted subcutaneously into a nude rat under anesthesia. After transplantation, blood was collected from the tail vein, plasma was collected, and human c-peptide concentration was measured by ELISA.
- iPIC 75x10 6 cells were fractionated into a 15 mL tube and centrifuged, the medium supernatant was removed, and 600 ⁇ L of fibrinogen + collagen solution was added and suspended.
- a fibrinogen + collagen solution in which iPIC was suspended was thinly applied onto a poly(glycolide-co-L-lactide) (size 40 x 60 mm) mesh impregnated with 300 ⁇ L of thrombin solution (125 U/mL) using a 3D printer.
- 600 ⁇ L of thrombin solution was applied to the entire surface and allowed to stand for 5 minutes to gel, thereby producing a thin layer of fibrin+collagen gel sheet containing iPIC.
- FIG. 9 shows the measurement results of the number of cells (concentration) in the fibrinogen+collagen solution discharged from the syringe.
- the cell concentration in the ejected liquid was almost constant regardless of the standing time, indicating that iPIC did not precipitate even though the syringe was held vertically, and a constant number of cells were ejected. . Therefore, it was shown that the dispersed state of cells could be maintained by using the fibrinogen + collagen solution.
- the iPIC was retained within the gel sheet, and no cell leakage was observed.
- Figure 10 shows the results of measuring the blood human c-peptide concentration over time in nude rats in which a fibrin + gelatin microsphere gel sheet containing iPIC or a fibrin + collagen gel sheet containing iPIC was subcutaneously implanted. No significant difference was observed in the amount of human c-peptide in blood between the fibrin+gelatin microsphere gel sheet and the fibrin+collagen gel sheet, indicating that both had equivalent performance.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Medicinal Chemistry (AREA)
- Zoology (AREA)
- Cell Biology (AREA)
- Transplantation (AREA)
- Dermatology (AREA)
- Oral & Maxillofacial Surgery (AREA)
- Biotechnology (AREA)
- Organic Chemistry (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Microbiology (AREA)
- Botany (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Dispersion Chemistry (AREA)
- Developmental Biology & Embryology (AREA)
- Gastroenterology & Hepatology (AREA)
- Nutrition Science (AREA)
- Physiology (AREA)
- Urology & Nephrology (AREA)
- Immunology (AREA)
- Hematology (AREA)
- Virology (AREA)
- Pharmacology & Pharmacy (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
人工多能性細胞や胚性幹細胞(ES細胞)等の多能性幹細胞から誘導される機能性分化細胞は、移植再生医療の細胞供給源として期待されている。今日では、そのような機能性分化細胞が生分解性ゲル中に内包されてなる各種構造体が開発され、それを利用した細胞移植治療の研究が進められている。
[1] 細胞がフィブリンゲルシートに均一に分散され内包されてなり、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有する、細胞移植用のフィブリンゲルシート。
[2] 細胞が1.5×106個超/cm2の量で内包される、[1]のフィブリンゲルシート。
[3] 細胞がスフェロイドの形態である、[1]又は[2]のフィブリンゲルシート。
[4] 細胞がiPS細胞由来膵島細胞である、[1]~[3]のいずれかのフィブリンゲルシート。
[5] さらに、生分解性ゲル化剤を含む、[1]~[4]のいずれかのフィブリンゲルシート。
[6] 生分解性ゲル化剤を0.2~2w/v%の量で含む、[5]のフィブリンゲルシート。
[7] 生分解性ゲル化剤がコラーゲンである、[5]又は[6]のフィブリンゲルシート。
