WO2014042290A1 - Novel use method for cyclic depsipeptide - Google Patents

Novel use method for cyclic depsipeptide Download PDF

Info

Publication number
WO2014042290A1
WO2014042290A1 PCT/JP2013/077279 JP2013077279W WO2014042290A1 WO 2014042290 A1 WO2014042290 A1 WO 2014042290A1 JP 2013077279 W JP2013077279 W JP 2013077279W WO 2014042290 A1 WO2014042290 A1 WO 2014042290A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cells
cell
cyclic depsipeptide
myocardial progenitor
Prior art date
Application number
PCT/JP2013/077279
Other languages
French (fr)
Japanese (ja)
Inventor
中尾 洋一
寛 前島
土井 隆行
山下 潤
英毅 魚崎
福島 弘之
Original Assignee
国立大学法人京都大学
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 国立大学法人京都大学 filed Critical 国立大学法人京都大学
Publication of WO2014042290A1 publication Critical patent/WO2014042290A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C12N5/0657Cardiomyocytes; Heart 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells

Definitions

  • the present invention relates to a pharmaceutical composition containing a cyclic depsipeptide and a method for efficiently differentiating and proliferating cardiomyocytes and / or myocardial progenitor cells using the cyclic depsipeptide.
  • Non-patent Document 1 Since adult cardiomyocytes have lost the ability to divide, it has been considered difficult to repair when the myocardium is damaged by myocardial infarction, myocarditis, aging, or the like. However, it was confirmed by Beltrami et al. That some cardiomyocytes also acquire regenerative ability after infarction (Non-patent Document 1). Furthermore, Laugwitz et al. Revealed the presence of myocardial progenitor cells in the heart of an individual after birth (Non-patent Document 2). However, in view of the decrease in cardiac function after myocardial infarction, it is considered that the normal differentiation and regeneration ability of myocardial progenitor cells present in the adult body is not sufficient to restore cardiac function. Therefore, a compound that proliferates cardiomyocytes has also been reported (Patent Document 1).
  • Patent Document 2 reagents for differentiating and proliferating myocardial progenitor cells (Patent Document 2) produced by mimicking the developmental stage of cardiomyocytes from pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) has been reported (Patent Document 3).
  • pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells)
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • Apratoxin A is a 25-membered ring depsipeptide isolated from cyanobacteria and structurally determined by Luesch et al. In 2001 (Non-Patent Document 3) and known to have antitumor activity. There is no known function related to differentiation and proliferation control.
  • the present invention is to provide a compound for efficiently differentiating and proliferating cardiomyocytes and / or myocardial progenitor cells.
  • the present inventors screened a drug that promotes the differentiation and proliferation of cardiomyocytes and / or myocardial progenitor cells induced to differentiate from mouse ES cells, and it is clear that the differentiation and proliferation of the cells are promoted by a plurality of cyclic depsipeptides. became.
  • the present invention has been completed based on such findings.
  • R 1 and R 3 are each H or C1-C6 alkyl group, R 2 is H, C1-C6 alkyl group, trimethylsilyl group and triethyl
  • R 4 is a substituent selected from the group consisting of silyl groups
  • R 4 is a substituent selected from the group consisting of H, OH, C1-C6 alkoxy groups, F, Cl, Br and I
  • R 1 is an isopropyl group or a t-butyl group
  • R 2 is H
  • R 3 is a methyl group
  • R 4 is a methoxy group
  • the pharmaceutical composition according to [1], wherein the cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII):
  • a method for producing cardiomyocytes comprising a step of adding a cyclic depsipeptide represented by (A) to a cardiac progenitor cell.
  • R 1 is an isopropyl group or a t-butyl group
  • R 2 is H
  • R 3 is a methyl group
  • R 4 is a methoxy group
  • cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII): [8] Any of [5] to [7], wherein the myocardial progenitor cells are in vivo myocardial progenitor cells including side population cells (SP cells), or myocardial progenitor cells derived from pluripotent stem cells. The method described in 1. [9] The method according to [8], wherein the myocardial progenitor cells derived from the pluripotent stem cells are Flk1 (KDR) positive cells.
  • SP cells side population cells
  • KDR Flk1
  • the induction efficiency of the myocardial cell can be remarkably improved. It is particularly useful for the differentiation and proliferation of cells, and is contained in a pharmaceutical composition used for the treatment of heart diseases such as myocardial infarction. Alternatively, it is particularly useful for efficiently obtaining cardiomyocytes by contacting cardiomyocyte progenitor cells with the compound.
  • the differentiation induction rate from myocardial progenitor cells (Flk1-positive cells) to cardiomyocytes ( ⁇ MHC-positive cells) by 21-2 and APT-1 at each concentration is shown.
  • the control is a negative control in the non-addition group, and cyclosporin A is a positive control.
  • the differentiation induction rate from myocardial progenitor cells (Flk1-positive cells) to cardiomyocytes ( ⁇ MHC-positive cells) by each concentration of APT-7, APT-8, APT-9 and APT-10 is shown.
  • the control is a negative control in the non-addition group, and cyclosporin A is a positive control.
  • A shows a protocol for induction from side-population (SP) cells into cardiomyocytes.
  • B shows immunostained images (photographs) of Day 21 cardiomyocytes supplemented with 21-2 using DAPI and anti-cardiac troponin antibody.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the cyclic depsipeptide represented by the above formula (I) or formula (II) or a salt thereof as an active ingredient. Furthermore, since the cyclic depsipeptide can be efficiently obtained by contacting with the myocardial progenitor cell, a method for producing a cardiomyocyte including the step of contacting the compound with the myocardial progenitor cell is provided.
