WO2014133090A1 - Pluripotent stem cell for treating brain tumor - Google Patents

Pluripotent stem cell for treating brain tumor Download PDF

Info

Publication number
WO2014133090A1
WO2014133090A1 PCT/JP2014/054910 JP2014054910W WO2014133090A1 WO 2014133090 A1 WO2014133090 A1 WO 2014133090A1 JP 2014054910 W JP2014054910 W JP 2014054910W WO 2014133090 A1 WO2014133090 A1 WO 2014133090A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
gene
negative
cell
cell preparation
Prior art date
Application number
PCT/JP2014/054910
Other languages
French (fr)
Japanese (ja)
Inventor
宏樹 難波
真理 出澤
正順 吉田
Original Assignee
国立大学法人浜松医科大学
国立大学法人東北大学
株式会社Clio
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 国立大学法人浜松医科大学, 国立大学法人東北大学, 株式会社Clio filed Critical 国立大学法人浜松医科大学
Priority to JP2015503024A priority Critical patent/JPWO2014133090A1/en
Publication of WO2014133090A1 publication Critical patent/WO2014133090A1/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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0695Stem cells; Progenitor cells; Precursor cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to a cell preparation containing pluripotent stem cells into which a suicide gene effective for treatment of brain tumors is introduced.
  • Glioma (glioma), which accounts for about 1 ⁇ 4 of brain tumors, grows infiltrating into the surrounding normal brain tissue, so that if the brain function is to be preserved, it is difficult to remove it by surgery. Furthermore, radiation therapy and anticancer drug treatment are often used after surgery, but the cure rate is low, and the average malignant glioblastoma (glioblastoma) has an average survival time of only about 1 year. Moreover, despite the development of various treatment strategies, this result has hardly changed for the past 30 years and is said to be one of the last malignant tumors left in the 21st century, and the development of new treatment strategies is urgently needed. It is.
  • suicide gene therapy introduced in the 1990s is a therapy that uses genes such as viruses to change harmful drugs in mammals into toxic substances (anticancer drugs) in transgenic cells and kill cancer cells. is there.
  • suicide gene therapy using the herpes simplex virus thymidine kinase gene (HSVtk gene) and the antiviral agent ganciclovir (GCV)
  • HSVtk gene herpes simplex virus thymidine kinase gene
  • GCV antiviral agent ganciclovir
  • Non-Patent Document 2 Attempts have been made to treat brain tumors by incorporating suicide genes into such migratory neural stem cells and expressing cytotoxic substances in the tumor site (Patent Document 1).
  • Non-patent Document 3 neural stem cells are extremely effective as a vector for carrying a suicide gene to a tumor site, but when considering clinical application, it is invasive and extremely difficult to remove neural stem cells from a patient. So far, the present inventors have used a mesenchymal stem cell collected from the bone marrow as a vector carrying a suicide gene to the tumor site as a more general-purpose vector having the same mobility as that of a neural stem cell. A treatment test was attempted (Patent Document 2). However, the mesenchymal stem cells used in this treatment test were heterogeneous and the effects were not constant.
  • SSEA-3 Stage-Specific Embryonic Antigen-3
  • Muse cells Multilineage-differentiating Stress Enduring cells; Muse cells
  • An object of the present invention is to provide a new medical use using pluripotent stem cells (Muse cells) in brain tumor treatment. More specifically, an object of the present invention is to provide a cell preparation for the prevention and / or treatment of brain tumor, comprising Muse cells into which a suicide gene has been introduced and a prodrug corresponding to the suicide gene.
  • Muse cells pluripotent stem cells
  • HSVtk herpes simplex virus thymidine kinase
  • GCV ganciclovir
  • the present invention is as follows.
  • a cell preparation for treating a brain tumor comprising a pluripotent stem cell into which a suicide gene has been introduced, wherein the pluripotent stem cell is derived from a mesenchymal tissue or a cultured mesenchymal cell in a living body.
  • Isolated SSEA-3-positive cells wherein the cell preparation is used with a prodrug of a drug that kills or inhibits the growth of a brain tumor, the prodrug being a substrate for an enzyme produced by expression of the suicide gene A cell preparation.
  • pluripotent stem cells are CD34 negative, CD117 negative, CD146 negative, CD271 negative, NG2 negative, vWF negative, Sox10 negative, Snai1 negative, Slug negative, Tyrp1 negative, and Dct negative.
  • the prodrug is gancyclovir (GCV), acyclovir, penciclovir, PMEA adefovir or PMPA tenofovir when the suicide gene is an HSVtk gene, and 5-fluorocytosine when the suicide gene is a cytosine deaminase gene, 5-fluorouracil when the suicide gene is a uracil phosphoribosyltransferase gene, 6-thioxanthine or 6-thioguanine when the suicide gene is a gpt gene, and a prodrug when the suicide gene is a nitroreductase (ntr) gene
  • GCV gancyclovir
  • penciclovir penciclovir
  • PMEA adefovir or PMPA tenofovir when the suicide gene is an HSVtk gene
  • 5-fluorocytosine when the suicide gene is a cytosine deaminase gene
  • 5-fluorouracil when the
  • the present invention relates to the expression of a suicide gene by administering a suicide gene-introduced Muse cell and a prodrug for the suicide gene from a vein or the like to a subject suffering from a brain tumor, thereby accumulating the Muse cell in the brain tumor. Based on the activation of the prodrug, tumor cells can be killed or their growth can be suppressed.
  • FIG. 2 shows the results of observing the culture state of glioma cells over time when glioma cells and Muse-TK are co-cultured at different cell ratios.
  • FIG. 3 is a diagram showing the decrease in glioma cells by counting the number of cells based on the results obtained in FIG. The result of having measured the migration ability of the Muse cell with respect to a glioma cell using the Boyden chamber is shown. The result of having verified the bystander effect by Muse-TK in vivo is shown.
  • the present invention relates to a cell preparation for treating a brain tumor, comprising pluripotent stem cells (Muse cells) into which a suicide gene has been introduced.
  • Muse cells pluripotent stem cells
  • the present invention aims at treatment of brain tumors, in particular, treatment of killing glioma cells or inhibiting their growth, using cell preparations containing pluripotent stem cells (Muse cells) into which a suicide gene has been introduced.
  • “brain tumor” refers to a state in which any type of nerve cell proliferates abnormally.
  • Examples of brain tumors include, but are not limited to, glioma, medulloblastoma, neuroblastoma, meningioma, pituitary adenoma, schwannoma, primary central nervous system lymphoma, sarcoma, and spinal cord tumor Can be mentioned.
  • the cell preparation of the present invention can target the above-mentioned various brain tumors, but it is preferable to treat gliomas that account for about 1/4 of the brain tumors as treatment targets.
  • Pluripotent stem cells The pluripotent stem cell used in the cell preparation of the present invention was found by Dezawa, one of the present inventors, in the human body and named “Muse (Multilineage-differentiating Stress Ending) cell”. It is. Muse cells can be obtained from skin tissues such as bone marrow fluid and dermal connective tissue, and are also scattered in connective tissues of each organ. In addition, this cell is a cell having the properties of both pluripotent stem cells and mesenchymal stem cells. For example, the cell surface markers “SSEA-3 (Stage-specific embryonic antigen-3)” and “ Identified as "CD105" double positive.
  • SSEA-3 Serial-specific embryonic antigen-3
  • Muse cells or cell populations containing Muse cells can be separated from living tissues using, for example, these antigen markers as indicators. Details such as a method for separating Muse cells, an identification method, and characteristics are disclosed in International Publication No. WO2011 / 007900. Also, as reported by Wakao et al. (2011, supra), when mesenchymal cells are cultured from bone marrow, skin, etc. and used as the population of Muse cells, all SSEA-3 positive cells are CD105 It is known to be a positive cell.
  • the Muse cells when separating Muse cells from living mesenchymal tissue or cultured mesenchymal stem cells, the Muse cells can be purified and used simply with SSEA-3 as an antigen marker.
  • pluripotent stem cells isolated from living mesenchymal tissue or cultured mesenchymal tissue using SSEA-3 as an antigen marker which can be used in cell preparations for treating brain tumors ( Muse cells) or a cell population containing Muse cells may be simply referred to as “SSEA-3 positive cells”.
  • “non-Muse cells” refer to cells other than “SSEA-3-positive cells”, which are cells contained in a mesenchymal tissue or cultured mesenchymal tissue in a living body.
  • Muse cells or cell populations containing Muse cells can be obtained from living tissue (eg, using antibodies against the cell surface marker SSEA-3 alone, or both antibodies against SSEA-3 and CD105, respectively) , Mesenchymal tissue).
  • living tissue eg, using antibodies against the cell surface marker SSEA-3 alone, or both antibodies against SSEA-3 and CD105, respectively
  • Mesenchymal tissue e.g., Mesenchymal tissue.
  • “living body” means a living body of a mammal. In the present invention, the living body does not include embryos whose developmental stage is earlier than the fertilized egg or blastocyst stage, but includes embryos in the developmental stage after the blastocyst stage including the fetus and blastocyst.
  • Mammals include, but are not limited to, primates such as humans and monkeys, rodents such as mice, rats, rabbits, guinea pigs, cats, dogs, sheep, pigs, cows, horses, donkeys, goats, ferrets, etc. It is done.
  • Muse cells used in the cell preparation of the present invention are clearly distinguished from embryonic stem cells (ES cells) and embryonic germ stem cells (EG cells) in that they are derived from living tissues.
  • ES cells embryonic stem cells
  • EG cells embryonic germ stem cells
  • “Mesenchymal tissue” refers to tissues existing in various organs such as bone, synovium, fat, blood, bone marrow, skeletal muscle, dermis, ligament, tendon, dental pulp, and umbilical cord.
  • Muse cells can be obtained from bone marrow or skin.
  • the Muse cell used may be autologous to the recipient who receives the cell transplant, or may be another family.
  • a Muse cell or a cell population containing a Muse cell can be separated from a living tissue using, for example, SSEA-3 positive and SSEA-3 and CD105 double positive as an index.
  • SSEA-3 positive and SSEA-3 and CD105 double positive are known to include various types of stem cells and progenitor cells.
  • Muse cells are not the same as these cells.
  • Such stem cells and progenitor cells include skin-derived progenitor cells (SKP), neural crest stem cells (NCSC), melanoblast (MB), perivascular cells (PC), endothelial progenitor cells (EP), adipose-derived stem cells (ADSC). ).
  • Muse cells can be isolated using “non-expression” of a marker unique to these cells as an index.
  • Muse cells are CD34 (EP and ADSC markers), CD117 (c-kit) (MB markers), CD146 (PC and ADSC markers), CD271 (NGFR) (NCSC markers), NG2 (PC marker), vWF factor (von Willebrand factor) (EP marker), Sox10 (NCSC marker), Snai1 (SKP marker), Slug (SKP marker), Tyrp1 (MB marker), and At least one of 11 markers selected from the group consisting of Dct (MB marker), for example 2, 3, 4, 5, 6, 7, 8, 9, 10 The non-expression of individual or eleven markers can be separated into indicators.
  • non-expression of CD117 and CD146 can be separated as an index
  • non-expression of CD117, CD146, NG2, CD34, vWF and CD271 can be separated as an index
  • the non-expression of 11 markers can be separated as an index.
  • the Muse cell having the above characteristics used in the cell preparation of the present invention is as follows: (I) low or no telomerase activity; (Ii) has the ability to differentiate into cells of any germ layer of the three germ layers; It may have at least one property selected from the group consisting of (iii) showing no neoplastic growth; and (iv) having a self-renewal capability.
  • the Muse cell used in the cell preparation of the present invention has all the above properties.
  • telomerase activity is low or absent means that, for example, when telomerase activity is detected using TRAPEZE XL telomerase detection kit (Millipore), it is low or cannot be detected.
  • “Low” telomerase activity means, for example, telomerase having a telomerase activity comparable to that of somatic human fibroblasts, or 1/5 or less, preferably 1/10 or less compared to Hela cells. It means having activity.
  • the Muse cell has the ability to differentiate into three germ layers (endoderm, mesodermal, and ectoderm) in vitro and in vivo, for example, induction culture in vitro Can be differentiated into hepatocytes, nerve cells, skeletal muscle cells, smooth muscle cells, bone cells, fat cells and the like. In addition, when transplanted to the testis in vivo, it may show the ability to differentiate into three germ layers.
  • Muse cells have the property of proliferating at a growth rate of about 1.3 days in suspension culture, but stop growing in about 10 days. Further, when transplanted to the testis, Muse cells have cancer for at least half a year. It has the property of not becoming Moreover, about said (iv), a Muse cell has self-renewal (self-replication) ability.
  • self-renewal refers to culturing cells contained in an embryoid body-like cell mass obtained by suspension culture of one Muse cell, and forming an embryoid body-like cell mass again. .
  • the self-renewal may be repeated once or multiple times.
  • Suicide gene means a gene that can kill itself when expressed in a cell, and typically includes a metabolic toxicity gene.
  • suicide genes include, but are not limited to, herpes simplex virus thymidine kinase (HSVtk) gene (Proc. Natl. Acad. Sci, USA, 78, 1441-1445 (1981)), cytosine deaminase gene (EG11326 codA 355395..356678, E.
  • the cell preparation of the present invention can be used together with a prodrug to treat a brain tumor.
  • the “prodrug” used together with the cell preparation of the present invention is a prodrug of a drug that kills the target tumor cell or inhibits its growth, and itself exhibits such cytotoxicity. It means a drug that you do not have.
  • This prodrug is converted into a drug having pharmacological activity (cytotoxicity) by an enzyme generated by expression of a suicide gene.
  • the prodrug include ganciclovir (GCV), acyclovir, pencyclovir, PMEA adefovir, PMPA tenofovir, etc., preferably GCV, acyclovir, and pencyclovir when the suicide gene is herpes simplex virus thymidine kinase (HSVtk) gene. it can.
  • guanine in the purine form of nucleic acid, and when used for DNA synthesis, DNA synthesis stops there and exerts an antiviral effect.
  • the suicide gene is a cytosine deaminase gene
  • 5-fluorocytosine can be used as the prodrug (Human Gene Therapy, 7, 713-720 (1996)).
  • the suicide gene is a uracil phosphoribosyltransferase gene
  • 5-fluorouracil can be used as a prodrug (Int. J. Oncol, 18, 117-120 (2001)).
  • the suicide gene is a guanine phosphoribosyltransferase gene
  • 6-thioxanthine or 6-thioguanine can be used as the prodrug (Human Gene Therapy, 8, 2043-2055 (1997)).
  • CB1954 can be used as a prodrug (Cancer Gene Therapy, 7, 721-731 (2000)).
  • the reaction mechanism of the enzyme and prodrug produced by the expression of the suicide gene will be described below using, for example, the HSVtk (herpes simplex virus thymidine kinase) gene and GCV (ganciclovir).
  • HSVtk herpes simplex virus thymidine kinase
  • GCV ganciclovir
  • the HSVtk gene is introduced into cancer cells and expressed, and GCV, which is an antiviral agent, is administered as a prodrug.
  • Cancer cells into which the HSVtk gene has been introduced phosphorylate GCV with HSVtk to form a monophosphorylated GCV (GCV-1P).
  • GCV-1P is then phosphorylated to triphosphate by its own thymidine kinase, and in order to inhibit DNA polymerase, cells that undergo DNA replication fall into apoptosis and die. Furthermore, GCV-1P is also taken into neighboring cells through a gap junction, and neighboring cells without gene transfer are also killed due to inhibition of DNA synthesis (bystander effect).
  • a Muse cell into which a suicide gene has been introduced is used.
  • a method of introducing the suicide gene into Muse cells a method of introducing a vector incorporating the gene is common.
  • Gene transfer by use of a vector includes, but is not limited to, viral or non-viral gene transfer (eg, plasmid transfer, phage integrase, transposon, adenovirus, adeno-associated virus, and lentivirus).
  • HSVtk herpes simplex virus thymidine kinase
  • PA317 mouse-derived HSVtk-retrovirus producing cell
  • cytosine deaminase gene it is incorporated into an adenovirus vector with cytomegalovirus early gene enhancer / promoter added to the cytosine deaminase gene cDNA of E. coli (Human Gene Therapy, 6, 1055-1063 (1995)). Can be introduced.
  • the gene When using a uracil phosphoribosyltransferase (UPRT) gene, the gene can be introduced using a retroviral vector LXSN incorporating the UPRT gene of E. coli.
  • the gene When the guanine phosphoribosyltransferase (gpt) gene is used, the gene can be introduced by infecting the cells with the virus using the supernatant of the E. coli gpt-retrovirus producing cell (GP / E86 gpt) culture.
  • a plasmid in which the cytomegalovirus early gene enhancer / promoter is added to the Escherichia coli ntr gene can be prepared, and the gene can be introduced into the cell by electroporation.
  • the cells may be appropriately proliferated.
  • the cell preparation of the present invention can be obtained by suspending Muse cells after introduction of a suicide gene in physiological saline or an appropriate buffer (for example, phosphate buffered saline).
  • physiological saline or an appropriate buffer for example, phosphate buffered saline.
  • the cell preparation may be cultured before being administered to the treatment subject and grown until a predetermined cell concentration is obtained.
  • Muse cells do not become tumors. Therefore, even if cells collected from living tissue remain undifferentiated, there is a possibility of canceration. Low and safe.
  • the culture of the collected Muse cells is not particularly limited, but can be performed in a normal growth medium (for example, ⁇ -minimal essential medium ( ⁇ -MEM) containing 10% calf serum). More specifically, referring to the above International Publication No. WO2011 / 007900, in the culture and proliferation of Muse cells, a medium, additives (for example, antibiotics, serum) and the like are appropriately selected, and Muse cells at a predetermined concentration are selected. A solution containing can be prepared.
  • a normal growth medium for example, ⁇ -minimal essential medium ( ⁇ -MEM) containing 10% calf serum.
  • DMSO dimethyl sulfoxide
  • serum albumin is used to protect the cells
  • antibiotics are used to prevent bacterial contamination and growth. You may make it contain in a cell formulation.
  • other pharmaceutically acceptable ingredients for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological saline, etc.
  • Cells or components other than Muse cells contained in mesenchymal stem cells may be contained in the cell preparation.
  • One skilled in the art can add these factors and agents to the cell preparation at appropriate concentrations.
  • the number of Muse cells into which the suicide gene contained in the cell preparation prepared above is introduced depends on the sex of the subject so that a desired effect (eg, disappearance of tumor, reduction of tumor size) can be obtained in the treatment of brain tumors. In consideration of the age, weight, state of the affected area, state of cells to be used, etc., it can be appropriately adjusted.
  • the cell preparation of the present invention can be used a plurality of times (for example, 2 to 10 times) at appropriate intervals (for example, twice a day, once a day, once a week) until a desired therapeutic effect is obtained. 2 times, once a week, once every two weeks).
  • the therapeutically effective dose is preferably, for example, 1 ⁇ 10 3 cells to 1 ⁇ 10 6 cells per individual and 1 to 10 doses.
  • the total dose in one individual includes, but is not limited to, 1 ⁇ 10 3 cells to 1 ⁇ 10 7 , 1 ⁇ 10 4 cells to 5 ⁇ 10 6 cells, and the like.
  • treatment of brain tumors with the cell preparation of the present invention is based on the expression of a suicide gene introduced into Muse cells, and the subsequent activation of the prodrug by the enzyme that is the gene product. Therefore, in the use of the cell preparation of the present invention, the administration site and administration method (local tumor administration, intracarotid artery administration, intravenous administration) of the cell preparation are not limited.
  • the prodrug used together with the cell preparation of the present invention may be intravenous administration as described above, or may be intraperitoneal administration.
  • a prodrug can be administered intravenously or intraperitoneally to obtain the desired therapeutic effect.
  • the administration time of the prodrug is not limited, but may be any time after administration of the cell preparation of the present invention, at the same time as administration, and before administration as long as it has the above effect.
  • a pharmaceutical composition comprising a Muse cell into which a suicide gene has been introduced, a prodrug, and optionally a carrier or excipient.
  • Example 1 Preparation of Muse cells into which herpes simplex virus thymidine kinase (HSVtk) gene was introduced
  • HSVtk herpes simplex virus thymidine kinase
  • HSVtk gene introduction HSVtk retrovirus-producing cells PA317, mouse fibroblasts, Genetic Therapy Inc.
  • Example 2 Detection of Muse cells around glioblastoma (glioblastoma) It was examined whether or not Muse cells were present in human glioblastoma collected from human subjects. A tissue section of the collected glioblastoma was prepared and immunostained using an antibody against the cell surface antigen (SSEA-3 ) of Muse cells according to a conventional method. As shown in FIG. 1, it was found that Muse cells exist around glioblastoma.
  • SSEA-3 cell surface antigen
  • Example 3 Confirmation of bystander effect in human glioma cells by Muse-TK cells to confirm the bystander effect by Muse-TK prepared in Example 1, co-culture of Muse-TK cells and glioma cells in vitro was examined.
  • 96-well human glioma cells A172 1.5 ⁇ 10 4 cells / well, Muse-TK cells (1/1), 1/4, 1/8, 1/16, 1/32 cells The cells were mixed as a number, cultured in the presence of 2 ⁇ g / ml ganciclovir (GCV) for a predetermined number of days, observed with a phase contrast microscope, and imaged.
  • GCV 2 ⁇ g / ml ganciclovir
  • FIG. 3 shows the results of actually counting glioma cells in culture on the 5th and 9th days after GCV addition based on the results of FIG. As can be seen from this result, cell proliferation of glioma cells was reduced to a cell ratio of 1: 1 to 1: 8. Thus, a bystander effect on glioma cells was observed in Muse-TK cells.
  • Example 4 Confirmation of migration ability to glioma cells by Muse-TK cells
  • Muse cells have the property of accumulating around tumors.
  • Muse cell migration was quantitatively measured using the Boyden chamber method (Boyden, S., J. Exp. Med., Vol. 115, p. 453-466 (1962)).
  • the Boyden chamber used was a QCM Chemotaxis Cell Migration Assay Kit (QCM 24 Well Colorimetric Cell Migration Assay) commercially available from Millipore.
  • the Boyden chamber includes an insert having a filter having uniform fine pores of 8 ⁇ m at the bottom inside the chamber.
  • the culture solution containing Muse cells or non-Muse cells was added to the upper part of the filter of the insert, the culture supernatant of glioma cells was added to the lower part of the insert, and after culturing for 18 hours, the number of cells that passed through the micropores of the filter was counted. .
  • the result is shown in FIG. "Muse A172 "and” Muse " In the “YKG-1” system, Muse cells passed through the micropores and moved significantly to the lower part of the insert.
  • the culture supernatant of glioma cells was used for non-Muse cells, and when DMEM was used instead of this culture supernatant, the ability to migrate Muse cells was not exhibited.
  • Example 5 Verification of tumor formation inhibitory effect in vivo
  • the bystander effect by Muse-TK prepared in Example 1 was confirmed in vivo.
  • U87-luc2 cells glioma cell line
  • Muse-TK cells and luciferase gene were introduced were co-transplanted into the right brain of nude mice (male 8 weeks old). More specifically, the numbers of Muse-TK cells and U87-luc2 cells to be transplanted were 2.5 ⁇ 10 4 and 10 ⁇ 10 4 (1: 4), respectively.
  • ganciclovir (GCV) Wired
  • Muse cells into which the suicide gene has been introduced accumulate around the brain tumor, and further, the cells constituting the brain tumor are killed by administration of a prodrug corresponding to the suicide gene. Can be applied to the treatment of brain tumors.

