WO2021065984A1 - Method for forming sheet of cardiomyocytes - Google Patents

Method for forming sheet of cardiomyocytes Download PDF

Info

Publication number
WO2021065984A1
WO2021065984A1 PCT/JP2020/037084 JP2020037084W WO2021065984A1 WO 2021065984 A1 WO2021065984 A1 WO 2021065984A1 JP 2020037084 W JP2020037084 W JP 2020037084W WO 2021065984 A1 WO2021065984 A1 WO 2021065984A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
culture
sheet
component
Prior art date
Application number
PCT/JP2020/037084
Other languages
French (fr)
Japanese (ja)
Inventor
芳樹 澤
繁 宮川
賢二 大山
文哉 大橋
Original Assignee
国立大学法人大阪大学
テルモ株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人大阪大学, テルモ株式会社 filed Critical 国立大学法人大阪大学
Priority to JP2021551353A priority Critical patent/JPWO2021065984A1/ja
Publication of WO2021065984A1 publication Critical patent/WO2021065984A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/34Muscles; Smooth muscle cells; Heart; Cardiac stem cells; Myoblasts; Myocytes; Cardiomyocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS 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
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS 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
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS 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
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials 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
    • A61L27/38Materials 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS 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
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms

Definitions

  • This disclosure is a commissioned research project of the Japan Medical Research and Development Organization, Regenerative Medicine Realization Center Network Program, Practical Application Research Center for Diseases and Tissues (Center A), "Center for the Creation of Myocardial Regenerative Therapy Using iPS Cells" in FY2018. It is a patent application to which Article 17 of the Development and Industrial Technology Enhancement Law applies, and transplants of various cells such as sheet-shaped cell cultures containing myocardial cells, especially pluripotent stem cell-derived myocardial cells.
  • the present invention relates to a method for producing, a transplanted piece manufactured by using the method, a method for treating a disease using the transplanted piece, and the like.
  • Non-Patent Document 1 a graft containing cardiomyocytes prepared by a cell engineering technique into an affected area.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • Non-Patent Documents 2 to 3 There are attempts to prepare sheet-like cell cultures containing such pluripotent stem cell-derived cardiomyocytes and therapeutic experiments in animals.
  • Non-Patent Documents 2 to 3 the development of sheet-like cell cultures containing pluripotent stem cell-derived cardiomyocytes has just begun, and there are still many unclear points about their functional characteristics and factors that influence them.
  • the graft-forming medium contains as little heterologous components other than humans as possible, that is, a so-called xeno-free state.
  • a sheet-shaped cell culture in a xeno-free state is often costly and laborious, and depending on the cells used, it is possible to form a sheet-shaped cell culture while maintaining the desired properties of the cells. It can be difficult.
  • the present disclosure is a method for producing a high-quality implant, which comprises various cells such as cardiomyocytes, particularly cardiomyocytes derived from pluripotent stem cells, while preserving the functions of the cells, using the method. It is an object of the present invention to provide a manufactured implant, a method for treating a disease using the implant, and the like.
  • a graft containing cardiomyocytes to be used for transplantation it is necessary to prepare with a xenofree graft-forming medium.
  • FBS fetal bovine serum
  • problems such as cardiomyocytes not beating sufficiently and grafts not being formed well have been found. ..
  • the present inventors are studying the preparation of sheet-shaped cell cultures for living body transplantation using cardiomyocytes derived from pluripotent stem cells, which have sufficient functions even when prepared with a Xenofree implant-forming medium.
  • a serum-free medium can be used to form a sheet. I found it. Based on this finding, further research was carried out, and it was found that it is possible to produce a high-quality graft that can withstand clinical application in the preparation of a graft containing various cells, and to complete the present invention. I arrived.
  • the present invention relates to the following: [1] A method for producing a graft containing somatic cells; (A) a step of seeding the cell population containing the somatic cells on a culture substrate, and (b) a step of seeding the seeded cell population in a transplantation piece-forming medium containing a cell adhesion component. Included, said method. [2] The method according to [1], wherein the culture substrate is further coated with a cell adhesion component and / or a masking component. [3] The method according to [1] or [2], wherein the content of the cell adhesion component in the graft-forming medium is 1/10 to 1/100 of the amount used for coating the culture substrate. ..
  • a method for producing a sheet-like cell culture containing cardiomyocytes (A) A step of seeding myocardial cells at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component, and (B) sheet-forming culture using a sheet-forming medium, and sheet-like cell culture.
  • a method for producing a sheet-shaped cell culture which comprises a step of forming a substance.
  • the sheet formation medium contains or does not contain a cell adhesion component.
  • [9] A method for evaluating the surface of a culture medium for producing a sheet-shaped cell culture.
  • the step of coating the surface of the culture substrate with the cell adhesion component thereby improving the adhesion of the cells to the surface of the culture substrate.
  • a step of coating the surface of the culture substrate with a masking component thereby reducing the adhesion of cells to the surface of the culture substrate.
  • the above-mentioned step including (iv) a step of forming a sheet-like cell culture by sheet-forming culture in a sheet-forming medium, and (v) a step of evaluating a peeling state of the sheet-like cell culture from the surface of a culture substrate.
  • transplants such as sheet-like cell cultures of higher quality than before can be efficiently produced from a cell population containing various cells, for example, cardiomyocytes, particularly cardiomyocytes induced to differentiate from pluripotent stem cells.
  • a cell population containing various cells for example, cardiomyocytes, particularly cardiomyocytes induced to differentiate from pluripotent stem cells.
  • cardiomyocytes particularly cardiomyocytes induced to differentiate from pluripotent stem cells.
  • the graft-forming medium does not use serum, that is, it does not contain impurities derived from the manufacturing process. It is particularly useful in regenerative medicine because it enables graft formation even when the graft is formed and cultured under the conditions.
  • a platelet lysate to the graft-forming medium, it is possible to prepare a graft having a desired property at a high level, and particularly to provide a very suitable graft for living-donor transplantation into humans. It becomes possible.
  • FIG. 1 is a photograph showing a state of sheet formation when laminin (iMatrix-511), vitronectin (VTN-N), and fibronectin (RetroNectin (R)) are used as cell adhesion components.
  • the upper row (A and B) is for laminin
  • the middle row (C and D) is for vitronectin
  • the lower row (E and F) is for fibronectin
  • the left column (A, C and E) is for use as a coating agent.
  • the right column (B, D and F) shows the result when used at 1/10 of the recommended concentration. In any case, it was confirmed that the sheet was formed.
  • FIG. 1 is a photograph showing a state of sheet formation when laminin (iMatrix-511), vitronectin (VTN-N), and fibronectin (RetroNectin (R)) are used as cell adhesion components.
  • the upper row (A and B) is for laminin
  • FIG. 2 shows a state in which the sheet-shaped cell culture is peeled off after sheeting with iMatrix-511 as a cell adhesion component and FBS as a masking component on a culture substrate coated with various concentrations. It is a photograph showing. By coating with FBS together with iMatrix-511, it was confirmed that peeling was possible even when iMatrix-511 with a wide concentration was used.
  • the present disclosure is a method of producing a graft containing the desired cells; (A) a step of seeding the cell population containing the cells on a culture substrate, and (b) a step of forming and culturing the seeded cell population in a medium containing a cell adhesion component.
  • the present invention relates to the above-mentioned method.
  • the "graft” means a structure for transplantation into a living body, and particularly means a structure for transplantation containing cells as a constituent component.
  • the implant is a structure for transplantation that is free of cells and structures other than cell-derived substances (eg, scaffolds, etc.).
  • the implants in the present disclosure include, but are not limited to, sheet-like cell cultures, spheroids, cell aggregates, and the like, preferably sheet-like cell cultures or spheroids, and more preferably sheet-like. It is a cell culture.
  • sheet-shaped cell culture refers to cells connected to each other to form a sheet.
  • the cells may be linked to each other directly (including those via cell elements such as adhesion molecules) and / or via intervening substances.
  • the intervening substance is not particularly limited as long as it is a substance capable of at least physically (mechanically) connecting cells to each other, and examples thereof include an extracellular matrix.
  • the mediator is preferably derived from cells, particularly from the cells that make up the sheet-like cell culture.
  • the cells are at least physically (mechanically) connected, but may be more functionally, for example, chemically or electrically connected.
  • the sheet-like cell culture may be composed of one cell layer (single layer) or two or more cell layers (laminate (multilayer), for example, two layers, three layers, etc. It may be 4 layers, 5 layers, 6 layers, etc.). Further, the sheet-shaped cell culture may have a three-dimensional structure having a thickness exceeding the thickness of one cell without showing a clear layered structure of the cells. For example, in the vertical cross section of a sheet-shaped cell culture, cells may be present in a non-uniformly (for example, mosaic-like) arrangement without being uniformly aligned in the horizontal direction.
  • a non-uniformly for example, mosaic-like
  • the grafts of the present disclosure preferably do not contain scaffolds (supports). Scaffolds may be used in the art to attach cells to and / or inside the scaffold to maintain the physical integrity of the implant, such as a sheet cell culture, eg, poly. Although membranes made of vinylidene fluoride (PVDF) and the like are known, the implants of the present disclosure can maintain their physical integrity without such scaffolds. In addition, the implants of the present disclosure preferably consist only of cell-derived substances constituting the implants and do not contain any other substances.
  • scaffolds supports
  • Scaffolds may be used in the art to attach cells to and / or inside the scaffold to maintain the physical integrity of the implant, such as a sheet cell culture, eg, poly.
  • PVDF vinylidene fluoride
  • the implants of the present disclosure can maintain their physical integrity without such scaffolds.
  • the implants of the present disclosure preferably consist only of cell-derived substances constituting the implants and do not contain any other substances.
  • the cell may be a heterologous cell or an allogeneic cell.
  • heterologous cell means a cell derived from an organism of a species different from the recipient when the graft is used for transplantation.
  • cells derived from monkeys and pigs correspond to heterologous cells.
  • homoogeneous cell means a cell derived from an organism of the same species as the recipient.
  • the human cell corresponds to an allogeneic cell. Allogeneic cells include autologous cells (also referred to as autologous cells or autologous cells), ie, recipient-derived cells and allogeneic non-autologous cells (also referred to as allogeneic cells).
  • Autologous cells are preferred in the present disclosure because they do not cause rejection when transplanted. However, it is also possible to utilize heterologous cells and allogeneic non-autologous cells. When using heterologous cells or allogeneic non-autologous cells, immunosuppressive treatment may be required to suppress rejection.
  • cells other than autologous cells that is, allogeneic non-self-derived cells and allogeneic non-self-derived cells may be collectively referred to as non-autologous cells.
  • the cell is an autologous cell or an allogeneic cell.
  • the cell is an autologous cell (including an autologous iPS cell).
  • the cell is an allogeneic cell (including an allogeneic iPS cell).
  • the cells constituting the implant of the present disclosure are not particularly limited as long as they can form the implant, and include, for example, adherent cells (adhesive cells).
  • Adhesive cells include, for example, adherent somatic cells (eg, myocardial cells, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, root membrane cells, gingival cells, bone membrane cells, skin.
  • iPS cells induced pluripotent stem cells
  • Somatic cells may be stem cells, particularly those differentiated from iPS cells (iPS cell-derived adherent cells).
  • Non-limiting examples of cells constituting the transplant include, for example, myoblasts (eg, skeletal myoblasts), mesenchymal stem cells (eg, bone marrow, adipose tissue, peripheral blood, skin, hair roots, muscle tissue, etc.) Endometrial, placenta, umbilical cord blood, etc.), myocardial cells, fibroblasts, cardiac stem cells, embryonic stem cells, iPS cells, synovial cells, chondrocytes, epithelial cells (eg, oral mucosal epithelial cells, retinal pigments) Epithelial cells, nasal mucosal epithelial cells, etc.), endothelial cells (eg, vascular endothelial cells, etc.), hepatocytes (eg, hepatic parenchymal cells, etc.), pancreatic cells (eg, pancreatic islet cells, etc.), kidney cells, adrenal cells, root membrane Examples include cells, gingival cells, bone membrane cells, skin cells
  • Non-limiting examples of iPS cell-derived adherent cells include iPS cell-derived myocardial cells, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, renal cells, adrenal cells, root membrane cells, gingival cells, and bone membrane cells. , Skin cells, synovial cells, cartilage cells and the like.
  • the cells that make up the implant can be derived from any organism that can be treated with the implant. Such organisms include, but are not limited to, for example, humans, non-human primates, dogs, cats, pigs, horses, goats, sheep, rodents (eg, mice, rats, hamsters, guinea pigs, etc.), rabbits, etc. Is included.
  • the number of types of cells constituting the transplant is not particularly limited, and may be composed of only one type of cells, or may be composed of two or more types of cells.
  • the content ratio (purity) of the most abundant cells is, for example, 50% or more, preferably 60% or more, more preferably 70 at the end of formation of the sheet-like cell culture. % Or more, more preferably 75% or more.
  • the culture substrate is not particularly limited as long as the cells can form a cell culture on the cells, and includes, for example, containers of various materials and / or shapes, solid or semi-solid surfaces in the containers, and the like. ..
  • the container preferably has a structure / material that does not allow a liquid such as a culture solution to permeate.
  • Such materials include, without limitation, for example, polyethylene, polypropylene, Teflon®, polyethylene terephthalate, polymethylmethacrylate, nylon 6,6, polyvinyl alcohol, cellulose, silicon, polystyrene, glass, polyacrylamide, polydimethyl.
  • Acrylamide, metals (eg, iron, stainless steel, aluminum, copper, brass) and the like can be mentioned.
  • the container preferably has at least one flat surface.
  • a culture container having a bottom surface made of a culture substrate capable of forming a cell culture and a liquid-impermeable side surface.
  • a culture vessel include, but are not limited to, a cell culture dish, a cell culture bottle, and the like.
  • the bottom surface of the container may be transparent or opaque. If the bottom surface of the container is transparent, cells can be observed and counted from the back side of the container.
  • the container may have a solid or semi-solid surface inside the container. Examples of the solid surface include plates and containers of various materials as described above, and examples of the semi-solid surface include gels and soft polymer matrices.
  • the culture substrate may be prepared using the above-mentioned materials, or a commercially available one may be used.
  • Preferred culture substrates include, without limitation, for example, a substrate having an adhesive surface suitable for forming a sheet-like cell culture, and a substrate having a low adhesive surface suitable for forming spheroids. And / or a substrate having a uniform well-like structure and the like.
  • a substrate coated with a hydrophilic compound such as corona discharge-treated polystyrene, collagen gel or hydrophilic polymer on the surface thereof, and further, collagen.
  • Fibronectin Fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan and other extracellular matrix, and base materials coated with cell adhesion factors such as cadoherin family, selectin family and integrin family on the surface.
  • cell adhesion factors such as cadoherin family, selectin family and integrin family on the surface.
  • substrates are commercially available (eg, Corning (R) TC-Treated Culture Dish, Corning, etc.).
  • spheroid formation for example, soft agar, temperature-responsive gel obtained by cross-linking poly (N-isopropylacrylamide) (PIPAAm) with polyethylene glycol (PEG), polyhydroxyethyl methacrylate (commercially available name: mebiol gel), polyhydroxyethyl methacrylate ( Examples include a base material coated with a non-cell adhesive compound such as a hydrogel such as poly-HEMA) and 2-methacryloyloxyethyl phosphorischoline (MPC) polymer and / or a base material having a uniform uneven structure on the surface. Be done. Such substrates are also commercially available (eg, EZSPHERE (R), etc.).
  • the culture medium may be transparent or opaque in whole or in part.
  • the surface of the culture substrate may be coated with a material whose physical properties change in response to irritation, for example, temperature or light.
  • a material whose physical properties change in response to irritation, for example, temperature or light.
  • Such materials include, but are not limited to, for example, (meth) acrylamide compounds, N-alkyl substituted (meth) acrylamide derivatives (eg, N-ethylacrylamide, Nn-propylacrylamide, Nn-propylmethacrylamide, etc.
  • Known materials such as a copolymer with a body and a photoresponsive material such as N-isopropylacrylamide gel containing spirobenzopyran can be used (see, for example, JP-A-2-21186 and JP-A-2003-33177). ). By giving a predetermined stimulus to these materials, their physical characteristics, for example, hydrophilicity and hydrophobicity can be changed, and the exfoliation of the cell culture adhering on the material can be promoted. Culture dishes coated with a temperature-responsive material are commercially available (eg, CellSeed Inc.'s UpCell (R) ) and can be used in the production methods of the present disclosure.
  • a temperature-responsive material are commercially available (eg, CellSeed Inc.'s UpCell (R) ) and can be used in the production methods of the present disclosure.
  • the culture medium may have various shapes.
  • the area thereof is not particularly limited, but may be, for example, about 1 cm 2 to about 200 cm 2 , about 2 cm 2 to about 100 cm 2 , about 3 cm 2 to about 50 cm 2 .
  • a circular culture dish having a diameter of 10 cm can be mentioned. In this case, the area is 56.7 cm 2 .
  • the culture surface may be flat or may have an uneven structure. When it has a concavo-convex structure, it is preferable that it has a uniform concavo-convex structure.
  • pluripotent stem cell is a well-known term in the art and has the ability to differentiate into three germ layers, i.e. cells of all lineages belonging to endoderm, mesoderm and ectoderm. Means cell.
  • pluripotent stem cells include, for example, embryonic stem cells (ES cells), nuclear-transplanted embryonic stem cells (ntES cells), induced pluripotent stem cells (iPS cells), and the like.
  • ES cells embryonic stem cells
  • ntES cells nuclear-transplanted embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • pluripotent stem cells are first suspended-cultured to form aggregates of any of the above three germ layers, and then cells that form aggregates. Induce differentiation into specific cells of interest.
  • pluripotent stem cell-derived differentiation-inducing cell means any cell that has been subjected to differentiation-inducing treatment so as to differentiate from a pluripotent stem cell into a specific type of cell.
  • differentiation-inducing cells include muscular cells such as myocardial cells and skeletal myoblasts, neural cells such as neuron cells, oligodendrocytes and dopamine-producing cells, retinal cells such as retinal pigment epithelial cells, and blood cells.
  • Hematopoietic cells such as cells and bone marrow cells, immune-related cells such as T cells, NK cells, NKT cells, dendritic cells and B cells, cells constituting organs such as hepatocytes, pancreatic ⁇ cells and renal cells, In addition to cartilage cells and germ cells, it also includes precursor cells and somatic stem cells that differentiate into these cells.
  • progenitor cells and somatic stem cells include mesenchymal stem cells in myocardial cells, pluripotent cardiac progenitor cells, monopoly cardiac progenitor cells, neural stem cells in neural cells, hematopoietic cells and immunity. Examples include hematopoietic stem cells and lymphoid stem cells in related cells.
  • Induction of differentiation of pluripotent stem cells can be performed using any known method.
  • the induction of differentiation of pluripotent stem cells into cardiomyocytes can be performed based on the methods described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 and WO2014 / 185358.
  • a differentiation-inducing cell derived from a pluripotent stem cell such as an iPS-derived cardiomyocyte
  • the undifferentiated cell may be removed after the differentiation induction.
  • the treatment for removing undifferentiated cells is known in the art, and the methods described in, for example, WO2017 / 038562, WO2016 / 072519, WO2007 / 088744, etc. can be used.
  • the differentiation-inducing cell may be a cell derived from an iPS cell into which any useful gene other than the gene for reprogramming has been introduced.
  • iPS cells into which the chimeric antigen receptor gene described in Themeli M. et al. Nature Biotechnology, vol. 31, no. 10, pp. 928-933, 2013 has been introduced. Examples include T cells derived from.
  • cells into which any useful gene has been introduced after induction of differentiation from pluripotent stem cells are also included in the differentiation-inducing cells of the present invention.
  • cell adhesion component means an isolated polypeptide having the property of adhering to cells, including extracellular matrix and cell adhesion factors described in the above description of the culture medium. To do. Such cell-adhesive components are generally used to coat the surface of the culture medium. Examples of cell-adhesive components include, but are not limited to, extracellular matrix such as collagen, fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan, and cells such as cadoherin family, selectin family, and integrin family.
  • these variants or functional equivalents such as laminin variants such as laminin 511 and laminin 221 and vitronectin variants such as VTN-N, Kimizuka et al., J. Biochem.
  • laminin variants such as laminin 511 and laminin 221
  • vitronectin variants such as VTN-N, Kimizuka et al., J. Biochem.
  • fibronectin such as retronectin (R) described in (1991), 110, pp.284-291 and the like.
  • R retronectin
  • the cell adhesion components of the present disclosure do not contain blood-derived components other than cell adhesion factors and / or their functional equivalents.
  • the present invention will be described in detail below, taking as an example the case where the desired cell is a cardiomyocyte and the implant is a sheet-like cell culture.
  • One aspect of the disclosure relates to a method for producing a high quality sheet cell culture containing cardiomyocytes.
  • the method of the present disclosure includes the following steps (a) and (b): (A) A step of seeding a cell population containing myocardial cells on a culture substrate, and (b) a step of sheeting and culturing the seeded cell population in a sheeting medium containing a cell adhesion component.
  • cardiomyocyte means a cell having the characteristics of cardiomyocyte.
  • the characteristics of cardiomyocytes include, but are not limited to, expression of cardiomyocyte markers, presence of autonomous pulsation, and the like.
  • Non-limiting examples of cardiomyocyte markers include, for example, c-TNT (cardiac troponin T), CD172a (also known as SIRPA or SHPS-1), KDR (also known as CD309, FLK1 or VEGFR2), PDGFRA, EMILIN2, VCAM and the like. ..
  • cardiomyocytes derived from pluripotent stem cells are c-TNT positive and / or CD172a positive.
  • the cardiomyocytes used in the method of the present disclosure may be directly obtained from a living body or derived from other cells, but are preferably derived from other cells. ..
  • Examples of the induction into cardiomyocytes include a method of introducing a myocardial inducing factor into fibroblasts and the like, and a method of inducing differentiation of cells having the property of differentiating into cardiomyocytes such as pluripotent stem cells and cardiac progenitor cells. ..
  • seeding on the culture substrate may be performed, for example, by injecting a cell suspension in which cells are suspended in a sheeting medium into a culture vessel provided with the culture substrate.
  • a cell suspension in which cells are suspended in a sheeting medium into a culture vessel provided with the culture substrate.
  • an instrument suitable for the injection operation of the cell suspension such as a dropper or a pipette, can be used.
  • the seeding density of cells is set to a density capable of forming a sheet-like cell culture, and the density may vary depending on the desired cells, but those skilled in the art select an appropriate density from methods known in the art. can do.
  • a sheet-shaped cell culture containing cardiomyocytes it can be, for example, 2.0 ⁇ 10 5 cells / cm 2 or more, but may be seeded at a higher density.
  • Examples of higher densities include, for example, a density that reaches confluence, that is, a density at which cells are expected to cover the entire adhesive surface of the culture vessel when seeded, for example, cells come into contact with each other when seeded. It can be as dense as expected, the density at which contact inhibition occurs, or the density at which cell proliferation is substantially stopped by contact inhibition or higher.
  • the upper limit of the seeding density is not particularly limited, but if the seeding density is excessively high, more cells will die, resulting in inefficiency. In one aspect of the present disclosure, the seeding density is, for example, about 1.0 ⁇ 10 6 / cm 2 to about 1.0 ⁇ 10 7 / cm 2 , about 1.0 ⁇ 10 6 / cm 2 to about 5.
  • the sowing density is from about 1.76 ⁇ 10 6 / cm 2 to about 2.33 ⁇ 10 6 / cm 2 .
  • the seeded cell population may contain other cells as long as they contain the desired cells (eg, myocardial cells), and when the desired cells are myocardial cells, for example, fibroblasts, vascular endothelial cells. And / or wall cells and the like may be further included.
  • the cell population the cell population collected from the tissue may be used as it is, or by using, for example, the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 or WO 2014/185358.
  • the cell population obtained by inducing differentiation from iPS cells may be used as it is, or may be used after cryopreservation, pre-culture, removal of undifferentiated cells, or the like.
  • the seeded cell population is a cell population that is induced to differentiate from iPS cells, seeded on a culture substrate (preferably on a flat culture substrate), adherently cultured, and then recovered. is there. Cryopreservation and thawing may be performed before or after such adhesive culture.
  • the culture conditions and the like may be the same as those for normal adhesive culture.
  • a commercially available culture container for adhesive culture may be used for culturing under 37 ° C. and 5% CO 2 conditions.
  • the seeding density of the cells may be any density as long as it does not interfere with the adhesion between the cells and / or the formation of the adhesion between the cells and the culture substrate, and may be, for example, a subconfluent density. It may be at or above a density that reaches confluence.
  • the culturing time may be such that adhesion between cells and / or adhesion between cells and the culture substrate is formed, and specifically, for example, 2 to 24 hours, 2 to 12 hours, and 2 to 6 hours. It may be about 2 to 4 hours.
  • the cell population does not include undifferentiated cells.
  • a cell population obtained by collecting from a living body or inducing differentiation from pluripotent stem cells may be subjected to an undifferentiated cell removal operation.
  • the "undifferentiated cell removal operation” refers to undifferentiated cells having tumorigenicity from a cell population, typically a cell population containing differentiation-inducing cells obtained by inducing differentiation of pluripotent stem cells. It means a removal operation and can be performed using any known method.
  • Non-limiting examples of such a method include various separation methods using markers specific to undifferentiated cells (for example, cell surface markers), for example, magnetic cell separation method (MACS), flow cytometry method, affinity separation.
  • markers specific to undifferentiated cells for example, cell surface markers
  • MCS magnetic cell separation method
  • Methods, methods for expressing selective markers (eg, antibiotic resistance genes, etc.) by specific promoters, factors necessary for the survival of undifferentiated cells nutrient sources (nutrient sources such as methionine, maintenance of undifferentiated state such as bFGF)
  • a method of exterminating undifferentiated cells by culturing in a medium excluding (factors, etc.) a method of culturing in the presence of factors that promote differentiation (VEGF, BMP, activin, etc.) and promoting undifferentiated cells, not yet Examples thereof include a method of treating the surface antigen of differentiated cells with a drug targeting the surface antigen.
  • WO2016 / 072519, WO2013 / 10080 examples thereof include the method described in JP-A-2016-093178 and the method using the heat treatment described in WO2017 / 038526.
  • the operation for removing undifferentiated cells is performed by culturing in a sugar-free medium as described in WO2007 / 088874.
  • the culture base material to be seeded is as described in detail above, but in a preferred embodiment, it is a culture base material whose surface is coated with a cell adhesion component such as an extracellular matrix or a cell adhesion factor.
  • the cell-adhesive component is not limited to this, for example, extracellular matrix such as collagen, fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan, and cell adhesion factors such as cadoherin family, selectin family, and integrin family.
  • these variants may be, for example, laminin 511 (a variant of laminin), VTN-N (a variant of vitronectin), retronectin (R) (a variant of fibronectin).
  • Coating of the cell adhesion component can be achieved by contacting and incubating a medium containing the cell adhesion component with the culture substrate.
  • the amount of the cell adhesion component that can be contained in such a medium may vary depending on the area of the substrate to be coated, the cell adhesion component used, the cell type to be cultured, and the like.
  • a person skilled in the art can set the optimum amount or concentration according to the product protocol, etc., and the amount or concentration can be set when coating with only the adhesive component without using the masking component, for example, laminin 511 or VTN-.
  • N it is about 0.1 to 1.0 ⁇ g / cm 2
  • retronectin (R) it is about 4 to 20 ⁇ g / cm 2 .
  • a culture substrate in which the surface is coated with a masking component such as a blood-derived component in addition to a cell adhesion component such as an extracellular matrix or a cell adhesion factor.
  • the "masking component” means a component capable of reducing the cell adhesion on the surface of the culture medium coated with the cell adhesion component.
  • Such components are typically blood-derived components, such as normal serum (eg, bovine serum such as bovine fetal serum, horse serum, human serum, etc.), as well as albumin and platelet lysates contained in normal serum. , Skim milk, albumin substitutes such as polyvinyl alcohol, and the like.
  • Coating of the medium containing the masking component may be performed simultaneously with or separately from the coating of the cell adhesion component, and is achieved by contacting and incubating the medium containing the cell adhesion component and / or the masking component with the culture substrate. obtain.
  • concentration of the masking component that can be contained in such a medium may vary depending on the concentration of the cell adhesion component or the cell adhesion component used, the area of the substrate to be coated, the cell type to be cultured, and the like.
  • the concentration of the masking component is 0.05%, 0.075%, 0.1%, 0.125%, 0.15%, 0.16%, 0.2% in the coating medium. , 0.3%, 0.4%, 0.5%, 0.6%, 0.8%, 1.0%, 1.25%, 1.5%, 1.75%, 2.0% 2.25%, 2.5%, 2.75%, 3.0%, 3.5%, 4.0%, 4.5%, 5.0%, 5.5%, 7.5% , 9%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 30%, 40% or more, and the upper limit is not particularly limited, but 45.
  • the amount or concentration of laminin 511 is 0.01 ⁇ g / cm 2 to 100 ⁇ g / cm 2 with respect to 0.1% to 30% FBS, laminin 511 0.
  • the incubation time is not particularly limited as long as the masking component can adhere to the culture substrate, for example, 1 to 72 hours, preferably 4 to 48 hours, more preferably 5 to 24 hours, still more preferably 6 to 12 hours. It's time.
  • the incubation temperature is also not particularly limited as long as the masking component can adhere to the culture substrate, and is, for example, 0 to 60 ° C., preferably 4 to 45 ° C., more preferably room temperature to 40 ° C.
  • the seeded cells are sheet-cultured.
  • the culture for forming the seeded cells as a transplant is referred to as "transplant formation culture”
  • the transplant is a sheet-like cell culture
  • the culture for forming the seeded cells into a sheet is referred to as a culture.
  • sheet culture it is referred to as "sheet culture”. Sheeting of seeded cells can be performed by any known method and condition. Non-limiting examples of such a method are described in, for example, Japanese Patent Application Laid-Open No. 2010-081829, Japanese Patent Application Laid-Open No. 2010-226991, Japanese Patent Application Laid-Open No. 2011-110368, Japanese Patent Application Laid-Open No.
  • Cell graft formation (eg, sheet formation) is believed to be achieved by the cells adhering to each other via adhesion molecules or intercellular adhesion mechanisms such as extracellular matrix.
  • the step of forming a graft of seeded cells can be accomplished, for example, by culturing the cells under conditions that form cell-cell adhesions. Such conditions may be any as long as they can form cell-cell adhesion, but usually, cell-cell adhesion can be formed under the same conditions as general cell culture conditions. Such conditions include, for example, culturing at about 37 ° C. and 5% CO 2.
  • the culture can be carried out under normal pressure (atmospheric pressure, non-pressurization).
  • the graft-forming culture sheet-forming culture
  • the graft-forming culture sheeted culture
  • Culturing can be carried out in containers of any size and shape. For the size and shape of the sheet-shaped cell culture, adjust the size and shape of the cell adhesion surface of the culture vessel, or install a mold of the desired size and shape on the cell adhesion surface of the culture vessel. It can be arbitrarily adjusted by culturing cells inside the cell.
  • the time for sheeting culture can vary depending on the type of cells to be seeded and the cell density.
  • the sheet may be formed by seeding at a density of, for example, about 2.1 ⁇ 10 5 cells / cm 2 and culturing for 4 days or more.
  • the seeding density reaches the confluence, that is, when the seeding is performed at a higher density, the period of sheeting culture can be shortened, and the culture time may be 2 to 4 days, more preferably 2 to 3 days. ..
  • the medium used for graft formation (for example, sheet formation) (the graft-forming medium, which may be referred to as a sheet-forming medium when the graft formation is sheet-forming) contains a cell adhesion component and transplants cells. It is not particularly limited as long as it enables fragment formation, and for example, physiological saline, various physiological buffer solutions (for example, PBS, WBSS, etc.), various basal medium-based liquids for cell culture, and the like can be used. You may use it.
  • Such basal medium is not limited to, for example, DMEM, MEM, F12, DME, RPMI1640, MCDB (MCDB102, 104, 107, 120, 131, 153, 199, etc.), L15, SkBM, RITC80-7, DMEM. / F12 and the like are included.
  • Many of these basal media are commercially available, and their compositions are also known.
  • the basal medium may be used as it has a standard composition (for example, as it is on the market), or the composition may be appropriately changed depending on the cell type and cell conditions. Therefore, the basal medium used in the present invention is not limited to those having a known composition, and includes those in which one or more components are added, removed, increased or decreased.
  • Transplantation media may also include additives such as normal serum (eg, bovine serum such as bovine fetal serum, horse serum, human serum, etc.) and various growth factors (eg, FGF, EGF, VEGF, HGF, etc.).
  • normal serum eg, bovine serum such as bovine fetal serum, horse serum, human serum, etc.
  • various growth factors eg, FGF, EGF, VEGF, HGF, etc.
  • the graft-forming medium is serum-free.
  • concentration of the cell adhesive component contained in the sheeting medium is not particularly limited as long as it does not adversely affect the cells, but may be equal to or less than the concentration used in the above coating.
  • concentration is, for example, about 0.01 to 10 ⁇ g / mL for laminin 511 or VTN-N, preferably about 0.1 to 1 ⁇ g / mL, and about 0.2 to 100 ⁇ g / mL for retronectin (R). , Preferably about 2-10 ⁇ g / mL and the like.
  • the sheeting medium further comprises a platelet lysate.
  • platelet lysate refers to a composition rich in growth factors and the like, which is obtained by repeatedly freezing and thawing platelets.
  • Such a composition is commercially available as a medium additive for cell culture, is known in the art, and can be prepared by, for example, the method described in Bieback et al., STEM CELLS, 2009; 27: 2331-2341. Is. In recent years, it is known to promote the proliferation of mesenchymal stem cells. In the production of sheet-shaped cell cultures containing cardiomyocytes, the present inventors have observed that by incorporating platelet lysates into the sheeting medium, strong autonomous pulsation is observed in a faster culture time than before. I found it for the first time.
  • the concentration of platelet lysate contained in the graft-forming medium may be such that it is usually used in the art, for example, 1%, 2.5%, 5%, 10%, 15%, 20% and the like. It may be there.
  • the platelet lysate is contained in the graft-forming medium in an amount of 1% to 20%, more preferably 2% to 10%, still more preferably 2.5% to 10%.
  • the graft-forming medium may be appropriately replaced during the graft-forming culture.
  • the composition of the medium may be changed according to the progress of graft formation.
  • the present inventors use a sheeted medium to which a Rho-kinase (ROCK) inhibitor is added as a medium for the first day of sheeted culture.
  • a Rho-kinase (ROCK) inhibitor is added as a medium for the first day of sheeted culture.
  • the sheeting medium used for the sheeting culture on day 1 comprises a Rho-kinase inhibitor.
  • the sheeting medium after the second day may or may not contain a Rho kinase inhibitor, but preferably does not contain a Rho kinase inhibitor.
  • the culture substrate may or may not be coated with a cell adhesive component.
  • the cell adhesion component coating the culture substrate may be the same as or different from the cell adhesion component contained in the sheeting medium. It may be, but preferably the same cell adhesion component.
  • the culture substrate is coated with only the cell adhesive component, preferably only the cell adhesive component contained in the sheeting medium. Therefore, in such an embodiment, the culture medium does not contain components other than the cell adhesive component, such as serum.
  • the culture substrate may be coated with another component in place of or in addition to the cell adhesion component. Examples of such other components include coating components exemplified in the above description of the culture medium, such as temperature-responsive materials.
  • the culture medium of the present disclosure is coated with serum, FBS or albumin in addition to the cell adhesive components.
  • the concentration of the cell adhesion component contained in the graft-forming medium may differ depending on the type of the cell adhesion component contained and the state of the cells forming the graft. For example, when cells with low viability, that is, weak activity are used, the content of the cell adhesion component should be low.
  • the concentration of the cell adhesion component contained in the sheeting medium is about 0.1%, about 0.%, based on the concentration used when the same cell adhesion component is used as the coating agent for the culture substrate (100%). It may be 5%, about 1%, about 5%, about 10%, about 20%, about 25%, about 50%, about 75%, about 100%, and the like.
  • the concentration range of the cell adhesion component contained in the sheeting medium is about 0 based on the concentration used when the same cell adhesion component is used as the coating agent for the culture substrate (100%). .1% to about 100%, about 0.1% to about 100%, about 0.1% to about 50%, about 0.1% to about 25%, about 0.1% to about 20%, about 0 .1% to about 10%, about 1% to about 100%, about 1% to about 100%, about 0.5% to about 100%, about 0.5% to about 100%, about 0.5% to About 50%, about 0.5% to about 25%, about 0.5% to about 20%, about 0.5% to about 10%, about 1% to about 50%, about 1% to about 25%, About 1% to about 20%, about 1% to about 10%, about 0.5% to about 100%, about 5% to about 100%, about 5% to about 50%, about 5% to about 25%, It may be about 5% to about 20%, about 5% to about 10%, and the like.
  • the cardiomyocytes used in the method of the present disclosure may be directly obtained from a living body or derived from other cells, but are preferably derived from other cells. It is a thing.
  • other cells to be induced include fibroblasts such as myocardial fibroblasts, progenitor cells that differentiate into myocardial cells such as cardiac progenitor cells, and pluripotent stem cells that can differentiate into arbitrary cells, which are preferable.
  • pluripotent stem cells more preferably iPS cells, and even more preferably human iPS cells.
  • Another aspect of the disclosure relates to a method of producing a sheet-like cell culture containing desired somatic cells, such as cardiomyocytes.
  • a method of producing a sheet-like cell culture containing desired somatic cells includes the following steps: (A) A step of seeding a cell population containing desired somatic cells (hereinafter, may be referred to as sheet-forming cells) at a density reaching confluence on a culture substrate coated with a cell-adhesive component and a masking component; And (B) a step of forming a sheet-like cell culture by sheet-forming and culturing the seeded cell population in a sheet-forming medium.
  • step (A) a cell population containing the desired somatic cells (sheet-forming cells) for forming a sheet-like cell culture is seeded on a culture substrate coated with a cell adhesion component.
  • the seeding on the culture substrate is as described in detail in the above-mentioned method for producing the implant.
  • the sheet-forming cells the cells described in detail in the cells constituting the implant can be used.
  • the culture medium is as described in detail above.
  • the method and concentration of coating with the cell adhesion component and the masking component are as detailed in the coating of the medium containing the masking component.
  • a cell population containing sheet-forming cells seeded on a culture substrate is sheet-cultured in a sheet-forming medium.
  • the sheet-forming culture is as described in detail in the above-mentioned graft-forming culture.
  • the sheeting medium in the method of this aspect is as detailed above, except that it may or may not contain cell adhesion components.
  • the sheeting medium comprises a cell adhesive component. In another embodiment, the sheeting medium is free of cell adhesive components.
  • Another aspect of the present disclosure relates to a method of evaluating the surface of a culture medium for producing a sheet-like cell culture.
  • Such a method includes the following steps: (I) The step of coating the surface of the culture substrate with the cell adhesion component, thereby improving the adhesion of the cells to the surface of the culture substrate. (Ii) Further, a step of coating the surface of the culture substrate with a masking component, thereby reducing the adhesion of cells to the surface of the culture substrate, (Iii) A step of seeding cardiomyocytes at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component. (Iv) A step of forming a sheet-like cell culture by sheet-forming culture in a sheet-forming medium, and (v) a step of evaluating a peeling state of the sheet-like cell culture from the surface of the culture substrate.
  • evaluating the surface of the culture substrate means that the sheet-like cell culture is cultured in a sheet using a culture medium coated with a medium containing a cell adhesive component and a masking component. It means that it is evaluated whether or not it has an appropriate substrate surface as a sheeted culture substrate through the evaluation of the peeled state. Since the cells used for producing the sheet-shaped cell culture have different peeling states from the culture base material for each lot, it is necessary to evaluate the surface of the culture base material for each lot of cells. According to the method of this aspect, the surface of the culture medium can be evaluated easily and accurately.
  • step (i) the surface of the culture substrate is coated with a cell adhesive component.
  • the cell adhesion component and coating are as described in detail in the above-mentioned method for producing a graft.
  • step (ii) the surface of the cell substrate coated in step (i) is further coated with a masking component.
  • Steps (iii) and (iv) are as described in detail in the method for producing a sheet-shaped cell culture.
  • step (v) the exfoliation state of the sheet-shaped cell culture from the surface of the culture substrate is evaluated.
  • evaluation the exfoliation state means evaluating the presence or absence of exfoliation of the sheet-like cell culture from the culture substrate and the type of exfoliation when sheet-forming culture and / or exfoliation work is performed using the coated culture substrate. Means to do. By evaluating such a peeled state, the surface of the culture medium can be evaluated.
  • exfoliation mainly include spontaneous exfoliation and exfoliation by exfoliation work, and those that can be exfoliated by exfoliation work are more likely to cause shrinkage of the sheet-like cell culture due to the intercellular binding force. It can be evaluated as a preferable surface of the culture substrate. For the evaluation, a person skilled in the art can select any method such as visual inspection or image analysis.
  • the sheet cell culture produced by the production method of the present disclosure comprises cardiomyocytes.
  • the sheet cell culture produced by the production method of the present disclosure comprises cardiomyocytes, fibroblasts, vascular endothelial cells and / or parietal cells.
  • the sheet cell cultures of the present disclosure are useful in treating diseases that are ameliorated by the application of sheet cell cultures, such as various diseases associated with tissue abnormalities. Therefore, in one aspect, the sheet cell cultures of the present disclosure are intended for use in the treatment of diseases that are ameliorated by the application of sheet cell cultures, particularly those associated with tissue abnormalities.
  • the sheet-shaped cell cultures of the present disclosure have similar properties peculiar to constituent cells except that they have higher mechanical strength than conventional sheet-shaped cell cultures, and therefore at least conventional myoblasts. It can be applied to tissues and diseases that can be treated with sheet-like cell cultures containing cells or fibroblasts.
  • the tissues to be treated include, for example, myocardium, cornea, retina, esophagus, skin, joints, cartilage, liver, pancreas, gingiva, kidney, thyroid, skeletal muscle, middle ear, bone marrow, stomach, etc.
  • Examples include the gastrointestinal tract such as the small intestine, duodenum, and large intestine.
  • the diseases to be treated are not limited, for example, heart diseases (for example, myocardial injury (myocardial infarction, cardiac trauma), cardiomyopathy, etc.), corneal diseases (for example, corneal epithelial stem cell exhaustion, corneal membrane).
  • heart diseases for example, myocardial injury (myocardial infarction, cardiac trauma), cardiomyopathy, etc.
  • corneal diseases for example, corneal epithelial stem cell exhaustion, corneal membrane.
  • Eye / chemical corrosion corneal ulcer, corneal opacification, corneal perforation, corneal scar, Stevens Johnson syndrome, ocular herbitis, etc.
  • retinal disease eg, retinal pigment degeneration, age-related yellow spot degeneration, etc.
  • Esophageal disease eg, prevention of esophageal inflammation / stenosis after esophageal surgery (removal of esophageal cancer)
  • skin disease eg, skin injury (trauma, burn), etc.
  • joint disease eg, osteoarthritis
  • Cartilage disease eg, cartilage damage
  • Liver disease eg, chronic liver disease
  • Pancreatic disease eg, diabetes
  • Dental disease eg, periodontal disease, etc.
  • Kidney disease eg, renal failure
  • thyroid disease eg, hypothyroidism
  • muscle disease eg, muscle injury, myitis, etc.
  • Patent Document 1 Non-Patent Document 1 Tanaka et al., J Gastroenterol. 2013; 48 (9): 1081-9.
  • the sheet-like cell cultures of the present disclosure can also be fragmented to an injectable size and injected at the site requiring treatment for greater efficacy than injection with a single cell suspension (Wang et. al., Cardiovasc Res. 2008; 77 (3): 515-24). Therefore, such a use is also possible for the sheet-shaped cell culture of the present disclosure.
  • Another aspect of the present disclosure relates to a method of treating a disease in a subject in need thereof, including applying an effective amount of the implant produced by the method of the present disclosure to a subject in need thereof.
  • the diseases to be treated are as described above.
  • the term “treatment” shall include all types of medically acceptable prophylactic and / or therapeutic interventions aimed at the cure, temporary remission or prevention of disease, etc.
  • the term “treatment” is medically acceptable for a variety of purposes, including delaying or stopping the progression of a disease associated with a tissue abnormality, regressing or eliminating a lesion, preventing the onset or recurrence of the disease, and the like. Including interventions to be performed.
  • an ingredient that enhances the viability, engraftment and / or function of the implant, other active ingredients useful for treating the target disease, etc. are used in combination with the implant of the present disclosure. be able to.
  • the treatment method of the present disclosure may further include the step of producing the implant of the present disclosure according to the production method of the present disclosure.
  • the treatment method of the present disclosure serves as a cell (for example, skin cells, blood cells, etc. when using iPS cells) or a source of cells for producing a graft from a subject before the step of producing the graft. It may further include the step of collecting tissue (for example, skin tissue, blood, etc. when using iPS cells).
  • the subject from which the cell or tissue from which the cell is source is collected is the same individual as the subject to whom the cell culture, composition, implant, or the like is administered.
  • the subject from which the cell or tissue from which the cell is sourced is harvested is a separate entity of the same species as the subject receiving the administration, such as a cell culture, composition, or implant.
  • the subject from which the cell or tissue that is the source of the cell is collected is an individual different from the subject to which the implant or the like is administered.
  • the effective amount is, for example, an amount capable of suppressing the onset or recurrence of a disease, reducing symptoms, or delaying or stopping the progression (for example, size, weight, number of sheet-like cell cultures, etc.).
  • the amount is preferably an amount that prevents the onset and recurrence of the disease or cures the disease.
  • an amount that does not cause an adverse effect exceeding the benefit of administration is preferable.
  • Such an amount can be appropriately determined by, for example, a test in an experimental animal such as a mouse, a rat, a dog or a pig, or a disease model animal, and such a test method is well known to those skilled in the art.
  • the size of the tissue lesion to be treated can be an important index for determining the effective amount.
  • Examples of the administration method include intravenous administration, intramuscular administration, intraosseous administration, intrathecal administration, and direct application to tissues.
  • the frequency of administration is typically once per treatment, but multiple doses can be administered if the desired effect is not obtained.
  • the cell culture, composition, sheet-like cell culture or the like of the present invention may be fixed to the target tissue by a locking means such as a suture or a staple.
  • human iPS cells maintained and cultured in a culture medium containing no feeder cells are cultured on EZSphere (Asahi Glass) for one day in StemFit AK03 medium (Ajinomoto) containing 10 ⁇ M Y27632 (Wako Pure Chemical Industries). Then, the obtained embryo-like body was cultured in a culture medium containing Actibin A, bone-forming protein (BMP) 4, and basic fibroblast growth factor (bFGF), and further, Wnt inhibitor (IWP3) and BMP4 inhibition were performed.
  • EZSphere Asahi Glass
  • StemFit AK03 medium Ajinomoto
  • bFGF basic fibroblast growth factor
  • Human myocardial cells derived from iPS cells were obtained by culturing in a culture medium containing a drug (Dorsomorphin) and a TGF ⁇ inhibitor (SB431542) and then in a culture medium containing VEGF and bFGF.
  • the proportion of cardiomyocytes in the resulting cell population ranged from 50% to 90%.
  • Example 1 Comparison between FBS-containing medium and PL-containing medium Using the cell population obtained above containing cardiomyocytes induced to differentiate from human iPS cells, the sheeting culture conditions were examined. 20% FBS or 5% human platelet lysate was added to DMEM / F12 medium as a sheeting medium. Laminin (iMatrix-511) as a cell adhesion component was further added to the sheeting medium containing the platelet lysate at 0.1 ⁇ g / mL, 0.25 ⁇ g / mL or 0.5 ⁇ g / mL, respectively. In addition, the Rho-kinase inhibitor Y27632 was added to the sheeting medium only on the first day of the sheeting culture.
  • the cell population containing cardiomyocytes was seeded in a temperature-responsive culture dish (UpCell (R) , CellSeed) at a density of 1.5 ⁇ 10 6 cells / cm 2 and placed in an environment of 37 ° C. and 5% CO 2 for 3 days. It was cultured. As the temperature-responsive culture dish, the same one as the culture solution (however, Y27632 was not included) was put therein, and the culture dish was incubated overnight at 37 ° C. and coated. After culturing, the sheet-shaped cell culture containing cardiomyocytes was exfoliated from the culture dish.
  • UpCell (R) UpCell (R) , CellSeed
  • Example 2 Sheeting in serum-free medium Similar to Test Example 1, 0.1 ⁇ g / mL of standard medium with 20% FBS added to DMEM / F12 medium and laminin (iMatrix-511) without adding blood-derived components was added. Sheet culture was performed using each of the medium (E6) as a sheet medium. The results are shown in the table below.
  • Example 3 Examination of cell adhesion components
  • laminin iMatrix-5111
  • vitronectin VTN
  • Sheeting culture was performed using -N) and fibronectin ( R) (Takarabio).
  • R fibronectin
  • the recommended concentrations are 0.5 ⁇ g / cm 2 , 0.5 ⁇ g / cm 2 and 8 ⁇ g / cm 2 , respectively.
  • sheet-forming culture was carried out using the recommended concentration and a concentration of 1/10 of the recommended concentration.
  • Example 4 Examination of cell condition and optimum concentration We investigated how the cell adhesion component affects the step of removing undifferentiated cells that can affect the activity of cells. In order to confirm whether the optimum concentration of the cell adhesion component that can be used for sheeting culture changes depending on the state of the cells, three types of cells were used for examination. As the highly active cells, a cell population containing myocardial cells induced to differentiate from the human iPS cells obtained above (no step of freezing and thawing the cells, no step of removing undifferentiated cells) was used as the above-mentioned medium-active cells.
  • the cell population containing the myocardial cells induced to differentiate from the human iPS cells obtained in 1 above was once frozen and thawed, and the cells containing the myocardial cells induced to differentiate from the human iPS cells obtained above were regarded as low-activity cells. Undifferentiated cells were removed from the population, then frozen and thawed.
  • the treatment for removing undifferentiated cells was carried out in order by using the heat treatment described in WO2017 / 038562, the sugar-free medium culture method described in WO2007 / 0888874, and the anti-CD30 antibody-binding drug treatment described in WO2016 / 072519.
  • Example 5 Examination of coating with cell adhesion component and masking component DMEM / F12 investigated how the peeling state of sheet-shaped cell culture changes when the cell culture dish is coated with a masking component such as FBS together with the cell adhesion component. 0%, 0.31%, 0.63%, 1.25%, 2.5%, 5%, 10%, 20% FBS and 0.076 ⁇ g / cm 2 , 0.76 ⁇ g / cm 2 , 7 A total of 40 coating solutions were prepared with .6 ⁇ g / cm 2 and 76 ⁇ g / cm 2 of iMatrix-511, which were added to each well of a temperature responsive culture dish and incubated overnight at 37 ° C. for coating.
  • a masking component such as FBS together with the cell adhesion component.
  • HBSS manufactured by Invitrogen
  • the culture solution was removed, HBSS (+) was added, and the mixture was allowed to stand at room temperature for 1 hour and 30 minutes for peeling work.
  • the exfoliated state of the sheet-shaped cell culture is shown in Table 3 and FIG. “Peeling” of the colorless cells in Table 3 indicates spontaneous peeling, and “peeling” of the gray cells indicates peeling by the peeling operation. X indicates that the material was not peeled off.
  • the peeling state of the sheet-shaped cell culture can be controlled so that the natural peeling can be suppressed by adjusting the concentrations of the masking component and the cell adhesion component, and the peeling can be performed only by the peeling operation.
  • the present invention it is possible to obtain a high-quality sheet-shaped cell culture when forming a sheet-shaped cell culture using cells or the like induced to differentiate from pluripotent stem cells.
  • a high-quality sheet-shaped cell culture even in the production of a xeno-free sheet-shaped cell culture used for clinical use, it becomes possible to easily form a high-quality sheet-shaped cell culture.