  • a depsipeptide is a peptide having one or more ester bonds
  • a cyclic depsipeptide is a depsipeptide having at least one ring structure.
  • a preferred cyclic depsipeptide in the present invention is a compound represented by formula (I) or formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • R 1 and R 3 are each H (hydrogen atom) or a C1-C6 alkyl group
  • R 2 is a substituent selected from the group consisting of H, C1-C6 alkyl group, a trimethylsilyl (TMS) group and triethylsilyl (TES) group
  • R 4 is H, OH, C1-C6 alkoxy group
  • F A substituent selected from the group consisting of Cl, Br and I
  • the “C1-C6 alkyl group” means a linear, branched or cyclic alkyl group, for example, methyl group, ethyl group, n-propyl group, isopropyl group, cyclopropyl group, n -Butyl group, isobutyl group, sec-butyl group, tert-butyl group, cyclobutyl group, n-pentyl group, isopentyl group, neopentyl group, tert-pentyl group, cyclopeentyl group, n-hexyl group, 2,2-dimethylbutyl Groups, 3,3-dimethylbutyl group, 2-ethylbutyl group, and cyclohexyl group.
  • C1-C6 alkoxy group means a linear, branched or cyclic alkoxy group, for example, a methoxy group, an ethoxy group, an n-propoxy group, an isopropoxy group, a cyclopropyloxy group.
  • the cyclic depsipeptide is a compound described in Formula (III) to Formula (VIII) having a substituent described in Table 1.
  • the cyclic depsipeptide includes an optical isomer or a racemate.
  • pharmaceutically acceptable salts of cyclic depsipeptides include, for example, inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, nitrate, formate, acetate, propionate.
  • a cyclic depsipeptide prodrug is a compound that is hydrolyzed in vivo to be converted into a cyclic depsipeptide.
  • a derivative in which an amino group is substituted with an alkanoyl group (acyl group) that is, an amidated derivative
  • hemiami examples thereof include nal ether derivatives, derivatives substituted with alkoxycarbonyloxymethyl groups, and N-oxide derivatives.
  • the cyclic depsipeptide is Doi T, et al. Chem. Asian J.6, 180, 2011, Doi T, et al. Org. Lett., 8, 531, 2006, Numajiri Y, et al. Chem Asian J, 4, 111, 2009, Zou B, et al. Org Lett, 5, 3503, 2003 and Qi-Yin Chen, et al. ACS Med. Chem. Lett., 2, 861-865, 2011 And can be synthesized by a method analogous thereto.
  • cyanobacteria can be lyophilized and extracted from a dissolved layer in an organic solvent.
  • the present invention also provides a pharmaceutical composition containing a cyclic depsipeptide.
  • the target disease in the present invention is a heart disease, and examples thereof include heart failure, ischemic heart disease or cardiomyopathy.
  • the cyclic depsipeptide of the present invention may be administered as it is alone or mixed with a pharmacologically acceptable carrier, excipient, diluent, etc., and administered orally or parenterally as a pharmaceutical composition of an appropriate dosage form. Can do.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups Agents, emulsions, suspensions and the like.
  • a composition for parenteral administration for example, injections, suppositories and the like are used, and injections include intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, and the like. Dosage forms may be included.
  • excipients eg sugar derivatives such as lactose, sucrose, sucrose, mannitol, sorbitol; starch derivatives such as corn starch, potato starch, alpha starch, dextrin; cellulose derivatives such as crystalline cellulose; Gum arabic; dextran; organic excipients such as pullulan; and silicate derivatives such as light anhydrous silicic acid, synthetic aluminum silicate, calcium silicate and magnesium metasilicate aluminate; phosphates such as calcium hydrogen phosphate; Carbonates such as calcium; inorganic excipients such as sulfates such as calcium sulfate), lubricants (eg, stearic acid metal salts such as stearic acid, calcium stearate, magnesium stearate; talc; Colloidal silica; wax like beeswax and gay wax Borax; adipic acid; sulfate such as sodium sulfate; glycol; fumedi
  • the dose of the cyclic depsipeptide in the present invention can vary depending on various conditions such as patient symptoms, age, weight and the like.
  • the dose varies depending on symptoms, age, etc.
  • the lower limit is 0.1 mg (preferably 0.5 mg) and the upper limit is 1000 mg (preferably 500 mg).
  • the dose may be increased or decreased depending on the symptoms.
  • the present invention also provides a method for producing cardiomyocytes, comprising the step of adding a cyclic depsipeptide to myocardial progenitor cells.
  • This step may be in vitro or in vivo, but is preferably an in vitro method in which a cyclic depsipeptide is added to isolated myocardial progenitor cells.
  • the myocardial cell means a myocardial cell having the characteristics of self-pulsation.
  • the myocardial progenitor cell is a progenitor cell of the cardiomyocyte and has the ability to generate a cardiomyocyte by culture or maturation.
  • a myocardial stem cell and a myocardial progenitor cell contained in a living heart Cells derived from sex stem cells, bone marrow cells, skeletal myoblasts, cells included in adipose-derived mesenchymal cells, or cells differentiated from these cells (JP 2006-115771, JP 2003-325169 and JP-A-2008-307205). Further, myocardial stem cells and myocardial progenitor cells are not particularly distinguished unless otherwise specified.
  • a myocardial progenitor cell contained in a living heart cells (SP cells) existing in a side population among cells contained in the heart are exemplified (Oyama T, et al. J Cell Biol.