[8] さらに、生分解性粒子を内包する、[1]~[4]のいずれかのフィブリンゲルシート。
[9] 生分解性粒子が目開き100μm~1000μmを通過できるサイズである、[8]のフィブリンゲルシート。
[10] 生分解性粒子を10~30w/v%の量で含む、[8]又は[9]のフィブリンゲルシート。
[11] 生分解性粒子がゼラチンゲル粒子である、[8]~[10]のいずれかのフィブリンゲルシート。
[12] 支持体をさらに備える、[1]~[11]のいずれかのフィブリンゲルシート。
[13] 細胞がフィブリンゲルシートに均一に分散され内包されてなり、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有する、細胞移植用のフィブリンゲルシートの製造方法であって、
細胞が懸濁されたフィブリノーゲン溶液をトロンビンと作用させて、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有するシート状に成型してゲル化させる工程、
を含む、製造方法。
[14] 細胞が、フィブリンゲルシートにおいて1.5×106個超/cm2となる量にて、フィブリノーゲン溶液に懸濁される、[13]の製造方法。
[15] 細胞がスフェロイドの形態である、[13]又は[14]の製造方法。
[16] 細胞がiPS細胞由来膵島細胞である、[13]~[15]のいずれかの製造方法。
[17] 細胞が懸濁されたフィブリノーゲン溶液がさらに、生分解性ゲル化剤を含む、[13]~[16]のいずれかの製造方法。
[18] 細胞が懸濁されたフィブリノーゲン溶液が、生分解性ゲル化剤を0.2~2w/v%の量で含む、[17]の製造方法。
[19] 生分解性ゲル化剤がコラーゲンである、[17]又は[18]の製造方法。
[20] 支持体上に塗布することによりシート状に成型する、[17]~[19]のいずれかの製造方法。
[21] バイオプリントにより支持体上に塗布する、[20]の製造方法。
[22] 支持体がトロンビンを含む、[20]又は[21]の製造方法。
[23] 細胞が懸濁されたフィブリノーゲン溶液がさらに、生分解性粒子を含む、[13]~[16]のいずれかの製造方法。
[24] 生分解性粒子が目開き100μm~1000μmを通過できるサイズである、[23]の製造方法。
[25] 細胞が懸濁されたフィブリノーゲン溶液が、生分解性粒子を10~30w/v%の量で含む、[23]又は[24]の製造方法。
[26] 生分解性粒子がゼラチンゲル粒子である、[23]~[25]のいずれかの製造方法。
[27] 支持体上に塗布することによりシート状に成型する、[23]~[26]のいずれかの製造方法。
[28] バイオプリントにより支持体上に塗布する、[27]の製造方法。
[29] 支持体がトロンビンを含む、[27]又は[28]の製造方法。
[30] トロンビンを、フィブリノーゲン1mgに対して0.4U以下の配合比となる量にて作用させる、[13]~[16]のいずれかの製造方法。
[31] 2~8℃の冷却下にてシート状に成型する、[30]の製造方法。
[32] さらに、支持体をフィブリンゲルシートと一体化させる工程を含む、[30]又は[31]の製造方法。
[33] [1]~[12]のいずれかのフィブリンゲルシートが複数積層されてなる、細胞移植用のフィブリンゲルシートの積層体。
[1a] 細胞がフィブリンゲルシートに均一に分散され内包されてなり、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有する、細胞移植治療方法において使用されるフィブリンゲルシート。
[2a] 細胞が1.5×106個超/cm2の量で内包される、[1a]のフィブリンゲルシート。
[3a] 細胞がスフェロイドの形態である、[1a]又は[2a]のフィブリンゲルシート。
[4a] 細胞がiPS細胞由来膵島細胞である、[1a]~[3a]のいずれかのフィブリンゲルシート。
[5a] さらに、生分解性ゲル化剤を含む、[1a]~[4a]のいずれかのフィブリンゲルシート。
[6a] 生分解性ゲル化剤を0.2~2w/v%の量で含む、[5a]のフィブリンゲルシート。
[7a] 生分解性ゲル化剤がコラーゲンである、[5a]又は[6a]のフィブリンゲルシート。
[8a] さらに、生分解性粒子を内包する、[1a]~[4a]のいずれかのフィブリンゲルシート。
[9a] 生分解性粒子が目開き100μm~1000μmを通過できるサイズである、[8a]のフィブリンゲルシート。
[10a] 生分解性粒子を10~30w/v%の量で含む、[8a]又は[9a]のフィブリンゲルシート。
[11a] 生分解性粒子がゼラチンゲル粒子である、[8a]~[10a]のいずれかのフィブリンゲルシート。
[12a] 支持体をさらに備える、[1a]~[10a]のいずれかのフィブリンゲルシート。
[33a] [1a]~[12a]のいずれかのフィブリンゲルシートが複数積層されてなる、細胞移植治療方法において使用されるフィブリンゲルシートの積層体。
本明細書は本願の優先権の基礎である2022年7月14日に出願された日本国特許出願2022-113546号の明細書及び/又は図面に記載される内容を包含する。
本明細書で引用した全ての刊行物、特許および特許出願をそのまま参考として本明細書にとりいれるものとする。
以下、本明細書において記載される用語について説明する。
本発明は、細胞がフィブリンゲルシートに均一に分散され内包されてなる、細胞移植用のフィブリンゲルシート(以下、単に「本発明のフィブリンゲルシート」と記載する場合がある)に関するものである。
工程1)多能性幹細胞から胚体内胚葉細胞へと分化誘導する;
工程2)胚体内胚葉細胞から原腸管細胞へと分化誘導する;
工程3)原腸管細胞から後方前腸細胞へと分化誘導する;
工程4)後方前腸細胞から膵前駆細胞へと分化誘導する;
工程5)膵前駆細胞から内分泌前駆細胞へと分化誘導する;
工程6)内分泌前駆細胞からiPICへと分化誘導する。
以下、各工程を説明するが、各細胞への分化誘導はこれらの手法に限定されない。
多能性幹細胞は、まず胚体内胚葉細胞に分化させる。多能性幹細胞から胚体内胚葉を誘導する方法は既に公知であり、そのいずれの方法を用いてもよい。好ましくは、多能性幹細胞は、アクチビンAを含む培地、より好ましくはアクチビンA、ROCK阻害剤、GSK3β阻害剤を含む培地で培養して、胚体内胚葉細胞に分化させる。培養開始時の細胞数としては、特に限定されず、22000~150000細胞/cm2、好ましくは22000~100000細胞/cm2、より好ましくは22000~80000細胞/cm2である。培養期間は1日~4日、好ましくは1日~3日、特に好ましくは3日である。
工程1)で得られた胚体内胚葉細胞を、さらに増殖因子を含む培地で培養して原腸管細胞に分化誘導する。培養期間は2日~8日、好ましくは約4日である。