Abstract

The present invention addresses the problem of providing: a method for producing a high-quality sheet-like cell culture containing a variety of cells such as cardiomyocytes, in particular, cardiomyocytes derived from pluripotent stem cells, the cells highly maintaining the function thereof; a sheet-like cell culture produced using the method; and a method for treating a disease using the sheet-like cell culture. The problem is solved by a sheet-like cell culture production method comprising: (a) a step of plating a cell population including cardiomyocytes on a culture substrate; and (b) a step of culturing the plated cell population with a sheet formation medium including a cell adhesive component to form a sheet.

Description

心筋細胞のシート化方法Cardiomyocyte sheeting method
 本開示は、平成30年度、国立研究開発法人日本医療研究開発機構 、再生医療実現拠点ネットワークプログラム 疾患・組織別実用化研究拠点(拠点A)「iPS細胞を用いた心筋再生治療創成拠点」委託研究開発、産業技術力強化法第17条の適用を受ける特許出願であって、種々の細胞、例えば心筋細胞、特に多能性幹細胞由来の心筋細胞を含む、シート状細胞培養物などの移植片を製造する方法、当該方法を用いて製造された移植片、当該移植片を用いた疾患の処置方法などに関する。 This disclosure is a commissioned research project of the Japan Medical Research and Development Organization, Regenerative Medicine Realization Center Network Program, Practical Application Research Center for Diseases and Tissues (Center A), "Center for the Creation of Myocardial Regenerative Therapy Using iPS Cells" in FY2018. It is a patent application to which Article 17 of the Development and Industrial Technology Enhancement Law applies, and transplants of various cells such as sheet-shaped cell cultures containing myocardial cells, especially pluripotent stem cell-derived myocardial cells. The present invention relates to a method for producing, a transplanted piece manufactured by using the method, a method for treating a disease using the transplanted piece, and the like.
 成体の心筋細胞は自己複製能に乏しく、心筋組織が損傷を受けた場合、その修復は極めて困難である。近年、損傷した心筋組織の修復のために、細胞工学的手法により作製した心筋細胞を含む移植片を患部に移植する試みが行われている(特許文献1、非特許文献1)。かかる移植片の作製に用いる心筋細胞の供給源として最近注目されているのが、胚性幹細胞(ES細胞)や人工多能性幹細胞(iPS細胞)などの多能性幹細胞から誘導した心筋細胞であり、このような多能性幹細胞由来の心筋細胞を含むシート状細胞培養物の作製や動物での治療実験が試みられている(非特許文献2~3)。しかしながら、多能性幹細胞由来の心筋細胞を含むシート状細胞培養物の開発は始まったばかりであり、その機能的特性や、それに影響する因子などについては依然不明な部分が多い。 Adult cardiomyocytes have poor self-renewal ability, and if myocardial tissue is damaged, it is extremely difficult to repair it. In recent years, in order to repair damaged myocardial tissue, attempts have been made to transplant a graft containing cardiomyocytes prepared by a cell engineering technique into an affected area (Patent Document 1, Non-Patent Document 1). Recently, attention has been paid to cardiomyocytes derived from pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) as a source of cardiomyocytes used for producing such transplants. There are attempts to prepare sheet-like cell cultures containing such pluripotent stem cell-derived cardiomyocytes and therapeutic experiments in animals (Non-Patent Documents 2 to 3). However, the development of sheet-like cell cultures containing pluripotent stem cell-derived cardiomyocytes has just begun, and there are still many unclear points about their functional characteristics and factors that influence them.
 再生医療に用いる臨床用のシート状細胞培養物を製造する場合、移植片形成媒体中に、なるべくヒト以外の異種由来成分を含まない状態、いわゆるゼノフリーな状態で製造されることが望ましい。しかしながら、ゼノフリーな状態でシート状細胞培養物を形成することはコストや手間がかかる場合が多く、また用いる細胞によっては該細胞の所望の性質を保ったままシート状細胞培養物を形成することが困難である場合もある。 When producing a clinical sheet-shaped cell culture used for regenerative medicine, it is desirable that the graft-forming medium contains as little heterologous components other than humans as possible, that is, a so-called xeno-free state. However, forming a sheet-shaped cell culture in a xeno-free state is often costly and laborious, and depending on the cells used, it is possible to form a sheet-shaped cell culture while maintaining the desired properties of the cells. It can be difficult.
特表2007-528755号公報Special Table 2007-528755
 本開示は、種々の細胞、例えば心筋細胞、特に多能性幹細胞由来の心筋細胞、を該細胞の機能を高度に有したまま含み、高品質な移植片を製造する方法、当該方法を用いて製造された移植片、当該移植片を用いた疾患の処置方法などを提供することを目的とする。 The present disclosure is a method for producing a high-quality implant, which comprises various cells such as cardiomyocytes, particularly cardiomyocytes derived from pluripotent stem cells, while preserving the functions of the cells, using the method. It is an object of the present invention to provide a manufactured implant, a method for treating a disease using the implant, and the like.
 移植の用に供する心筋細胞を含む移植片を調製するにあたっては、ゼノフリーの移植片形成媒体で調製される必要がある。また、移植片形成媒体中にウシ胎児血清(FBS)を添加したり、無血清培地を用いたりすると、心筋細胞が十分に拍動しない、うまく移植片形成されないなどの問題が見出されていた。 When preparing a graft containing cardiomyocytes to be used for transplantation, it is necessary to prepare with a xenofree graft-forming medium. In addition, when fetal bovine serum (FBS) is added to the graft-forming medium or a serum-free medium is used, problems such as cardiomyocytes not beating sufficiently and grafts not being formed well have been found. ..
 本発明者らは、ゼノフリーの移植片形成媒体で調製しても十分な機能を有する多能性幹細胞由来の心筋細胞を用いた生体移植用のシート状細胞培養物の調製について研究する中で、無血清培地でシート化培養してもうまくシート化されないという課題に直面した。かかる課題を解決すべく研究を続ける中で、無血清の移植片形成媒体に細胞接着性成分を添加してシート化することにより、例えば無血清培地を用いてもシート化できるという新たな知見を見出した。かかる知見に基づいてさらに研究を進め、種々の細胞を含有する移植片の調製において、臨床応用に耐え得る高品質な移植片を製造することが可能であることを見出し、本発明を完成させるに至った。 The present inventors are studying the preparation of sheet-shaped cell cultures for living body transplantation using cardiomyocytes derived from pluripotent stem cells, which have sufficient functions even when prepared with a Xenofree implant-forming medium. We faced the problem that sheeting culture was not successful even when sheeted and cultured in a serum-free medium. While continuing research to solve this problem, we have found a new finding that by adding a cell adhesion component to a serum-free graft-forming medium to form a sheet, for example, a serum-free medium can be used to form a sheet. I found it. Based on this finding, further research was carried out, and it was found that it is possible to produce a high-quality graft that can withstand clinical application in the preparation of a graft containing various cells, and to complete the present invention. I arrived.
 すなわち、本発明は下記に掲げるものに関する:
[1] 体細胞を含む移植片を製造する方法であって;
(a)前記体細胞を含む細胞集団を、培養基材上に播種する工程、および(b)播種した細胞集団を、細胞接着性成分を含む移植片形成媒体で移植片形成培養する工程、を含む、前記方法。
[2] 培養基材が、さらに細胞接着性成分および/またはマスキング成分でコーティングされている、[1]に記載の方法。
[3] 移植片形成媒体中の細胞接着性成分の含有量が、培養基材のコーティングに用いる量の1/10~1/100の濃度である、[1]または[2]に記載の方法。
[4] (a)の前に、細胞集団から未分化細胞を除去する工程を含む、[1]~[3]のいずれか1つに記載の方法。
[5] 移植片形成媒体が、血清を含まない、[1]~[4]のいずれか1つに記載の方法。
[6] 移植片形成媒体が、血小板溶解物を含む、[1]~[5]のいずれか1つに記載の方法。
That is, the present invention relates to the following:
[1] A method for producing a graft containing somatic cells;
(A) a step of seeding the cell population containing the somatic cells on a culture substrate, and (b) a step of seeding the seeded cell population in a transplantation piece-forming medium containing a cell adhesion component. Included, said method.
[2] The method according to [1], wherein the culture substrate is further coated with a cell adhesion component and / or a masking component.
[3] The method according to [1] or [2], wherein the content of the cell adhesion component in the graft-forming medium is 1/10 to 1/100 of the amount used for coating the culture substrate. ..
[4] The method according to any one of [1] to [3], which comprises a step of removing undifferentiated cells from a cell population before (a).
[5] The method according to any one of [1] to [4], wherein the graft-forming medium does not contain serum.
[6] The method according to any one of [1] to [5], wherein the graft-forming medium contains a platelet lysate.
[7] 心筋細胞を含むシート状細胞培養物を製造する方法であって;
(A)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で心筋細胞を播種する工程、および(B)シート化形成媒体でシート形成培養し、シート状細胞培養物を形成する工程、を含む、シート状細胞培養物の製造方法。
[8] シート化形成媒体が細胞接着性成分を含む、または含まない、[7]に記載の方法。
[7] A method for producing a sheet-like cell culture containing cardiomyocytes;
(A) A step of seeding myocardial cells at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component, and (B) sheet-forming culture using a sheet-forming medium, and sheet-like cell culture. A method for producing a sheet-shaped cell culture, which comprises a step of forming a substance.
[8] The method according to [7], wherein the sheet formation medium contains or does not contain a cell adhesion component.
[9] シート状細胞培養物を製造するための培養基材表面を評価する方法であって、
(i)細胞接着性成分で培養基材表面をコーティングする工程、それにより細胞の培養基材表面への細胞の接着性を向上させ、
(ii)さらにマスキング成分で培養基材表面をコーティングする工程、それにより培養基材表面への細胞の接着性を低下させ、
(iii)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で体細胞を播種する工程、
(iv)シート化媒体中でシート化培養し、シート状細胞培養物を形成する工程、および
(v)シート状細胞培養物の培養基材表面からの剥離状態を評価する工程、を含む、前記方法。
[9] A method for evaluating the surface of a culture medium for producing a sheet-shaped cell culture.
(I) The step of coating the surface of the culture substrate with the cell adhesion component, thereby improving the adhesion of the cells to the surface of the culture substrate.
(Ii) Further, a step of coating the surface of the culture substrate with a masking component, thereby reducing the adhesion of cells to the surface of the culture substrate,
(Iii) A step of seeding somatic cells at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component.
The above-mentioned step including (iv) a step of forming a sheet-like cell culture by sheet-forming culture in a sheet-forming medium, and (v) a step of evaluating a peeling state of the sheet-like cell culture from the surface of a culture substrate. Method.
[10] シート化媒体が、血清を含まない、[7]~[9]のいずれか1つに記載の方法。
[11] シート化媒体が、血小板溶解物を含む、[7]~[10]のいずれか1つに記載の方法。
[12] マスキング成分が、血液由来成分である、[1]~[11]のいずれか1つに記載の方法。
[13] マスキング成分が、血清、血小板溶解物またはアルブミンである、[1]~[12]のいずれか1つに記載の方法。
[14] マスキング成分が、0.05%以上の濃度のFBSである、[1]~[13]のいずれか1つに記載の方法。
[15] 体細胞が、多能性幹細胞由来の心筋細胞である、[1]~[14]のいずれか1つに記載の方法。
[16] [1]~[6]のいずれか1つに記載の方法によって製造された移植片または[7]もしくは[8]に記載の方法によって製造されたシート状細胞培養物の治療有効量を、それを必要とする対象に投与する工程を含む、前記対象における心疾患の治療方法。
[10] The method according to any one of [7] to [9], wherein the sheeting medium does not contain serum.
[11] The method according to any one of [7] to [10], wherein the sheeting medium contains a platelet lysate.
[12] The method according to any one of [1] to [11], wherein the masking component is a blood-derived component.
[13] The method according to any one of [1] to [12], wherein the masking component is serum, platelet lysate or albumin.
[14] The method according to any one of [1] to [13], wherein the masking component is FBS having a concentration of 0.05% or more.
[15] The method according to any one of [1] to [14], wherein the somatic cell is a cardiomyocyte derived from a pluripotent stem cell.
[16] A therapeutically effective amount of the graft produced by the method according to any one of [1] to [6] or the sheet-shaped cell culture produced by the method according to [7] or [8]. A method for treating a heart disease in a subject, comprising the step of administering the drug to the subject in need thereof.
 本発明によれば、種々の細胞、例えば心筋細胞、特に多能性幹細胞から分化誘導した心筋細胞を含む細胞集団から、従来よりも高品質なシート状細胞培養物などの移植片を高効率に製造することができる。特に多能性幹細胞から分化誘導した心筋細胞を含むシート状細胞培養物など、再生医療に用いる移植片の調製において、移植片形成媒体中に血清を用いない、すなわち製造工程由来の不純物を含まない条件で移植片形成培養した場合であっても、うまく移植片形成可能となるため、再生医療において特に有用である。また、さらに移植片形成媒体に血小板溶解物を加えることにより、所望の性質を高いレベルで保持した移植片を調製可能であり、とくにヒトへの生体移植用に非常に好適な移植片を提供することが可能となる。 According to the present invention, transplants such as sheet-like cell cultures of higher quality than before can be efficiently produced from a cell population containing various cells, for example, cardiomyocytes, particularly cardiomyocytes induced to differentiate from pluripotent stem cells. Can be manufactured. In particular, in the preparation of grafts used for regenerative medicine, such as sheet-shaped cell cultures containing myocardial cells induced to differentiate from pluripotent stem cells, the graft-forming medium does not use serum, that is, it does not contain impurities derived from the manufacturing process. It is particularly useful in regenerative medicine because it enables graft formation even when the graft is formed and cultured under the conditions. Further, by adding a platelet lysate to the graft-forming medium, it is possible to prepare a graft having a desired property at a high level, and particularly to provide a very suitable graft for living-donor transplantation into humans. It becomes possible.
図1は、細胞接着性成分としてラミニン(iMatrix-511)、ビトロネクチン(VTN-N)、フィブロネクチン(RetroNectin(R))を用いた場合のシート化の様子を表す写真である。上段(AおよびB)はラミニン、中段(CおよびD)はビトロネクチン、下段(EおよびF)はフィブロネクチンを用いた場合であり、左列(A、CおよびE)はコーティング剤として使用する場合の推奨濃度で用いた場合、右列(B、DおよびF)は推奨濃度の1/10で用いた場合の結果を表す。いずれの場合においてもシート化が確認できた。FIG. 1 is a photograph showing a state of sheet formation when laminin (iMatrix-511), vitronectin (VTN-N), and fibronectin (RetroNectin (R)) are used as cell adhesion components. The upper row (A and B) is for laminin, the middle row (C and D) is for vitronectin, the lower row (E and F) is for fibronectin, and the left column (A, C and E) is for use as a coating agent. When used at the recommended concentration, the right column (B, D and F) shows the result when used at 1/10 of the recommended concentration. In any case, it was confirmed that the sheet was formed. 図2は、細胞接着性成分としてiMatrix-511、および、マスキング成分としてFBSを、様々な濃度でコーティングした培養基材でシート化を行い、剥離作業後のシート状細胞培養物の剥離状態の様子を表す写真である。iMatrix-511ととともにFBSでコーティングすることにより幅広い濃度のiMatrix-511を使用しても剥離可能であることが確認できた。FIG. 2 shows a state in which the sheet-shaped cell culture is peeled off after sheeting with iMatrix-511 as a cell adhesion component and FBS as a masking component on a culture substrate coated with various concentrations. It is a photograph showing. By coating with FBS together with iMatrix-511, it was confirmed that peeling was possible even when iMatrix-511 with a wide concentration was used.
 以下、本発明を詳細に説明する。
 本明細書において別様に定義されない限り、本明細書で用いる全ての技術用語および科学用語は、当業者が通常理解しているものと同じ意味を有する。本明細書中で参照する全ての特許、出願および他の出版物や情報は、その全体を参照により本明細書に援用する。また本明細書において参照された出版物と本明細書の記載に矛盾が生じた場合は、本明細書の記載が優先されるものとする。
Hereinafter, the present invention will be described in detail.
Unless defined otherwise herein, all technical and scientific terms used herein have the same meaning as those commonly understood by one of ordinary skill in the art. All patents, applications and other publications and information referenced herein are hereby incorporated by reference in their entirety. In the event of a conflict between the publications referred to herein and the description herein, the description herein shall prevail.
 本開示は、所望の細胞を含む移植片を製造する方法であって;
(a)前記細胞を含む細胞集団を、培養基材上に播種する工程、および
(b)播種した細胞集団を、細胞接着性成分を含む培地で移植片形成培養する工程、
を含む、前記方法に関する。
The present disclosure is a method of producing a graft containing the desired cells;
(A) a step of seeding the cell population containing the cells on a culture substrate, and (b) a step of forming and culturing the seeded cell population in a medium containing a cell adhesion component.
The present invention relates to the above-mentioned method.
 本開示において、「移植片」とは、生体内へ移植するための構造物を意味し、特に細胞を構成成分として含む移植用構造物を意味する。好ましい一態様においては、移植片は、細胞および細胞由来の物質以外の構造物(例えばスキャフォールドなど)を含まない移植用構造物である。本開示における移植片としては、これに限定するものではないが、例えばシート状細胞培養物、スフェロイド、細胞凝集塊、などが挙げられ、好ましくはシート状細胞培養物またはスフェロイド、より好ましくはシート状細胞培養物である。 In the present disclosure, the "graft" means a structure for transplantation into a living body, and particularly means a structure for transplantation containing cells as a constituent component. In a preferred embodiment, the implant is a structure for transplantation that is free of cells and structures other than cell-derived substances (eg, scaffolds, etc.). The implants in the present disclosure include, but are not limited to, sheet-like cell cultures, spheroids, cell aggregates, and the like, preferably sheet-like cell cultures or spheroids, and more preferably sheet-like. It is a cell culture.
 本開示において、「シート状細胞培養物」は、細胞が互いに連結してシート状になったものをいう。細胞同士は、直接(接着分子などの細胞要素を介するものを含む)および/または介在物質を介して、互いに連結していてもよい。介在物質としては、細胞同士を少なくとも物理的(機械的)に連結し得る物質であれば特に限定されないが、例えば、細胞外マトリックスなどが挙げられる。介在物質は、好ましくは細胞由来のもの、特に、シート状細胞培養物を構成する細胞に由来するものである。細胞は少なくとも物理的(機械的)に連結されるが、さらに機能的、例えば、化学的、電気的に連結されてもよい。シート状細胞培養物は、1の細胞層から構成されるもの(単層)であっても、2以上の細胞層から構成されるもの(積層体(多層)、例えば、2層、3層、4層、5層、6層など)であってもよい。また、シート状細胞培養物は、細胞が明確な層構造を示すことなく、細胞1個分の厚みを超える厚みを有する3次元構造を有してもよい。例えば、シート状細胞培養物の垂直断面において、細胞が水平方向に均一に整列することなく、不均一に(例えば、モザイク状に)配置された状態で存在していてもよい。 In the present disclosure, "sheet-shaped cell culture" refers to cells connected to each other to form a sheet. The cells may be linked to each other directly (including those via cell elements such as adhesion molecules) and / or via intervening substances. The intervening substance is not particularly limited as long as it is a substance capable of at least physically (mechanically) connecting cells to each other, and examples thereof include an extracellular matrix. The mediator is preferably derived from cells, particularly from the cells that make up the sheet-like cell culture. The cells are at least physically (mechanically) connected, but may be more functionally, for example, chemically or electrically connected. The sheet-like cell culture may be composed of one cell layer (single layer) or two or more cell layers (laminate (multilayer), for example, two layers, three layers, etc. It may be 4 layers, 5 layers, 6 layers, etc.). Further, the sheet-shaped cell culture may have a three-dimensional structure having a thickness exceeding the thickness of one cell without showing a clear layered structure of the cells. For example, in the vertical cross section of a sheet-shaped cell culture, cells may be present in a non-uniformly (for example, mosaic-like) arrangement without being uniformly aligned in the horizontal direction.
 本開示の移植片、特にシート状細胞培養物は、好ましくはスキャフォールド(支持体)を含まない。スキャフォールドは、その表面上および/またはその内部に細胞を付着させ、シート状細胞培養物などの移植片の物理的一体性を維持するために当該技術分野において用いられることがあり、例えば、ポリビニリデンジフルオリド(PVDF)製の膜等が知られているが、本開示の移植片は、かかるスキャフォールドがなくともその物理的一体性を維持することができる。また、本開示の移植片は、好ましくは、移植片を構成する細胞由来の物質のみからなり、それら以外の物質を含まない。 The grafts of the present disclosure, particularly sheet cell cultures, preferably do not contain scaffolds (supports). Scaffolds may be used in the art to attach cells to and / or inside the scaffold to maintain the physical integrity of the implant, such as a sheet cell culture, eg, poly. Although membranes made of vinylidene fluoride (PVDF) and the like are known, the implants of the present disclosure can maintain their physical integrity without such scaffolds. In addition, the implants of the present disclosure preferably consist only of cell-derived substances constituting the implants and do not contain any other substances.
 細胞は異種由来細胞であっても同種由来細胞であってもよい。ここで「異種由来細胞」は、移植片が移植に用いられる場合、そのレシピエントとは異なる種の生物に由来する細胞を意味する。例えば、レシピエントがヒトである場合、サルやブタに由来する細胞などが異種由来細胞に該当する。また、「同種由来細胞」は、レシピエントと同一の種の生物に由来する細胞を意味する。例えば、レシピエントがヒトである場合、ヒト細胞が同種由来細胞に該当する。同種由来細胞は、自己由来細胞(自己細胞または自家細胞ともいう)、すなわち、レシピエントに由来する細胞と、同種非自己由来細胞(他家細胞ともいう)を含む。自己由来細胞は、移植しても拒絶反応が生じないため、本開示においては好ましい。しかしながら、異種由来細胞や同種非自己由来細胞を利用することも可能である。異種由来細胞や同種非自己由来細胞を利用する場合は、拒絶反応を抑制するため、免疫抑制処置が必要となることがある。なお、本明細書中で、自己由来細胞以外の細胞、すなわち、異種由来細胞と同種非自己由来細胞を非自己由来細胞と総称することもある。本開示の一態様において、細胞は自家細胞または他家細胞である。本開示の一態様において、細胞は自家細胞(自家iPS細胞を含む)である。本開示の別の態様において、細胞は他家細胞(他家iPS細胞を含む)である。 The cell may be a heterologous cell or an allogeneic cell. Here, "heterologous cell" means a cell derived from an organism of a species different from the recipient when the graft is used for transplantation. For example, when the recipient is a human, cells derived from monkeys and pigs correspond to heterologous cells. In addition, "homogeneous cell" means a cell derived from an organism of the same species as the recipient. For example, when the recipient is a human, the human cell corresponds to an allogeneic cell. Allogeneic cells include autologous cells (also referred to as autologous cells or autologous cells), ie, recipient-derived cells and allogeneic non-autologous cells (also referred to as allogeneic cells). Autologous cells are preferred in the present disclosure because they do not cause rejection when transplanted. However, it is also possible to utilize heterologous cells and allogeneic non-autologous cells. When using heterologous cells or allogeneic non-autologous cells, immunosuppressive treatment may be required to suppress rejection. In the present specification, cells other than autologous cells, that is, allogeneic non-self-derived cells and allogeneic non-self-derived cells may be collectively referred to as non-autologous cells. In one aspect of the disclosure, the cell is an autologous cell or an allogeneic cell. In one aspect of the disclosure, the cell is an autologous cell (including an autologous iPS cell). In another aspect of the disclosure, the cell is an allogeneic cell (including an allogeneic iPS cell).