Abstract

The purpose of the present invention is to provide a novel medical use using a pluripotent stem cell (Muse cell). Provided is a cell preparation for treating a brain tumor, said cell preparation comprising SSEA-3 positive pluripotent stem cells that are separated from a mesenchymal tissue in an organism or cultured mesenchymal cells. The cell preparation according to the present invention is based on a mechanism in which Muse cells carrying a suicide gene transferred thereinto is intravenously administered to a subject having a brain tumor, and a prodrug corresponding to the suicide gene is further administered so that the Muse cells are allowed to selectively accumulate around the brain tumor and thus cells constituting the brain tumor are exterminated or the growth of the cells is reduced through the activation of the prodrug accompanied with the expression of the suicide gene.

Description

脳腫瘍治療のための多能性幹細胞Pluripotent stem cells for brain tumor treatment
 本発明は、脳腫瘍の治療に有効な自殺遺伝子を導入した多能性幹細胞を含有する細胞製剤に関する。 The present invention relates to a cell preparation containing pluripotent stem cells into which a suicide gene effective for treatment of brain tumors is introduced.
 脳腫瘍の約1/4を占めるグリオーマ(神経膠腫)は、周囲の正常脳組織に浸潤性に発育するため、脳機能を温存しようとすれば手術による全摘出は困難である。更に、術後には放射線治療や抗癌剤治療を併用することが多いが、治癒率は低く、最も悪性のグリオブラストーマ(膠芽腫)といわれる腫瘍では平均生存期間は約1年にすぎない。しかもこの成績はさまざまな治療戦略の開発にもかかわらず、過去30年間ほとんど変わっておらず、21世紀に残された最後の悪性腫瘍の1つと言われており、新たな治療戦略の開発が急務である。 Glioma (glioma), which accounts for about ¼ of brain tumors, grows infiltrating into the surrounding normal brain tissue, so that if the brain function is to be preserved, it is difficult to remove it by surgery. Furthermore, radiation therapy and anticancer drug treatment are often used after surgery, but the cure rate is low, and the average malignant glioblastoma (glioblastoma) has an average survival time of only about 1 year. Moreover, despite the development of various treatment strategies, this result has hardly changed for the past 30 years and is said to be one of the last malignant tumors left in the 21st century, and the development of new treatment strategies is urgently needed. It is.
 1990年代に導入された「自殺遺伝子治療」という遺伝子治療は、ウイルスなどの遺伝子を用いて、哺乳類では無害な薬物を遺伝子導入細胞中で毒物(抗癌剤)に変化させ、癌細胞を殺す治療法である。中でも単純ヘルペスウイルスチミジンキナーゼ遺伝子(HSVtk遺伝子)と抗ウイルス剤ガンシクロビル(GCV)を用いた自殺遺伝子治療においては、遺伝子導入細胞が全体の10%程度でもすべての腫瘍が消失するバイスタンダー効果(bystander effect)がある(非特許文献1)。しかし、この治療法はラットなどの動物実験では顕著な効果が見られたが、臨床成績は必ずしも満足のいくものではなかった。その理由の1つとして、遺伝子導入のために用いたレトロウイルス産生線維芽細胞の移動能が低く、浸潤性に発育するグリオーマ細胞をカバーしきれなかったことが挙げられる。 The gene therapy called “suicide gene therapy” introduced in the 1990s is a therapy that uses genes such as viruses to change harmful drugs in mammals into toxic substances (anticancer drugs) in transgenic cells and kill cancer cells. is there. In particular, in the suicide gene therapy using the herpes simplex virus thymidine kinase gene (HSVtk gene) and the antiviral agent ganciclovir (GCV), the bystander effect that all tumors disappear even if the transgenic cells are about 10% of the whole. (Non-Patent Document 1). However, although this treatment showed remarkable effects in animal experiments such as rats, the clinical results were not always satisfactory. One reason for this is that the retrovirus-producing fibroblasts used for gene transfer have a low migration ability and could not cover glioma cells that grow invasively.
 一方、近年発見され神経再生の研究が盛んな神経幹細胞は脳内で極めて活発な移動能を持ち、ラットの実験では脳腫瘍と神経幹細胞を別々に左右の脳に移植して観察すると神経幹細胞が対側の脳腫瘍まで移動し、腫瘍周辺に集積することが知られている(非特許文献2)。このような移動能のある神経幹細胞に自殺遺伝子を組み込み、腫瘍部で細胞障害性物質を発現させることにより脳腫瘍を治療する方法が試みられている(特許文献1)。本発明者らは、HSVtk/GCV遺伝子を導入した神経幹細胞を脳腫瘍の周辺に移植したところ、バイスタンダー効果による抗腫瘍効果を認め、神経幹細胞が悪性グリオーマの治療に適した細胞であることを見出している(非特許文献3)。しかしながら、神経幹細胞は自殺遺伝子を腫瘍部に運搬するベクターとしては極めて有効であるが、臨床応用を考えた場合に、神経幹細胞を患者から取り出すのは侵襲的で極めて困難である。これまで、本発明者らは、神経幹細胞と同様の移動能を持ちかつより汎用的なベクターとして、骨髄より採取された間葉系幹細胞を自殺遺伝子を腫瘍部に運搬するベクターとして用いて脳腫瘍の治療試験を試みた(特許文献2)。しかしながら、この治療試験に使用した間葉系幹細胞は不均一であり効果が一定でなかった。 On the other hand, neural stem cells that have been discovered recently and are actively researched for nerve regeneration have extremely active migration ability in the brain, and in rat experiments, when brain tumors and neural stem cells are transplanted to the left and right brains separately and observed, It is known that it moves to the side brain tumor and accumulates around the tumor (Non-Patent Document 2). Attempts have been made to treat brain tumors by incorporating suicide genes into such migratory neural stem cells and expressing cytotoxic substances in the tumor site (Patent Document 1). The inventors of the present invention transplanted a neural stem cell into which HSVtk / GCV gene has been introduced around a brain tumor, found an antitumor effect due to the bystander effect, and found that the neural stem cell is a cell suitable for the treatment of malignant glioma. (Non-patent Document 3). However, neural stem cells are extremely effective as a vector for carrying a suicide gene to a tumor site, but when considering clinical application, it is invasive and extremely difficult to remove neural stem cells from a patient. So far, the present inventors have used a mesenchymal stem cell collected from the bone marrow as a vector carrying a suicide gene to the tumor site as a more general-purpose vector having the same mobility as that of a neural stem cell. A treatment test was attempted (Patent Document 2). However, the mesenchymal stem cells used in this treatment test were heterogeneous and the effects were not constant.
 本発明者らの一人である出澤の研究により、間葉系細胞画分に存在し、誘導操作なしに得られる、SSEA-3(Stage-Specific Embryonic Antigen-3)を表面抗原として発現している多能性幹細胞(Multilineage-differentiating Stress Enduring cells;Muse細胞)が間葉系細胞画分の有する多能性を担っており、組織再生を目指した疾患治療に応用できる可能性があることが分かってきた(特許文献3;非特許文献4;非特許文献5)。しかしながら、脳腫瘍の予防及び/又は治療にMuse細胞を使用し、期待される治療効果が得られることを明らかにした例はない。 According to research by Dezawa, one of the present inventors, SSEA-3 (Stage-Specific Embryonic Antigen-3), which is present in the mesenchymal cell fraction and obtained without induction, is expressed as a surface antigen. It has been found that pluripotent stem cells (Multilineage-differentiating Stress Enduring cells; Muse cells) are responsible for the pluripotency of the mesenchymal cell fraction and can be applied to disease treatment aimed at tissue regeneration. (Patent Document 3; Non-Patent Document 4; Non-Patent Document 5). However, there are no examples that show that the expected therapeutic effect can be obtained by using Muse cells for the prevention and / or treatment of brain tumors.
特表2002-526104号公報Special Table 2002-526104 特開2006-345726号公報JP 2006-345726 A 国際公開第WO2011/007900号International Publication No. WO2011 / 007900
 本発明は、脳腫瘍治療において、多能性幹細胞(Muse細胞)を用いた新たな医療用途を提供することを目的とする。より具体的には、本発明は、自殺遺伝子を導入したMuse細胞及び該自殺遺伝子に対応するプロドラッグを含む、脳腫瘍の予防及び/又は治療のための細胞製剤を提供することを目的とする。 An object of the present invention is to provide a new medical use using pluripotent stem cells (Muse cells) in brain tumor treatment. More specifically, an object of the present invention is to provide a cell preparation for the prevention and / or treatment of brain tumor, comprising Muse cells into which a suicide gene has been introduced and a prodrug corresponding to the suicide gene.
 本発明者らは、単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子を導入したヒトMuse細胞をガンシクロビル(GCV)とともに、グリオーマ細胞の培養系に添加することによって、該グリオーマ細胞へのMuse細胞のバイスタンダー効果及び遊走能を確認し、グリオーマ細胞の増殖が顕著に抑制されることを見出し、本発明を完成させるに至った。 By adding human Muse cells into which the herpes simplex virus thymidine kinase (HSVtk) gene has been introduced together with ganciclovir (GCV) to the culture system of glioma cells, the bystander effect of Muse cells on the glioma cells And the migration ability was confirmed, it was found that the proliferation of glioma cells was remarkably suppressed, and the present invention was completed.
 すなわち、本発明は、以下の通りである。
 [1]自殺遺伝子を導入した多能性幹細胞を含む、脳腫瘍を治療するための細胞製剤であって、ここで、該多能性幹細胞は、生体の間葉系組織又は培養間葉系細胞から分離されたSSEA-3陽性細胞であり、該細胞製剤は、脳腫瘍を死滅させる又はその増殖を阻害する薬物のプロドラッグとともに使用され、該プロドラッグは、前記自殺遺伝子の発現によって生成する酵素に対する基質である細胞製剤。
 [2]脳腫瘍を構成するグリオーマ細胞を治療対象とする、上記[1]に記載の細胞製剤。
 [3]前記多能性幹細胞が、CD105陽性である、上記[1]及び[2]に記載の細胞製剤。
 [4]前記多能性幹細胞が、CD117陰性及びCD146陰性である、上記[1]~[3]に記載の細胞製剤。
 [5]前記多能性幹細胞が、CD117陰性、CD146陰性、NG2陰性、CD34陰性、vWF陰性、及びCD271陰性である、上記[1]~[4]に記載の細胞製剤。
 [6]前記多能性幹細胞が、CD34陰性、CD117陰性、CD146陰性、CD271陰性、NG2陰性、vWF陰性、Sox10陰性、Snai1陰性、Slug陰性、Tyrp1陰性、及びDct陰性である、上記[1]~[5]に記載の細胞製剤。
 [7]前記多能性幹細胞が、以下の性質の全てを有する多能性幹細胞である、上記[1]~[6]に記載の細胞製剤:
(i)テロメラーゼ活性が低いか又は無い;
(ii)三胚葉のいずれの胚葉の細胞に分化する能力を持つ;
(iii)腫瘍性増殖を示さない;及び
(iv)セルフリニューアル能を持つ。
 [8]前記多能性幹細胞が、脳腫瘍部位に集積する能力を有する、上記[1]~[7]に記載の細胞製剤。
 [9]前記自殺遺伝子が、単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子、シトシンデアミナーゼ遺伝子、ウラシルホスホリボシルトランスフェラーゼ遺伝子、グアニンホスホリボシルトランスフェラーゼ(gpt)遺伝子又はニトロレダクターゼ遺伝子である、上記[1]~[8]に記載の細胞製剤。
 [10]前記プロドラッグが、自殺遺伝子がHSVtk遺伝子である場合はガンシクロビル(GCV)、アシクロビル、ペンシクロビル、PMEAアデフォビル又はPMPAテノフォビルであり、自殺遺伝子がシトシンデアミナーゼ遺伝子の場合は5-フルオロシトシンであり、自殺遺伝子がウラシルホスホリボシルトランスフェラーゼ遺伝子の場合は5-フルオロウラシルであり、自殺遺伝子がgpt遺伝子の場合は6-チオキサンチン又は6-チオグアニンであり、自殺遺伝子がニトロレダクターゼ(ntr)遺伝子の場合はプロドラッグはCB1954である、上記[1]~[8]に記載の細胞製剤。
That is, the present invention is as follows.
[1] A cell preparation for treating a brain tumor comprising a pluripotent stem cell into which a suicide gene has been introduced, wherein the pluripotent stem cell is derived from a mesenchymal tissue or a cultured mesenchymal cell in a living body. Isolated SSEA-3-positive cells, wherein the cell preparation is used with a prodrug of a drug that kills or inhibits the growth of a brain tumor, the prodrug being a substrate for an enzyme produced by expression of the suicide gene A cell preparation.
[2] The cell preparation described in [1] above, wherein glioma cells constituting a brain tumor are treated.
[3] The cell preparation according to [1] and [2] above, wherein the pluripotent stem cell is CD105 positive.
[4] The cell preparation described in [1] to [3] above, wherein the pluripotent stem cells are CD117 negative and CD146 negative.
[5] The cell preparation described in [1] to [4] above, wherein the pluripotent stem cells are CD117 negative, CD146 negative, NG2 negative, CD34 negative, vWF negative, and CD271 negative.