  • the SP cell means a cell having a high excretion ability with respect to a DNA fluorescent dye called Hoechst33342. Cardiomyocytes and myocardial progenitor cells may be mixed with each other or isolated.
  • cardiomyocytes are also characterized by being positive for myocardial markers cardiac troponin (cTNT or troponin T type 2) and / or ⁇ MHC ( ⁇ myosin heavy chain).
  • myocardial progenitor cells are not particularly limited, but by being positive for Flk1 / KDR, islet1, Wt1 and / or N-Cadherin and / or present in the side population (SP cell fraction) in FACS analysis Characterized.
  • cardiac progenitor cells may be prepared by differentiating pluripotent stem cells in vitro.
  • the pluripotent stem cell is a stem cell having pluripotency that can be differentiated into all cells existing in a living body and also having proliferative ability, and is not particularly limited, for example, Embryonic stem (ES) cells, embryonic stem (ntES) cells derived from cloned embryos obtained by nuclear transfer, sperm stem cells (“GS cells”), embryonic germ cells (“EG cells”), induced pluripotent stems (iPS) cells, cultured fibroblasts, pluripotent cells derived from bone marrow stem cells (Muse cells) and the like are included.
  • ES Embryonic stem
  • ntES embryonic stem
  • GS cells sperm stem cells
  • EG cells embryonic germ cells
  • iPS induced pluripotent stems
  • cultured fibroblasts pluripotent cells derived from bone marrow stem cells (Muse cells) and the like are
  • Embryonic stem cells ES cells are stem cells established from the inner cell mass of early embryos (for example, blastocysts) of mammals such as humans and mice, and having pluripotency and proliferation ability by self-replication.
  • ES cells are embryonic stem cells derived from the inner cell mass of the blastocyst, the embryo after the morula, in the 8-cell stage of a fertilized egg, and have the ability to differentiate into any cell that constitutes an adult, so-called differentiation. And ability to proliferate by self-replication.
  • ES cells were discovered in mice in 1981 (MJ Evans and MH Kaufman (1981), Nature 292: 154-156), and then ES cell lines were established in primates such as humans and monkeys (JA Thomson et al. (1998), Science 282: 1145-1147; JA Thomson et al. (1995), Proc. Natl. Acad. Sci. USA, 92: 7844-7848; JA Thomsonet al. (1996), Biol. Reprod. 55: 254-259; JA Thomson and VS Marshall (1998), Curr. Top. Dev. Biol., 38: 133-165).
  • ES cells can be established by taking an inner cell mass from a blastocyst of a fertilized egg of a target animal and culturing the inner cell mass on a fibroblast feeder. In addition, maintenance of cells by subculture is performed using a culture solution to which substances such as leukemia inhibitory factor (LIF) and basic fibroblast growth factor (basic fibroblast growth factor (bFGF)) are added. It can be carried out.
  • LIF leukemia inhibitory factor
  • bFGF basic fibroblast growth factor
  • DMEM / F-12 culture medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM non-essential amino acid, 2 mM L-glutamic acid, 20% KSR and 4 ng / ml bFGF is used as the culture medium for ES cell production.
  • Human ES cells can be maintained in a humid atmosphere of 37 ° C., 2% CO 2 /98% air (O. Fumitaka et al. (2008), Nat. Biotechnol., 26: 215-224).
  • ES cells also need to be passaged every 3-4 days, where passage is eg 0.25% trypsin and 0.1 mg / ml collagenase IV in PBS containing 1 mM CaCl 2 and 20% KSR. Can be used.
  • ES cells can be generally selected by Real-Time PCR using the expression of gene markers such as alkaline phosphatase, Oct-3 / 4, Nanog as an index.
  • gene markers such as alkaline phosphatase, Oct-3 / 4, Nanog
  • OCT-3 / 4, NANOG, and ECAD can be used as an index (E. Kroon et al. (2008), Nat. Biotechnol., 26: 443). -452).
  • Human ES cell lines for example, WA01 (H1) and WA09 (H9) are obtained from the WiCell Research Institute, and KhES-1, KhES-2 and KhES-3 are obtained from the Institute of Regenerative Medicine (Kyoto, Japan), Kyoto University Is possible.
  • sperm stem cells are testis-derived pluripotent stem cells that are the origin of spermatogenesis. Like ES cells, these cells can be induced to differentiate into various types of cells, and have characteristics such as the ability to create chimeric mice when transplanted into mouse blastocysts (M. Kanatsu-Shinohara et al. ( 2003) Biol. Reprod., 69: 612-616; K. Shinohara et al. (2004), Cell, 119: 1001-1012).
  • GDNF glial cell line-derived neurotrophic factor
  • Embryonic germ cells are cells that are established from embryonic primordial germ cells and have the same pluripotency as ES cells, such as LIF, bFGF, stem cell factor, etc. It can be established by culturing primordial germ cells in the presence of these substances (Y. Matsui et al. (1992), Cell, 70: 841-847; JL Resnick et al. (1992), Nature, 359: 550 -551).
  • iPS Artificial pluripotent stem cells
  • somatic cells in the form of DNA or protein, which is almost equivalent to ES cells
  • It is an artificial stem cell derived from a somatic cell having the characteristics of, for example, differentiation pluripotency and proliferation ability by self-replication (K. Takahashi and S. Yamanaka (2006) Cell, 126: 663-676; K. Takahashi et al (2007), Cell, 131: 861-872; J. Yu et al. (2007), Science, 318: 1917-1920; Nakagawa, M. et al., Nat. Biotechnol.