 本開示の移植片を構成する細胞は、移植片を形成し得るものであれば特に限定されず、例えば、接着細胞(付着性細胞)を含む。接着細胞は、例えば、接着性の体細胞(例えば、心筋細胞、線維芽細胞、上皮細胞、内皮細胞、肝細胞、膵細胞、腎細胞、副腎細胞、歯根膜細胞、歯肉細胞、骨膜細胞、皮膚細胞、滑膜細胞、軟骨細胞など)および幹細胞(例えば、筋芽細胞、心臓幹細胞などの組織幹細胞、胚性幹細胞、人工多能性幹細胞(iPS:induced pluripotent stem)細胞などの多能性幹細胞、間葉系幹細胞等)などを含む。本開示において、人工多能性幹細胞(iPS細胞)は遺伝子を導入して誘導された細胞である。体細胞は、幹細胞、特にiPS細胞から分化させたもの(iPS細胞由来接着細胞)であってもよい。移植片を構成する細胞の非限定例としては、例えば、筋芽細胞(例えば、骨格筋芽細胞など)、間葉系幹細胞(例えば、骨髄、脂肪組織、末梢血、皮膚、毛根、筋組織、子宮内膜、胎盤、臍帯血由来のものなど)、心筋細胞、線維芽細胞、心臓幹細胞、胚性幹細胞、iPS細胞、滑膜細胞、軟骨細胞、上皮細胞(例えば、口腔粘膜上皮細胞、網膜色素上皮細胞、鼻粘膜上皮細胞など)、内皮細胞(例えば、血管内皮細胞など)、肝細胞(例えば、肝実質細胞など)、膵細胞(例えば、膵島細胞など)、腎細胞、副腎細胞、歯根膜細胞、歯肉細胞、骨膜細胞、皮膚細胞等が挙げられる。iPS細胞由来接着細胞の非限定例としては、iPS細胞由来の心筋細胞、線維芽細胞、上皮細胞、内皮細胞、肝細胞、膵細胞、腎細胞、副腎細胞、歯根膜細胞、歯肉細胞、骨膜細胞、皮膚細胞、滑膜細胞、軟骨細胞などが挙げられる。 The cells constituting the implant of the present disclosure are not particularly limited as long as they can form the implant, and include, for example, adherent cells (adhesive cells). Adhesive cells include, for example, adherent somatic cells (eg, myocardial cells, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, root membrane cells, gingival cells, bone membrane cells, skin. Cells, synovial cells, chondrocytes, etc.) and stem cells (eg, tissue stem cells such as myoblasts, cardiac stem cells, embryonic stem cells, pluripotent stem cells such as induced pluripotent stem (iPS) cells, It includes mesenchymal stem cells, etc.). In the present disclosure, induced pluripotent stem cells (iPS cells) are cells induced by introducing a gene. Somatic cells may be stem cells, particularly those differentiated from iPS cells (iPS cell-derived adherent cells). Non-limiting examples of cells constituting the transplant include, for example, myoblasts (eg, skeletal myoblasts), mesenchymal stem cells (eg, bone marrow, adipose tissue, peripheral blood, skin, hair roots, muscle tissue, etc.) Endometrial, placenta, umbilical cord blood, etc.), myocardial cells, fibroblasts, cardiac stem cells, embryonic stem cells, iPS cells, synovial cells, chondrocytes, epithelial cells (eg, oral mucosal epithelial cells, retinal pigments) Epithelial cells, nasal mucosal epithelial cells, etc.), endothelial cells (eg, vascular endothelial cells, etc.), hepatocytes (eg, hepatic parenchymal cells, etc.), pancreatic cells (eg, pancreatic islet cells, etc.), kidney cells, adrenal cells, root membrane Examples include cells, gingival cells, bone membrane cells, skin cells and the like. Non-limiting examples of iPS cell-derived adherent cells include iPS cell-derived myocardial cells, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, renal cells, adrenal cells, root membrane cells, gingival cells, and bone membrane cells. , Skin cells, synovial cells, cartilage cells and the like.
 移植片を構成する細胞は、移植片による治療が可能な任意の生物に由来し得る。かかる生物には、限定されずに、例えば、ヒト、非ヒト霊長類、イヌ、ネコ、ブタ、ウマ、ヤギ、ヒツジ、げっ歯目動物(例えば、マウス、ラット、ハムスター、モルモットなど)、ウサギなどが含まれる。また、移植片を構成する細胞の種類の数は特に限定されず、1種類のみの細胞で構成されていてもよいが、2種類以上の細胞を用いたものであってもよい。移植片を形成する細胞が2種類以上ある場合、最も多い細胞の含有比率(純度)は、シート状細胞培養物の形成終了時において、例えば50%以上、好ましくは60%以上、より好ましくは70%以上、さらに好ましくは75%以上であり得る。 The cells that make up the implant can be derived from any organism that can be treated with the implant. Such organisms include, but are not limited to, for example, humans, non-human primates, dogs, cats, pigs, horses, goats, sheep, rodents (eg, mice, rats, hamsters, guinea pigs, etc.), rabbits, etc. Is included. The number of types of cells constituting the transplant is not particularly limited, and may be composed of only one type of cells, or may be composed of two or more types of cells. When there are two or more types of cells forming a graft, the content ratio (purity) of the most abundant cells is, for example, 50% or more, preferably 60% or more, more preferably 70 at the end of formation of the sheet-like cell culture. % Or more, more preferably 75% or more.
 培養基材は、細胞がその上で細胞培養物を形成し得るものであれば特に限定されず、例えば、種々の材質および/または形状の容器、容器中の固形もしくは半固形の表面などを含む。容器は、培養液などの液体を透過させない構造・材料が好ましい。かかる材料としては、限定することなく、例えば、ポリエチレン、ポリプロピレン、テフロン(登録商標)、ポリエチレンテレフタレート、ポリメチルメタクリレート、ナイロン6,6、ポリビニルアルコール、セルロース、シリコン、ポリスチレン、ガラス、ポリアクリルアミド、ポリジメチルアクリルアミド、金属(例えば、鉄、ステンレス、アルミニウム、銅、真鍮)等が挙げられる。また、容器は、少なくとも1つの平坦な面を有することが好ましい。かかる容器の例としては、限定することなく、例えば、細胞培養物の形成が可能な培養基材で構成された底面と、液体不透過性の側面とを備えた培養容器が挙げられる。かかる培養容器の特定の例としては、限定されずに、細胞培養皿、細胞培養ボトルなどが挙げられる。容器の底面は透明であっても不透明であってもよい。容器の底面が透明であると、容器の裏側から細胞の観察、計数などが可能となる。また、容器は、その内部に固形もしくは半固形の表面を有してもよい。固形の表面としては、上記のごとき種々の材料のプレートや容器などが、半固形の表面としては、ゲル、軟質のポリマーマトリックスなどが挙げられる。培養基材は、上記材料を用いて作製してもよいし、市販のものを利用してもよい。 The culture substrate is not particularly limited as long as the cells can form a cell culture on the cells, and includes, for example, containers of various materials and / or shapes, solid or semi-solid surfaces in the containers, and the like. .. The container preferably has a structure / material that does not allow a liquid such as a culture solution to permeate. Such materials include, without limitation, for example, polyethylene, polypropylene, Teflon®, polyethylene terephthalate, polymethylmethacrylate, nylon 6,6, polyvinyl alcohol, cellulose, silicon, polystyrene, glass, polyacrylamide, polydimethyl. Acrylamide, metals (eg, iron, stainless steel, aluminum, copper, brass) and the like can be mentioned. Also, the container preferably has at least one flat surface. Examples of such a container include, without limitation, a culture container having a bottom surface made of a culture substrate capable of forming a cell culture and a liquid-impermeable side surface. Specific examples of such a culture vessel include, but are not limited to, a cell culture dish, a cell culture bottle, and the like. The bottom surface of the container may be transparent or opaque. If the bottom surface of the container is transparent, cells can be observed and counted from the back side of the container. Further, the container may have a solid or semi-solid surface inside the container. Examples of the solid surface include plates and containers of various materials as described above, and examples of the semi-solid surface include gels and soft polymer matrices. The culture substrate may be prepared using the above-mentioned materials, or a commercially available one may be used.
 好ましい培養基材としては、限定することなく、例えば、シート状細胞培養物の形成に適した、接着性の表面を有する基材、スフェロイドの形成に適した、低接着性の表面を有する基材および/または均一なウェル状構造を有する基材などが挙げられる。具体的には、シート状細胞培養物の形成の場合であれば、例えば、コロナ放電処理したポリスチレン、コラーゲンゲルや親水性ポリマーなどの親水性化合物を該表面にコーティングした基材、さらには、コラーゲン、フィブロネクチン、ラミニン、ビトロネクチン、プロテオグリカン、グリコサミノグリカンなどの細胞外マトリックスや、カドヘリンファミリー、セレクチンファミリー、インテグリンファミリーなどの細胞接着因子などを表面にコーティングした基材などが挙げられる。また、かかる基材は市販されている(例えば、Corning(R) TC-Treated Culture Dish、Corningなど)。またスフェロイドの形成の場合であれば、例えば軟寒天、ポリ(N-イソプロピルアクリルアミド)(PIPAAm)をポリエチレングリコール(PEG)で架橋した温度応答性ゲル(市販名:メビオールゲル)、ポリメタクリル酸ヒドロキシエチル(ポリHEMA)、2-メタクリロイルオキシエチルホスホリスコリン(MPC)ポリマーなどのハイドロゲルなどの非細胞接着性化合物を表面にコーティングした基材および/または均一な凹凸構造を表面に有する基材などが挙げられる。かかる基材もまた市販されている(例えば、EZSPHERE(R)など)。培養基材は全体または部分が透明であっても不透明であってもよい。 Preferred culture substrates include, without limitation, for example, a substrate having an adhesive surface suitable for forming a sheet-like cell culture, and a substrate having a low adhesive surface suitable for forming spheroids. And / or a substrate having a uniform well-like structure and the like. Specifically, in the case of forming a sheet-like cell culture, for example, a substrate coated with a hydrophilic compound such as corona discharge-treated polystyrene, collagen gel or hydrophilic polymer on the surface thereof, and further, collagen. , Fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan and other extracellular matrix, and base materials coated with cell adhesion factors such as cadoherin family, selectin family and integrin family on the surface. In addition, such substrates are commercially available (eg, Corning (R) TC-Treated Culture Dish, Corning, etc.). In the case of spheroid formation, for example, soft agar, temperature-responsive gel obtained by cross-linking poly (N-isopropylacrylamide) (PIPAAm) with polyethylene glycol (PEG), polyhydroxyethyl methacrylate (commercially available name: mebiol gel), polyhydroxyethyl methacrylate ( Examples include a base material coated with a non-cell adhesive compound such as a hydrogel such as poly-HEMA) and 2-methacryloyloxyethyl phosphorischoline (MPC) polymer and / or a base material having a uniform uneven structure on the surface. Be done. Such substrates are also commercially available (eg, EZSPHERE (R), etc.). The culture medium may be transparent or opaque in whole or in part.
 培養基材は、刺激、例えば、温度や光に応答して物性が変化する材料で表面が被覆されていてもよい。かかる材料としては、限定されずに、例えば、(メタ)アクリルアミド化合物、N-アルキル置換(メタ)アクリルアミド誘導体(例えば、N-エチルアクリルアミド、N-n-プロピルアクリルアミド、N-n-プロピルメタクリルアミド、N-イソプロピルアクリルアミド、N-イソプロピルメタクリルアミド、N-シクロプロピルアクリルアミド、N-シクロプロピルメタクリルアミド、N-エトキシエチルアクリルアミド、N-エトキシエチルメタクリルアミド、N-テトラヒドロフルフリルアクリルアミド、N-テトラヒドロフルフリルメタクリルアミド等)、N,N-ジアルキル置換(メタ)アクリルアミド誘導体(例えば、N,N-ジメチル(メタ)アクリルアミド、N,N-エチルメチルアクリルアミド、N,N-ジエチルアクリルアミド等)、環状基を有する(メタ)アクリルアミド誘導体(例えば、1-(1-オキソ-2-プロペニル)-ピロリジン、1-(1-オキソ-2-プロペニル)-ピペリジン、4-(1-オキソ-2-プロペニル)-モルホリン、1-(1-オキソ-2-メチル-2-プロペニル)-ピロリジン、1-(1-オキソ-2-メチル-2-プロペニル)-ピペリジン、4-(1-オキソ-2-メチル-2-プロペニル)-モルホリン等)、またはビニルエーテル誘導体(例えば、メチルビニルエーテル)のホモポリマーまたはコポリマーからなる温度応答性材料、アゾベンゼン基を有する光吸収性高分子、トリフェニルメタンロイコハイドロオキシドのビニル誘導体とアクリルアミド系単量体との共重合体、および、スピロベンゾピランを含むN-イソプロピルアクリルアミドゲル等の光応答性材料などの公知のものを用いることができる(例えば、特開平2-211865、特開2003-33177参照)。これらの材料に所定の刺激を与えることによりその物性、例えば、親水性や疎水性を変化させ、同材料上に付着した細胞培養物の剥離を促進することができる。温度応答性材料で被覆された培養皿は市販されており(例えば、CellSeed Inc.のUpCell(R))、これらを本開示の製造方法に使用することができる。  The surface of the culture substrate may be coated with a material whose physical properties change in response to irritation, for example, temperature or light. Such materials include, but are not limited to, for example, (meth) acrylamide compounds, N-alkyl substituted (meth) acrylamide derivatives (eg, N-ethylacrylamide, Nn-propylacrylamide, Nn-propylmethacrylamide, etc. N-isopropylacrylamide, N-isopropylmethacrylamide, N-cyclopropylacrylamide, N-cyclopropylmethacrylamide, N-ethoxyethylacrylamide, N-ethoxyethylmethacrylamide, N-tetrahydrofurfurylacrylamide, N-tetrahydrofurfurylmethacryl (Amid, etc.), N, N-dialkyl-substituted (meth) acrylamide derivatives (eg, N, N-dimethyl (meth) acrylamide, N, N-ethylmethylacrylamide, N, N-diethylacrylamide, etc.), having cyclic groups (eg, N, N-dialkylacrylamide, N, N-diethylacrylamide, etc.) Meta) Acrylamide derivatives (eg 1- (1-oxo-2-propenyl) -pyrrolidine, 1- (1-oxo-2-propenyl) -piperidin, 4- (1-oxo-2-propenyl) -morpholin, 1 -(1-oxo-2-methyl-2-propenyl) -pyrrolidine, 1- (1-oxo-2-methyl-2-propenyl) -piperidin, 4- (1-oxo-2-methyl-2-propenyl) -A temperature-responsive material consisting of a homopolymer or copolymer of a vinyl ether derivative (eg, methylvinyl ether), a photoabsorbable polymer having an azobenzene group, a vinyl derivative of triphenylmethane leucohydrooxide and an acrylamide-based single amount. Known materials such as a copolymer with a body and a photoresponsive material such as N-isopropylacrylamide gel containing spirobenzopyran can be used (see, for example, JP-A-2-21186 and JP-A-2003-33177). ). By giving a predetermined stimulus to these materials, their physical characteristics, for example, hydrophilicity and hydrophobicity can be changed, and the exfoliation of the cell culture adhering on the material can be promoted. Culture dishes coated with a temperature-responsive material are commercially available (eg, CellSeed Inc.'s UpCell (R) ) and can be used in the production methods of the present disclosure.
 培養基材は、種々の形状であってもよい。また、その面積は特に限定されないが、例えば、約1cm~約200cm、約2cm~約100cm、約3cm~約50cmなどであってよい。例えば、培養基材として直径10cmの円形の培養皿が挙げられる。この場合、面積は56.7cmとなる。培養表面は平坦であってもよいし、凹凸構造を有していてもよい。凹凸構造を有する場合、均一な凹凸構造であることが好ましい。 The culture medium may have various shapes. The area thereof is not particularly limited, but may be, for example, about 1 cm 2 to about 200 cm 2 , about 2 cm 2 to about 100 cm 2 , about 3 cm 2 to about 50 cm 2 . For example, as a culture substrate, a circular culture dish having a diameter of 10 cm can be mentioned. In this case, the area is 56.7 cm 2 . The culture surface may be flat or may have an uneven structure. When it has a concavo-convex structure, it is preferable that it has a uniform concavo-convex structure.
 本開示において、「多能性幹細胞」は、当該技術分野で周知の用語であり、三胚葉、すなわち内胚葉、中胚葉および外胚葉に属する全ての系列の細胞に分化することができる能力を有する細胞を意味する。多能性幹細胞の非限定例としては、例えば、胚性幹細胞(ES細胞)、核移植胚性幹細胞(ntES細胞)、人工多能性幹細胞(iPS細胞)などが挙げられる。通常多能性幹細胞を特定の細胞に分化誘導する際には、まず多能性幹細胞を浮遊培養して、上記三胚葉のいずれかの細胞の凝集体を形成し、その後凝集体を形成する細胞を目的とする特定の細胞に分化誘導させる。 In the present disclosure, "pluripotent stem cell" is a well-known term in the art and has the ability to differentiate into three germ layers, i.e. cells of all lineages belonging to endoderm, mesoderm and ectoderm. Means cell. Non-limiting examples of pluripotent stem cells include, for example, embryonic stem cells (ES cells), nuclear-transplanted embryonic stem cells (ntES cells), induced pluripotent stem cells (iPS cells), and the like. Normally, when inducing differentiation of pluripotent stem cells into specific cells, pluripotent stem cells are first suspended-cultured to form aggregates of any of the above three germ layers, and then cells that form aggregates. Induce differentiation into specific cells of interest.
 本開示において、「多能性幹細胞由来の分化誘導細胞」は、多能性幹細胞から特定の種類の細胞に分化するように分化誘導処理された任意の細胞を意味する。分化誘導細胞の非限定例は、心筋細胞、骨格筋芽細胞などの筋肉系の細胞、ニューロン細胞、オリゴデンドロサイト、ドーパミン産生細胞などの神経系の細胞、網膜色素上皮細胞などの網膜細胞、血球細胞、骨髄細胞などの造血系の細胞、T細胞、NK細胞、NKT細胞、樹状細胞、B細胞などの免疫関連の細胞、肝細胞、膵β細胞、腎細胞などの臓器を構成する細胞、軟骨細胞、生殖細胞などの他、これらの細胞に分化する前駆細胞や体性幹細胞などを含む。かかる前駆細胞や体性幹細胞の典型例としては、例えば心筋細胞における間葉系幹細胞、多分化性心臓前駆細胞、単能性心臓前駆細胞、神経系の細胞における神経幹細胞、造血系の細胞や免疫関連の細胞における造血幹細胞およびリンパ系幹細胞などが挙げられる。多能性幹細胞の分化誘導は、既知の任意の手法を用いて行うことができる。例えば、多能性幹細胞から心筋細胞への分化誘導は、Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358に記載の手法に基づいて行うことができる。所望の細胞として、iPS由来心筋細胞等の多能性幹細胞由来の分化誘導細胞を用いる場合、分化誘導後に未分化細胞の除去処理を行ってもよい。未分化細胞の除去処理は、当該技術分野において知られており、例えばWO2017/038562、WO2016/072519およびWO2007/088874等に記載された方法を用いることができる。 In the present disclosure, "pluripotent stem cell-derived differentiation-inducing cell" means any cell that has been subjected to differentiation-inducing treatment so as to differentiate from a pluripotent stem cell into a specific type of cell. Non-limiting examples of differentiation-inducing cells include muscular cells such as myocardial cells and skeletal myoblasts, neural cells such as neuron cells, oligodendrocytes and dopamine-producing cells, retinal cells such as retinal pigment epithelial cells, and blood cells. Hematopoietic cells such as cells and bone marrow cells, immune-related cells such as T cells, NK cells, NKT cells, dendritic cells and B cells, cells constituting organs such as hepatocytes, pancreatic β cells and renal cells, In addition to cartilage cells and germ cells, it also includes precursor cells and somatic stem cells that differentiate into these cells. Typical examples of such progenitor cells and somatic stem cells include mesenchymal stem cells in myocardial cells, pluripotent cardiac progenitor cells, monopoly cardiac progenitor cells, neural stem cells in neural cells, hematopoietic cells and immunity. Examples include hematopoietic stem cells and lymphoid stem cells in related cells. Induction of differentiation of pluripotent stem cells can be performed using any known method. For example, the induction of differentiation of pluripotent stem cells into cardiomyocytes can be performed based on the methods described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 and WO2014 / 185358. When a differentiation-inducing cell derived from a pluripotent stem cell such as an iPS-derived cardiomyocyte is used as the desired cell, the undifferentiated cell may be removed after the differentiation induction. The treatment for removing undifferentiated cells is known in the art, and the methods described in, for example, WO2017 / 038562, WO2016 / 072519, WO2007 / 088744, etc. can be used.
 また分化誘導細胞は、リプログラミングのための遺伝子以外の任意の有用な遺伝子が導入されたiPS細胞から誘導された細胞であってもよい。かかる細胞の非限定例としては、例えば、Themeli M. et al. Nature Biotechnology, vol. 31, no. 10, pp. 928-933, 2013に記載のキメラ抗原受容体の遺伝子が導入されたiPS細胞から誘導されるT細胞などが挙げられる。また、多能性幹細胞から分化誘導された後、任意の有用な遺伝子が導入された細胞もまた、本発明の分化誘導細胞に包含される。 Further, the differentiation-inducing cell may be a cell derived from an iPS cell into which any useful gene other than the gene for reprogramming has been introduced. Non-limiting examples of such cells include, for example, iPS cells into which the chimeric antigen receptor gene described in Themeli M. et al. Nature Biotechnology, vol. 31, no. 10, pp. 928-933, 2013 has been introduced. Examples include T cells derived from. In addition, cells into which any useful gene has been introduced after induction of differentiation from pluripotent stem cells are also included in the differentiation-inducing cells of the present invention.
 本開示において、「細胞接着性成分」は、上記培養基材の説明において記載される、細胞外マトリクスおよび細胞接着性因子などを含む、細胞と接着する性質を有する単離されたポリペプチドを意味する。かかる細胞接着性成分は、一般的には培養基材の表面を被覆(コート)するために用いられる。細胞接着性成分の例としては、これに限定するものではないが、例えばコラーゲン、フィブロネクチン、ラミニン、ビトロネクチン、プロテオグリカン、グリコサミノグリカンなどの細胞外マトリックス、カドヘリンファミリー、セレクチンファミリー、インテグリンファミリーなどの細胞接着因子などが挙げられるほか、これらの改変物または機能的同等物、例えばラミニン511、ラミニン221などのラミニンの改変物、VTN-Nなどのビトロネクチンの改変物、Kimizuka et al., J. Biochem. (1991), 110, pp.284-291などに記載のレトロネクチン(R)などのフィブロネクチンの改変物などが挙げられる。これらが少なくとも一つ含まれているか、または二つ以上の混合物であってもよい。本開示の細胞接着性成分は、好ましい一態様において、細胞接着因子および/またはその機能的同等物以外の血液由来成分を含まない。 In the present disclosure, "cell adhesion component" means an isolated polypeptide having the property of adhering to cells, including extracellular matrix and cell adhesion factors described in the above description of the culture medium. To do. Such cell-adhesive components are generally used to coat the surface of the culture medium. Examples of cell-adhesive components include, but are not limited to, extracellular matrix such as collagen, fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan, and cells such as cadoherin family, selectin family, and integrin family. In addition to such as adhesion factors, these variants or functional equivalents, such as laminin variants such as laminin 511 and laminin 221 and vitronectin variants such as VTN-N, Kimizuka et al., J. Biochem. Examples thereof include variants of fibronectin such as retronectin (R) described in (1991), 110, pp.284-291 and the like. These may be contained at least one or a mixture of two or more. In a preferred embodiment, the cell adhesion components of the present disclosure do not contain blood-derived components other than cell adhesion factors and / or their functional equivalents.
 以下に所望の細胞が心筋細胞であり、移植片がシート状細胞培養物である場合を例として、本発明を詳述する。
 本開示の一側面は、心筋細胞を含む高品質なシート状細胞培養物を製造する方法に関する。本開示の方法は、以下の工程(a)および(b)を含む:
(a)心筋細胞を含む細胞集団を、培養基材上に播種する工程、および
(b)播種した細胞集団を、細胞接着性成分を含むシート化媒体でシート化培養する工程。
The present invention will be described in detail below, taking as an example the case where the desired cell is a cardiomyocyte and the implant is a sheet-like cell culture.
One aspect of the disclosure relates to a method for producing a high quality sheet cell culture containing cardiomyocytes. The method of the present disclosure includes the following steps (a) and (b):
(A) A step of seeding a cell population containing myocardial cells on a culture substrate, and (b) a step of sheeting and culturing the seeded cell population in a sheeting medium containing a cell adhesion component.
 本開示において、「心筋細胞」とは、心筋細胞の特徴を有する細胞を意味する。心筋細胞の特徴としては、限定されずに、例えば、心筋細胞マーカーの発現、自律的拍動の存在などが挙げられる。心筋細胞マーカーの非限定例としては、例えば、c-TNT(cardiac troponin T)、CD172a(別名SIRPAまたはSHPS-1)、KDR(別名CD309、FLK1またはVEGFR2)、PDGFRA、EMILIN2、VCAMなどが挙げられる。一態様において、多能性幹細胞由来の心筋細胞は、c-TNT陽性かつ/またはCD172a陽性である。 In the present disclosure, "cardiomyocyte" means a cell having the characteristics of cardiomyocyte. The characteristics of cardiomyocytes include, but are not limited to, expression of cardiomyocyte markers, presence of autonomous pulsation, and the like. Non-limiting examples of cardiomyocyte markers include, for example, c-TNT (cardiac troponin T), CD172a (also known as SIRPA or SHPS-1), KDR (also known as CD309, FLK1 or VEGFR2), PDGFRA, EMILIN2, VCAM and the like. .. In one embodiment, cardiomyocytes derived from pluripotent stem cells are c-TNT positive and / or CD172a positive.
 本開示の方法に用いられる心筋細胞は、生体から直接得られたものであっても、他の細胞から誘導されたものであってもよいが、好ましくは他の細胞から誘導されたものである。心筋細胞への誘導としては、線維芽細胞等に心筋誘導因子を導入する方法や、多能性幹細胞、心臓前駆細胞などの心筋細胞に分化する性質を有する細胞を分化誘導する方法などが挙げられる。 The cardiomyocytes used in the method of the present disclosure may be directly obtained from a living body or derived from other cells, but are preferably derived from other cells. .. Examples of the induction into cardiomyocytes include a method of introducing a myocardial inducing factor into fibroblasts and the like, and a method of inducing differentiation of cells having the property of differentiating into cardiomyocytes such as pluripotent stem cells and cardiac progenitor cells. ..