工程2)で得られた原腸管細胞を、さらに増殖因子、シクロパミン、ノギン等を含む培地で培養し、後方前腸細胞に分化誘導する。培養期間は1日~5日、好ましくは約2日程度である。培養は2次元培養及び3次元培養のいずれで行ってもよい。
工程3)で得られた後方前腸細胞を、さらにCDK8/19阻害活性を有する因子を含む培地、好ましくはCDK8/19阻害活性を有する因子と増殖因子を含む培地で培養し、膵前駆細胞に分化誘導する。培養期間は2日~10日、好ましくは約5日程度である。培養は2次元培養及び3次元培養のいずれで行ってもよい。
工程4)で得られた膵前駆細胞を、さらに増殖因子を含む培地で培養して内分泌前駆細胞に分化誘導する。培養は2次元培養及び3次元培養のいずれで行ってもよい。2次元培養の場合には、工程4)で得られた膵前駆細胞を、0.25%トリプシン-EDTA溶液で処理し、ピペッティングすることにより当該液中に分散させて細胞分散液を得、得られた分散液を遠心分離に付し、回収した細胞を少量の新たな培地に再懸濁し、その細胞懸濁液を工程5)の新しい培地に再播種する。培養期間は2日~3日、好ましくは約2日である。
工程5)で得られた内分泌前駆細胞を、さらに増殖因子を含む培地で培養してiPICに分化誘導する。培養期間は10日~30日、好ましくは約10~20日である。
本発明はまた、前記本発明のフィブリンゲルシートの製造方法(以下、「本発明方法」と記載する場合がある)に関するものであり、当該方法は、細胞が懸濁されたフィブリノーゲン溶液をトロンビンと作用させて、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有するシート状に成型・ゲル化(固化)させる工程を含む。フィブリンゲルは、適当な溶媒(例えば、水、生理食塩水等)中にてフィブリノーゲンとトロンビンと合わせ、ゲル化(固化)させることにより得ることができる。
本発明のフィブリンゲルシートは、分散され内包される上述の細胞に応じて、例えば、表皮、神経、脳、脊髄、食道、胃、小腸、大腸、膀胱、尿道、肺、甲状腺、膵臓、肝臓、筋肉、骨格、心臓、血管、脾臓、腎臓、血液等(これらに限定はされない)の器官や組織を構成する細胞やその前駆細胞と同等又は類似する機能を有し、それら器官や組織の機能を増強又は維持するために、あるいは、疾患や障害等の原因により低減又は消失したそれら器官や組織の機能を補助又は補充するための細胞治療方法において利用することができ、それを必要とする患者に移植するために用いられる。
iPICは、上記工程1)-6)、既報(Stem Cell Research(2015)14、185-197;Nature Biotechnology 2014;32:1121-1133)に従い、ヒトiPS細胞(Ff-I14-s04株)から分化誘導して調製した。
<方法>
・フィブリンゲル塊の作製
iPIC 3x106cells(凝集塊にして約6000個)を1.5mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン溶液(80mg/mL)25μLを添加し懸濁した。さらにトロンビン溶液(125U/mL)25μLを添加し5分間静置しゲル化させ、iPICを内包したフィブリンゲル塊(直径約5mm)を作製した。
iPIC 3x106cellsを1.5mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン溶液(80mg/mL)25μLを添加し懸濁した。トロンビン溶液(125U/mL)12.5μLを含侵させたポリエステル製メッシュ(開口率69%、10x10mm)の上にiPICを懸濁したフィブリノーゲン溶液を薄く塗布した後、トロンビン溶液25μLを全体に塗布し5分間静置しゲル化させ、iPICを内包した薄層のフィブリンゲルシートを作製した。
麻酔下にてヌードラットの皮下に、iPICを内包したフィブリンゲル塊、iPICを内包したフィブリンゲルシートを移植した。移植後、尾静脈より採血し血漿を分取し、ELISA法にてヒトc-ペプチド濃度を測定した。
図1にフィブリンゲル塊とフィブリンゲルシートの外観を示す。いずれもiPICがゲル内に保持されており、細胞の漏出は認められなかった。
<方法>
・ゲル化時間の評価
フィブリノーゲン溶液(40mg/mL)とトロンビン溶液(31U/mL,15.6U/mL,10.4U/mL,5.2U/mL,3.1U/mL)を調製し、1.5mLチューブ内でそれぞれ440μLずつ合わせ、1~2回ピペッティングして混合した後、ゲル化の状態を以下の基準で評価して、トロンビン濃度とゲル化時間の関係を解析した。
〇:チューブ内でピペッティング操作が可能
△:ピペッティング操作が可能であるが、部分的にゲル化している
×:全体的にゲル化して、ピペッティング操作を行うことができない
iPIC 45x106cellsを15mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン溶液(40mg/mL)360μLを添加し懸濁した。その後、トロンビン溶液(15.6U/mL)360μLを添加し懸濁した後、予め4℃に冷却した金属製の型(40x60x0.4mm)に素早く広げ、さらにポリエステル製メッシュ(開口率69%、サイズ40x10mm)をのせ、室温で6分間静置しゲル化させた。
トロンビン濃度とゲル化時間の関係を解析した結果、トロンビン濃度が低下するに従って、ゲル化時間が遅延し、15.6U/mL以下の濃度であれば、40x60x0.4mmの大きさのフィブリンゲルシートを成型するのに必要な時間が確保できることが示された(図4)。
<方法>
・細胞分散性保持の確認
ゼラチンを2w/v%となるように生理食塩水を用いて溶解し、4℃冷蔵庫内にて冷却しゲル化させた後、ゼラチンゲルを目開き200μmのメッシュに通すことにより、約200μmのゼラチンゲルのマイクロスフェア(以下、「ゼラチンマイクロスフェア」と記載する)を作製した。ゼラチンマイクロスフェアを20w/v%となるようにフィブリノーゲン溶液(40mg/mL)に懸濁し、フィブリノーゲン+ゼラチンマイクロスフェア溶液を調製した。iPICをフィブリノーゲン溶液(40mg/mL)およびフィブリノーゲン+ゼラチンマイクロスフェア溶液に懸濁したのち静置し、沈殿の形成の有無により、細胞の分散状態の保持を評価した。
iPIC 3x106cellsを1.5mLチューブに分取し遠心分離した後、培地上清を除去し、上記のとおり調製したフィブリノーゲン+ゼラチンマイクロスフェア溶液25μLを添加し懸濁した。トロンビン溶液(62.5U/mL)12.5μLを含侵させたポリエステル製メッシュ(開口率69%、サイズ10x10mm)の上にiPICを懸濁したフィブリノーゲン+ゼラチンマイクロスフェア溶液を薄く塗布した後、トロンビン溶液25μLを全体に塗布し5分間静置してゲル化させ、iPICを内包した薄層のフィブリン+ゼラチンマイクロスフェアゲルシートを作製した。