[6] The above [1], wherein the pluripotent stem cells are CD34 negative, CD117 negative, CD146 negative, CD271 negative, NG2 negative, vWF negative, Sox10 negative, Snai1 negative, Slug negative, Tyrp1 negative, and Dct negative. The cell preparation according to [5].
[7] The cell preparation described in [1] to [6] above, wherein the pluripotent stem cell is a pluripotent stem cell having all of the following properties:
(I) low or no telomerase activity;
(Ii) has the ability to differentiate into cells of any germ layer of the three germ layers;
(Iii) no neoplastic growth; and (iv) self-renewal ability.
[8] The cell preparation described in [1] to [7] above, wherein the pluripotent stem cell has an ability to accumulate at a brain tumor site.
[9] The above [1] to [8], wherein the suicide gene is herpes simplex virus thymidine kinase (HSVtk) gene, cytosine deaminase gene, uracil phosphoribosyltransferase gene, guanine phosphoribosyltransferase (gpt) gene or nitroreductase gene. ] The cell preparation as described in.
[10] The prodrug is gancyclovir (GCV), acyclovir, penciclovir, PMEA adefovir or PMPA tenofovir when the suicide gene is an HSVtk gene, and 5-fluorocytosine when the suicide gene is a cytosine deaminase gene, 5-fluorouracil when the suicide gene is a uracil phosphoribosyltransferase gene, 6-thioxanthine or 6-thioguanine when the suicide gene is a gpt gene, and a prodrug when the suicide gene is a nitroreductase (ntr) gene The cell preparation according to [1] to [8] above, wherein is CB1954.
 本発明は、脳腫瘍を患っている対象に対し、自殺遺伝子を導入したMuse細胞及び該自殺遺伝子に対するプロドラッグを静脈等から投与することにより、Muse細胞を脳腫瘍に集積させ、自殺遺伝子の発現に伴うプロドラッグの活性化に基づいて、腫瘍細胞を死滅又はその増殖を抑制することができる。 The present invention relates to the expression of a suicide gene by administering a suicide gene-introduced Muse cell and a prodrug for the suicide gene from a vein or the like to a subject suffering from a brain tumor, thereby accumulating the Muse cell in the brain tumor. Based on the activation of the prodrug, tumor cells can be killed or their growth can be suppressed.
ヒトグリア芽腫(膠芽細胞種)へのMuse細胞の集積を示す。The accumulation of Muse cells in human glioblastoma (glioblastoma cell type) is shown. 自殺遺伝子を導入したMuse細胞(Muse-TK)とヒトグリオーマ(A172)におけるバイスタンダー効果を示す。図2は、グリオーマ細胞とMuse-TKとを異なる細胞比で同時培養した場合のグリオーマ細胞の培養状態を経時的に観察した結果を示す。The bystander effect in Muse cells (Muse-TK) and human glioma (A172) introduced with a suicide gene is shown. FIG. 2 shows the results of observing the culture state of glioma cells over time when glioma cells and Muse-TK are co-cultured at different cell ratios. 図3は、図2で得られた結果に基づいて、グリオーマ細胞の減少を細胞数をカウントすることによって示した図である。FIG. 3 is a diagram showing the decrease in glioma cells by counting the number of cells based on the results obtained in FIG. グリオーマ細胞に対するMuse細胞の遊走能をボイデンチャンバーを用いて測定した結果を示す。The result of having measured the migration ability of the Muse cell with respect to a glioma cell using the Boyden chamber is shown. インビボにおけるMuse-TKによるバイスタンダー効果を検証した結果を示す。The result of having verified the bystander effect by Muse-TK in vivo is shown.
 本発明は、自殺遺伝子を導入した多能性幹細胞(Muse細胞)を含む、脳腫瘍を治療するための細胞製剤に関する。本発明を以下に詳細に説明する。 The present invention relates to a cell preparation for treating a brain tumor, comprising pluripotent stem cells (Muse cells) into which a suicide gene has been introduced. The present invention is described in detail below.
1.適用疾患
 本発明は、自殺遺伝子を導入した多能性幹細胞(Muse細胞)を含む細胞製剤を用いて、脳腫瘍の治療、特に、グリオーマ細胞の死滅又はその増殖を阻害する治療を目指す。ここで、「脳腫瘍」とは、任意のタイプの神経細胞が異常に増殖する状態を指す。脳腫瘍の例としては、限定されないが、グリオーマ(膠腫)、髄芽腫、神経芽細胞腫、髄膜腫、下垂体腺腫、神経鞘腫、原発性中枢神経系リンパ腫、肉腫、及び脊髄腫瘍が挙げられる。本発明の細胞製剤は、上記各種の脳腫瘍を対象とすることができるが、脳腫瘍の約1/4を占めるグリオーマを治療対象とすることが好ましい。
1. Applicable Diseases The present invention aims at treatment of brain tumors, in particular, treatment of killing glioma cells or inhibiting their growth, using cell preparations containing pluripotent stem cells (Muse cells) into which a suicide gene has been introduced. Here, “brain tumor” refers to a state in which any type of nerve cell proliferates abnormally. Examples of brain tumors include, but are not limited to, glioma, medulloblastoma, neuroblastoma, meningioma, pituitary adenoma, schwannoma, primary central nervous system lymphoma, sarcoma, and spinal cord tumor Can be mentioned. The cell preparation of the present invention can target the above-mentioned various brain tumors, but it is preferable to treat gliomas that account for about 1/4 of the brain tumors as treatment targets.
2.細胞製剤
(1)多能性幹細胞(Muse細胞)
 本発明の細胞製剤に使用される多能性幹細胞は、本発明者らの一人である出澤が、ヒト生体内にその存在を見出し、「Muse(Multilineage-differentiating Stress Enduring)細胞」と命名した細胞である。Muse細胞は、骨髄液や真皮結合組織等の皮膚組織から得ることができ、各臓器の結合組織にも散在する。また、この細胞は、多能性幹細胞と間葉系幹細胞の両方の性質を有する細胞であり、例えば、それぞれの細胞表面マーカーである「SSEA-3(Stage-specific embryonic antigen-3)」と「CD105」のダブル陽性として同定される。したがって、Muse細胞又はMuse細胞を含む細胞集団は、例えば、これらの抗原マーカーを指標として生体組織から分離することができる。Muse細胞の分離法、同定法、及び特徴などの詳細は、国際公開第WO2011/007900号に開示されている。また、Wakaoら(2011、上述)によって報告されているように、骨髄、皮膚などから間葉系細胞を培養し、それをMuse細胞の母集団として用いる場合、SSEA-3陽性細胞の全てがCD105陽性細胞であることが分かっている。したがって、本発明における細胞製剤においては、生体の間葉系組織又は培養間葉系幹細胞からMuse細胞を分離する場合は、単にSSEA-3を抗原マーカーとしてMuse細胞を精製し、使用することができる。なお、本明細書においては、脳腫瘍を治療するための細胞製剤において使用され得る、SSEA-3を抗原マーカーとして、生体の間葉系組織又は培養間葉系組織から分離された多能性幹細胞(Muse細胞)又はMuse細胞を含む細胞集団を単に「SSEA-3陽性細胞」と記載することがある。また、本明細書においては、「非Muse細胞」とは、生体の間葉系組織又は培養間葉系組織に含まれる細胞であって、「SSEA-3陽性細胞」以外の細胞を指す。
2. Cell preparation (1) Pluripotent stem cells (Muse cells)
The pluripotent stem cell used in the cell preparation of the present invention was found by Dezawa, one of the present inventors, in the human body and named “Muse (Multilineage-differentiating Stress Ending) cell”. It is. Muse cells can be obtained from skin tissues such as bone marrow fluid and dermal connective tissue, and are also scattered in connective tissues of each organ. In addition, this cell is a cell having the properties of both pluripotent stem cells and mesenchymal stem cells. For example, the cell surface markers “SSEA-3 (Stage-specific embryonic antigen-3)” and “ Identified as "CD105" double positive. Accordingly, Muse cells or cell populations containing Muse cells can be separated from living tissues using, for example, these antigen markers as indicators. Details such as a method for separating Muse cells, an identification method, and characteristics are disclosed in International Publication No. WO2011 / 007900. Also, as reported by Wakao et al. (2011, supra), when mesenchymal cells are cultured from bone marrow, skin, etc. and used as the population of Muse cells, all SSEA-3 positive cells are CD105 It is known to be a positive cell. Accordingly, in the cell preparation of the present invention, when separating Muse cells from living mesenchymal tissue or cultured mesenchymal stem cells, the Muse cells can be purified and used simply with SSEA-3 as an antigen marker. . In the present specification, pluripotent stem cells isolated from living mesenchymal tissue or cultured mesenchymal tissue using SSEA-3 as an antigen marker, which can be used in cell preparations for treating brain tumors ( Muse cells) or a cell population containing Muse cells may be simply referred to as “SSEA-3 positive cells”. In the present specification, “non-Muse cells” refer to cells other than “SSEA-3-positive cells”, which are cells contained in a mesenchymal tissue or cultured mesenchymal tissue in a living body.
 簡単には、Muse細胞又はMuse細胞を含む細胞集団は、細胞表面マーカーであるSSEA-3に対する抗体を単独で用いて、又はSSEA-3及びCD105に対するそれぞれの抗体を両方用いて、生体組織(例えば、間葉系組織)から分離することができる。ここで、「生体」とは、哺乳動物の生体をいう。本発明において、生体には、受精卵や胞胚期より発生段階が前の胚は含まれないが、胎児や胞胚を含む胞胚期以降の発生段階の胚は含まれる。哺乳動物には、限定されないが、ヒト、サル等の霊長類、マウス、ラット、ウサギ、モルモット等のげっ歯類、ネコ、イヌ、ヒツジ、ブタ、ウシ、ウマ、ロバ、ヤギ、フェレット等が挙げられる。本発明の細胞製剤に使用されるMuse細胞は、生体の組織由来である点で、胚性幹細胞(ES細胞)や胚性生殖幹細胞(EG細胞)と明確に区別される。また、「間葉系組織」とは、骨、滑膜、脂肪、血液、骨髄、骨格筋、真皮、靭帯、腱、歯髄、臍帯などの組織及び各種臓器に存在する組織をいう。例えば、Muse細胞は、骨髄や皮膚から得ることができる。例えば、生体の間葉系組織を採取し、この組織からMuse細胞を分離し、利用することが好ましい。また、上記分離手段を用いて、培養間葉系細胞からMuse細胞を分離してもよい。なお、本発明の細胞製剤においては、使用されるMuse細胞は、細胞移植を受けるレシピエントに対して自家であってもよく、又は他家であってもよい。 Briefly, Muse cells or cell populations containing Muse cells can be obtained from living tissue (eg, using antibodies against the cell surface marker SSEA-3 alone, or both antibodies against SSEA-3 and CD105, respectively) , Mesenchymal tissue). Here, “living body” means a living body of a mammal. In the present invention, the living body does not include embryos whose developmental stage is earlier than the fertilized egg or blastocyst stage, but includes embryos in the developmental stage after the blastocyst stage including the fetus and blastocyst. Mammals include, but are not limited to, primates such as humans and monkeys, rodents such as mice, rats, rabbits, guinea pigs, cats, dogs, sheep, pigs, cows, horses, donkeys, goats, ferrets, etc. It is done. Muse cells used in the cell preparation of the present invention are clearly distinguished from embryonic stem cells (ES cells) and embryonic germ stem cells (EG cells) in that they are derived from living tissues. “Mesenchymal tissue” refers to tissues existing in various organs such as bone, synovium, fat, blood, bone marrow, skeletal muscle, dermis, ligament, tendon, dental pulp, and umbilical cord. For example, Muse cells can be obtained from bone marrow or skin. For example, it is preferable to collect a mesenchymal tissue of a living body and separate and use Muse cells from this tissue. Moreover, you may isolate | separate a Muse cell from a cultured mesenchymal cell using the said isolation | separation means. In the cell preparation of the present invention, the Muse cell used may be autologous to the recipient who receives the cell transplant, or may be another family.