 工程(a)において、培養基材への播種は、例えば、細胞をシート化媒体に懸濁した細胞懸濁液を、培養基材を備えた培養容器に注入することなどにより行ってもよい。細胞懸濁液の注入には、スポイトやピペットなど、細胞懸濁液の注入操作に適した器具を用いることができる。細胞の播種密度は、シート状細胞培養物を形成し得る密度で行われ、かかる密度は所望の細胞により異なり得るが、当業者であれば当該技術分野において公知の手法などから適切な密度を選択することができる。例えば心筋細胞を含むシート状細胞培養物である場合、例えば2.0×10個/cm以上などであり得るが、より高密度で播種してもよい。 In step (a), seeding on the culture substrate may be performed, for example, by injecting a cell suspension in which cells are suspended in a sheeting medium into a culture vessel provided with the culture substrate. For injection of the cell suspension, an instrument suitable for the injection operation of the cell suspension, such as a dropper or a pipette, can be used. The seeding density of cells is set to a density capable of forming a sheet-like cell culture, and the density may vary depending on the desired cells, but those skilled in the art select an appropriate density from methods known in the art. can do. For example, in the case of a sheet-shaped cell culture containing cardiomyocytes, it can be, for example, 2.0 × 10 5 cells / cm 2 or more, but may be seeded at a higher density.
 より高密度の例としては、例えばコンフルエントに達する密度、すなわち播種した際に細胞が培養容器の接着表面一面を覆うことが想定される程度の密度、例えば、播種した際に、細胞が互いに接触することが想定される程度の密度、接触阻害が発生する密度、または接触阻害により細胞の増殖を実質的に停止する密度あるいはそれ以上であり得る。播種密度の上限は、特に制限されないが、密度が過度に高い場合には、死滅する細胞が多くなり、非効率となる。本開示の一態様において、播種密度は、例えば約1.0×10個/cm~約1.0×10個/cm、約1.0×10個/cm~約5.0×10個/cm、約1.0×10個/cm~約3.0×10個/cm、約1.5×10個/cm~約1.0×10個/cm、約1.5×10個/cm~約5.0×10個/cm、約1.5×10個/cm~約3.0×10個/cm、約2.0×10個/cm~約1.0×10個/cm、約2.0×10個/cm~約5.0×10個/cm、約2.0×10個/cm~約3.0×10個/cmなどであり得る。好ましい一態様において、播種密度は、約1.76×10個/cm~約2.33×10個/cmである。 Examples of higher densities include, for example, a density that reaches confluence, that is, a density at which cells are expected to cover the entire adhesive surface of the culture vessel when seeded, for example, cells come into contact with each other when seeded. It can be as dense as expected, the density at which contact inhibition occurs, or the density at which cell proliferation is substantially stopped by contact inhibition or higher. The upper limit of the seeding density is not particularly limited, but if the seeding density is excessively high, more cells will die, resulting in inefficiency. In one aspect of the present disclosure, the seeding density is, for example, about 1.0 × 10 6 / cm 2 to about 1.0 × 10 7 / cm 2 , about 1.0 × 10 6 / cm 2 to about 5. .0 x 10 6 pieces / cm 2 , about 1.0 x 10 6 pieces / cm 2 to about 3.0 x 10 6 pieces / cm 2 , about 1.5 x 10 6 pieces / cm 2 to about 1.0 × 10 7 pieces / cm 2 , about 1.5 × 10 6 pieces / cm 2 to about 5.0 × 10 6 pieces / cm 2 , about 1.5 × 10 6 pieces / cm 2 to about 3.0 × 10 6 pieces / cm 2 , about 2.0 x 10 6 pieces / cm 2 to about 1.0 x 10 7 pieces / cm 2 , about 2.0 x 10 6 pieces / cm 2 to about 5.0 x 10 6 pieces It can be / cm 2 , about 2.0 × 10 6 pieces / cm 2 to about 3.0 × 10 6 pieces / cm 2 . In a preferred embodiment, the sowing density is from about 1.76 × 10 6 / cm 2 to about 2.33 × 10 6 / cm 2 .
 播種される細胞集団は、所望の細胞(例えば心筋細胞)を含んでいれば、他の細胞を含んでいてもよく、所望の細胞が心筋細胞である場合は、例えば線維芽細胞、血管内皮細胞および/または壁細胞などがさらに含まれ得る。細胞集団は、組織から採取した細胞集団をそのまま用いてもよいし、例えば上記Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358に記載の手法などを用いてiPS細胞から分化誘導して得られた細胞集団をそのまま用いてもよいし、凍結保存やプレ培養、未分化細胞除去などを実施した後に用いてもよい。好ましい一態様において、播種される細胞集団は、iPS細胞から分化誘導後、培養基材上(好ましくは平面状の培養基材上)に播種して接着培養を行い、その後回収された細胞集団である。かかる接着培養の前または後に、凍結保存および解凍を実施してもよい。接着培養を行うことにより、その後の移植片の形成において、高品質な移植片の形成を、高確率で達成することが可能となる。
 かかる接着培養ステップにおいて、培養条件などは、通常の接着培養を行う場合の条件に準じてよい。例えば、市販の接着培養用培養容器を用いて、37℃、5%CO条件下での培養などであってよい。細胞の播種密度は、細胞同士の接着および/または細胞と培養基材との接着の形成を妨げない密度であればいかなる密度であってもよく、例えばサブコンフルエントな密度であってもよいし、コンフルエントに達する密度またはそれ以上であってもよい。培養時間は、細胞同士の接着および/または細胞と培養基材との接着が形成される程度の時間であればよく、具体的には例えば2~24時間、2~12時間、2~6時間、2~4時間程度であればよい。
The seeded cell population may contain other cells as long as they contain the desired cells (eg, myocardial cells), and when the desired cells are myocardial cells, for example, fibroblasts, vascular endothelial cells. And / or wall cells and the like may be further included. As the cell population, the cell population collected from the tissue may be used as it is, or by using, for example, the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 or WO 2014/185358. The cell population obtained by inducing differentiation from iPS cells may be used as it is, or may be used after cryopreservation, pre-culture, removal of undifferentiated cells, or the like. In a preferred embodiment, the seeded cell population is a cell population that is induced to differentiate from iPS cells, seeded on a culture substrate (preferably on a flat culture substrate), adherently cultured, and then recovered. is there. Cryopreservation and thawing may be performed before or after such adhesive culture. By performing adhesive culture, it is possible to achieve high-quality graft formation with a high probability in the subsequent formation of the graft.
In such an adhesive culture step, the culture conditions and the like may be the same as those for normal adhesive culture. For example, a commercially available culture container for adhesive culture may be used for culturing under 37 ° C. and 5% CO 2 conditions. The seeding density of the cells may be any density as long as it does not interfere with the adhesion between the cells and / or the formation of the adhesion between the cells and the culture substrate, and may be, for example, a subconfluent density. It may be at or above a density that reaches confluence. The culturing time may be such that adhesion between cells and / or adhesion between cells and the culture substrate is formed, and specifically, for example, 2 to 24 hours, 2 to 12 hours, and 2 to 6 hours. It may be about 2 to 4 hours.
 移植片形成のための細胞集団に未分化細胞が含まれる場合、体内に移植された後に腫瘍化などのリスクが生じ得る。したがって、好ましい一態様において、細胞集団は未分化細胞を含まない。未分化細胞を含まない細胞集団を得るために、生体から採取または多能性幹細胞から分化誘導して得られた細胞集団に対し、未分化細胞除去操作を施してよい。本開示において、「未分化細胞除去操作」は、細胞集団、典型的には多能性幹細胞を分化誘導して得られた分化誘導細胞を含む細胞集団から、腫瘍形成能を有する未分化細胞を除去する操作を意味し、既知の任意の手法を用いて行うことができる。かかる手法の非限定例としては、未分化細胞に特異的なマーカー(例えば、細胞表面マーカーなど)を用いた種々の分離法、例えば、磁気細胞分離法(MACS)、フローサイトメトリー法、アフィニティ分離法や、特異的プロモーターにより選択マーカー(例えば、抗生物質耐性遺伝子など)を発現させる方法、未分化細胞の生存に必要な因子(栄養源(メチオニン等の栄養源やbFGF等の未分化状態の維持因子など)を除いた培地で培養して未分化細胞を駆逐する方法、分化を促進する因子(VEGF、BMP、activin、など)の存在下で培養し、未細胞の分化を促進させる方法、未分化細胞の表面抗原をターゲットにした薬剤で処理する方法などが挙げられる。また、WO2014/126146、WO2012/056997に記載の方法、WO2012/147992に記載の方法、WO2012/133674に記載の方法、WO2012/012803(特表2013-535194)に記載の方法、WO2012/078153(特表2014-501518)に記載の方法、特開2013-143968およびTohyama S. et al., Cell Stem Cell Vol.12 January 2013, Page 127-137に記載の方法、Lee MO et al., PNAS 2013 Aug 27;110(35):E3281-90に記載の方法、WO2016/072519に記載の方法、WO2013/100080に記載の方法、特開2016-093178に記載の方法、WO2017/038526に記載の熱処理を用いる方法なども挙げられる。好ましくは、未分化細胞の除去操作は、WO2007/088874に記載されるような無糖培地で培養する方法、WO2016/072519に記載されるような特異抗体を用いる方法およびWO2017/038526に記載されるような熱処理を用いる方法などが挙げられる。 If undifferentiated cells are included in the cell population for graft formation, there may be a risk of tumorigenesis after transplantation into the body. Therefore, in a preferred embodiment, the cell population does not include undifferentiated cells. In order to obtain a cell population that does not contain undifferentiated cells, a cell population obtained by collecting from a living body or inducing differentiation from pluripotent stem cells may be subjected to an undifferentiated cell removal operation. In the present disclosure, the "undifferentiated cell removal operation" refers to undifferentiated cells having tumorigenicity from a cell population, typically a cell population containing differentiation-inducing cells obtained by inducing differentiation of pluripotent stem cells. It means a removal operation and can be performed using any known method. Non-limiting examples of such a method include various separation methods using markers specific to undifferentiated cells (for example, cell surface markers), for example, magnetic cell separation method (MACS), flow cytometry method, affinity separation. Methods, methods for expressing selective markers (eg, antibiotic resistance genes, etc.) by specific promoters, factors necessary for the survival of undifferentiated cells (nutrient sources (nutrient sources such as methionine, maintenance of undifferentiated state such as bFGF) A method of exterminating undifferentiated cells by culturing in a medium excluding (factors, etc.), a method of culturing in the presence of factors that promote differentiation (VEGF, BMP, activin, etc.) and promoting undifferentiated cells, not yet Examples thereof include a method of treating the surface antigen of differentiated cells with a drug targeting the surface antigen. Further, the method described in WO2014 / 126146, WO2012 / 056997, the method described in WO2012 / 147992, the method described in WO2012 / 133674, WO2012. / 012803 (Special Table 2013-535194), WO2012 / 078153 (Special Table 2014-501518), Japanese Patent Application Laid-Open No. 2013-143966 and Tohyama S. et al., Cell Stem Cell Vol.12 January 2013 , Page 127-137, Lee MO et al., PNAS 2013 Aug 27; 110 (35): E3281-90, WO2016 / 072519, WO2013 / 10080, Examples thereof include the method described in JP-A-2016-093178 and the method using the heat treatment described in WO2017 / 038526. Preferably, the operation for removing undifferentiated cells is performed by culturing in a sugar-free medium as described in WO2007 / 088874. , A method using a specific antibody as described in WO2016 / 072519, a method using a heat treatment as described in WO2017 / 038526, and the like.
 播種される培養基材は、上記で詳述したとおりであるが、好ましい一態様において、細胞外マトリクスや細胞接着因子などの細胞接着性成分を表面にコーティングした培養基材である。細胞接着性成分としては、これに限定するものではないが、例えばコラーゲン、フィブロネクチン、ラミニン、ビトロネクチン、プロテオグリカン、グリコサミノグリカンなどの細胞外マトリックス、カドヘリンファミリー、セレクチンファミリー、インテグリンファミリーなどの細胞接着因子などが挙げられるほか、これらの改変物、例えばラミニン511(ラミニンの改変物)、VTN-N(ビトロネクチンの改変物)、レトロネクチン(R)(フィブロネクチンの改変物)であってもよい。細胞接着性成分のコーティングは、細胞接着性成分を含む媒体を培養基材と接触させ、インキュベートすることにより達成し得る。かかる媒体に含まれ得る細胞接着性成分の量は、コーティングする基材の面積、用いる細胞接着性成分や、培養する細胞種などにより異なり得る。当業者であれば、製品のプロトコルなどに従って最適な量または濃度を設定することができ、かかる量または濃度としては、マスキング成分を使用せず接着成分だけでコーティングする場合、例えばラミニン511やVTN-Nであれば約0.1~1.0μg/cm、レトロネクチン(R)であれば約4~20μg/cmなどが挙げられる。 The culture base material to be seeded is as described in detail above, but in a preferred embodiment, it is a culture base material whose surface is coated with a cell adhesion component such as an extracellular matrix or a cell adhesion factor. The cell-adhesive component is not limited to this, for example, extracellular matrix such as collagen, fibronectin, laminin, vitronectin, proteoglycan, glycosaminoglycan, and cell adhesion factors such as cadoherin family, selectin family, and integrin family. In addition to these, these variants may be, for example, laminin 511 (a variant of laminin), VTN-N (a variant of vitronectin), retronectin (R) (a variant of fibronectin). Coating of the cell adhesion component can be achieved by contacting and incubating a medium containing the cell adhesion component with the culture substrate. The amount of the cell adhesion component that can be contained in such a medium may vary depending on the area of the substrate to be coated, the cell adhesion component used, the cell type to be cultured, and the like. A person skilled in the art can set the optimum amount or concentration according to the product protocol, etc., and the amount or concentration can be set when coating with only the adhesive component without using the masking component, for example, laminin 511 or VTN-. For N, it is about 0.1 to 1.0 μg / cm 2 , and for retronectin (R) , it is about 4 to 20 μg / cm 2 .
 さらに別の一態様において、細胞外マトリクスや細胞接着因子などの細胞接着性成分の他に血液由来成分などのマスキング成分を表面にコーティングした培養基材である。「マスキング成分」は、細胞接着性成分でコーティングされた培養基材表面の細胞接着性を低減させることができる成分を意味する。かかる成分としては、典型的には、血液由来成分であり、例えば通常血清(例えば、ウシ胎仔血清などのウシ血清、ウマ血清、ヒト血清等)の他、通常血清に含まれるアルブミン、血小板溶解物、スキムミルク、ポリビニルアルコールなどのアルブミン代替物質などが挙げられる。 In yet another embodiment, it is a culture substrate in which the surface is coated with a masking component such as a blood-derived component in addition to a cell adhesion component such as an extracellular matrix or a cell adhesion factor. The "masking component" means a component capable of reducing the cell adhesion on the surface of the culture medium coated with the cell adhesion component. Such components are typically blood-derived components, such as normal serum (eg, bovine serum such as bovine fetal serum, horse serum, human serum, etc.), as well as albumin and platelet lysates contained in normal serum. , Skim milk, albumin substitutes such as polyvinyl alcohol, and the like.
 マスキング成分を含む媒体のコーティングは、細胞接着性成分のコーティングと同時または別々に行なってよく、細胞接着性成分および/またはマスキング成分を含む媒体を培養基材と接触させ、インキュベートすることにより達成し得る。かかる媒体に含まれ得るマスキング成分の濃度は、用いる細胞接着性成分や細胞接着性成分の濃度、コーティングする基材の面積、培養する細胞種などにより異なり得る。 Coating of the medium containing the masking component may be performed simultaneously with or separately from the coating of the cell adhesion component, and is achieved by contacting and incubating the medium containing the cell adhesion component and / or the masking component with the culture substrate. obtain. The concentration of the masking component that can be contained in such a medium may vary depending on the concentration of the cell adhesion component or the cell adhesion component used, the area of the substrate to be coated, the cell type to be cultured, and the like.
 マスキング成分の濃度としては、例えばFBSであればコーティング媒体中に0.05%、0.075%、0.1%、0.125%、0.15%、0.16%、0.2%、0.3%、0.4%、0.5%、0.6%、0.8%、1.0%、1.25%、1.5%、1.75%、2.0%、2.25%、2.5%、2.75%、3.0%、3.5%、4.0%、4.5%、5.0%、5.5%、7.5%、9%、10%、12.5%、15%、17.5%、20%、22.5%、25%、30%、40%以上であってよく、上限値は特に限定されないが45%以下30%、25%、22.5%、20%、17.5%、15%、12.5%、10%、9%、7.5%、5.5%、5.0%、4.5%、4.0%、3.5%、3.0%、2.75%、2.5%、2.0%、1.5%、1.25%、1.0%、0.8%、0.6%、0.5%、0.31%、0.16%、0.125%、0.1%以下であてよい。 For example, in the case of FBS, the concentration of the masking component is 0.05%, 0.075%, 0.1%, 0.125%, 0.15%, 0.16%, 0.2% in the coating medium. , 0.3%, 0.4%, 0.5%, 0.6%, 0.8%, 1.0%, 1.25%, 1.5%, 1.75%, 2.0% 2.25%, 2.5%, 2.75%, 3.0%, 3.5%, 4.0%, 4.5%, 5.0%, 5.5%, 7.5% , 9%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 30%, 40% or more, and the upper limit is not particularly limited, but 45. % Or less 30%, 25%, 22.5%, 20%, 17.5%, 15%, 12.5%, 10%, 9%, 7.5%, 5.5%, 5.0%, 4.5%, 4.0%, 3.5%, 3.0%, 2.75%, 2.5%, 2.0%, 1.5%, 1.25%, 1.0%, It may be 0.8%, 0.6%, 0.5%, 0.31%, 0.16%, 0.125%, 0.1% or less.
 細胞接着性成分としてラミニン511と組み合わせてコーティングする場合、ラミニン511の量または濃度として0.01μg/cm~100μg/cmに対して0.1%~30%のFBS、ラミニン511の0.05μg/cm~80μg/cmに対して0.15%~25%のFBS、ラミニン511の0.05μg/cm~100μg/cmに対して0.7.5%~20%、ラミニン511の0.05μg/cm~1.0μg/cmに対して0.05%~25%のFBS、剥離作業により剥離可能となる観点からラミニン511の0.5μg/cm~80μg/cmに対して0.25%~7.5%のFBS、より好ましくはラミニン511の0.5μg/cm~80μg/cmに対して0.75%~12.5%のFBSである。FBSなどの異種血清でコーティングした場合、培養基材を任意の方法で臨床上障害とならないレベルまで洗浄することが好ましい。 When coated in combination with laminin 511 as a cell adhesion component, the amount or concentration of laminin 511 is 0.01 μg / cm 2 to 100 μg / cm 2 with respect to 0.1% to 30% FBS, laminin 511 0. 05μg / cm 2 ~ 80μg / cm 2 with respect to 0.15% to 25% of the FBS, 0.7.5% 20% with respect to 0.05μg / cm 2 ~ 100μg / cm 2 laminin 511, laminin 0.05% to 25% FBS with respect to 0.05 μg / cm 2 to 1.0 μg / cm 2 of 511, 0.5 μg / cm 2 to 80 μg / cm of laminin 511 from the viewpoint of peeling by peeling work 0.25% to 7.5% FBS for two, more preferably from 0.75% to 12.5% FBS relative to 0.5μg / cm 2 ~ 80μg / cm 2 laminin 511. When coated with heterologous serum such as FBS, it is preferable to wash the culture substrate by any method to a level that does not cause clinical damage.
 インキュベート時間は、マスキング成分が培養基材上に付着することができれば特に限定されず、例えば、1~72時間、好ましくは4~48時間、より好ましくは5~24時間、さらに好ましくは6~12時間である。インキュベート温度も、マスキング成分が培養基材上に付着することができれば特に限定されず、例えば、0~60℃、好ましくは4~45℃、より好ましくは室温~40℃である。 The incubation time is not particularly limited as long as the masking component can adhere to the culture substrate, for example, 1 to 72 hours, preferably 4 to 48 hours, more preferably 5 to 24 hours, still more preferably 6 to 12 hours. It's time. The incubation temperature is also not particularly limited as long as the masking component can adhere to the culture substrate, and is, for example, 0 to 60 ° C., preferably 4 to 45 ° C., more preferably room temperature to 40 ° C.
 工程(b)において、播種した細胞をシート化培養する。本明細書においては、播種した細胞を移植片として形成するための培養を「移植片形成培養」と称し、移植片がシート状細胞培養物であり、播種した細胞をシート化するための培養を特に、「シート化培養」と称する。播種した細胞のシート化は、既知の任意の手法および条件で行うことができる。かかる手法の非限定例は、例えば、特開2010-081829、特開2010-226991、特開2011-110368、特開2011-172925、WO2014/185517などに記載されている。細胞の移植片形成(例えばシート化)は、細胞同士が接着分子や、細胞外マトリックスなどの細胞間接着機構を介して互いに接着することにより達成されると考えられている。したがって、播種した細胞を移植片形成するステップは、例えば、細胞を、細胞間接着を形成する条件下で培養することにより達成することができる。かかる条件は、細胞間接着を形成することができればいかなるものであってもよいが、通常は一般的な細胞培養条件と同様の条件であれば細胞間接着を形成することができる。かかる条件としては、例えば、約37℃、5%COでの培養が挙げられる。また、培養は通常の圧力下(大気圧下、非加圧下)で行うことができる。移植片形成培養(シート化培養)においては、細胞間接着が形成されればよいため、必ずしも細胞が増殖する必要はない。好ましい一態様において、移植片形成培養(シート化培養)は、細胞を増殖させずに行われる。培養は任意の大きさおよび形状の容器で行うことができる。シート状細胞培養物の大きさや形状は、培養容器の細胞付着面の大きさ・形状を調整すること、または、培養容器の細胞付着面に、所望の大きさ・形状の型枠を設置し、その内部で細胞を培養することなどにより任意に調節することができる。 In step (b), the seeded cells are sheet-cultured. In the present specification, the culture for forming the seeded cells as a transplant is referred to as "transplant formation culture", and the transplant is a sheet-like cell culture, and the culture for forming the seeded cells into a sheet is referred to as a culture. In particular, it is referred to as "sheet culture". Sheeting of seeded cells can be performed by any known method and condition. Non-limiting examples of such a method are described in, for example, Japanese Patent Application Laid-Open No. 2010-081829, Japanese Patent Application Laid-Open No. 2010-226991, Japanese Patent Application Laid-Open No. 2011-110368, Japanese Patent Application Laid-Open No. 2011-172925, WO2014 / 185517 and the like. Cell graft formation (eg, sheet formation) is believed to be achieved by the cells adhering to each other via adhesion molecules or intercellular adhesion mechanisms such as extracellular matrix. Thus, the step of forming a graft of seeded cells can be accomplished, for example, by culturing the cells under conditions that form cell-cell adhesions. Such conditions may be any as long as they can form cell-cell adhesion, but usually, cell-cell adhesion can be formed under the same conditions as general cell culture conditions. Such conditions include, for example, culturing at about 37 ° C. and 5% CO 2. In addition, the culture can be carried out under normal pressure (atmospheric pressure, non-pressurization). In the graft-forming culture (sheet-forming culture), it is sufficient that cell-cell adhesion is formed, so that the cells do not necessarily have to proliferate. In a preferred embodiment, the graft-forming culture (sheeted culture) is performed without cell proliferation. Culturing can be carried out in containers of any size and shape. For the size and shape of the sheet-shaped cell culture, adjust the size and shape of the cell adhesion surface of the culture vessel, or install a mold of the desired size and shape on the cell adhesion surface of the culture vessel. It can be arbitrarily adjusted by culturing cells inside the cell.
 シート化培養の時間は、播種する細胞の種類や細胞密度により異なり得る。例えばiPS細胞から心筋細胞を調製してシート化する場合、例えば約2.1×10個/cmなどの密度で播種し、4日以上培養することによりシート化を行ってよい。また、播種密度をコンフルエントに達する密度、すなわちより高密度で播種する場合、シート化培養の期間を短縮することができ、培養時間は2~4日、より好ましくは2~3日であってよい。 The time for sheeting culture can vary depending on the type of cells to be seeded and the cell density. For example, when cardiomyocytes are prepared from iPS cells and made into a sheet, the sheet may be formed by seeding at a density of, for example, about 2.1 × 10 5 cells / cm 2 and culturing for 4 days or more. Further, when the seeding density reaches the confluence, that is, when the seeding is performed at a higher density, the period of sheeting culture can be shortened, and the culture time may be 2 to 4 days, more preferably 2 to 3 days. ..
 移植片形成(例えばシート化)に用いる媒体(移植片形成媒体、移植片形成がシート化である場合は特にシート化媒体と称する場合もある)としては、細胞接着性成分を含み、細胞の移植片形成を可能にするものであれば特に限定されず、例えば、生理食塩水、種々の生理緩衝液(例えば、PBS、HBSS等)、種々の細胞培養用の基礎培地をベースにした液体などを使用してもよい。かかる基礎培地には、限定されずに、例えば、DMEM、MEM、F12、DME、RPMI1640、MCDB(MCDB102、104、107、120、131、153、199など)、L15、SkBM、RITC80-7、DMEM/F12などが含まれる。これらの基礎培地の多くは市販されており、その組成も公知となっている。基礎培地は、標準的な組成のまま(例えば、市販されたままの状態で)用いてもよいし、細胞種や細胞条件に応じてその組成を適宜変更してもよい。したがって、本発明に用いる基礎培地は、公知の組成のものに限定されず、1または2以上の成分が追加、除去、増量もしくは減量されたものを含む。移植片形成媒体は、通常血清(例えば、ウシ胎仔血清などのウシ血清、ウマ血清、ヒト血清等)、種々の成長因子(例えば、FGF、EGF、VEGF、HGF等)などの添加物を含んでもよいが、シート状細胞培養物をゼノフリー条件下で製造する場合、特にウシ血清、ウマ血清などの異種血清を含まないことが好ましい。本開示は、細胞接着性成分を含む移植片形成媒体を用いて移植片形成培養することを特徴とし、これにより移植片形成媒体が無血清であっても高品質で移植片形成可能であるという効果を奏するものである。