iPIC 75x106cellsを15mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン+ゼラチンマイクロスフェア溶液600μLを添加し懸濁した。トロンビン溶液(62.5U/mL)300μLを含侵させたポリエステル製メッシュ(開口率69%、サイズ40x60mm)及びグリコール酸/乳酸ポリエステル(90/10、ポリ(グリコリド-co-L-ラクチド)、サイズ40x60mm)メッシュの上にiPICを懸濁したフィブリノーゲン+ゼラチンマイクロスフェア溶液をピペット又は3Dプリンターを用いてそれぞれ薄く塗布した後、トロンビン溶液600μLを全体に塗布し5分間静置しゲル化させ、iPICを内包した薄層のフィブリン+ゼラチンマイクロスフェアゲルシートを作製した。
図6にフィブリノーゲン溶液およびフィブリノーゲン+ゼラチンマイクロスフェア溶液に懸濁し静置したiPICの状態を示す。フィブリノーゲン溶液(図6(A))では静置10分後にはiPICの沈殿が認められた(矢頭)。一方、フィブリノーゲン+ゼラチンマイクロスフェア溶液(図6(B))では静置30分後でもiPICの分散状態が維持されていた。
<方法>
・細胞分散性保持の確認
フィブリノーゲン溶液(80mg/mL)とコラーゲン溶液(3w/v%)を3:1の比率で混合し、フィブリノーゲン+コラーゲン溶液を調製した。iPICをフィブリノーゲン+コラーゲン溶液中に懸濁した後、シリンジ内に充填した。シリンジを、筒先を下にして垂直に保持し、静置0、10、20、30分後に排出液の一定量を分取し細胞数(濃度)を計測することによって、フィブリノーゲン+コラーゲン溶液の細胞分散性保持機能を評価した。
フィブリノーゲン溶液(80mg/mL)とコラーゲン溶液(3w/v%)を3:1の比率で混合し、フィブリノーゲン+コラーゲン溶液を調製した。iPIC 3x106cellsを1.5mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン+コラーゲン溶液25μLを添加し懸濁した。トロンビン溶液(125U/mL)12.5μLを含侵させたグリコール酸/乳酸ポリエステル(90/10、ポリ(グリコリド-co-L-ラクチド))メッシュの上にiPICを懸濁したフィブリノーゲン+コラーゲン溶液を薄く塗布した後、トロンビン溶液25μLを全体に塗布し5分間静置しゲル化させ、iPICを内包した薄層のフィブリン+コラーゲンゲルシートを作製した。
iPIC 75x106cellsを15mLチューブに分取し遠心分離した後、培地上清を除去し、フィブリノーゲン+コラーゲン溶液600μLを添加し懸濁した。トロンビン溶液(125U/mL)300μLを含侵させたポリ(グリコリド-co-L-ラクチド)、サイズ40x60mm)メッシュの上にiPICを懸濁したフィブリノーゲン+コラーゲン溶液を3Dプリンターを用いて薄く塗布した後、トロンビン溶液600μLを全体に塗布し5分間静置しゲル化させ、iPICを内包した薄層のフィブリン+コラーゲンゲルシートを作製した。
図9にシリンジから排出されたフィブリノーゲン+コラーゲン溶液中の細胞数(濃度)の計測結果を示した。排出液中の細胞濃度は静置時間にかかわらずほぼ一定であり、シリンジを垂直に保持したにもかかわらずiPICが沈殿することなく、ほぼ一定数の細胞数が排出されたことが示された。従って、フィブリノーゲン+コラーゲン溶液を用いることにより、細胞の分散状態を維持できることが示された。
Claims (15)
- 細胞がフィブリンゲルシートに均一に分散され内包されてなり、
該フィブリンゲルシートがさらに、0.2~2w/v%の量のコラーゲン、及び/又は10~30w/v%の量の100μm~1000μmを通過できるゼラチンゲル粒子を含む、
細胞移植用のフィブリンゲルシート。 - 一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有する、請求項1に記載のフィブリンゲルシート。
- 前記細胞が1.5×106個超/cm2の量で内包される、請求項1に記載のフィブリンゲルシート。
- 前記細胞がスフェロイドの形態である、請求項1に記載のフィブリンゲルシート。
- 前記細胞がiPS細胞由来膵島細胞である、請求項1に記載のフィブリンゲルシート。
- 支持体をさらに備える、請求項1に記載のフィブリンゲルシート。
- 細胞がフィブリンゲルシートに均一に分散され内包されてなる細胞移植用のフィブリンゲルシートの製造方法であって、
該細胞が懸濁されたフィブリノーゲン溶液をトロンビンと作用させて、シート状に成型してゲル化させる工程、を含み、
該細胞が懸濁されたフィブリノーゲン溶液がさらに、0.2~2w/v%の量のコラーゲン、及び/又は10~30w/v%の量の100μm~1000μmを通過できるゼラチンゲル粒子を含む、製造方法。 - 前記ゲル化させる工程が、前記細胞が懸濁されたフィブリノーゲン溶液をトロンビンと作用させて、一面の表面積が2.25cm2以上のサイズを有し、1mm以下の厚さを有するシート状に成型してゲル化させる、請求項7に記載の製造方法。
- 前記細胞が、フィブリンゲルシートにおいて1.5×106個超/cm2となる量にて、フィブリノーゲン溶液に懸濁される、請求項7に記載の製造方法。
- 前記細胞がスフェロイドの形態である、請求項7に記載の製造方法。
- 前記細胞がiPS細胞由来膵島細胞である、請求項7に記載の製造方法。
- 支持体上に塗布することによりシート状に成型する、請求項7に記載の製造方法。
- バイオプリントにより支持体上に塗布する、請求項12に記載の製造方法。
- 支持体がトロンビンを含む、請求項12に記載の製造方法。
- 請求項1に記載のフィブリンゲルシートが複数積層されてなる、細胞移植用のフィブリンゲルシートの積層体。
Priority Applications (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23839669.1A EP4556555A1 (en) | 2022-07-14 | 2023-07-13 | Fibrin gel sheet for cell transplantation |
| JP2024533746A JPWO2024014497A1 (ja) | 2022-07-14 | 2023-07-13 | |
| KR1020257003710A KR20250036837A (ko) | 2022-07-14 | 2023-07-13 | 세포 이식용의 피브린 겔 시트 |
| AU2023307842A AU2023307842A1 (en) | 2022-07-14 | 2023-07-13 | Fibrin gel sheet for cell transplantation |
| CN202380053547.5A CN119546748A (zh) | 2022-07-14 | 2023-07-13 | 细胞移植用纤维蛋白凝胶片 |