  • the reprogramming factor is a gene specifically expressed in ES cells, its gene product or non-cording RNA, a gene that plays an important role in maintaining undifferentiation of ES cells, its gene product or non-coding RNA, or It may be constituted by a low molecular compound.
  • genes included in the reprogramming factor include Oct3 / 4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15 -2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3 or Glis1 etc. are exemplified, and these reprogramming factors may be used alone or in combination.
  • the reprogramming factors include histone deacetylase (HDAC) inhibitors [for example, small molecule inhibitors such as valproate (VPA), trichostatin A, sodium butyrate, MC 1293, M344, siRNA and shRNA against HDAC (eg , Nucleic acid expression inhibitors such as HDAC1 siRNA Smartpool ((Millipore), HuSH 29mer shRNA Constructs against HDAC1 (OriGene), etc.)], MEK inhibitors (eg PD184352, PD98059, U0126, SL327 and PD0325901), Glycogen synthase kin -3 inhibitors (eg, Bio and CHIR99021), DNA methyltransferase inhibitors (eg, 5-azacytidine), histone methyltransferase inhibitors (eg, small molecule inhibitors such as BIX-01294, Suv39hl, Suv39h2, SetDBl and G9a Nucleic acid expression inhibitors such as si
  • the reprogramming factor may be introduced into a somatic cell by a technique such as lipofection, fusion with a cell membrane-permeable peptide (for example, HIV-derived TAT and polyarginine), or microinjection.
  • a cell membrane-permeable peptide for example, HIV-derived TAT and polyarginine
  • Virus vectors include retrovirus vectors, lentivirus vectors (cell, 126, pp.663-676, 2006; Cell, 131, pp.861-872, 2007; Science, 318, pp.1917-1920, 2007 ), Adenovirus vectors (Science, 322, 945-949, 2008), adeno-associated virus vectors, Sendai virus vectors (WO 2010/008054) and the like.
  • artificial chromosome vectors examples include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), and bacterial artificial chromosomes (BAC, PAC).
  • HAC human artificial chromosomes
  • YAC yeast artificial chromosomes
  • BAC bacterial artificial chromosomes
  • a plasmid a plasmid for mammalian cells can be used (Science, 322: 949-953, 2008).
  • the vector can contain regulatory sequences such as a promoter, enhancer, ribosome binding sequence, terminator, polyadenylation site, etc. so that a nuclear reprogramming substance can be expressed.
  • Selective marker sequences such as kanamycin resistance gene, ampicillin resistance gene, puromycin resistance gene, thymidine kinase gene, diphtheria toxin gene, reporter gene sequences such as green fluorescent protein (GFP), ⁇ -glucuronidase (GUS), FLAG, etc.
  • GFP green fluorescent protein
  • GUS ⁇ -glucuronidase
  • FLAG FLAG
  • the above vector has a LoxP sequence before and after the introduction of the gene into a somatic cell in order to excise the gene or promoter encoding the reprogramming factor and the gene encoding the reprogramming factor that binds to it. May be.
  • RNA it may be introduced into somatic cells by techniques such as lipofection and microinjection, and in order to suppress degradation, RNA incorporating 5-methylcytidine and pseudouridine® (TriLink® Biotechnologies) is used. Yes (Warren L, (2010) Cell Stem Cell. 7: 618-630).
  • Examples of the culture medium for inducing iPS cells include DMEM, DMEM / F12 or DME culture medium containing 10 to 15% FBS (these culture media include LIF, penicillin / streptomycin, puromycin, L-glutamine). , Non-essential amino acids, ⁇ -mercaptoethanol, etc.) or commercially available culture media (eg, culture media for mouse ES cell culture (TX-WES culture solution, Thrombo X), primate ES cells) Culture medium for culture (primate ES / iPS cell culture medium, Reprocell), serum-free medium (mTeSR, Stemcell Technology).
  • DMEM DMEM / F12 or DME culture medium containing 10 to 15% FBS
  • these culture media include LIF, penicillin / streptomycin, puromycin, L-glutamine). , Non-essential amino acids, ⁇ -mercaptoethanol, etc.
  • commercially available culture media eg, culture media for mouse ES cell culture (TX
  • a somatic cell is brought into contact with a reprogramming factor on a DMEM or DMEM / F12 medium containing 10% FBS at 37 ° C. in the presence of 5% CO 2 for about 4 to 7 days. Then, re-spread the cells on feeder cells (for example, mitomycin C-treated STO cells, SNL cells, etc.), and use bFGF-containing primate ES cell culture medium about 10 days after contact between the somatic cells and the reprogramming factor. Culturing and generating iPS-like colonies about 30 to about 45 days or more after the contact.
  • feeder cells for example, mitomycin C-treated STO cells, SNL cells, etc.
  • 10% FBS-containing DMEM medium including LIF, penicillin / streptomycin, etc.
  • feeder cells eg, mitomycin C-treated STO cells, SNL cells, etc.
  • 5% CO 2 at 37 ° C. can be suitably included with puromycin, L-glutamine, non-essential amino acids, ⁇ -mercaptoethanol, etc.
  • ES-like colonies after about 25 to about 30 days or more .
  • somatic cells to be reprogrammed themselves are used (Takahashi K, et al. (2009), PLoS One. 4: e8067 or WO2010 / 137746), or extracellular matrix (eg, Laminin- 5 (WO2009 / 123349) and Matrigel (BD)) are exemplified.