したがって好ましい一態様において、移植片形成媒体は血清を含まない。
 シート化媒体に含まれる細胞接着性成分の濃度は、細胞に悪影響を及ぼさない限り特に限定されないが、上記コーティングの際に用いられる濃度と同等またはそれ以下でよい。かかる濃度としては、例えばラミニン511やVTN-Nであれば約0.01~10μg/mL、好ましくは約0.1~1μg/mL、レトロネクチンで(R)あれば約0.2~100μg/mL、好ましくは約2~10μg/mLなどが挙げられる。
The medium used for graft formation (for example, sheet formation) (the graft-forming medium, which may be referred to as a sheet-forming medium when the graft formation is sheet-forming) contains a cell adhesion component and transplants cells. It is not particularly limited as long as it enables fragment formation, and for example, physiological saline, various physiological buffer solutions (for example, PBS, WBSS, etc.), various basal medium-based liquids for cell culture, and the like can be used. You may use it. Such basal medium is not limited to, for example, DMEM, MEM, F12, DME, RPMI1640, MCDB (MCDB102, 104, 107, 120, 131, 153, 199, etc.), L15, SkBM, RITC80-7, DMEM. / F12 and the like are included. Many of these basal media are commercially available, and their compositions are also known. The basal medium may be used as it has a standard composition (for example, as it is on the market), or the composition may be appropriately changed depending on the cell type and cell conditions. Therefore, the basal medium used in the present invention is not limited to those having a known composition, and includes those in which one or more components are added, removed, increased or decreased. Transplantation media may also include additives such as normal serum (eg, bovine serum such as bovine fetal serum, horse serum, human serum, etc.) and various growth factors (eg, FGF, EGF, VEGF, HGF, etc.). However, when the sheet-shaped cell culture is produced under xenofree conditions, it is particularly preferable that it does not contain heterologous sera such as bovine serum and horse serum. The present disclosure is characterized in that a graft-forming culture is performed using a graft-forming medium containing a cell adhesion component, whereby a high-quality graft can be formed even if the graft-forming medium is serum-free. It is effective.
Therefore, in a preferred embodiment, the graft-forming medium is serum-free.
The concentration of the cell adhesive component contained in the sheeting medium is not particularly limited as long as it does not adversely affect the cells, but may be equal to or less than the concentration used in the above coating. Such a concentration is, for example, about 0.01 to 10 μg / mL for laminin 511 or VTN-N, preferably about 0.1 to 1 μg / mL, and about 0.2 to 100 μg / mL for retronectin (R). , Preferably about 2-10 μg / mL and the like.
 別の好ましい一態様において、シート化媒体は、さらに血小板溶解物を含む。本開示において、「血小板溶解物」(Platelet lysate:PL)は、血小板に対して凍結融解を繰り返すことにより得られる、成長因子等を豊富に含む組成物をいう。かかる組成物は、細胞培養用の培地添加物として市販されており、当該技術分野において公知であり、例えばBieback et al., STEM CELLS, 2009;27:2331-2341に記載の方法などにより調製可能である。近年では、間葉系幹細胞の増殖を促進することなどが知られている。本発明者らは、心筋細胞を含むシート状細胞培養物の製造において、シート化媒体に血小板溶解物を含有せしめることにより、従来よりも早い培養日数で、強い自律拍動が観察されることを初めて見出した。 In another preferred embodiment, the sheeting medium further comprises a platelet lysate. In the present disclosure, "platelet lysate" (PL) refers to a composition rich in growth factors and the like, which is obtained by repeatedly freezing and thawing platelets. Such a composition is commercially available as a medium additive for cell culture, is known in the art, and can be prepared by, for example, the method described in Bieback et al., STEM CELLS, 2009; 27: 2331-2341. Is. In recent years, it is known to promote the proliferation of mesenchymal stem cells. In the production of sheet-shaped cell cultures containing cardiomyocytes, the present inventors have observed that by incorporating platelet lysates into the sheeting medium, strong autonomous pulsation is observed in a faster culture time than before. I found it for the first time.
 移植片形成媒体中に含まれる血小板溶解物の濃度は、当該技術分野において通常用いられる程度であればよく、例えば1%、2.5%、5%、10%、15%、20%などであってよい。好ましい一態様において、血小板溶解物は、移植片形成媒体中に1%~20%、より好ましくは2%~10%、さらに好ましくは2.5%~10%程度含有される。 The concentration of platelet lysate contained in the graft-forming medium may be such that it is usually used in the art, for example, 1%, 2.5%, 5%, 10%, 15%, 20% and the like. It may be there. In a preferred embodiment, the platelet lysate is contained in the graft-forming medium in an amount of 1% to 20%, more preferably 2% to 10%, still more preferably 2.5% to 10%.
 移植片形成媒体は、移植片形成培養中に適宜入れ替えてよい。また、移植片形成の進行に合わせて媒体の組成を変化させてもよい。本発明者らは、iPS細胞から誘導された心筋細胞を含むシート状細胞培養物の製造において、Rhoキナーゼ(ROCK)阻害剤を添加したシート化媒体を、シート化培養1日目の媒体として用いることにより、効果的にシート状細胞培養物が形成されることを新たに見出した。したがって本開示の好適な一態様において、1日目のシート化培養に用いるシート化媒体は、Rhoキナーゼ阻害剤を含む。かかる態様においては、2日目以降のシート化媒体にはRhoキナーゼ阻害剤を含んでも含まなくてもよいが、好ましくはRhoキナーゼ阻害剤を含まない。 The graft-forming medium may be appropriately replaced during the graft-forming culture. In addition, the composition of the medium may be changed according to the progress of graft formation. In the production of sheet-like cell cultures containing cardiomyocytes derived from iPS cells, the present inventors use a sheeted medium to which a Rho-kinase (ROCK) inhibitor is added as a medium for the first day of sheeted culture. As a result, it was newly found that sheet-shaped cell cultures are effectively formed. Therefore, in a preferred embodiment of the present disclosure, the sheeting medium used for the sheeting culture on day 1 comprises a Rho-kinase inhibitor. In such an embodiment, the sheeting medium after the second day may or may not contain a Rho kinase inhibitor, but preferably does not contain a Rho kinase inhibitor.
 培養基材は細胞接着性成分でコーティングされていてもコーティングされていなくてもよい。培養基材が細胞接着性成分でコーティングされている場合、培養基材をコーティングしている細胞接着性成分は、シート化媒体に含まれる細胞接着性成分と同一であってもよいし異なっていてもよいが、好ましくは同一の細胞接着性成分である。一態様において、培養基材は、細胞接着性成分のみ、好ましくはシート化媒体に含まれる細胞接着性成分のみでコーティングされている。したがってかかる態様においては、培養基材は細胞接着性成分以外の成分、例えば血清などを含まない。また、培養基材は、細胞接着性成分に代えてまたは加えて別の成分でコーティングされていてもよい。かかる別の成分としては、例えば上記培養基材の説明において例示したコーティング成分、例えば温度応答性材料などが挙げられる。一態様において、本開示の培養基材は、細胞接着性成分に加えて、血清、FBSまたはアルブミンでコーティングされている。 The culture substrate may or may not be coated with a cell adhesive component. When the culture substrate is coated with the cell adhesion component, the cell adhesion component coating the culture substrate may be the same as or different from the cell adhesion component contained in the sheeting medium. It may be, but preferably the same cell adhesion component. In one embodiment, the culture substrate is coated with only the cell adhesive component, preferably only the cell adhesive component contained in the sheeting medium. Therefore, in such an embodiment, the culture medium does not contain components other than the cell adhesive component, such as serum. In addition, the culture substrate may be coated with another component in place of or in addition to the cell adhesion component. Examples of such other components include coating components exemplified in the above description of the culture medium, such as temperature-responsive materials. In one aspect, the culture medium of the present disclosure is coated with serum, FBS or albumin in addition to the cell adhesive components.
 移植片形成媒体に含まれる細胞接着性成分の濃度は、含まれる細胞接着性成分の種類や移植片形成する細胞の状態などにより異なり得る。例えば、バイアビリティの低い、すなわち活性が弱い細胞を用いている場合、細胞接着性成分の含有量は少ない方がよい。シート化媒体に含まれる細胞接着性成分の濃度は、培養基材のコーティング剤として同じ細胞接着性成分を用いる場合に使用する濃度を基準(100%)として、約0.1%、約0.5%、約1%、約5%、約10%、約20%、約25%、約50%、約75%、約100%などであってよい。したがって好ましい一態様において、シート化媒体に含まれる細胞接着性成分の濃度範囲は、培養基材のコーティング剤として同じ細胞接着性成分を用いる場合に使用する濃度を基準(100%)として、約0.1%~約100%、約0.1%~約100%、約0.1%~約50%、約0.1%~約25%、約0.1%~約20%、約0.1%~約10%、約1%~約100%、約1%~約100%、約0.5%~約100%、約0.5%~約100%、約0.5%~約50%、約0.5%~約25%、約0.5%~約20%、約0.5%~約10%、約1%~約50%、約1%~約25%、約1%~約20%、約1%~約10%、約0.5%~約100%、約5%~約100%、約5%~約50%、約5%~約25%、約5%~約20%、約5%~約10%などであってよい。 The concentration of the cell adhesion component contained in the graft-forming medium may differ depending on the type of the cell adhesion component contained and the state of the cells forming the graft. For example, when cells with low viability, that is, weak activity are used, the content of the cell adhesion component should be low. The concentration of the cell adhesion component contained in the sheeting medium is about 0.1%, about 0.%, based on the concentration used when the same cell adhesion component is used as the coating agent for the culture substrate (100%). It may be 5%, about 1%, about 5%, about 10%, about 20%, about 25%, about 50%, about 75%, about 100%, and the like. Therefore, in a preferred embodiment, the concentration range of the cell adhesion component contained in the sheeting medium is about 0 based on the concentration used when the same cell adhesion component is used as the coating agent for the culture substrate (100%). .1% to about 100%, about 0.1% to about 100%, about 0.1% to about 50%, about 0.1% to about 25%, about 0.1% to about 20%, about 0 .1% to about 10%, about 1% to about 100%, about 1% to about 100%, about 0.5% to about 100%, about 0.5% to about 100%, about 0.5% to About 50%, about 0.5% to about 25%, about 0.5% to about 20%, about 0.5% to about 10%, about 1% to about 50%, about 1% to about 25%, About 1% to about 20%, about 1% to about 10%, about 0.5% to about 100%, about 5% to about 100%, about 5% to about 50%, about 5% to about 25%, It may be about 5% to about 20%, about 5% to about 10%, and the like.
 上述のとおり、本開示の方法に用いられる心筋細胞は、生体から直接得られたものであっても、他の細胞から誘導されたものであってもよいが、好ましくは他の細胞から誘導されたものである。誘導する他の細胞としては、心筋線維芽細胞などの線維芽細胞、心臓前駆細胞などの心筋細胞に分化する前駆細胞の他、任意の細胞に分化し得る多能性幹細胞などが挙げられ、好ましくは多能性幹細胞、より好ましくはiPS細胞、さらに好ましくはヒトiPS細胞である。 As described above, the cardiomyocytes used in the method of the present disclosure may be directly obtained from a living body or derived from other cells, but are preferably derived from other cells. It is a thing. Examples of other cells to be induced include fibroblasts such as myocardial fibroblasts, progenitor cells that differentiate into myocardial cells such as cardiac progenitor cells, and pluripotent stem cells that can differentiate into arbitrary cells, which are preferable. Are pluripotent stem cells, more preferably iPS cells, and even more preferably human iPS cells.
 本開示の別の側面は、所望の体細胞、例えば心筋細胞を含むシート状細胞培養物を製造する方法に関する。かかる方法は、以下の工程を含む:
(A)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で所望の体細胞(以下、シート形成細胞と称する場合がある)を含む細胞集団を播種する工程;および
(B)播種した細胞集団を、シート化媒体中でシート化培養し、シート状細胞培養物を形成する工程。
 工程(A)において、シート状細胞培養物を形成するための所望の体細胞(シート形成細胞)を含む細胞集団を、細胞接着性成分でコーティングされた培養基材上に播種する。培養基材への播種については、上記移植片の製造方法において詳述したとおりである。シート形成細胞は、上記移植片を構成する細胞において詳述した細胞を用いることができる。
Another aspect of the disclosure relates to a method of producing a sheet-like cell culture containing desired somatic cells, such as cardiomyocytes. Such a method includes the following steps:
(A) A step of seeding a cell population containing desired somatic cells (hereinafter, may be referred to as sheet-forming cells) at a density reaching confluence on a culture substrate coated with a cell-adhesive component and a masking component; And (B) a step of forming a sheet-like cell culture by sheet-forming and culturing the seeded cell population in a sheet-forming medium.
In step (A), a cell population containing the desired somatic cells (sheet-forming cells) for forming a sheet-like cell culture is seeded on a culture substrate coated with a cell adhesion component. The seeding on the culture substrate is as described in detail in the above-mentioned method for producing the implant. As the sheet-forming cells, the cells described in detail in the cells constituting the implant can be used.
 培養基材は、上記において詳述したとおりである。細胞接着性成分およびマスキング成分でのコーティングの方法および濃度はマスキング成分を含む媒体のコーティングにおいて詳述したとおりである。 The culture medium is as described in detail above. The method and concentration of coating with the cell adhesion component and the masking component are as detailed in the coating of the medium containing the masking component.
 工程(B)において、培養基材上に播種されたシート形成細胞を含む細胞集団を、シート化媒体中でシート化培養する。ここでシート化培養は、上記移植片形成培養において詳述したとおりである。本側面の方法におけるシート化媒体は、細胞接着性成分を含んでも含まなくてもよいこと以外は、上記において詳述したとおりである。ある一態様において、シート化媒体は、細胞接着性成分を含む。別の一態様において、シート化媒体は、細胞接着性成分を含まない。 In step (B), a cell population containing sheet-forming cells seeded on a culture substrate is sheet-cultured in a sheet-forming medium. Here, the sheet-forming culture is as described in detail in the above-mentioned graft-forming culture. The sheeting medium in the method of this aspect is as detailed above, except that it may or may not contain cell adhesion components. In one embodiment, the sheeting medium comprises a cell adhesive component. In another embodiment, the sheeting medium is free of cell adhesive components.
 本開示の別の側面は、シート状細胞培養物を製造するための培養基材表面を評価する方法に関する。かかる方法は、以下の工程を含む:
(i)細胞接着性成分で培養基材表面をコーティングする工程、それにより細胞の培養基材表面への細胞の接着性を向上させ、
(ii)さらにマスキング成分で培養基材表面をコーティングする工程、それにより培養基材表面への細胞の接着性を低下させ、
(iii)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で心筋細胞を播種する工程、
(iv)シート化媒体中でシート化培養し、シート状細胞培養物を形成する工程、および
(v)シート状細胞培養物の培養基材表面からの剥離状態を評価する工程。
Another aspect of the present disclosure relates to a method of evaluating the surface of a culture medium for producing a sheet-like cell culture. Such a method includes the following steps:
(I) The step of coating the surface of the culture substrate with the cell adhesion component, thereby improving the adhesion of the cells to the surface of the culture substrate.
(Ii) Further, a step of coating the surface of the culture substrate with a masking component, thereby reducing the adhesion of cells to the surface of the culture substrate,
(Iii) A step of seeding cardiomyocytes at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component.
(Iv) A step of forming a sheet-like cell culture by sheet-forming culture in a sheet-forming medium, and (v) a step of evaluating a peeling state of the sheet-like cell culture from the surface of the culture substrate.
 本側面の方法において、「培養基材表面を評価する」とは、細胞接着性成分およびマスキング成分を含む媒体でコーティングした培養基材を用いてシート化培養する際に、シート状細胞培養物の剥離状態の評価を通じてシート化培養基材として適切な基材表面を有しているか評価することを意味する。シート状細胞培養物の製造に用いる細胞は、ロットごとに培養基材からの剥離状態が異なるため、細胞のロットごとに培養基材表面を評価する必要がある。本側面の方法によれば、培養基材表面の評価を簡便かつ的確に行うことができる。 In the method of this aspect, "evaluating the surface of the culture substrate" means that the sheet-like cell culture is cultured in a sheet using a culture medium coated with a medium containing a cell adhesive component and a masking component. It means that it is evaluated whether or not it has an appropriate substrate surface as a sheeted culture substrate through the evaluation of the peeled state. Since the cells used for producing the sheet-shaped cell culture have different peeling states from the culture base material for each lot, it is necessary to evaluate the surface of the culture base material for each lot of cells. According to the method of this aspect, the surface of the culture medium can be evaluated easily and accurately.
 工程(i)において、培養基材表面を細胞接着性成分でコーティングする。細胞接着性成分およびコーティングについては、上記移植片の製造方法において詳述したとおりである。
 工程(ii)において、工程(i)でコーティングした細胞基材表面を、さらにマスキング成分でコーティングする。
In step (i), the surface of the culture substrate is coated with a cell adhesive component. The cell adhesion component and coating are as described in detail in the above-mentioned method for producing a graft.
In step (ii), the surface of the cell substrate coated in step (i) is further coated with a masking component.
 工程(iii)および(iv)については、上記シート状細胞培養物の製造方法において詳述したとおりである。
 工程(v)において、シート状細胞培養物の培養基材表面からの剥離状態を評価する。「剥離状態を評価する」とは、コーティングした培養基材を用いてシート化培養および/または剥離作業をした際のシート状細胞培養物の培養基材からの剥離の有無および剥離の種類を評価することを意味する。かかる剥離状態を評価することにより、培養基材表面を評価することができる。剥離の種類には、主に自然剥離と剥離作業による剥離が存在し、自然剥離したものは細胞間結合力によるシート状細胞培養物の収縮が生じ得るため、剥離作業により剥離可能なものがより好ましい培養基材表面であると評価することができる。評価は、当業者であれば目視や画像解析など任意の方法を選択することができる。
Steps (iii) and (iv) are as described in detail in the method for producing a sheet-shaped cell culture.
In step (v), the exfoliation state of the sheet-shaped cell culture from the surface of the culture substrate is evaluated. "Evaluating the exfoliation state" means evaluating the presence or absence of exfoliation of the sheet-like cell culture from the culture substrate and the type of exfoliation when sheet-forming culture and / or exfoliation work is performed using the coated culture substrate. Means to do. By evaluating such a peeled state, the surface of the culture medium can be evaluated. The types of exfoliation mainly include spontaneous exfoliation and exfoliation by exfoliation work, and those that can be exfoliated by exfoliation work are more likely to cause shrinkage of the sheet-like cell culture due to the intercellular binding force. It can be evaluated as a preferable surface of the culture substrate. For the evaluation, a person skilled in the art can select any method such as visual inspection or image analysis.
 本開示の別の側面は、本開示の製造方法により製造されたシート状細胞培養物に関する。本発明の一態様において、本開示の製造方法により製造されたシート状細胞培養物は、心筋細胞からなる。本発明の別の態様において、本開示の製造方法により製造されたシート状細胞培養物は、心筋細胞、線維芽細胞、血管内皮細胞および/または壁細胞を含む。
 本開示のシート状細胞培養物は、シート状細胞培養物の適用により改善される疾患、例えば、組織の異常に関連する種々の疾患の処置に有用である。したがって、一態様において、本開示のシート状細胞培養物は、シート状細胞培養物の適用により改善される疾患、特に、組織の異常に関連する疾患の処置に用いるためのものである。本開示のシート状細胞培養物は、従来のシート状細胞培養物に比べて高い機械的強度を有する以外は、これと同様の構成細胞固有の性質を有しているため、少なくとも従来の筋芽細胞または線維芽細胞を含むシート状細胞培養物による処置が可能な組織や疾患に適用することができる。処置の対象となる組織としては、限定されずに、例えば、心筋、角膜、網膜、食道、皮膚、関節、軟骨、肝臓、膵臓、歯肉、腎臓、甲状腺、骨格筋、中耳、骨髄、胃、小腸、十二指腸、大腸などの消化管などが挙げられる。また、処置の対象となる疾患としては、限定されずに、例えば、心疾患(例えば、心筋傷害(心筋梗塞、心外傷)、心筋症など)、角膜疾患(例えば、角膜上皮幹細胞疲弊症、角膜損傷(熱・化学腐食)、角膜潰瘍、角膜混濁、角膜穿孔、角膜瘢痕、スティーブンス・ジョンソン症候群、眼類天疱瘡など)、網膜疾患(例えば、網膜色素変性症、加齢黄斑変性症など)、食道疾患(例えば、食道手術(食道ガン除去)後の食道の炎症・狭窄の予防など)、皮膚疾患(例えば、皮膚損傷(外傷、熱傷)など)、関節疾患(例えば、変形性関節炎など)、軟骨疾患(例えば、軟骨の損傷など)、肝疾患(例えば、慢性肝疾患など)、膵臓疾患(例えば、糖尿病など)、歯科疾患(例えば、歯周病など)、腎臓疾患(例えば、腎不全、腎性貧血、腎性骨異栄養症など)、甲状腺疾患(例えば、甲状腺機能低下症など)、筋疾患(例えば、筋損傷、筋炎など)、中耳疾患(例えば、中耳炎など)、骨髄疾患(例えば、白血病、再生不良性貧血、免疫不全疾患など)が挙げられる。本開示のシート状細胞培養物が上記疾患に有用であることは、例えば、特許文献1、非特許文献1、Tanaka et al., J Gastroenterol. 2013;48(9):1081-9.などに記載されている。本開示のシート状細胞培養物は、注射可能な大きさに断片化し、これを処置が必要な部位に注射することで、単細胞懸濁液による注射よりも高い効果を得ることもできる(Wang et al., Cardiovasc Res. 2008;77(3):515-24)。したがって、本開示のシート状細胞培養物についても、このような利用法が可能である。
Another aspect of the present disclosure relates to sheet cell cultures produced by the production methods of the present disclosure. In one aspect of the invention, the sheet cell culture produced by the production method of the present disclosure comprises cardiomyocytes. In another aspect of the invention, the sheet cell culture produced by the production method of the present disclosure comprises cardiomyocytes, fibroblasts, vascular endothelial cells and / or parietal cells.
The sheet cell cultures of the present disclosure are useful in treating diseases that are ameliorated by the application of sheet cell cultures, such as various diseases associated with tissue abnormalities. Therefore, in one aspect, the sheet cell cultures of the present disclosure are intended for use in the treatment of diseases that are ameliorated by the application of sheet cell cultures, particularly those associated with tissue abnormalities. The sheet-shaped cell cultures of the present disclosure have similar properties peculiar to constituent cells except that they have higher mechanical strength than conventional sheet-shaped cell cultures, and therefore at least conventional myoblasts. It can be applied to tissues and diseases that can be treated with sheet-like cell cultures containing cells or fibroblasts. The tissues to be treated include, for example, myocardium, cornea, retina, esophagus, skin, joints, cartilage, liver, pancreas, gingiva, kidney, thyroid, skeletal muscle, middle ear, bone marrow, stomach, etc. Examples include the gastrointestinal tract such as the small intestine, duodenum, and large intestine. In addition, the diseases to be treated are not limited, for example, heart diseases (for example, myocardial injury (myocardial infarction, cardiac trauma), cardiomyopathy, etc.), corneal diseases (for example, corneal epithelial stem cell exhaustion, corneal membrane). Injury (heat / chemical corrosion), corneal ulcer, corneal opacification, corneal perforation, corneal scar, Stevens Johnson syndrome, ocular herbitis, etc.), retinal disease (eg, retinal pigment degeneration, age-related yellow spot degeneration, etc.) , Esophageal disease (eg, prevention of esophageal inflammation / stenosis after esophageal surgery (removal of esophageal cancer)), skin disease (eg, skin injury (trauma, burn), etc.), joint disease (eg, osteoarthritis) , Cartilage disease (eg, cartilage damage), Liver disease (eg, chronic liver disease), Pancreatic disease (eg, diabetes), Dental disease (eg, periodontal disease, etc.), Kidney disease (eg, renal failure) , Renal anemia, renal osteodystrophy, etc.), thyroid disease (eg, hypothyroidism), muscle disease (eg, muscle injury, myitis, etc.), middle ear disease (eg, middle ear inflammation, etc.), bone marrow disease (For example, leukemia, poor regeneration anemia, immunodeficiency disease, etc.). The usefulness of the sheet-shaped cell culture of the present disclosure for the above-mentioned diseases is described in, for example, Patent Document 1, Non-Patent Document 1, Tanaka et al., J Gastroenterol. 2013; 48 (9): 1081-9. Are listed. The sheet-like cell cultures of the present disclosure can also be fragmented to an injectable size and injected at the site requiring treatment for greater efficacy than injection with a single cell suspension (Wang et. al., Cardiovasc Res. 2008; 77 (3): 515-24). Therefore, such a use is also possible for the sheet-shaped cell culture of the present disclosure.