| IL318270A IL318270A (en) | 2022-07-14 | 2023-07-13 | Fibrin gel sheet for cell transplantation |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2022-113546 | 2022-07-14 | ||
| JP2022113546 | 2022-07-14 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024014497A1 true WO2024014497A1 (ja) | 2024-01-18 |
Family
ID=89536775
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2023/025809 Ceased WO2024014497A1 (ja) | 2022-07-14 | 2023-07-13 | 細胞移植用のフィブリンゲルシート |
Country Status (7)
| Country | Link |
|---|---|
| EP (1) | EP4556555A1 (ja) |
| JP (1) | JPWO2024014497A1 (ja) |
| KR (1) | KR20250036837A (ja) |
| CN (1) | CN119546748A (ja) |
| AU (1) | AU2023307842A1 (ja) |
| IL (1) | IL318270A (ja) |
| WO (1) | WO2024014497A1 (ja) |
Citations (27)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2007069666A1 (ja) | 2005-12-13 | 2007-06-21 | Kyoto University | 核初期化因子 |
| WO2008118220A2 (en) | 2006-11-28 | 2008-10-02 | Veritainer Corporation | Radiation detection unit for mounting a radiation sensor to a container crane |
| WO2008124133A1 (en) | 2007-04-07 | 2008-10-16 | Whitehead Institute For Biomedical Research | Reprogramming of somatic cells |
| WO2008151058A2 (en) | 2007-05-30 | 2008-12-11 | The General Hospital Corporation | Methods of generating pluripotent cells from somatic cells |
| JP2008307007A (ja) | 2007-06-15 | 2008-12-25 | Bayer Schering Pharma Ag | 出生後のヒト組織由来未分化幹細胞から誘導したヒト多能性幹細胞 |
| WO2009012428A2 (en) | 2007-07-18 | 2009-01-22 | Lifescan, Inc. | Differentiation of human embryonic stem cells |
| US20090047263A1 (en) | 2005-12-13 | 2009-02-19 | Kyoto University | Nuclear reprogramming factor and induced pluripotent stem cells |
| US20120071477A1 (en) | 2009-11-30 | 2012-03-22 | Senex Biotechnology, Inc. | Cdki pathway inhibitors and uses thereof |
| WO2013116786A1 (en) | 2012-02-02 | 2013-08-08 | Senex Biotechnology Inc. | Cdk8/cdk19 selective inhibitors and their use in anti-metastatic and chemopreventative methods for cancer |
| WO2014038958A1 (en) | 2012-09-10 | 2014-03-13 | Besseggen 1896 As | Balance chair |
| WO2014134169A1 (en) | 2013-02-26 | 2014-09-04 | Senex Biotechnology, Inc. | Inhibitors of cdk8/19 for use in treating estrogen receptor positive breast cancer |
| US20150274726A1 (en) | 2012-11-08 | 2015-10-01 | Selvita Sa | Substituted tricyclic benzimidazoles as kinase inhibitors |
| WO2015159938A1 (ja) | 2014-04-18 | 2015-10-22 | 武田薬品工業株式会社 | 複素環化合物 |
| WO2015159937A1 (ja) | 2014-04-18 | 2015-10-22 | 武田薬品工業株式会社 | 縮合複素環化合物 |
| WO2016009076A1 (en) | 2014-07-17 | 2016-01-21 | Merck Patent Gmbh | Novel naphthryidines and isoquinolines and their use as cdk8/19 inhibitors |
| WO2016016951A1 (ja) | 2014-07-29 | 2016-02-04 | 櫻護謨株式会社 | 安全弁機能を備えた結合金具 |
| WO2016018511A2 (en) | 2014-06-10 | 2016-02-04 | University Of South Carolina | Methods and compositions for treatment of her-positive cancers |
| WO2016021734A1 (ja) | 2014-08-04 | 2016-02-11 | 武田薬品工業株式会社 | 膵前駆細胞の増殖方法 |
| WO2016100782A1 (en) | 2014-12-18 | 2016-06-23 | University Of South Carolina | Suppression of neointimal formation following vascular surgery using cdk8 inhibitors |