 上記のように、Muse細胞又はMuse細胞を含む細胞集団は、例えば、SSEA-3陽性、及びSSEA-3とCD105の二重陽性を指標にして生体組織から分離することができるが、ヒト成人皮膚には、種々のタイプの幹細胞及び前駆細胞を含むことが知られている。しかしながら、Muse細胞は、これらの細胞と同じではない。このような幹細胞及び前駆細胞には、皮膚由来前駆細胞(SKP)、神経堤幹細胞(NCSC)、メラノブラスト(MB)、血管周囲細胞(PC)、内皮前駆細胞(EP)、脂肪由来幹細胞(ADSC)が挙げられる。これらの細胞に固有のマーカーの「非発現」を指標として、Muse細胞を分離することができる。より具体的には、Muse細胞は、CD34(EP及びADSCのマーカー)、CD117(c-kit)(MBのマーカー)、CD146(PC及びADSCのマーカー)、CD271(NGFR)(NCSCのマーカー)、NG2(PCのマーカー)、vWF因子(フォンビルブランド因子)(EPのマーカー)、Sox10(NCSCのマーカー)、Snai1(SKPのマーカー)、Slug(SKPのマーカー)、Tyrp1(MBのマーカー)、及びDct(MBのマーカー)からなる群から選択される11個のマーカーのうち少なくとも1個、例えば、2個、3個、4個、5個、6個、7個、8個、9個、10個又は11個のマーカーの非発現を指標に分離することができる。例えば、限定されないが、CD117及びCD146の非発現を指標に分離することができ、さらに、CD117、CD146、NG2、CD34、vWF及びCD271の非発現を指標に分離することができ、さらに、上記の11個のマーカーの非発現を指標に分離することができる。 As described above, a Muse cell or a cell population containing a Muse cell can be separated from a living tissue using, for example, SSEA-3 positive and SSEA-3 and CD105 double positive as an index. Are known to include various types of stem cells and progenitor cells. However, Muse cells are not the same as these cells. Such stem cells and progenitor cells include skin-derived progenitor cells (SKP), neural crest stem cells (NCSC), melanoblast (MB), perivascular cells (PC), endothelial progenitor cells (EP), adipose-derived stem cells (ADSC). ). Muse cells can be isolated using “non-expression” of a marker unique to these cells as an index. More specifically, Muse cells are CD34 (EP and ADSC markers), CD117 (c-kit) (MB markers), CD146 (PC and ADSC markers), CD271 (NGFR) (NCSC markers), NG2 (PC marker), vWF factor (von Willebrand factor) (EP marker), Sox10 (NCSC marker), Snai1 (SKP marker), Slug (SKP marker), Tyrp1 (MB marker), and At least one of 11 markers selected from the group consisting of Dct (MB marker), for example 2, 3, 4, 5, 6, 7, 8, 9, 10 The non-expression of individual or eleven markers can be separated into indicators. For example, without limitation, non-expression of CD117 and CD146 can be separated as an index, and non-expression of CD117, CD146, NG2, CD34, vWF and CD271 can be separated as an index, and The non-expression of 11 markers can be separated as an index.
 また、本発明の細胞製剤に使用される上記特徴を有するMuse細胞は、以下: 
(i)テロメラーゼ活性が低いか又は無い;
(ii)三胚葉のいずれの胚葉の細胞に分化する能力を持つ;
(iii)腫瘍性増殖を示さない;及び
(iv)セルフリニューアル能を持つ
からなる群から選択される少なくとも1つの性質を有してもよい。本発明の一局面では、本発明の細胞製剤に使用されるMuse細胞は、上記性質を全て有する。ここで、上記(i)について、「テロメラーゼ活性が低いか又は無い」とは、例えば、TRAPEZE XL telomerase detection kit(Millipore社)を用いてテロメラーゼ活性を検出した場合に、低いか又は検出できないことをいう。テロメラーゼ活性が「低い」とは、例えば、体細胞であるヒト線維芽細胞と同程度のテロメラーゼ活性を有しているか、又はHela細胞に比べて1/5以下、好ましくは1/10以下のテロメラーゼ活性を有していることをいう。上記(ii)について、Muse細胞は、in vitro及びin vivoにおいて、三胚葉(内胚葉系、中胚葉系、及び外胚葉系)に分化する能力を有し、例えば、in vitroで誘導培養することにより、肝細胞、神経細胞、骨格筋細胞、平滑筋細胞、骨細胞、脂肪細胞等に分化し得る。また、in vivoで精巣に移植した場合にも三胚葉に分化する能力を示す場合がある。さらに、静注により生体に移植することで損傷を受けた臓器(心臓、皮膚、脊髄、肝、筋肉等)に遊走及び生着し、分化する能力を有する。上記(iii)について、Muse細胞は、浮遊培養では増殖速度約1.3日で増殖するが、10日間程度で増殖が止まるという性質を有し、さらに精巣に移植した場合、少なくとも半年間は癌化しないという性質を有する。また、上記(iv)について、Muse細胞は、セルフリニューアル(自己複製)能を有する。ここで、「セルフリニューアル」とは、1個のMuse細胞を浮遊培養することにより得られる胚様体様細胞塊に含まれる細胞を培養し、再度胚様体様細胞塊を形成させることをいう。セルフリニューアルは1回又は複数回のサイクルを繰り返せばよい。
Further, the Muse cell having the above characteristics used in the cell preparation of the present invention is as follows:
(I) low or no telomerase activity;
(Ii) has the ability to differentiate into cells of any germ layer of the three germ layers;
It may have at least one property selected from the group consisting of (iii) showing no neoplastic growth; and (iv) having a self-renewal capability. In one aspect of the present invention, the Muse cell used in the cell preparation of the present invention has all the above properties. Here, with regard to (i) above, “telomerase activity is low or absent” means that, for example, when telomerase activity is detected using TRAPEZE XL telomerase detection kit (Millipore), it is low or cannot be detected. Say. “Low” telomerase activity means, for example, telomerase having a telomerase activity comparable to that of somatic human fibroblasts, or 1/5 or less, preferably 1/10 or less compared to Hela cells. It means having activity. Regarding the above (ii), the Muse cell has the ability to differentiate into three germ layers (endoderm, mesodermal, and ectoderm) in vitro and in vivo, for example, induction culture in vitro Can be differentiated into hepatocytes, nerve cells, skeletal muscle cells, smooth muscle cells, bone cells, fat cells and the like. In addition, when transplanted to the testis in vivo, it may show the ability to differentiate into three germ layers. Furthermore, it has the ability to migrate, engraft and differentiate into organs (heart, skin, spinal cord, liver, muscle, etc.) that have been damaged by implantation into a living body by intravenous injection. As for (iii) above, Muse cells have the property of proliferating at a growth rate of about 1.3 days in suspension culture, but stop growing in about 10 days. Further, when transplanted to the testis, Muse cells have cancer for at least half a year. It has the property of not becoming Moreover, about said (iv), a Muse cell has self-renewal (self-replication) ability. Here, “self-renewal” refers to culturing cells contained in an embryoid body-like cell mass obtained by suspension culture of one Muse cell, and forming an embryoid body-like cell mass again. . The self-renewal may be repeated once or multiple times.
(2)細胞製剤の調製及び使用
 本発明は、自殺遺伝子を導入したMuse細胞を含む、脳腫瘍を治療するための細胞製剤であって、該自殺遺伝子に対応するプロドラッグとともに使用する細胞製剤を提供する。
 (a)自殺遺伝子
 本明細書において使用するとき、「自殺遺伝子」とは、細胞内で発現すると自己を殺傷することができる遺伝子を意味し、典型的には、代謝毒性遺伝子が挙げられる。自殺遺伝子には、限定されないが、単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子(Proc.Natl.Acad.Sci,USA,78,1441-1445(1981))、シトシンデアミナーゼ遺伝子(EG11326 codA 355395..356678,大腸菌)、ウラシルホスホリボシルトランスフェラーゼ遺伝子(EG11332 upp 2618894..2618268,大腸菌)、グアニンホスホリボシルトランスフェラーゼ(gpt)遺伝子(EG10414 gpt 255977..256435,大腸菌)、ニトロレダクターゼ(ntr)遺伝子(EG11261 nfsA 890407..891129,大腸菌)が含まれる。
 (b)プロドラッグ
 本発明の細胞製剤は、プロドラッグとともに使用することにより脳腫瘍を治療することができる。ここで、本発明の細胞製剤とともに使用される「プロドラッグ」とは、目的とする腫瘍細胞を死滅させる又はその増殖を阻害する薬物のプロドラッグであって、それ自体ではこのような細胞毒性を持たない薬物を意味する。このプロドラッグは、自殺遺伝子の発現によって生成する酵素により薬理活性(細胞毒性)をもつ薬物に変換される。プロドラッグとしては、自殺遺伝子が単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子の場合、ガンシクロビル(GCV)、アシクロビル、ペンシクロビル、PMEAアデフォビル、PMPAテノフォビルなどが挙げられ、好ましくはGCV、アシクロビル、ペンシクロビルを用いることができる。これらは、いずれも核酸のプリン体のグアニンの類似物質でDNA合成に使われると、そこでDNA合成が停止し、抗ウイルス効果を発揮する。また、自殺遺伝子がシトシンデアミナーゼ遺伝子の場合、プロドラッグは5-フルオロシトシンを用いることができる(Human Gene Therapy,7,713-720(1996))。自殺遺伝子がウラシルホスホリボシルトランスフェラーゼ遺伝子の場合、プロドラッグは5-フルオロウラシルを用いることができる(Int.J.Oncol,18,117-120(2001))。自殺遺伝子がグアニンホスホリボシルトランスフェラーゼ遺伝子の場合、プロドラッグは6-チオキサンチン又は6-チオグアニンを用いることができる(Human Gene Therapy,8,2043-2055(1997))。自殺遺伝子がニトロレダクターゼ遺伝子の場合、プロドラッグはCB1954を用いることができる(Cancer Gene Therapy,7,721-731(2000))。
(2) Preparation and use of cell preparation The present invention provides a cell preparation for treating brain tumors, including Muse cells into which a suicide gene has been introduced, which is used together with a prodrug corresponding to the suicide gene. To do.
(A) Suicide gene As used herein, “suicide gene” means a gene that can kill itself when expressed in a cell, and typically includes a metabolic toxicity gene. Examples of suicide genes include, but are not limited to, herpes simplex virus thymidine kinase (HSVtk) gene (Proc. Natl. Acad. Sci, USA, 78, 1441-1445 (1981)), cytosine deaminase gene (EG11326 codA 355395..356678, E. coli), uracil phosphoribosyltransferase gene (EG11332 upp 2618894..2618268, E. coli), guanine phosphoribosyltransferase (gpt) gene (EG10414 gpt 255597..256435, E. coli), nitroreductase (ntr) gene (EG11261 nfs7.40 891129, E. coli).
(B) Prodrug The cell preparation of the present invention can be used together with a prodrug to treat a brain tumor. Here, the “prodrug” used together with the cell preparation of the present invention is a prodrug of a drug that kills the target tumor cell or inhibits its growth, and itself exhibits such cytotoxicity. It means a drug that you do not have. This prodrug is converted into a drug having pharmacological activity (cytotoxicity) by an enzyme generated by expression of a suicide gene. Examples of the prodrug include ganciclovir (GCV), acyclovir, pencyclovir, PMEA adefovir, PMPA tenofovir, etc., preferably GCV, acyclovir, and pencyclovir when the suicide gene is herpes simplex virus thymidine kinase (HSVtk) gene. it can. These are all analogs of guanine in the purine form of nucleic acid, and when used for DNA synthesis, DNA synthesis stops there and exerts an antiviral effect. When the suicide gene is a cytosine deaminase gene, 5-fluorocytosine can be used as the prodrug (Human Gene Therapy, 7, 713-720 (1996)). When the suicide gene is a uracil phosphoribosyltransferase gene, 5-fluorouracil can be used as a prodrug (Int. J. Oncol, 18, 117-120 (2001)). When the suicide gene is a guanine phosphoribosyltransferase gene, 6-thioxanthine or 6-thioguanine can be used as the prodrug (Human Gene Therapy, 8, 2043-2055 (1997)). When the suicide gene is a nitroreductase gene, CB1954 can be used as a prodrug (Cancer Gene Therapy, 7, 721-731 (2000)).
 また、自殺遺伝子の発現により生成する酵素とプロドラッグの反応機構について、例えば、HSVtk(単純ヘルペスウイルスチミジンキナーゼ)遺伝子とGCV(ガンシクロビル)を用いて、以下に説明する。例えば癌細胞にHSVtk遺伝子を導入して発現させ、抗ウイルス剤であるGCVをプロドラッグとして投与する。HSVtk遺伝子を導入された癌細胞はHSVtkによってGCVをリン酸化し、一リン酸化型のGCV(GCV-1P)を形成する。GCV-1Pは、その後、自身のチミジンキナーゼによって三リン酸までリン酸化が進み、DNAポリメラーゼを阻害するために、DNA複製をおこす細胞はアポトーシスに陥り死滅する。さらにGCV-1Pはギャップジャンクションを通って隣接細胞へも取り込まれ、遺伝子導入がない隣接細胞もDNA合成が阻害され死滅する(バイスタンダー効果)。 Also, the reaction mechanism of the enzyme and prodrug produced by the expression of the suicide gene will be described below using, for example, the HSVtk (herpes simplex virus thymidine kinase) gene and GCV (ganciclovir). For example, the HSVtk gene is introduced into cancer cells and expressed, and GCV, which is an antiviral agent, is administered as a prodrug. Cancer cells into which the HSVtk gene has been introduced phosphorylate GCV with HSVtk to form a monophosphorylated GCV (GCV-1P). GCV-1P is then phosphorylated to triphosphate by its own thymidine kinase, and in order to inhibit DNA polymerase, cells that undergo DNA replication fall into apoptosis and die. Furthermore, GCV-1P is also taken into neighboring cells through a gap junction, and neighboring cells without gene transfer are also killed due to inhibition of DNA synthesis (bystander effect).
 (c)自殺遺伝子のMuse細胞への導入と細胞製剤の調製