 本開示の別の側面は、本開示の方法により製造された移植片の有効量を、それを必要とする対象に適用することを含む、前記対象における疾患を処置する方法に関する。処置の対象となる疾患は、上記したとおりである。 Another aspect of the present disclosure relates to a method of treating a disease in a subject in need thereof, including applying an effective amount of the implant produced by the method of the present disclosure to a subject in need thereof. The diseases to be treated are as described above.
 本開示において、用語「処置」は、疾患の治癒、一時的寛解または予防などを目的とする医学的に許容される全ての種類の予防的および/または治療的介入を包含するものとする。例えば、「処置」の用語は、組織の異常に関連する疾患の進行の遅延または停止、病変の退縮または消失、当該疾患発症の予防または再発の防止などを含む、種々の目的の医学的に許容される介入を包含する。 In the present disclosure, the term "treatment" shall include all types of medically acceptable prophylactic and / or therapeutic interventions aimed at the cure, temporary remission or prevention of disease, etc. For example, the term "treatment" is medically acceptable for a variety of purposes, including delaying or stopping the progression of a disease associated with a tissue abnormality, regressing or eliminating a lesion, preventing the onset or recurrence of the disease, and the like. Including interventions to be performed.
 本開示の処置方法においては、移植片の生存性、生着性および/または機能などを高める成分や、対象疾患の処置に有用な他の有効成分などを、本開示の移植片等と併用することができる。 In the treatment method of the present disclosure, an ingredient that enhances the viability, engraftment and / or function of the implant, other active ingredients useful for treating the target disease, etc. are used in combination with the implant of the present disclosure. be able to.
 本開示の処置方法は、本開示の製造方法に従って、本開示の移植片を製造するステップをさらに含んでもよい。本開示の処置方法は、移植片を製造するステップの前に、対象から移植片を製造するための細胞(iPS細胞を用いる場合は、例えば、皮膚細胞、血球等)または細胞の供給源となる組織(iPS細胞を用いる場合は、例えば、皮膚組織、血液等)を採取するステップをさらに含んでもよい。一態様において、細胞または細胞の供給源となる組織を採取する対象は、細胞培養物、組成物、または移植片等の投与を受ける対象と同一の個体である。別の態様において、細胞または細胞の供給源となる組織を採取する対象は、細胞培養物、組成物、または移植片等の投与を受ける対象とは同種の別個体である。別の態様において、細胞または細胞の供給源となる組織を採取する対象は、移植片等の投与を受ける対象とは異種の個体である。 The treatment method of the present disclosure may further include the step of producing the implant of the present disclosure according to the production method of the present disclosure. The treatment method of the present disclosure serves as a cell (for example, skin cells, blood cells, etc. when using iPS cells) or a source of cells for producing a graft from a subject before the step of producing the graft. It may further include the step of collecting tissue (for example, skin tissue, blood, etc. when using iPS cells). In one embodiment, the subject from which the cell or tissue from which the cell is source is collected is the same individual as the subject to whom the cell culture, composition, implant, or the like is administered. In another embodiment, the subject from which the cell or tissue from which the cell is sourced is harvested is a separate entity of the same species as the subject receiving the administration, such as a cell culture, composition, or implant. In another embodiment, the subject from which the cell or tissue that is the source of the cell is collected is an individual different from the subject to which the implant or the like is administered.
 本開示において、有効量とは、例えば、疾患の発症や再発を抑制し、症状を軽減し、または進行を遅延もしくは停止し得る量(例えば、シート状細胞培養物のサイズ、重量、枚数等)であり、好ましくは、当該疾患の発症および再発を予防し、または当該疾患を治癒する量である。また、投与による利益を超える悪影響が生じない量が好ましい。かかる量は、例えば、マウス、ラット、イヌまたはブタなどの実験動物や疾患モデル動物における試験などにより適宜決定することができ、このような試験法は当業者によく知られている。また、処置の対象となる組織病変の大きさは、有効量決定のための重要な指標となり得る。 In the present disclosure, the effective amount is, for example, an amount capable of suppressing the onset or recurrence of a disease, reducing symptoms, or delaying or stopping the progression (for example, size, weight, number of sheet-like cell cultures, etc.). The amount is preferably an amount that prevents the onset and recurrence of the disease or cures the disease. In addition, an amount that does not cause an adverse effect exceeding the benefit of administration is preferable. Such an amount can be appropriately determined by, for example, a test in an experimental animal such as a mouse, a rat, a dog or a pig, or a disease model animal, and such a test method is well known to those skilled in the art. In addition, the size of the tissue lesion to be treated can be an important index for determining the effective amount.
 投与方法としては、例えば、静脈投与、筋肉内投与、骨内投与、髄腔内投与、組織への直接的な適用などが挙げられる。投与頻度は、典型的には1回の処置につき1回であるが、所望の効果が得られない場合には、複数回投与することも可能である。組織に適用する際、本発明の細胞培養物、組成物、またはシート状細胞培養物等を対象の組織に縫合糸やステープルなどの係止手段により固定してもよい。 Examples of the administration method include intravenous administration, intramuscular administration, intraosseous administration, intrathecal administration, and direct application to tissues. The frequency of administration is typically once per treatment, but multiple doses can be administered if the desired effect is not obtained. When applied to a tissue, the cell culture, composition, sheet-like cell culture or the like of the present invention may be fixed to the target tissue by a locking means such as a suture or a staple.
 本発明を以下の例を参照してより詳細に説明するが、これらは本発明の特定の具体例を示すものであり、本発明はこれらに限定されるものではない。 The present invention will be described in more detail with reference to the following examples, but these show specific specific examples of the present invention, and the present invention is not limited thereto.
 以下の実施例において、多能性幹細胞として、京都大学iPS細胞研究所(CiRA)で樹立された臨床用ヒトiPS細胞を用いた。M. Nakagawa et al., Scientific Reports, 4:3594 (2014)を参考に、ヒトiPS細胞をフィーダーフリー法で維持した。ついで、Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358およびWO2017/038562の記載を参考にして、ヒトiPS細胞を心筋細胞へと分化誘導して胚様体を得た。具体的には、フィーダー細胞を含まない培養液で維持培養したヒトiPS細胞を、EZ Sphere(旭硝子)上で10μMのY27632(和光純薬)を含有するStemFit AK03培地(味の素)中で1日培養し、得られた胚様体をアクチビンA、骨形成タンパク質(BMP)4および塩基性線維芽細胞増殖因子(bFGF)を含有する培養液中で培養し、さらにWnt阻害剤(IWP3)およびBMP4阻害剤(Dorsomorphin)およびTGFβ阻害剤(SB431542)を含む培養液中で培養し、その後VEGFおよびbFGFを含む培養液中で培養を行うことで、iPS細胞由来のヒト心筋細胞を得た。得られた細胞集団における心筋細胞の割合は50%~90%であった。 In the following examples, clinical human iPS cells established at the Center for iPS Cell Research and Application (CiRA), Kyoto University were used as pluripotent stem cells. Human iPS cells were maintained by the feeder-free method with reference to M. Nakagawa et al., Scientific Reports, 4: 3594 (2014). Then, referring to the description of Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015, WO2014 / 185358 and WO2017 / 038562, the embryoid body is induced to differentiate human iPS cells into cardiomyocytes. Got Specifically, human iPS cells maintained and cultured in a culture medium containing no feeder cells are cultured on EZSphere (Asahi Glass) for one day in StemFit AK03 medium (Ajinomoto) containing 10 μM Y27632 (Wako Pure Chemical Industries). Then, the obtained embryo-like body was cultured in a culture medium containing Actibin A, bone-forming protein (BMP) 4, and basic fibroblast growth factor (bFGF), and further, Wnt inhibitor (IWP3) and BMP4 inhibition were performed. Human myocardial cells derived from iPS cells were obtained by culturing in a culture medium containing a drug (Dorsomorphin) and a TGFβ inhibitor (SB431542) and then in a culture medium containing VEGF and bFGF. The proportion of cardiomyocytes in the resulting cell population ranged from 50% to 90%.
例1.FBS含有培地とPL含有培地との比較
 上記で得られた、ヒトiPS細胞から分化誘導した心筋細胞を含む細胞集団を用い、シート化培養条件を検討した。DMEM/F12培地に20%FBSまたは5%ヒト血小板溶解物をそれぞれ加えたものをシート化媒体とした。血小板溶解物を加えたシート化媒体には、さらに細胞接着性成分としてラミニン(iMatrix-511)をそれぞれ0.1μg/mL、0.25μg/mLまたは0.5μg/mL加えた。また、シート化培養の1日目のみ、シート化媒体にRhoキナーゼ阻害剤Y27632を加えた。心筋細胞を含む細胞集団は、1.5×10個/cmの密度で温度応答性培養皿(UpCell(R)、セルシード)に播種し、37℃、5%COの環境で3日間培養した。温度応答性培養皿は、培養液と同じもの(ただしY27632は含まない)を入れて、37℃で一晩インキュベートしてコーティングしたものを用いた。培養の後、培養皿から心筋細胞を含むシート状細胞培養物を剥離した。
Example 1. Comparison between FBS-containing medium and PL-containing medium Using the cell population obtained above containing cardiomyocytes induced to differentiate from human iPS cells, the sheeting culture conditions were examined. 20% FBS or 5% human platelet lysate was added to DMEM / F12 medium as a sheeting medium. Laminin (iMatrix-511) as a cell adhesion component was further added to the sheeting medium containing the platelet lysate at 0.1 μg / mL, 0.25 μg / mL or 0.5 μg / mL, respectively. In addition, the Rho-kinase inhibitor Y27632 was added to the sheeting medium only on the first day of the sheeting culture. The cell population containing cardiomyocytes was seeded in a temperature-responsive culture dish (UpCell (R) , CellSeed) at a density of 1.5 × 10 6 cells / cm 2 and placed in an environment of 37 ° C. and 5% CO 2 for 3 days. It was cultured. As the temperature-responsive culture dish, the same one as the culture solution (however, Y27632 was not included) was put therein, and the culture dish was incubated overnight at 37 ° C. and coated. After culturing, the sheet-shaped cell culture containing cardiomyocytes was exfoliated from the culture dish.
 結果を下表に示す。
Figure JPOXMLDOC01-appb-T000001
 FBSを入れたシート化媒体を用いた場合は、3日間シート化培養した時点では、拍動が観察されなかったり、また観察されても弱い拍動であったのに対し、ラミニンおよび血小板溶解物を入れたシート化媒体を用いたものは、いずれも3日間の培養で強い拍動が観察された。
The results are shown in the table below.
Figure JPOXMLDOC01-appb-T000001
When a sheeted medium containing FBS was used, no pulsation was observed at the time of sheeting and culturing for 3 days, and even if it was observed, the pulsation was weak, whereas laminin and platelet lysate were observed. Strong pulsation was observed in all the media using the sheet-containing medium containing the above, after culturing for 3 days.
例2.無血清培地でのシート化試験
 例1と同様に、DMEM/F12培地に20%FBSを加えた標準的な培地と、血液由来成分を加えずラミニン(iMatrix-511)を0.1μg/mL加えた培地(E6)とをそれぞれシート化媒体として、シート化培養を行った。
 結果を下表に示す。
Figure JPOXMLDOC01-appb-T000002
Example 2. Sheeting in serum-free medium Similar to Test Example 1, 0.1 μg / mL of standard medium with 20% FBS added to DMEM / F12 medium and laminin (iMatrix-511) without adding blood-derived components was added. Sheet culture was performed using each of the medium (E6) as a sheet medium.
The results are shown in the table below.
Figure JPOXMLDOC01-appb-T000002
 ラミニンを含有する無血清培地(血液由来成分を含まない培地)でシート化した場合、培養2日目までは拍動が観察されなかったものの、3日目には自律拍動が観察されシート化することができた。培地に血清や血小板溶解物などの血液由来成分を含まなくとも細胞接着性成分(ラミニン)を含有することで自立拍動する心筋細胞シート状細胞培養物を得ることができることが明らかとなった。 When sheeted with a serum-free medium containing laminin (medium containing no blood-derived components), pulsation was not observed until the second day of culture, but autonomous pulsation was observed and sheeted on the third day. We were able to. It was clarified that a cardiomyocyte sheet-like cell culture that beats independently can be obtained by containing a cell adhesion component (laminin) even if the medium does not contain a blood-derived component such as serum or platelet lysate.
例3.細胞接着性成分の検討
 ラミニン以外の細胞接着性成分を用いた場合であってもシート化が可能であるか否かを調べるため、細胞接着性成分として、ラミニン(iMatrix-511)、ビトロネクチン(VTN-N)およびフィブロネクチン(RetroNectin(R))(タカラバイオ)を用いてシート化培養を行った。iMatrix-511、VTN-NおよびRetroNectin(R)を培養基材のコーティングに用いる場合の推奨濃度はそれぞれ0.5μg/cm、0.5μg/cmおよび8μg/cmであり、シート化培養においては、推奨濃度および推奨濃度の1/10の濃度を用いてシート化培養を行った。例2で用いたE6培地のラミニン0.1μg/cmに代えて上記の細胞接着性成分をそれぞれ加えて、例2と同様にシート化培養を行った。
 結果を図1に示す。いずれの培地を用いた場合であってもシート状細胞培養物を調製可能であった。
Example 3. Examination of cell adhesion components In order to investigate whether sheeting is possible even when cell adhesion components other than laminin are used, laminin (iMatrix-511) and vitronectin (VTN) are used as cell adhesion components. Sheeting culture was performed using -N) and fibronectin ( R) (Takarabio). When iMatrix-511, VTN-N and RetroNectin (R) are used for coating the culture substrate, the recommended concentrations are 0.5 μg / cm 2 , 0.5 μg / cm 2 and 8 μg / cm 2 , respectively. In, sheet-forming culture was carried out using the recommended concentration and a concentration of 1/10 of the recommended concentration. The above cell adhesion components were added in place of laminin 0.1 μg / cm 2 in the E6 medium used in Example 2, and sheet-forming culture was carried out in the same manner as in Example 2.
The results are shown in FIG. Sheet-shaped cell cultures could be prepared regardless of which medium was used.
例4.細胞の状態と至適濃度の検討
 細胞の活性に影響を与え得る未分化細胞除去の工程を行った場合に細胞接着性成分がどのように影響するかを調べた。細胞の状態によってシート化培養に用い得る細胞接着性成分の至適濃度が変化するかどうかを確認するため、3種類の細胞を用いて検討した。高活性の細胞として、上記で得られたヒトiPS細胞から分化誘導した心筋細胞を含む細胞集団(細胞の凍結解凍の工程なし、未分化細胞除去の工程なし)を、中活性の細胞として、上記で得られたヒトiPS細胞から分化誘導した心筋細胞を含む細胞集団を一度凍結し、解凍したものを、低活性の細胞として、上記で得られたヒトiPS細胞から分化誘導した心筋細胞を含む細胞集団から未分化細胞の除去処理を行い、その後凍結し、解凍したものを用いた。未分化細胞の除去処理は、WO2017/038562に記載の熱処理、WO2007/088874に記載の無糖培地培養法、WO2016/072519に記載の抗CD30抗体結合薬剤処理を順に用いて行なった。
Example 4. Examination of cell condition and optimum concentration We investigated how the cell adhesion component affects the step of removing undifferentiated cells that can affect the activity of cells. In order to confirm whether the optimum concentration of the cell adhesion component that can be used for sheeting culture changes depending on the state of the cells, three types of cells were used for examination. As the highly active cells, a cell population containing myocardial cells induced to differentiate from the human iPS cells obtained above (no step of freezing and thawing the cells, no step of removing undifferentiated cells) was used as the above-mentioned medium-active cells. The cell population containing the myocardial cells induced to differentiate from the human iPS cells obtained in 1 above was once frozen and thawed, and the cells containing the myocardial cells induced to differentiate from the human iPS cells obtained above were regarded as low-activity cells. Undifferentiated cells were removed from the population, then frozen and thawed. The treatment for removing undifferentiated cells was carried out in order by using the heat treatment described in WO2017 / 038562, the sugar-free medium culture method described in WO2007 / 0888874, and the anti-CD30 antibody-binding drug treatment described in WO2016 / 072519.
 上記3種の細胞を用い、細胞接着性成分として、iMatrix-511を推奨濃度の1/10の濃度加えたE6培地を用いて、例2と同様にシート化培養を行った。
 その結果、高活性の細胞を用いた場合は、シート化できたが、細胞間接着力が強くなりシート状細胞培養物が自然剥離してしまった。中活性の細胞を用いた場合は、シート化でき、剥離作業により剥離可能であった。低活性の細胞を用いた場合は、剥離作業を行うと細胞間接着力が弱く、シートとして剥離することができなかった。そこで、低活性の細胞を用いて、iMatrix-511を推奨濃度の1/100の濃度加えたE6培地を用いて同様にシート化培養してみたところ、シート化でき、剥離作業により剥離可能であった。
Using the above three types of cells, sheet culture was carried out in the same manner as in Example 2 using E6 medium to which iMatrix-511 was added at a concentration of 1/10 of the recommended concentration as a cell adhesion component.
As a result, when highly active cells were used, the cells could be formed into sheets, but the cell-cell adhesion became stronger and the sheet-like cell cultures spontaneously exfoliated. When medium-active cells were used, they could be made into sheets and could be peeled off by peeling work. When low-activity cells were used, the cell-cell adhesion was weak when the peeling operation was performed, and the cells could not be peeled as a sheet. Therefore, when iMatrix-511 was similarly sheeted and cultured using E6 medium to which 1/100 of the recommended concentration was added using low-activity cells, it was possible to form a sheet and peel it off by peeling work. It was.
 以上のことから、細胞の活性により細胞間接着力が変化するため、それに合わせた濃度の細胞接着性成分を添加する必要があることがわかった。すなわち、凍結・解凍の工程や未分化細胞除去の工程を行う場合、細胞の活性が低下して細胞間接着力が低下するため、細胞接着性成分を補うことが必要であることが明らかとなった。 From the above, it was found that the cell-cell adhesion force changes depending on the cell activity, so it is necessary to add a cell adhesion component at a concentration corresponding to that. That is, it was clarified that it is necessary to supplement the cell adhesion component because the cell activity is reduced and the cell-cell adhesion is reduced when the freezing / thawing step and the step of removing undifferentiated cells are performed. ..
例5.細胞接着性成分およびマスキング成分によるコーティングの検討
 細胞培養皿を細胞接着性成分とともにFBSなどのマスキング成分でコーティングした際にシート状細胞培養物の剥離状態がどのように変化するか調べた
 DMEM/F12に0%、0.31%、0.63%、1.25%、2.5%、5%、10%、20%のFBSおよび0.076μg/cm、0.76μg/cm、7.6μg/cm、76μg/cmのiMatrix-511を加えたコーティング溶液計40種類を作成し、これを温度応答性培養皿の各ウェルに添加し37℃で一晩インキュベートしてコーティングした。コーティング溶液除去後、培養皿にHBSS(Invitrogen製)を1mL添加し、20秒間ゆっくりと撹拌洗浄を行った後ピペッティングにより廃棄し、これを2回繰り返し行った。例4の未分化細胞の除去処理を行なった低活性細胞を用いてiMatrix-511を推奨濃度の1/10の濃度加えたE6培地を用いてコーティングした培養皿を用いて例2と同様にシート化培養を行った。
Example 5. Examination of coating with cell adhesion component and masking component DMEM / F12 investigated how the peeling state of sheet-shaped cell culture changes when the cell culture dish is coated with a masking component such as FBS together with the cell adhesion component. 0%, 0.31%, 0.63%, 1.25%, 2.5%, 5%, 10%, 20% FBS and 0.076 μg / cm 2 , 0.76 μg / cm 2 , 7 A total of 40 coating solutions were prepared with .6 μg / cm 2 and 76 μg / cm 2 of iMatrix-511, which were added to each well of a temperature responsive culture dish and incubated overnight at 37 ° C. for coating. After removing the coating solution, 1 mL of HBSS (manufactured by Invitrogen) was added to the culture dish, and the mixture was slowly stirred and washed for 20 seconds and then discarded by pipetting, and this was repeated twice. Sheets similar to Example 2 using a culture dish coated with E6 medium containing iMatrix-511 at a concentration of 1/10 of the recommended concentration using low-activity cells treated to remove undifferentiated cells of Example 4. Chemical culture was performed.
 培養液を除去し、HBSS(+)を加え、室温で1時間30分間静置し、剥離作業を行なった。シート状細胞培養物の剥離状態を表3および図2に示す。表3の無色セルの「剥離」は自然剥離を示し、灰色セルの「剥離」は剥離作業によって剥離されたものを示す。×は剥離されなかったものを示す。 The culture solution was removed, HBSS (+) was added, and the mixture was allowed to stand at room temperature for 1 hour and 30 minutes for peeling work. The exfoliated state of the sheet-shaped cell culture is shown in Table 3 and FIG. “Peeling” of the colorless cells in Table 3 indicates spontaneous peeling, and “peeling” of the gray cells indicates peeling by the peeling operation. X indicates that the material was not peeled off.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
 FBSなどのマスキング成分を含むことで高濃度の細胞接着性成分を含む溶液でコーティングしても剥離可能であることが明らかとなった。これは細胞接着性成分による培養皿表面の細胞接着作用がマスキング成分により低減されたものと推測される。例4において低活性細胞は、細胞接着性成分を添加しなければ剥離できなかったが、細胞接着性成分およびマスキング成分のコーティングにより剥離可能となった。したがって、当該コーティングにより細胞の活性に応じた濃度の細胞接着性成分を補うことなくシート状細胞培養物を剥離することが出来ることが明らかとなった。さらにマスキング成分と細胞接着性成分の濃度調整をすることで自然剥離を抑え、剥離作業によってはじめて剥離可能となるようにシート状細胞培養物の剥離状態をコントロールできることが明らかとなった。 It was clarified that by containing a masking component such as FBS, it can be peeled off even if it is coated with a solution containing a high concentration of cell adhesion component. It is presumed that this is because the cell adhesion action on the surface of the culture dish due to the cell adhesion component was reduced by the masking component. In Example 4, the low-activity cells could not be exfoliated without the addition of the cell adhesion component, but could be exfoliated by coating the cell adhesion component and the masking component. Therefore, it was clarified that the sheet-like cell culture can be exfoliated without supplementing the cell adhesion component at a concentration corresponding to the cell activity by the coating. Furthermore, it was clarified that the peeling state of the sheet-shaped cell culture can be controlled so that the natural peeling can be suppressed by adjusting the concentrations of the masking component and the cell adhesion component, and the peeling can be performed only by the peeling operation.
 本発明により、多能性幹細胞から分化誘導した細胞等を用いてシート状細胞培養物を形成する際に、高品質なシート状細胞培養物を得ることができる。特に臨床用に用いるゼノフリーなシート状細胞培養物の製造においても、高品質なシート状細胞培養物を簡便に形成することが可能となる。 According to the present invention, it is possible to obtain a high-quality sheet-shaped cell culture when forming a sheet-shaped cell culture using cells or the like induced to differentiate from pluripotent stem cells. In particular, even in the production of a xeno-free sheet-shaped cell culture used for clinical use, it becomes possible to easily form a high-quality sheet-shaped cell culture.