  • iPS cells may be established under hypoxic conditions (oxygen concentration of 0.1% or more and 15% or less) (Yoshida Y, et al. (2009), Cell Stem Cell. 5: 237 -241 or WO2010 / 013845).
  • the culture medium is exchanged with a fresh culture medium once a day from the second day onward.
  • the number of somatic cells used for nuclear reprogramming is not limited, but ranges from about 5 ⁇ 10 3 to about 5 ⁇ 10 6 cells per 100 cm 2 of culture dish.
  • IPS cells can be selected according to the shape of the formed colonies.
  • a drug resistance gene that is expressed in conjunction with a gene that is expressed when somatic cells are initialized for example, Oct3 / 4, Nanog
  • a culture solution containing the corresponding drug selection The established iPS cells can be selected by culturing with the culture medium.
  • the marker gene is a fluorescent protein gene
  • iPS cells are selected by observing with a fluorescence microscope, in the case of a luminescent enzyme gene, by adding a luminescent substrate, and in the case of a chromogenic enzyme gene, by adding a chromogenic substrate can do.
  • the term “somatic cell” refers to any animal cell (preferably, a mammalian cell including a human) except a germ line cell such as an egg, oocyte, ES cell, or totipotent cell.
  • Somatic cells include, but are not limited to, fetal (pup) somatic cells, neonatal (pup) somatic cells, and mature healthy or diseased somatic cells. , Passage cells, and established cell lines.
  • somatic cells include, for example, (1) neural stem cells, hematopoietic stem cells, mesenchymal stem cells, tissue stem cells such as dental pulp stem cells (somatic stem cells), (2) tissue progenitor cells, (3) lymphocytes, epithelium Cells, endothelial cells, muscle cells, fibroblasts (skin cells, etc.), hair cells, hepatocytes, gastric mucosal cells, enterocytes, spleen cells, pancreatic cells (exocrine pancreas cells, etc.), brain cells, lung cells, kidney cells Examples thereof include differentiated cells such as fat cells.
  • somatic cells having the same or substantially the same HLA genotype as the transplant destination individual from the viewpoint that rejection does not occur.
  • substantially the same means that the HLA genotype matches the transplanted cells to such an extent that an immune response can be suppressed by an immunosuppressive agent.
  • HLA-A, HLA-B And somatic cells having an HLA type in which 3 loci of HLA-DR or 4 loci plus HLA-C are matched.
  • E Cloned embryo-derived ES cells obtained by nuclear transfer nt ES cells are cloned embryo-derived ES cells produced by nuclear transfer technology and have almost the same characteristics as ES cells derived from fertilized eggs (T. Wakayama et al. (2001), Science, 292: 740-743; S. Wakayama et al. (2005), Biol. Reprod., 72: 932-936; J. Byrne et al. (2007) , Nature, 450: 497-502).
  • an ES cell established from an inner cell mass of a clonal embryo-derived blastocyst obtained by replacing the nucleus of an unfertilized egg with the nucleus of a somatic cell is an nt ES (nuclear transfer ES) cell.
  • nt ES nuclear transfer ES
  • nuclear transfer technology JB Cibelli et al. (1998), Nature Biotechnol., 16: 642-646) and ES cell production technology (above) is used (Wakayama). Seika et al. (2008), Experimental Medicine, Vol. 26, No. 5 (extra number), 47-52).
  • Nuclear transfer can be initialized by injecting a somatic cell nucleus into a mammal's enucleated unfertilized egg and culturing for several hours.
  • Muse cells are pluripotent stem cells produced by the method described in WO2011 / 007900. Specifically, fibroblasts or bone marrow stromal cells are treated with trypsin for a long time, preferably 8 or 16 hours. It is a pluripotent cell obtained by suspension culture after treatment, and is positive for SSEA-3 and CD105.
  • the method for inducing differentiation from pluripotent stem cells to myocardial progenitor cells is not particularly limited.
  • the following method can be used.
  • Pluripotent stem cells may be separated by any method, and adhesion culture or / and co-culture with feeder cells may be performed using a culture dish subjected to suspension culture or coating treatment. Further, suspension culture and adhesion culture may be performed in combination.
  • a separation method a separation solution having mechanical, protease activity and collagenase activity (for example, Accutase (TM) and Accumax (TM)) or a separation solution having only collagenase activity may be used.
  • TM Accutase
  • TM Accutase
  • TM Accutase activity
  • TM Accutase activity
  • TM Accutase activity
  • TM Accutase activity
  • TM Accumax
  • the surface of the culture dish is not artificially treated (for example, coated with an extracellular matrix or the like) for the purpose of improving adhesion to cells, or artificially adhered.
  • poly-HEMA polyhydroxyethyl methacrylic acid
  • a culture dish coated with matrigel (BD) type I collagen, type IV collagen, gelatin, laminin, heparan sulfate proteoglycan, or entactin, and combinations thereof can be used.
  • the cells used for co-culture are OP9 cells (Nishikawa, SI et al, Development 125, 1747-1757 (1998)) or END-2 cells (Mummery C, et al, Circulation. 107: 2733-40 (2003 )) Is exemplified.
  • the medium in this step can be prepared using a medium used for animal cell culture as a basal medium.
  • the basal medium include IMDM medium, Medium ⁇ 199 medium, Eagle's'Minimum Essential Medium (EMEM) medium, ⁇ MEM medium, Doulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium Etc. are included. ⁇ MEM or DMEM is preferable.
  • the medium may contain serum or may be serum-free.