 本発明の細胞製剤においては、自殺遺伝子を導入したMuse細胞が使用される。Muse細胞に前記自殺遺伝子を導入する方法としては、該遺伝子を組み込んだベクターを導入する方法が一般的である。ベクターの使用による遺伝子導入としては、限定されないが、ウイルス性又は非ウイルス性遺伝子導入(例えば、プラスミド導入、ファージインテグラーゼ、トランスポゾン、アデノウイルス、アデノ随伴ウイルス、及びレンチウイスルなど)が挙げられる。より具体的には、自殺遺伝子として、単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子を用いる場合、マウス由来のHSVtk-レトロウイルス産生細胞(PA317)培養の上清液により、ウイルスを細胞に感染させて遺伝子を導入することができる。シトシンデアミナーゼ遺伝子を用いる場合には、大腸菌のシトシンデアミナーゼ遺伝子のcDNAにサイトメガロウイルス初期遺伝子エンハンサー/プロモーターを付したアデノウイルスベクターに組み込み(Human Gene Therapy,6,1055-1063(1995))、細胞に導入することができる。また、ウラシルホスホリボシルトランスフェラーゼ(UPRT)遺伝子を用いる場合には、大腸菌のUPRT遺伝子を組み込んだレトロウイルスベクターLXSNを用いて遺伝子を導入することができる。グアニンホスホリボシルトランスフェラーゼ(gpt)遺伝子を用いる場合には、大腸菌のgpt-レトロウイルス産生細胞(GP/E86gpt)培養の上清液により、ウイルスを細胞に感染させて遺伝子を導入することができる。また、ニトロレダクターゼ(ntr)遺伝子を用いる場合には、大腸菌のntr遺伝子にサイトメガロウイルス初期遺伝子エンハンサー/プロモーターを付したプラスミドを作成し、エレクトロポレーションにより細胞内に遺伝子を導入することができる。なお、遺伝子導入前に所定の細胞数を確保するために、適宜、細胞を増殖させてもよい。
(C) Introduction of suicide gene into Muse cell and preparation of cell preparation In the cell preparation of the present invention, a Muse cell into which a suicide gene has been introduced is used. As a method of introducing the suicide gene into Muse cells, a method of introducing a vector incorporating the gene is common. Gene transfer by use of a vector includes, but is not limited to, viral or non-viral gene transfer (eg, plasmid transfer, phage integrase, transposon, adenovirus, adeno-associated virus, and lentivirus). More specifically, when the herpes simplex virus thymidine kinase (HSVtk) gene is used as the suicide gene, the virus is infected to the cells with the supernatant of the mouse-derived HSVtk-retrovirus producing cell (PA317) culture. Can be introduced. When the cytosine deaminase gene is used, it is incorporated into an adenovirus vector with cytomegalovirus early gene enhancer / promoter added to the cytosine deaminase gene cDNA of E. coli (Human Gene Therapy, 6, 1055-1063 (1995)). Can be introduced. When using a uracil phosphoribosyltransferase (UPRT) gene, the gene can be introduced using a retroviral vector LXSN incorporating the UPRT gene of E. coli. When the guanine phosphoribosyltransferase (gpt) gene is used, the gene can be introduced by infecting the cells with the virus using the supernatant of the E. coli gpt-retrovirus producing cell (GP / E86 gpt) culture. When using a nitroreductase (ntr) gene, a plasmid in which the cytomegalovirus early gene enhancer / promoter is added to the Escherichia coli ntr gene can be prepared, and the gene can be introduced into the cell by electroporation. In order to secure a predetermined number of cells before gene introduction, the cells may be appropriately proliferated.
 本発明の細胞製剤は、自殺遺伝子導入後のMuse細胞を生理食塩水や適切な緩衝液(例えば、リン酸緩衝生理食塩水)に懸濁させることによって得られる。この場合、自家又は他家の組織から分離したMuse細胞数が少ない場合には、細胞製剤を治療対象への投与前に細胞を培養して、所定の細胞濃度が得られるまで増殖させてもよい。なお、すでに報告されているように(国際公開第WO2011/007900号)、Muse細胞は、腫瘍化しないため、生体組織から回収した細胞が未分化のまま含まれていても癌化の可能性が低く安全である。また、回収したMuse細胞の培養は、特に限定されないが、通常の増殖培地(例えば、10%仔牛血清を含むα-最少必須培地(α-MEM))において行うことができる。より詳しくは、上記国際公開第WO2011/007900号を参照して、Muse細胞の培養及び増殖において、適宜、培地、添加物(例えば、抗生物質、血清)等を選択し、所定濃度のMuse細胞を含む溶液を調製することができる。 The cell preparation of the present invention can be obtained by suspending Muse cells after introduction of a suicide gene in physiological saline or an appropriate buffer (for example, phosphate buffered saline). In this case, when the number of Muse cells isolated from the tissue of the home or other family is small, the cell preparation may be cultured before being administered to the treatment subject and grown until a predetermined cell concentration is obtained. . In addition, as already reported (International Publication No. WO2011 / 007900), Muse cells do not become tumors. Therefore, even if cells collected from living tissue remain undifferentiated, there is a possibility of canceration. Low and safe. The culture of the collected Muse cells is not particularly limited, but can be performed in a normal growth medium (for example, α-minimal essential medium (α-MEM) containing 10% calf serum). More specifically, referring to the above International Publication No. WO2011 / 007900, in the culture and proliferation of Muse cells, a medium, additives (for example, antibiotics, serum) and the like are appropriately selected, and Muse cells at a predetermined concentration are selected. A solution containing can be prepared.
 (d)細胞製剤の使用
 Muse細胞の細胞製剤への使用においては、該細胞を保護するためにジメチルスルフォキシド(DMSO)や血清アルブミン等を、細菌の混入及び増殖を防ぐために抗生物質等を細胞製剤に含有させてもよい。さらに、製剤上許容される他の成分(例えば、担体、賦形剤、崩壊剤、緩衝剤、乳化剤、懸濁剤、無痛化剤、安定剤、保存剤、防腐剤、生理食塩水など)や間葉系幹細胞に含まれるMuse細胞以外の細胞又は成分を細胞製剤に含有させてもよい。当業者は、これら因子及び薬剤を適切な濃度で細胞製剤に添加することができる。
(D) Use of cell preparations When using Muse cells in cell preparations, dimethyl sulfoxide (DMSO) or serum albumin is used to protect the cells, and antibiotics are used to prevent bacterial contamination and growth. You may make it contain in a cell formulation. Furthermore, other pharmaceutically acceptable ingredients (for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological saline, etc.) Cells or components other than Muse cells contained in mesenchymal stem cells may be contained in the cell preparation. One skilled in the art can add these factors and agents to the cell preparation at appropriate concentrations.
 上記で調製される細胞製剤中に含有する自殺遺伝子を導入したMuse細胞の数は、脳腫瘍の治療において所望の効果(例えば、腫瘍の消滅、腫瘍サイズの減少)が得られるように、対象の性別、年齢、体重、患部の状態、使用する細胞の状態等を考慮して、適宜、調整することができる。また、本発明の細胞製剤は、所望の治療効果が得られるまで、複数回(例えば、2~10回)、適宜、間隔(例えば、1日に2回、1日に1回、1週間に2回、1週間に1回、2週間に1回)をおいて投与されてもよい。したがって、対象の状態にもよるが、治療上有効量としては、例えば、一個体あたり1×10細胞~1×10細胞で1~10回の投与量が好ましい。一個体における投与総量としては、限定されないが、1×10細胞~1×10、1×10細胞~5×10細胞などが挙げられる。 The number of Muse cells into which the suicide gene contained in the cell preparation prepared above is introduced depends on the sex of the subject so that a desired effect (eg, disappearance of tumor, reduction of tumor size) can be obtained in the treatment of brain tumors. In consideration of the age, weight, state of the affected area, state of cells to be used, etc., it can be appropriately adjusted. In addition, the cell preparation of the present invention can be used a plurality of times (for example, 2 to 10 times) at appropriate intervals (for example, twice a day, once a day, once a week) until a desired therapeutic effect is obtained. 2 times, once a week, once every two weeks). Therefore, although depending on the condition of the subject, the therapeutically effective dose is preferably, for example, 1 × 10 3 cells to 1 × 10 6 cells per individual and 1 to 10 doses. The total dose in one individual includes, but is not limited to, 1 × 10 3 cells to 1 × 10 7 , 1 × 10 4 cells to 5 × 10 6 cells, and the like.
 上記した通り、本発明の細胞製剤による脳腫瘍の治療は、Muse細胞に導入された自殺遺伝子の発現、それに続く該遺伝子産物である酵素によるプロドラッグの活性化に基づく。そこで、本発明の細胞製剤の使用においては、該細胞製剤の投与部位、投与方法(腫瘍局所投与、頚動脈内投与、静脈内投与)は限定されない。また、本発明の細胞製剤とともに使用されるプロドラッグは、上記記載の静脈投与であってもよく、また腹腔内投与であってもよい。例えば、本発明によれば、限定されないが、Muse細胞の遊走能(腫瘍周辺への集積脳)を利用して、例えば、脳腫瘍を有する対象に対して、該細胞製剤を静脈投与後、プロドラッグを静脈内又は腹腔内投与することによって、所望の治療効果を得ることができる。なお、プロドラッグの投与時期は、限定されないが、上記効果を有する限り、本発明の細胞製剤の投与後、投与と同時、及び投与前のいずれであってもよい。本発明の一態様においては、自殺遺伝子を導入したMuse細胞、プロドラッグ、及び場合により担体、賦形剤を含む医薬組成物が提供される。 As described above, treatment of brain tumors with the cell preparation of the present invention is based on the expression of a suicide gene introduced into Muse cells, and the subsequent activation of the prodrug by the enzyme that is the gene product. Therefore, in the use of the cell preparation of the present invention, the administration site and administration method (local tumor administration, intracarotid artery administration, intravenous administration) of the cell preparation are not limited. The prodrug used together with the cell preparation of the present invention may be intravenous administration as described above, or may be intraperitoneal administration. For example, according to the present invention, but not limited to, by using the migration ability of Muse cells (accumulated brain around the tumor), for example, to a subject having a brain tumor, after the intravenous administration of the cell preparation, a prodrug Can be administered intravenously or intraperitoneally to obtain the desired therapeutic effect. The administration time of the prodrug is not limited, but may be any time after administration of the cell preparation of the present invention, at the same time as administration, and before administration as long as it has the above effect. In one aspect of the present invention, there is provided a pharmaceutical composition comprising a Muse cell into which a suicide gene has been introduced, a prodrug, and optionally a carrier or excipient.
 以下の実施例により、本発明をさらに具体的に説明するが、本発明はこれら実施例により何ら限定されるものではない。 The following examples further illustrate the present invention, but the present invention is not limited to these examples.
実施例1:単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子を導入したMuse細胞の作製
(1)ヒトMuse細胞の調製
 ヒトMuse細胞の調製は、国際公開第WO2011/007900号に記載された方法に従って行った。より具体的には、ヒト骨髄液から接着性を有する間葉系細胞を培養し、増殖を経て、Muse細胞又はMuse細胞を含む細胞集団をSSEA-3陽性細胞としてFACSにて分離した。また、非Muse細胞は、上記間葉系細胞のうち、SSEA-3陰性の細胞群であり、対照として用いた。その後、リン酸緩衝生理食塩水又は培養液を用いて、所定濃度に調整し、以下のバイスタンダー効果及び細胞遊走能の評価に使用した。なお、骨髄間葉系細胞などの間葉系細胞を培養して得たものをMuse細胞の母集団として用いる場合、Wakaoら(2011、上述)によって報告されているように、SSEA-3陽性細胞は全て、CD105陽性細胞であることが分かっている。
(2)HSVtk遺伝子導入
 HSVtkレトロウイルス産生細胞(PA317、マウス線維芽細胞、Genetic Therapy Inc.)(Gaithersburg,MDより提供)をMSC培地中で48時間培養した後、その上清よりHSVtkレトロウイルスを得た。これを8μg/mlポリブレン(Aldrich Chemical Company Inc.,Milwaukee,WI)とともに培養中のMuse細胞に添加し、さらに5時間培養し、続いて洗浄後に新鮮な培養液と交換した。150μg/ml G418(Sigma-Aldrich Japan K.K.,Tokyo,Japan)とともに1週間培養し、薬剤耐性細胞を選択することにより遺伝子導入細胞のみを得た。これらの細胞株の中からガンシクロビルGCV感受性の高い株を選び、さらに増殖させ、充分量のHSVtk遺伝子導入細胞(Muse-TK細胞)を得た。
Example 1: Preparation of Muse cells into which herpes simplex virus thymidine kinase (HSVtk) gene was introduced (1) Preparation of human Muse cells Human Muse cells were prepared according to the method described in International Publication No. WO2011 / 007900. . More specifically, mesenchymal cells having adhesiveness were cultured from human bone marrow fluid, and after proliferation, Muse cells or a cell population containing Muse cells were separated as SSEA-3 positive cells by FACS. Non-Muse cells are a group of SSEA-3 negative cells among the above mesenchymal cells and used as controls. Then, it adjusted to the predetermined density | concentration using the phosphate buffered saline or a culture solution, and used for the evaluation of the following bystander effects and cell migration ability. When cells obtained by culturing mesenchymal cells such as bone marrow mesenchymal cells are used as the population of Muse cells, SSEA-3-positive cells are reported as reported by Wakao et al. (2011, supra). Are all known to be CD105 positive cells.