| WO2016182904A1 (en) | 2015-05-08 | 2016-11-17 | President And Fellows Of Harvard College | Targeted selection of patients for treatment with cortistatin derivatives |
| WO2018155622A1 (ja) | 2017-02-23 | 2018-08-30 | 日本酢ビ・ポバール株式会社 | 細胞又は組織包埋デバイス |
| WO2020203579A1 (ja) * | 2019-04-01 | 2020-10-08 | 凸版印刷株式会社 | 三次元組織体及びその製造方法並びに細胞含有組成物の製造方法 |
| WO2020209389A1 (ja) * | 2019-04-10 | 2020-10-15 | 国立大学法人京都大学 | 生体組織様構造体の製造方法 |
| JP2021052663A (ja) * | 2019-09-30 | 2021-04-08 | 国立大学法人大阪大学 | 多能性幹細胞由来の分化誘導細胞を含有するシート状物の作製方法 |
| WO2021065395A1 (ja) * | 2019-10-01 | 2021-04-08 | 国立大学法人大阪大学 | フィブリンシートの製造方法 |
| WO2022114128A1 (ja) * | 2020-11-26 | 2022-06-02 | 凸版印刷株式会社 | 立体的細胞組織の製造方法及び立体的細胞組織 |
| JP2022113546A (ja) | 2021-01-25 | 2022-08-04 | 株式会社藤商事 | 遊技機 |
-
2023
- 2023-07-13 IL IL318270A patent/IL318270A/en unknown
- 2023-07-13 AU AU2023307842A patent/AU2023307842A1/en active Pending
- 2023-07-13 JP JP2024533746A patent/JPWO2024014497A1/ja active Pending
- 2023-07-13 EP EP23839669.1A patent/EP4556555A1/en active Pending
- 2023-07-13 KR KR1020257003710A patent/KR20250036837A/ko active Pending
- 2023-07-13 CN CN202380053547.5A patent/CN119546748A/zh active Pending
- 2023-07-13 WO PCT/JP2023/025809 patent/WO2024014497A1/ja not_active Ceased
Patent Citations (33)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20090047263A1 (en) | 2005-12-13 | 2009-02-19 | Kyoto University | Nuclear reprogramming factor and induced pluripotent stem cells |
| JP2008283972A (ja) | 2005-12-13 | 2008-11-27 | Kyoto Univ | 誘導多能性幹細胞の製造方法 |
| WO2007069666A1 (ja) | 2005-12-13 | 2007-06-21 | Kyoto University | 核初期化因子 |
| WO2008118220A2 (en) | 2006-11-28 | 2008-10-02 | Veritainer Corporation | Radiation detection unit for mounting a radiation sensor to a container crane |
| WO2008124133A1 (en) | 2007-04-07 | 2008-10-16 | Whitehead Institute For Biomedical Research | Reprogramming of somatic cells |
| WO2008151058A2 (en) | 2007-05-30 | 2008-12-11 | The General Hospital Corporation | Methods of generating pluripotent cells from somatic cells |
| WO2009006930A1 (en) | 2007-06-15 | 2009-01-15 | Izumi Bio, Inc. | Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue |
| WO2009007852A2 (en) | 2007-06-15 | 2009-01-15 | Izumi Bio, Inc | Multipotent/pluripotent cells and methods |
| WO2009006997A1 (en) | 2007-06-15 | 2009-01-15 | Izumi Bio, Inc. | Human pluripotent stem cells and their medical use |
| JP2008307007A (ja) | 2007-06-15 | 2008-12-25 | Bayer Schering Pharma Ag | 出生後のヒト組織由来未分化幹細胞から誘導したヒト多能性幹細胞 |
| WO2009012428A2 (en) | 2007-07-18 | 2009-01-22 | Lifescan, Inc. | Differentiation of human embryonic stem cells |
| US20120071477A1 (en) | 2009-11-30 | 2012-03-22 | Senex Biotechnology, Inc. | Cdki pathway inhibitors and uses thereof |
| JP2015506376A (ja) | 2012-02-02 | 2015-03-02 | セネックス バイオテクノロジー インク.Senex Biotechnology Inc. | Cdk8/cdk19選択的阻害剤、ならびに癌のための抗転移および化学防御の方法におけるそれらの使用 |
| WO2013116786A1 (en) | 2012-02-02 | 2013-08-08 | Senex Biotechnology Inc. | Cdk8/cdk19 selective inhibitors and their use in anti-metastatic and chemopreventative methods for cancer |