Claims (16)

  1.  体細胞を含む移植片を製造する方法であって;
    (a)前記体細胞を含む細胞集団を、培養基材上に播種する工程、および
    (b)播種した細胞集団を、細胞接着性成分を含む移植片形成媒体で移植片形成培養する工程、
    を含む、前記方法。
    A method of producing a graft containing somatic cells;
    (A) a step of seeding the cell population containing the somatic cells on a culture substrate, and (b) a step of culturing the seeded cell population in a graft-forming medium containing a cell adhesion component.
    The method described above.
  2.  培養基材が、さらに細胞接着性成分および/またはマスキング成分でコーティングされている、請求項1に記載の方法。 The method according to claim 1, wherein the culture substrate is further coated with a cell adhesion component and / or a masking component.
  3.  移植片形成媒体中の細胞接着性成分の含有量が、培養基材のコーティングに用いる量の1/10~1/100の濃度である、請求項1または2に記載の方法。 The method according to claim 1 or 2, wherein the content of the cell adhesion component in the graft-forming medium is 1/10 to 1/100 of the amount used for coating the culture substrate.
  4.  (a)の前に、細胞集団から未分化細胞を除去する工程を含む、請求項1~3のいずれか一項に記載の方法。 The method according to any one of claims 1 to 3, which comprises a step of removing undifferentiated cells from the cell population before (a).
  5.  移植片形成媒体が、血清を含まない、請求項1~4のいずれか一項に記載の方法。 The method according to any one of claims 1 to 4, wherein the graft-forming medium does not contain serum.
  6.  移植片形成媒体が、血小板溶解物を含む、請求項1~5のいずれか一項に記載の方法。 The method according to any one of claims 1 to 5, wherein the graft-forming medium contains a platelet lysate.
  7.  心筋細胞を含むシート状細胞培養物を製造する方法であって;
    (A)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で心筋細胞を播種する工程、および
    (B)シート化形成媒体でシート形成培養し、シート状細胞培養物を形成する工程、
    を含む、シート状細胞培養物の製造方法。
    A method for producing sheet-like cell cultures containing cardiomyocytes;
    (A) A step of seeding cardiomyocytes at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component, and (B) sheet-forming culture using a sheet-forming medium, and sheet-like cell culture. The process of forming things,
    A method for producing a sheet-shaped cell culture, including.
  8.  シート化形成媒体が細胞接着性成分を含む、または含まない、請求項7に記載の方法。 The method according to claim 7, wherein the sheet formation medium contains or does not contain a cell adhesive component.
  9.  シート状細胞培養物を製造するための培養基材表面を評価する方法であって、
    (i)細胞接着性成分で培養基材表面をコーティングする工程、それにより細胞の培養基材表面への細胞の接着性を向上させ、
    (ii)さらにマスキング成分で培養基材表面をコーティングする工程、それにより培養基材表面への細胞の接着性を低下させ、
    (iii)細胞接着性成分およびマスキング成分でコーティングされた培養基材上に、コンフルエントに達する密度で体細胞を播種する工程、
    (iv)シート化媒体中でシート化培養し、シート状細胞培養物を形成する工程、および
    (v)シート状細胞培養物の培養基材表面からの剥離状態を評価する工程、
    を含む、前記方法。
    A method for evaluating the surface of a culture medium for producing a sheet-like cell culture.
    (I) The step of coating the surface of the culture substrate with the cell adhesion component, thereby improving the adhesion of the cells to the surface of the culture substrate.
    (Ii) Further, a step of coating the surface of the culture substrate with a masking component, thereby reducing the adhesion of cells to the surface of the culture substrate,
    (Iii) A step of seeding somatic cells at a density reaching confluence on a culture substrate coated with a cell adhesion component and a masking component.
    (Iv) A step of forming a sheet-like cell culture by sheet-forming culture in a sheet-forming medium, and (v) a step of evaluating the peeling state of the sheet-like cell culture from the surface of the culture substrate.
    The method described above.
  10.  シート化媒体が、血清を含まない、請求項7~9のいずれか一項に記載の方法。 The method according to any one of claims 7 to 9, wherein the sheeting medium does not contain serum.
  11.  シート化媒体が、血小板溶解物を含む、請求項7~10のいずれか一項に記載の方法。 The method according to any one of claims 7 to 10, wherein the sheeting medium contains a platelet lysate.
  12.  マスキング成分が、血液由来成分である、請求項1~11のいずれか一項に記載の方法。 The method according to any one of claims 1 to 11, wherein the masking component is a blood-derived component.
  13.  マスキング成分が、血清、血小板溶解物またはアルブミンである、請求項1~12のいずれか一項に記載の方法。 The method according to any one of claims 1 to 12, wherein the masking component is serum, platelet lysate or albumin.
  14.  マスキング成分が、0.05%以上の濃度のFBSである、請求項1~13のいずれか一項に記載の方法。 The method according to any one of claims 1 to 13, wherein the masking component is FBS having a concentration of 0.05% or more.
  15.  体細胞が、多能性幹細胞由来の心筋細胞である、請求項1~14のいずれか一項に記載の方法。 The method according to any one of claims 1 to 14, wherein the somatic cell is a cardiomyocyte derived from a pluripotent stem cell.
  16.  請求項1~6のいずれか一項に記載の方法によって製造された移植片または請求項7もしくは8に記載の方法によって製造されたシート状細胞培養物の治療有効量を、それを必要とする対象に投与する工程を含む、前記対象における心疾患の治療方法。 A therapeutically effective amount of the implant produced by the method according to any one of claims 1 to 6 or the sheet cell culture produced by the method according to claim 7 or 8 is required. A method of treating a heart disease in a subject, comprising the step of administering to the subject.
PCT/JP2020/037084 2019-09-30 2020-09-30 Method for forming sheet of cardiomyocytes WO2021065984A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2021551353A JPWO2021065984A1 (en) 2019-09-30 2020-09-30