(2) HSVtk gene introduction HSVtk retrovirus-producing cells (PA317, mouse fibroblasts, Genetic Therapy Inc.) (provided by Gaithersburg, MD) were cultured in MSC medium for 48 hours, and then HSVtk retrovirus was transferred from the supernatant. Obtained. This was added to Muse cells in culture together with 8 μg / ml polybrene (Aldrich Chemical Company Inc., Milwaukee, Wis.), Further cultured for 5 hours, and then replaced with fresh medium after washing. By culturing with 150 μg / ml G418 (Sigma-Aldrich Japan KK, Tokyo, Japan) for 1 week and selecting drug-resistant cells, only transgenic cells were obtained. Among these cell lines, a ganciclovir GCV-sensitive line was selected and further expanded to obtain a sufficient amount of HSVtk gene-introduced cells (Muse-TK cells).
実施例2:グリオブラストーマ(膠芽腫)周辺におけるMuse細胞の検出
 ヒト対象から採取したヒトグリオブラストーマにMuse細胞が存在しているかどうかを検討した。採取したグリオブラストーマの組織切片を作製し、Muse細胞の細胞表面抗原(SSEA-3に対する抗体を用いて、常法に従って免疫染色を行った。図1に示すように、Muse細胞はグリオブラストーマの周辺に存在することが分かった。
Example 2: Detection of Muse cells around glioblastoma (glioblastoma) It was examined whether or not Muse cells were present in human glioblastoma collected from human subjects. A tissue section of the collected glioblastoma was prepared and immunostained using an antibody against the cell surface antigen (SSEA-3 ) of Muse cells according to a conventional method. As shown in FIG. 1, it was found that Muse cells exist around glioblastoma.
実施例3:Muse-TK細胞によるヒトグリオーマ細胞におけるバイスタンダー効果の確認
 実施例1において作製したMuse-TKによるバイスタンダー効果を確認するために、インビトロにおいてMuse-TK細胞とグリオーマ細胞との共培養により検討した。96ウェルにヒトグリオーマ細胞(A172)を1.5×10細胞/ウェル、Muse-TK細胞をそれと同数(1/1)、1/4、1/8、1/16、1/32の細胞数として混ぜ、2μg/mlのガンシクロビル(GCV)の存在下に所定日数で培養し、位相差顕微鏡で観察し、撮像した。図2に示すように、GCV添加後の第9日目には、Muse-TK細胞とグリオーマ細胞の割合が1:1、1:4及び1:8の培養系において、グリオーマ細胞の死滅が観察された。さらに、図2の結果に基づいて、GCV添加後の第5日目と第9日目に培養中のグリオーマ細胞を実際にカウントした結果を図3に示す。この結果からも分かるように、1:1~1:8の細胞比までグリオーマ細胞の細胞増殖を減退させた。このように、Muse-TK細胞において、グリオーマ細胞に対するバイスタンダー効果が認められた。
Example 3: Confirmation of bystander effect in human glioma cells by Muse-TK cells To confirm the bystander effect by Muse-TK prepared in Example 1, co-culture of Muse-TK cells and glioma cells in vitro Was examined. 96-well human glioma cells (A172) 1.5 × 10 4 cells / well, Muse-TK cells (1/1), 1/4, 1/8, 1/16, 1/32 cells The cells were mixed as a number, cultured in the presence of 2 μg / ml ganciclovir (GCV) for a predetermined number of days, observed with a phase contrast microscope, and imaged. As shown in FIG. 2, on the ninth day after the addition of GCV, the death of glioma cells was observed in the culture systems in which the ratio of Muse-TK cells to glioma cells was 1: 1, 1: 4, and 1: 8. It was done. Further, FIG. 3 shows the results of actually counting glioma cells in culture on the 5th and 9th days after GCV addition based on the results of FIG. As can be seen from this result, cell proliferation of glioma cells was reduced to a cell ratio of 1: 1 to 1: 8. Thus, a bystander effect on glioma cells was observed in Muse-TK cells.
実施例4:Muse-TK細胞によるグリオーマ細胞への遊走能の確認
 実施例2に示されるように、Muse細胞は腫瘍周辺に集積する性質を有する。そこで、グリオーマ細胞(A172及びYKG1)の培養上清を用いて、インビトロにおけるMuse細胞の遊走能を検討した。Muse細胞の遊走性をボイデンチャンバー法(Boyden,S.,J.Exp.Med.,Vol.115,p.453-466(1962))を用いて定量的に測定した。使用したボイデンチャンバーは、Milliporeから市販されているQCM Chemotaxis Cell Migration Assay Kit(QCM 24 Well Colorimetric Cell Migration Assay)を使用した。このボイデンチャンバーは、チャンバー内部に、8μmの均一な微細孔を有するフィルターを底部に有するインサートを含む。インサートのフィルター上部にMuse細胞又は非Muse細胞を含む培養液を添加し、インサートの下部にグリオーマ細胞の培養上清を添加し、18時間培養後、フィルターの微細孔を通過した細胞数をカウントした。その結果を図4に示す。「Muse A172」及び「Muse YKG-1」の系において、Muse細胞は、微細孔を通過して、顕著にインサート下部に移動した。一方、非Muse細胞に対してグリオーマ細胞の培養上清を用いた場合、及びこの培養上清の代わりにDMEMを用いた場合には、Muse細胞の移動能は発揮されなかった。このように、グリオーマ細胞の培養上清中には、Muse細胞の遊走能を活性化する何らかの因子が存在することが示唆され、インビボにおいては、この因子によってMuse細胞の脳腫瘍への集積を促しているものと考えられる。
Example 4: Confirmation of migration ability to glioma cells by Muse-TK cells As shown in Example 2, Muse cells have the property of accumulating around tumors. Thus, the in vitro migration ability of Muse cells was examined using culture supernatants of glioma cells (A172 and YKG1). Muse cell migration was quantitatively measured using the Boyden chamber method (Boyden, S., J. Exp. Med., Vol. 115, p. 453-466 (1962)). The Boyden chamber used was a QCM Chemotaxis Cell Migration Assay Kit (QCM 24 Well Colorimetric Cell Migration Assay) commercially available from Millipore. The Boyden chamber includes an insert having a filter having uniform fine pores of 8 μm at the bottom inside the chamber. The culture solution containing Muse cells or non-Muse cells was added to the upper part of the filter of the insert, the culture supernatant of glioma cells was added to the lower part of the insert, and after culturing for 18 hours, the number of cells that passed through the micropores of the filter was counted. . The result is shown in FIG. "Muse   A172 "and" Muse "   In the “YKG-1” system, Muse cells passed through the micropores and moved significantly to the lower part of the insert. On the other hand, when the culture supernatant of glioma cells was used for non-Muse cells, and when DMEM was used instead of this culture supernatant, the ability to migrate Muse cells was not exhibited. In this way, it is suggested that some factor that activates the migration ability of Muse cells is present in the culture supernatant of glioma cells, and this factor promotes the accumulation of Muse cells in brain tumors in vivo. It is thought that there is.
実施例5:インビボにおける腫瘍形成抑制効果の検証
 実施例1において作製したMuse-TKによるバイスタンダー効果をインビボにおいて確認した。Muse-TK細胞とルシフェラーゼ遺伝子が導入されたU87-luc2細胞(グリオーマ細胞株)をヌードマウス(雄性8週齢)の右脳内に同時移植した。より具体的には、移植されるMuse-TK細胞及びU87-luc2細胞の細胞数は、それぞれ2.5×10個及び10×10個(1:4)とした。上記細胞を脳内に移植後(0日)、翌日(1日目)から、ガンシクロビル(GCV)(Wako)をPBSにより希釈して5mg/mlとし、マウス1匹あたり1回1mg/200μl PBSを1日2回、10日間、腹腔内に投与した。対照として、GCVの代わりにPBSを投与した群、及びMuse-TK細胞の非移植群(U87-luc2細胞のみ)を用いた。細胞移植後から1日目、及び移植後から7日目ごとに、IVIS200を使用し、Bioluminescence imaging(BLI)にて腫瘍形成(増大又は縮小)過程を観察した。その結果を図5に示す。腫瘍形成については35日目まで観察したが、対照群であるPBS投与群(2段目及び4段目)、及びMuse-TK細胞の非移植群(3段目)では、経時的に脳腫瘍の増大が見られた。一方、Muse-TK細胞+GCV群(1段目)では、移植後の7日目より脳腫瘍の形成が抑制され、35日目では脳腫瘍はほとんど消滅していた。実験した4群のそれぞれについて、ルシフェラーゼによる蛍光強度を経時的に測定した場合においも、上記と同様に、Muse-TK細胞+GCV群において蛍光強度の顕著な低下が見られた(データ示さず)。さらに、上記マウスの生存率を検討するためにKaplan-Meier曲線を作製した。その結果、移植後の54日目において、Muse-TK細胞+GCV群では、実験に使用した全てのマウスは生存していたが、その他の3群については、31~37日目において全てのマウスは腫瘍死した(データ示さず)。これらの結果より、インビボにおいてもMuse-TK細胞によるグリオーマ細胞に対するバイスタンダー効果が確認された。
Example 5: Verification of tumor formation inhibitory effect in vivo The bystander effect by Muse-TK prepared in Example 1 was confirmed in vivo. U87-luc2 cells (glioma cell line) into which Muse-TK cells and luciferase gene were introduced were co-transplanted into the right brain of nude mice (male 8 weeks old). More specifically, the numbers of Muse-TK cells and U87-luc2 cells to be transplanted were 2.5 × 10 4 and 10 × 10 4 (1: 4), respectively. After transplanting the cells into the brain (day 0), the next day (day 1), ganciclovir (GCV) (Wako) is diluted with PBS to 5 mg / ml, and 1 mg / 200 μl PBS once per mouse. It was administered intraperitoneally twice a day for 10 days. As a control, a group administered with PBS instead of GCV and a non-transplanted group of Muse-TK cells (U87-luc2 cells only) were used. On the 1st day after cell transplantation and every 7th day after transplantation, the process of tumor formation (increase or decrease) was observed with Bioluminescence imaging (BLI) using IVIS200. The result is shown in FIG. Tumor formation was observed up to day 35, but in the control group PBS administration group (2nd and 4th stage) and the non-transplanted group of Muse-TK cells (3rd stage), brain tumors were observed over time. An increase was seen. On the other hand, in the Muse-TK cell + GCV group (first stage), the formation of brain tumor was suppressed from the 7th day after transplantation, and the brain tumor almost disappeared on the 35th day. When the fluorescence intensity by luciferase was measured over time for each of the four groups tested, a marked decrease in fluorescence intensity was observed in the Muse-TK cell + GCV group as described above (data not shown). Furthermore, a Kaplan-Meier curve was prepared to examine the survival rate of the mice. As a result, on the 54th day after transplantation, in the Muse-TK cell + GCV group, all the mice used in the experiment were alive, but in the other 3 groups, all the mice were observed on the 31st to 37th days. The tumor died (data not shown). From these results, a bystander effect on glioma cells by Muse-TK cells was confirmed also in vivo.
 本発明の細胞製剤は、脳腫瘍の対象に投与することにより、自殺遺伝子を導入したMuse細胞が脳腫瘍周辺に集積し、さらに自殺遺伝子に対応するプロドラッグの投与により、脳腫瘍を構成する細胞を死滅させることができ、脳腫瘍の治療に応用できる。 When the cell preparation of the present invention is administered to a brain tumor subject, Muse cells into which the suicide gene has been introduced accumulate around the brain tumor, and further, the cells constituting the brain tumor are killed by administration of a prodrug corresponding to the suicide gene. Can be applied to the treatment of brain tumors.
 本明細書に引用する全ての刊行物及び特許文献は、参照により全体として本明細書中に援用される。なお、例示を目的として、本発明の特定の実施形態を本明細書において説明したが、本発明の精神及び範囲から逸脱することなく、種々の改変が行われる場合があることは、当業者に容易に理解されるであろう。 All publications and patent literature cited herein are hereby incorporated by reference in their entirety. While specific embodiments of the invention have been described herein for purposes of illustration, it will be apparent to those skilled in the art that various modifications may be made without departing from the spirit and scope of the invention. It will be easily understood.