  • albumin transferrin, Knockout Serum Replacement (KSR) (serum substitute for FBS during ES cell culture), fatty acid, insulin, collagen precursor, trace element, 2-mercaptoethanol, 3'-thiol
  • KSR Knockout Serum Replacement
  • fatty acid insulin, collagen precursor, trace element, 2-mercaptoethanol, 3'-thiol
  • serum replacements such as glycerol, ITS-supplements, B27-supplements, N2-supplements, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), non-essential amino acids, vitamins, cytokines, Wnt It may also contain one or more substances such as signal inhibitors, antibiotics, antioxidants, pyruvate, buffers, inorganic salts. Examples of cytokines include activin A and BMP4.
  • XAV939 As Wnt signal inhibitors, XAV939 (Shih-Min A. Huang, et al, Nature 461, 614-620, 2009), vitamin A (retinoic acid), lithium, flavonoid, Dickkopf1 (Dkk1), insulin-like growth factor binding protein Examples include (IGFBP) (WO2009 / 131166), siRNA for ⁇ -catenin, and the like.
  • the culture temperature is not limited to the following, but is about 30 to 40 ° C., preferably about 37 ° C.
  • the culture is performed in an atmosphere of CO 2 -containing air, and the CO 2 concentration is preferably about 2 to 5%. is there.
  • the culture time is the number of days required for Flk1 / KDR to be expressed, for example, 4 days or longer.
  • the step of adding a cyclic depsipeptide for producing cardiomyocytes to myocardial progenitor cells can be performed under the same conditions as in the differentiation induction method from pluripotent stem cells to myocardial progenitor cells.
  • the period during which the cyclic depsipeptide is added is the number of days required for cTNT or ⁇ MHC to be expressed, for example, 6 days or longer.
  • the concentration at which cyclic depsipeptide is added is appropriately selected according to the type of progenitor cells and cyclic depsipeptide to be used, but is preferably 0.25 nM to 25 nM.
  • APT-7 (formula (V)
  • APT-8 (formula (VI))
  • APT-9 (formula (VII)
  • APT-10 (formula (VIII)
  • APT-7, APT-8, APT-9 and APT-10 were able to confirm remarkable activity in the concentration ranges of 10 nM, 7.5 nM, 7.5 nM to 20 nM and 1 nM to 1.5 nM, respectively. These activities were higher than cyclosporin A.
  • SP cells Side population cells in cardiomyocytes were isolated using the method of Oyama et al. (Oyama T, et al. J Cell Biol. 176, 329-341, 2007). Specifically, cardiomyocytes extracted from newborn rats were suspended at 1.0 ⁇ 10 6 cells / ml in PBS containing 3% FBS, 1 ⁇ g / ml Hoechst 33342 was added, and incubated at 37 ° C. for 60 minutes in the dark. Thereafter, the cells were isolated as SP cells using a flow cytometer.
  • SP cells were excited as Hoechst 33342 with a 350 nm UV laser and separated as cells detected at specific locations at 450 nm (Hoechst blue) and 675 nm (Hoechst red) (Goodell MA, et al. J Exp Med. 183 : 1797-1806, 1996). At this time, cells stained with Propidium iodide were removed as dead cells. The obtained SP cells were cultured in IMDM containing 10% FBS, and after 21 hours, 21-2 was added and cultivation was continued for 72 hours. Furthermore, the culture was continued under the condition of no addition of 21-2, and the cells were observed by cTNT antibody staining 3 weeks after SP cell isolation (FIG. 3A). The result is shown in FIG. 3B.
  • cardiomyocytes with sarcomere structure were induced and partly pulsated. Based on the above, it is presumed that 21-2 has at least the same function as oxytocin and trichostatin A, which have been reported so far, capable of inducing differentiation from myocardial SP cells to cardiomyocytes.
  • cyclic depsipeptide shown here has the ability to induce cardiomyocyte differentiation from myocardial progenitor cells.
  • the efficiency of inducing cardiomyocytes from myocardial progenitor cells can be remarkably improved, which is particularly useful for the treatment of heart diseases or cell transplantation using pluripotent stem cells.

Abstract

 A pharmaceutical composition containing at least one compound selected from the group consisting of a cyclic depsipeptide represented by formula (I) or (II), medically acceptable salts of same, solvates of same and prodrugs of same, and a manufacturing method for cardiomyocytes, said method including a step for bringing the above-mentioned compound into contact with cardiac progenitor cells. (In the formulas: R1 and R3 are each independently H or a C1-C6 alkyl group; R2 is a substituent group selected from the group consisting of H, a C1-C6 alkyl group, a trimethylsilyl group and a triethylsilyl group; R4 is a substituent group selected from the group consisting of H, OH, a C1-C6 alkoxy group, F, Cl, Br and I; L-M has a structure selected from the group consisting of CH=C(Me), CH=CH, CH2-CH2 and CH2-CH(Me); X is O or S; and n is 1 or 2.)

Description

環状デプシペプチドの新規使用方法Novel usage of cyclic depsipeptide
 本発明は、環状デプシペプチドを含有する医薬組成物ならびに環状デプシペプチドを用いて心筋細胞および/または心筋前駆細胞を効率よく分化増殖させるための方法に関する。 The present invention relates to a pharmaceutical composition containing a cyclic depsipeptide and a method for efficiently differentiating and proliferating cardiomyocytes and / or myocardial progenitor cells using the cyclic depsipeptide.