| WO2014038958A1 (en) | 2012-09-10 | 2014-03-13 | Besseggen 1896 As | Balance chair |
| US20150274726A1 (en) | 2012-11-08 | 2015-10-01 | Selvita Sa | Substituted tricyclic benzimidazoles as kinase inhibitors |
| WO2014134169A1 (en) | 2013-02-26 | 2014-09-04 | Senex Biotechnology, Inc. | Inhibitors of cdk8/19 for use in treating estrogen receptor positive breast cancer |
| US20160000787A1 (en) | 2013-02-26 | 2016-01-07 | Senex Biotechnology, Inc. | Inhibitors of cdk8/19 for use in treating estrogen receptor positive breast cancer |
| WO2015159938A1 (ja) | 2014-04-18 | 2015-10-22 | 武田薬品工業株式会社 | 複素環化合物 |
| WO2015159937A1 (ja) | 2014-04-18 | 2015-10-22 | 武田薬品工業株式会社 | 縮合複素環化合物 |
| WO2016018511A2 (en) | 2014-06-10 | 2016-02-04 | University Of South Carolina | Methods and compositions for treatment of her-positive cancers |
| WO2016009076A1 (en) | 2014-07-17 | 2016-01-21 | Merck Patent Gmbh | Novel naphthryidines and isoquinolines and their use as cdk8/19 inhibitors |
| WO2016016951A1 (ja) | 2014-07-29 | 2016-02-04 | 櫻護謨株式会社 | 安全弁機能を備えた結合金具 |
| WO2016021734A1 (ja) | 2014-08-04 | 2016-02-11 | 武田薬品工業株式会社 | 膵前駆細胞の増殖方法 |
| WO2016100782A1 (en) | 2014-12-18 | 2016-06-23 | University Of South Carolina | Suppression of neointimal formation following vascular surgery using cdk8 inhibitors |
| WO2016182904A1 (en) | 2015-05-08 | 2016-11-17 | President And Fellows Of Harvard College | Targeted selection of patients for treatment with cortistatin derivatives |
| WO2018155622A1 (ja) | 2017-02-23 | 2018-08-30 | 日本酢ビ・ポバール株式会社 | 細胞又は組織包埋デバイス |
| WO2020203579A1 (ja) * | 2019-04-01 | 2020-10-08 | 凸版印刷株式会社 | 三次元組織体及びその製造方法並びに細胞含有組成物の製造方法 |
| WO2020209389A1 (ja) * | 2019-04-10 | 2020-10-15 | 国立大学法人京都大学 | 生体組織様構造体の製造方法 |
| JP2021052663A (ja) * | 2019-09-30 | 2021-04-08 | 国立大学法人大阪大学 | 多能性幹細胞由来の分化誘導細胞を含有するシート状物の作製方法 |
| WO2021065395A1 (ja) * | 2019-10-01 | 2021-04-08 | 国立大学法人大阪大学 | フィブリンシートの製造方法 |
| WO2022114128A1 (ja) * | 2020-11-26 | 2022-06-02 | 凸版印刷株式会社 | 立体的細胞組織の製造方法及び立体的細胞組織 |
| JP2022113546A (ja) | 2021-01-25 | 2022-08-04 | 株式会社藤商事 | 遊技機 |
Non-Patent Citations (14)
| Title |
|---|
| HIROTAKE KOMATSU ET AL., TRANSPLANT INTERNATIONAL, vol. 33, 2020, pages 806 - 818 |
| HUANGFU D.MELTON, DA. ET AL., NATURE BIOTECHNOLOGY, vol. 26, no. 7, 2008, pages 795 - 797 |
| KIM JB.SCHOLER HR ET AL., NATURE, vol. 454, 2008, pages 646 - 650 |
| MEIRIGENG QI ET AL., BIOMATERIALS, vol. 25, no. 27, December 2004 (2004-12-01), pages 5885 - 92 |
| NATURE BIOTECHNOLOGY, vol. 32, 2014, pages 1121 - 1133 |
| OKITA K ET AL., NAT. METHODS, vol. 8, no. 5, May 2011 (2011-05-01), pages 409 - 12 |
| OKITA K ET AL., STEM CELLS., vol. 31, no. 3, pages 458 - 66 |
| OKITA, K., ICHISAKA, T., YAMANAKA, S., NATURE, vol. 451, 2007, pages 141 - 146 |
| SHI Y.DING S. ET AL., CELL STEM CELL, vol. 3, 2008, pages 568 - 574 |
| STEM CELL RESEARCH, vol. 14, 2015, pages 185 - 197 |
| TAKAHASHI KYAMANAKA S. ET AL., CELL, vol. 131, 2007, pages 861 - 872 |
| TAKAHASHI KYAMANAKA S., CELL, vol. 126, 2006, pages 663 - 676 |