Claims (10)

  1.  自殺遺伝子を導入した多能性幹細胞を含む、脳腫瘍を治療するための細胞製剤であって、ここで、該多能性幹細胞は、生体の間葉系組織又は培養間葉系細胞から分離されたSSEA-3陽性細胞であり、該細胞製剤は、脳腫瘍を死滅させる又はその増殖を阻害する薬物のプロドラッグとともに使用され、該プロドラッグは、前記自殺遺伝子の発現によって生成する酵素の基質である細胞製剤。 A cell preparation for treating a brain tumor comprising pluripotent stem cells introduced with a suicide gene, wherein the pluripotent stem cells are isolated from mesenchymal tissue or cultured mesenchymal cells in a living body SSEA-3-positive cells, wherein the cell preparation is used with a prodrug of a drug that kills or inhibits the growth of a brain tumor, and the prodrug is a cell that is a substrate for an enzyme produced by expression of the suicide gene Formulation.
  2.  脳腫瘍を構成するグリオーマ細胞を治療対象とする、請求項1に記載の細胞製剤。 The cell preparation according to claim 1, wherein glioma cells constituting a brain tumor are treated.
  3.  前記多能性幹細胞が、CD105陽性である、請求項1又は2に記載の細胞製剤。 The cell preparation according to claim 1 or 2, wherein the pluripotent stem cells are CD105 positive.
  4.  前記多能性幹細胞が、CD117陰性及びCD146陰性である、請求項1~3のいずれか1項に記載の細胞製剤。 The cell preparation according to any one of claims 1 to 3, wherein the pluripotent stem cells are CD117 negative and CD146 negative.
  5.  前記多能性幹細胞が、CD117陰性、CD146陰性、NG2陰性、CD34陰性、vWF陰性、及びCD271陰性である、請求項1~4のいずれか1項に記載の細胞製剤。 The cell preparation according to any one of claims 1 to 4, wherein the pluripotent stem cells are CD117 negative, CD146 negative, NG2 negative, CD34 negative, vWF negative, and CD271 negative.
  6.  前記多能性幹細胞が、CD34陰性、CD117陰性、CD146陰性、CD271陰性、NG2陰性、vWF陰性、Sox10陰性、Snai1陰性、Slug陰性、Tyrp1陰性、及びDct陰性である、請求項1~5のいずれか1項に記載の細胞製剤。 The pluripotent stem cell is CD34 negative, CD117 negative, CD146 negative, CD271 negative, NG2 negative, vWF negative, Sox10 negative, Snai1 negative, Slug negative, Tyrp1 negative, and Dct negative. The cell preparation according to claim 1.
  7.  前記多能性幹細胞が、以下の性質の全てを有する多能性幹細胞である、請求項1~6のいずれか1項に記載の細胞製剤:
    (i)テロメラーゼ活性が低いか又は無い;
    (ii)三胚葉のいずれの胚葉の細胞に分化する能力を持つ;
    (iii)腫瘍性増殖を示さない;及び
    (iv)セルフリニューアル能を持つ。
    The cell preparation according to any one of claims 1 to 6, wherein the pluripotent stem cell is a pluripotent stem cell having all of the following properties:
    (I) low or no telomerase activity;
    (Ii) has the ability to differentiate into cells of any germ layer of the three germ layers;
    (Iii) no neoplastic growth; and (iv) self-renewal ability.
  8.  前記多能性幹細胞が、脳腫瘍部位に集積する能力を有する、請求項1~7のいずれか1項に記載の細胞製剤。 The cell preparation according to any one of claims 1 to 7, wherein the pluripotent stem cells have an ability to accumulate at a brain tumor site.
  9.  前記自殺遺伝子が、単純ヘルペスウイルスチミジンキナーゼ(HSVtk)遺伝子、シトシンデアミナーゼ遺伝子、ウラシルホスホリボシルトランスフェラーゼ遺伝子、グアニンホスホリボシルトランスフェラーゼ(gpt)遺伝子又はニトロレダクターゼ遺伝子である、請求項1~8のいずれか1項に記載の細胞製剤。 9. The suicide gene according to claim 1, wherein the herpes simplex virus thymidine kinase (HSVtk) gene, cytosine deaminase gene, uracil phosphoribosyltransferase gene, guanine phosphoribosyltransferase (gpt) gene, or nitroreductase gene. The cell preparation described in 1.
  10.  前記プロドラッグが、自殺遺伝子がHSVtk遺伝子である場合はガンシクロビル(GCV)、アシクロビル、ペンシクロビル、PMEAアデフォビル又はPMPAテノフォビルであり、自殺遺伝子がシトシンデアミナーゼ遺伝子の場合は5-フルオロシトシンであり、自殺遺伝子がウラシルホスホリボシルトランスフェラーゼ遺伝子の場合は5-フルオロウラシルであり、自殺遺伝子がgpt遺伝子の場合は6-チオキサンチン又は6-チオグアニンであり、自殺遺伝子がニトロレダクターゼ(ntr)遺伝子の場合はプロドラッグはCB1954である、請求項1~8のいずれか1項に記載の細胞製剤。 The prodrug is ganciclovir (GCV), acyclovir, pencyclovir, PMEA adefovir or PMPA tenofovir when the suicide gene is an HSVtk gene, and 5-fluorocytosine when the suicide gene is a cytosine deaminase gene. The uracil phosphoribosyltransferase gene is 5-fluorouracil, the suicide gene is 6-thioxanthine or 6-thioguanine when the suicide gene is gpt gene, and the prodrug is CB1954 when the suicide gene is nitroreductase (ntr) gene The cell preparation according to any one of claims 1 to 8, wherein
PCT/JP2014/054910 2013-02-28 2014-02-27 Pluripotent stem cell for treating brain tumor WO2014133090A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2015503024A JPWO2014133090A1 (en) 2013-02-28 2014-02-27 Pluripotent stem cells for brain tumor treatment