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2019-179085 2019-09-30
JP2019179085 2019-09-30

Publications (1)

Publication Number Publication Date
WO2021065984A1 true WO2021065984A1 (en) 2021-04-08

Family

ID=75336980

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2020/037084 WO2021065984A1 (en) 2019-09-30 2020-09-30 Method for forming sheet of cardiomyocytes

Country Status (2)

Country Link
JP (1) JPWO2021065984A1 (en)
WO (1) WO2021065984A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112272699A (en) * 2018-07-06 2021-01-26 株式会社迈傲锐治 Method for producing cell sheet, myocardial cell sheet, and kit for producing myocardial cell sheet

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016076368A1 (en) * 2014-11-12 2016-05-19 テルモ株式会社 Myocardial cell sheet

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016076368A1 (en) * 2014-11-12 2016-05-19 テルモ株式会社 Myocardial cell sheet

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BURNOUF, T. ET AL.: "Human platelet lysate: Replacing fetal bovine serum as a gold standard for human cell propagation", BIOMATERIALS, vol. 76, 2016, pages 371 - 387, XP029317322, DOI: 10.1016/j.biomaterials.2015.10.065 *
SAPORITO, FRANCESCA, SANDRI GIUSEPPINA; BONFERONI MARIA CRISTINA; ROSSI SILVIA; MALAVASI LORENZO; FANTE CLAUDIA DEL; VIGANI BARBAR: "Electrospun gelatin-chondroitin sulfate scaffolds loaded with platelet lysate promote immature cardiomyocyte proliferation", POLYMERS, vol. 10, no. 208, 21 February 2018 (2018-02-21), pages 1 - 20, XP055701574, DOI: 10.3390/polym10020208 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112272699A (en) * 2018-07-06 2021-01-26 株式会社迈傲锐治 Method for producing cell sheet, myocardial cell sheet, and kit for producing myocardial cell sheet

Also Published As

Publication number Publication date
JPWO2021065984A1 (en) 2021-04-08

Similar Documents

Publication Publication Date Title
Baldwin et al. In vitro pre-vascularisation of tissue-engineered constructs A co-culture perspective
JP6599095B2 (en) Method for producing sheet cell culture
JP2022105168A (en) Method for modifying cell culture in adhered state
Poornejad et al. Current cell-based strategies for whole kidney regeneration
JP7256818B2 (en) Sheeting method of cardiomyocytes
WO2021065984A1 (en) Method for forming sheet of cardiomyocytes
JP2023175787A (en) Method for forming pluripotent stem cell-derived cell into sheet
US20200109368A1 (en) Method for preparing differentiation-induced cells
WO2020067443A1 (en) Somatic cell sheet-forming method
WO2020067436A1 (en) Method for forming graft of pluripotent stem cell-derived cells
JP2021052661A (en) Buffer for diluting cryopreserved cell including pluripotent stem cell
JP7471069B2 (en) Methods for enhancing graft activity
WO2021065976A1 (en) Method for enhancing activity in graft
WO2021065989A1 (en) Method for increasing proportion of cd56+ cells
WO2021065971A1 (en) Dilution buffer solution for cryopreserved cells
JP2021052662A (en) Method for increasing activity of graft
EP3739040B1 (en) Sheeting method for pluripotent stem cell-derived cells
US20200390936A1 (en) Method for cryopreserving cells
JP2021132597A (en) Methods for producing cell culture comprising cd56 positive cells
WO2020067434A1 (en) Method for cryopreserving cells
JP2016096732A (en) Production method of sheet-like cell culture
JPWO2019177135A1 (en) Method for producing sheet-shaped cell culture
JPWO2019177146A1 (en) Method for producing sheet-shaped cell culture
Soong Development of a novel technology to engineer heart muscle for contractile and paracrine support in heart failure
Smith Engineering Poly (ethylene glycol) Materials to Promote Cardiogenesis

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: 20870897

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021551353

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20870897

Country of ref document: EP

Kind code of ref document: A1