 成体の心筋細胞は分裂能を失っていることから、心筋梗塞、心筋炎または老化等により心筋が損傷された場合、修復をすることが困難と考えられていた。しかし、Beltramiらにより、心筋細胞の中にも梗塞後に再生能を獲得する細胞があることが確認された(非特許文献1)。さらに、Laugwitzらが、出生後の個体の心臓に心筋前駆細胞の存在を明らかとした(非特許文献2)。しかしながら、心筋梗塞後の心機能の低下を鑑みると、成体内に存在する心筋前駆細胞の通常の分化再生能のみでは、心機能を回復するには十分でないと考えられる。そこで、心筋細胞を増殖させる化合物なども報告されている(特許文献1)。 Since adult cardiomyocytes have lost the ability to divide, it has been considered difficult to repair when the myocardium is damaged by myocardial infarction, myocarditis, aging, or the like. However, it was confirmed by Beltrami et al. That some cardiomyocytes also acquire regenerative ability after infarction (Non-patent Document 1). Furthermore, Laugwitz et al. Revealed the presence of myocardial progenitor cells in the heart of an individual after birth (Non-patent Document 2). However, in view of the decrease in cardiac function after myocardial infarction, it is considered that the normal differentiation and regeneration ability of myocardial progenitor cells present in the adult body is not sufficient to restore cardiac function. Therefore, a compound that proliferates cardiomyocytes has also been reported (Patent Document 1).
 近年、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心筋細胞の発生段階を模倣して製造した心筋前駆細胞(特許文献2)を分化増殖させる試薬が報告されている(特許文献3)。 In recent years, reagents for differentiating and proliferating myocardial progenitor cells (Patent Document 2) produced by mimicking the developmental stage of cardiomyocytes from pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) Has been reported (Patent Document 3).
 アプラトキシンA は、2001 年にLueschらによって、シアノバクテリアより単離され、構造決定された25員環デプシペプチドであり(非特許文献3)、抗腫瘍活性を有することが知られているが、心筋分化増殖制御に関する機能については知られていない。 Apratoxin A is a 25-membered ring depsipeptide isolated from cyanobacteria and structurally determined by Luesch et al. In 2001 (Non-Patent Document 3) and known to have antitumor activity. There is no known function related to differentiation and proliferation control.
WO2007/048352WO2007 / 048352 WO2009/118928WO2009 / 118928 WO2012/067266WO2012 / 067266
 本発明は心筋細胞および/または心筋前駆細胞を効率よく分化増殖させるための化合物を提供することである。 The present invention is to provide a compound for efficiently differentiating and proliferating cardiomyocytes and / or myocardial progenitor cells.
 本発明者らはマウスES細胞から分化誘導した心筋細胞および/または心筋前駆細胞の分化増殖を促進させる薬剤をスクリーニングしたところ、複数の環状デプシペプチドにより該細胞の分化増殖が促進されることが明らかとなった。本発明はこのような知見に基づき完成するに至ったものである。 The present inventors screened a drug that promotes the differentiation and proliferation of cardiomyocytes and / or myocardial progenitor cells induced to differentiate from mouse ES cells, and it is clear that the differentiation and proliferation of the cells are promoted by a plurality of cyclic depsipeptides. became. The present invention has been completed based on such findings.
 すなわち、本発明は以下の特徴を有する。
[1] 以下の式(I)または式(II)(式中、R1およびR3はそれぞれHまたはC1-C6アルキル基であり、R2はH、C1-C6アルキル基、トリメチルシリル基およびトリエチルシリル基からなる群から選択される置換基であり、R4はH、OH、C1-C6アルコキシ基、F、Cl、BrおよびIからなる群から選択される置換基であり、L-MはCH=C(Me)、CH=CH、CH2-CH2およびCH2-CH(Me)からなる群から選択される構造を有し、Xは、OまたはSであり、nは、1または2である)で表される環状デプシペプチドまたはその薬学上許容される塩、溶媒和物もしくはプロドラッグを有効成分として含有する医薬組成物。
Figure JPOXMLDOC01-appb-C000017
Figure JPOXMLDOC01-appb-C000018
[2] R1がイソプロピル基またはt-ブチル基であり、R2がHであり、R3がメチル基であり、R4がメトキシ基であり、L-MがCH=C(Me)であり、XがOまたはSであり、nが1である、[1]に記載の医薬組成物。
[3] 環状デプシペプチドが、次の式(III)から式(VIII)で表される化合物から成る群より選択される、[1]に記載の医薬組成物;
Figure JPOXMLDOC01-appb-C000019
Figure JPOXMLDOC01-appb-C000020
Figure JPOXMLDOC01-appb-C000021
Figure JPOXMLDOC01-appb-C000022
Figure JPOXMLDOC01-appb-C000023
Figure JPOXMLDOC01-appb-C000024
[4] 心不全、虚血性心疾患または心筋症から選択される心疾患の治療に用いられる[1]~[3]のいずれかに記載の医薬組成物。
[5] 以下の式(I)または式(II)(式中、R1およびR3はそれぞれHまたはC1-C6アルキル基であり、R2はH、C1-C6アルキル基、トリメチルシリル基およびトリエチルシリル基からなる群から選択される置換基であり、R4はH、OH、C1-C6アルコキシ基、F、Cl、BrおよびIからなる群から選択される置換基であり、L-MはCH=C(Me)、CH=CH、CH2-CH2およびCH2-CH(Me)からなる群から選択される構造を有し、Xは、OまたはSであり、nは、1または2である)で表される環状デプシペプチドを心筋前駆細胞へ添加する工程を含む、心筋細胞の製造方法。
Figure JPOXMLDOC01-appb-C000025
Figure JPOXMLDOC01-appb-C000026
[6] R1がイソプロピル基またはt-ブチル基であり、R2がHであり、R3がメチル基であり、R4がメトキシ基であり、L-MがCH=C(Me)であり、XがOまたはSであり、nが1である、[5]に記載の方法。
[7] 環状デプシペプチドが、次の式(III)から式(VIII)で表される化合物から成る群より選択される、[5]に記載の方法。
Figure JPOXMLDOC01-appb-C000027
Figure JPOXMLDOC01-appb-C000028
Figure JPOXMLDOC01-appb-C000029
Figure JPOXMLDOC01-appb-C000030
Figure JPOXMLDOC01-appb-C000031
Figure JPOXMLDOC01-appb-C000032
[8] 前記心筋前駆細胞が、side population細胞(SP細胞)を含む生体内の心筋前駆細胞、または多能性幹細胞から誘導された心筋前駆細胞である、[5]~[7]のいずれかに記載の方法。
[9] 前記多能性幹細胞から誘導された心筋前駆細胞が、Flk1 (KDR)陽性の細胞である、[8]に記載の方法。
That is, the present invention has the following features.