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2013-039732 2013-02-28
JP2013039732 2013-02-28

Publications (1)

Publication Number Publication Date
WO2014133090A1 true WO2014133090A1 (en) 2014-09-04

Family

ID=51428349

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2014/054910 WO2014133090A1 (en) 2013-02-28 2014-02-27 Pluripotent stem cell for treating brain tumor

Country Status (2)

Country Link
JP (1) JPWO2014133090A1 (en)
WO (1) WO2014133090A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107073041A (en) * 2014-09-05 2017-08-18 国立大学法人东京大学 The multi-functional stem cell of Diabetic Skin Ulcer treatment
EP3459553A4 (en) * 2016-05-16 2020-01-15 National University Corporation Nagoya University Amelioration and treatment of perinatal brain damage with pluripotent stem cells
CN110869034A (en) * 2017-06-20 2020-03-06 国立大学法人名古屋大学 Amelioration and treatment of brain damage with fetal growth retardation using pluripotent stem cells
EP3622961A4 (en) * 2017-05-09 2021-01-13 Keio University Cell formulation for treating brain tumor

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002526104A (en) * 1998-10-07 2002-08-20 ザ・チルドレンズ・ホスピタル・メデイカル・センター・コーポレーシヨン Transplantable neural progenitors and stem cells for treatment of brain tumors
JP2006345726A (en) * 2005-06-14 2006-12-28 Hamamatsu Univ School Of Medicine Expression vector for treatment of brain tumor
WO2011007900A1 (en) * 2009-07-15 2011-01-20 Dezawa Mari Pluripotent stem cell that can be isolated from body tissue

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002526104A (en) * 1998-10-07 2002-08-20 ザ・チルドレンズ・ホスピタル・メデイカル・センター・コーポレーシヨン Transplantable neural progenitors and stem cells for treatment of brain tumors
JP2006345726A (en) * 2005-06-14 2006-12-28 Hamamatsu Univ School Of Medicine Expression vector for treatment of brain tumor
WO2011007900A1 (en) * 2009-07-15 2011-01-20 Dezawa Mari Pluripotent stem cell that can be isolated from body tissue

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DACHS, GU: "From bench to bedside for gene - directed enzyme prodrug therapy of cancer", ANTICANCER DRUGS, vol. 16, no. 4, 2005, pages 349 - 359 *
HIROKI NANBA ET AL.: "Muse Saibo o Vector to suru Glioma no HSVtk Idenshi Saibo Chiryo", DAI 31 KAI THE JAPAN SOCIETY FOR NEURO-ONCOLOGY GAKUJUTSU SHUKAI PROGRAM SHOROKUSHU, December 2013 (2013-12-01), pages 123 *
HIROKI NANBA: "Akusei Noshuyo Chiryo-yo no Idenshi Hatsugen Vector Saibo no Kaihatsu", CHEMICAL ENGINEERING, vol. 54, no. 11, 2009, pages 813 - 818 *
MARI IDESAWA: "Aratani Hakken sareta Hito Seitai Yurai no Tanosei Kansaibo Muse Saibo: Shinkei Saisei Iryo eno Kanosei", DAI 23 KAI JAPANESE PERIPHERAL NERVE SOCIETY GAKUJUTSU SHUKAI TOKUBETSU KOEN 2, PERIPHERAL NERVE, vol. 23, no. 2, 2012, pages 135 - 139 *
SHIN'ICHIRO KOIZUMI ET AL.: "in vitro, in vivo ni Okeru Akusei Shinkei Koshu eno Mouse iPS Saibo no Idono no Kiso Hyoka", DAI 28 KAI THE JAPAN SOCIETY FOR NEURO-ONCOLOGY PROGRAM SHOROKUSHU, 2010, pages 113 *
SHINJI AMANO ET AL.: "Kotsuzui Kan'yokei Kansaibo o Mochiita glioma ni Taisuru tk Jisatsu Idenshi Ryoho", DAI 27 KAI THE JAPAN SOCIETY FOR NEURO-ONCOLOGY PROGRAM SHOROKUSHU, 2009, pages 72 *
SHINJI AMANO ET AL.: "Kotsuzui Kan'yokei Kansaibo o Mochiita glioma ni Taisuru tk Jisatsu Idenshi Ryoho", DAI 68 KAI ANNUAL MEETING OF THE JAPAN NEUROSURGICAL SOCIETY, 2009, pages 21-0065-06 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107073041A (en) * 2014-09-05 2017-08-18 国立大学法人东京大学 The multi-functional stem cell of Diabetic Skin Ulcer treatment
EP3189844A4 (en) * 2014-09-05 2018-05-02 The University of Tokyo Pluripotent stem cell for treating diabetic skin ulcer
US11000552B2 (en) 2014-09-05 2021-05-11 The University Of Tokyo Pluripotent stem cell for treating diabetic skin ulcer
EP3459553A4 (en) * 2016-05-16 2020-01-15 National University Corporation Nagoya University Amelioration and treatment of perinatal brain damage with pluripotent stem cells
EP3622961A4 (en) * 2017-05-09 2021-01-13 Keio University Cell formulation for treating brain tumor
CN110869034A (en) * 2017-06-20 2020-03-06 国立大学法人名古屋大学 Amelioration and treatment of brain damage with fetal growth retardation using pluripotent stem cells

Also Published As

Publication number Publication date
JPWO2014133090A1 (en) 2017-02-02

Similar Documents

Publication Publication Date Title
Li et al. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity
Li et al. Genetic engineering of platelets to neutralize circulating tumor cells
Yu et al. Overexpression of CXCR4 in mesenchymal stem cells promotes migration, neuroprotection and angiogenesis in a rat model of stroke
WO2014027474A1 (en) Pluripotent stem cell that induces repair and regeneration after myocardial infarction
CN108025026A (en) The purposes of anti-third party's central type memory T cell
KR101507174B1 (en) Immune privileged and modulatory progenitor cells
KR101846464B1 (en) Natural killer cell comprising foreign mitochondria and pharmaceutical composition comprising same
WO1999016863A1 (en) Generation of hematopoietic cells from multipotent neural stem cells
Alvarez-Dolado Cell fusion: biological perspectives and potential for regenerative medicine
WO2014133090A1 (en) Pluripotent stem cell for treating brain tumor
KR20160075676A (en) Method
Huang et al. Class II transactivator knockdown limits major histocompatibility complex II expression, diminishes immune rejection, and improves survival of allogeneic bone marrow stem cells in the infarcted heart
JP2022527932A (en) Method for producing NK cells into which the CAR gene has been introduced and its use
KR102162727B1 (en) Novel cell therapeutics composition by simultaneous administration of human cell-derived extracellular vesicles and cells
WO2015171852A2 (en) Methods and compositions for non-cytotoxic stem cell transplantation
US20130171115A1 (en) Cell-mediated gene therapy for cancer using mesenchymal stem cells expressing a suicide gene
JP2015160820A (en) Pluripotent stem cell for treating chronic renal damage
WO2018207808A1 (en) Cell formulation for treating brain tumor
KR20150112890A (en) Composition for preventing or treating B-cell lymphoma comprising IL-21 expressing mesenchymal stem cells
JP7072777B2 (en) Pluripotent stem cells for the treatment of chronic nephropathy
JP4834291B2 (en) STAT3 activated stem cells
Jung et al. Hematopoietic stem cell engraftment: a direct comparison between intramarrow and intravenous injection in nonhuman primates
Fang et al. Intermittent dosing of G-CSF to ameliorate carbon tetrachloride-induced liver fibrosis in mice
WO2019027298A9 (en) Pharmaceutical composition comprising mesenchymal stem cell expressing trail and cd as effective ingredient for prevention or treatment of cancer
WO2018187469A1 (en) Methods and compositions for non-cytotoxic stem cell transplantation

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: 14756328

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2015503024

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: 14756328

Country of ref document: EP

Kind code of ref document: A1