| TOYODA ET AL., STEM CELL RESEARCH, vol. 14, 2015, pages 185 - 197 |
| YU J.THOMSON JA. ET AL., SCIENCE, vol. 318, 2007, pages 1917 - 1920 |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2024014497A1 (ja) | 2024-01-18 |
| AU2023307842A1 (en) | 2025-02-06 |
| EP4556555A1 (en) | 2025-05-21 |
| CN119546748A (zh) | 2025-02-28 |
| IL318270A (en) | 2025-03-01 |
| KR20250036837A (ko) | 2025-03-14 |
| AU2023307842A2 (en) | 2025-03-06 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP2025000878A (ja) | 生体組織様構造体の製造方法 | |
| EP4249587A1 (en) | Maturation agent | |
| JPWO2020027316A1 (ja) | 細胞製造法 | |
| EP4293105A1 (en) | Maturation agent | |
| WO2024014497A1 (ja) | 細胞移植用のフィブリンゲルシート | |
| JP7709737B2 (ja) | 増殖抑制剤 | |
| RU2815583C2 (ru) | Способ получения биологической тканеподобной структуры | |
| JPWO2019182157A1 (ja) | ハイドロゲルカプセル | |
| WO2023210578A1 (ja) | Alk5阻害活性とcdk8/19阻害活性とを有する成熟化剤 |
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: 23839669 Country of ref document: EP Kind code of ref document: A1 |
|
| DPE1 | Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101) | ||
| WWE | Wipo information: entry into national phase |
Ref document number: 2024533746 Country of ref document: JP |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 318270 Country of ref document: IL |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 202380053547.5 Country of ref document: CN Ref document number: 18993681 Country of ref document: US |
|
| WWE | Wipo information: entry into national phase |
Ref document number: AU2023307842 Country of ref document: AU |
|
| REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112024027442 Country of ref document: BR |
|
| ENP | Entry into the national phase |
Ref document number: 20257003710 Country of ref document: KR Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 1020257003710 Country of ref document: KR |
|
| ENP | Entry into the national phase |
Ref document number: 2023307842 Country of ref document: AU Date of ref document: 20230713 Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025103122 Country of ref document: RU Ref document number: 2023839669 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2023839669 Country of ref document: EP Effective date: 20250214 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 11202500214V Country of ref document: SG |
|
| WWP | Wipo information: published in national office |
Ref document number: 11202500214V Country of ref document: SG |
|
| WWP | Wipo information: published in national office |
Ref document number: 202380053547.5 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 2025103122 Country of ref document: RU |
|
| WWP | Wipo information: published in national office |
Ref document number: 1020257003710 Country of ref document: KR |
|
| WWP | Wipo information: published in national office |
Ref document number: 2023839669 Country of ref document: EP |
|
| ENP | Entry into the national phase |
Ref document number: 112024027442 Country of ref document: BR Kind code of ref document: A2 Effective date: 20241227 |