[1] The following formula (I) or formula (II) (wherein R 1 and R 3 are each H or C1-C6 alkyl group, R 2 is H, C1-C6 alkyl group, trimethylsilyl group and triethyl) R 4 is a substituent selected from the group consisting of silyl groups, R 4 is a substituent selected from the group consisting of H, OH, C1-C6 alkoxy groups, F, Cl, Br and I, and LM is CH = Having a structure selected from the group consisting of C (Me), CH = CH, CH 2 -CH 2 and CH 2 -CH (Me), X is O or S, and n is 1 or 2. Or a pharmaceutically acceptable salt, solvate or prodrug thereof as an active ingredient.
Figure JPOXMLDOC01-appb-C000017
Figure JPOXMLDOC01-appb-C000018
[2] R 1 is an isopropyl group or a t-butyl group, R 2 is H, R 3 is a methyl group, R 4 is a methoxy group, LM is CH = C (Me), The pharmaceutical composition according to [1], wherein X is O or S, and n is 1.
[3] The pharmaceutical composition according to [1], wherein the cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII):
Figure JPOXMLDOC01-appb-C000019
Figure JPOXMLDOC01-appb-C000020
Figure JPOXMLDOC01-appb-C000021
Figure JPOXMLDOC01-appb-C000022
Figure JPOXMLDOC01-appb-C000023
Figure JPOXMLDOC01-appb-C000024
[4] The pharmaceutical composition according to any one of [1] to [3], which is used for the treatment of a heart disease selected from heart failure, ischemic heart disease or cardiomyopathy.
[5] The following formula (I) or formula (II) (wherein R 1 and R 3 are each H or C1-C6 alkyl group, R 2 is H, C1-C6 alkyl group, trimethylsilyl group and triethyl R 4 is a substituent selected from the group consisting of silyl groups, R 4 is a substituent selected from the group consisting of H, OH, C1-C6 alkoxy groups, F, Cl, Br and I, and LM is CH = Having a structure selected from the group consisting of C (Me), CH = CH, CH 2 -CH 2 and CH 2 -CH (Me), X is O or S, and n is 1 or 2. A method for producing cardiomyocytes, comprising a step of adding a cyclic depsipeptide represented by (A) to a cardiac progenitor cell.
Figure JPOXMLDOC01-appb-C000025
Figure JPOXMLDOC01-appb-C000026
[6] R 1 is an isopropyl group or a t-butyl group, R 2 is H, R 3 is a methyl group, R 4 is a methoxy group, LM is CH = C (Me), The method according to [5], wherein X is O or S, and n is 1.
[7] The method according to [5], wherein the cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII):
Figure JPOXMLDOC01-appb-C000027
Figure JPOXMLDOC01-appb-C000028
Figure JPOXMLDOC01-appb-C000029
Figure JPOXMLDOC01-appb-C000030
Figure JPOXMLDOC01-appb-C000031
Figure JPOXMLDOC01-appb-C000032
[8] Any of [5] to [7], wherein the myocardial progenitor cells are in vivo myocardial progenitor cells including side population cells (SP cells), or myocardial progenitor cells derived from pluripotent stem cells. The method described in 1.
[9] The method according to [8], wherein the myocardial progenitor cells derived from the pluripotent stem cells are Flk1 (KDR) positive cells.
 心筋前駆細胞に、上記式(I)または式(II)で表される化合物を接触させると、心筋細胞の誘導効率を顕著に向上させることができるので、これらの化合物は、心筋前駆細胞または心筋細胞の分化増殖のために特に有用であり、心筋梗塞等の心疾患の治療に用いられる医薬組成物へ含有される。または、心筋前駆細胞を当該化合物と接触させることで、心筋細胞を効率良く得るために特に有用である。 When the compound represented by the above formula (I) or formula (II) is brought into contact with the myocardial progenitor cell, the induction efficiency of the myocardial cell can be remarkably improved. It is particularly useful for the differentiation and proliferation of cells, and is contained in a pharmaceutical composition used for the treatment of heart diseases such as myocardial infarction. Alternatively, it is particularly useful for efficiently obtaining cardiomyocytes by contacting cardiomyocyte progenitor cells with the compound.
各濃度の21-2およびAPT-1による心筋前駆細胞(Flk1陽性細胞)から心筋細胞(αMHC陽性細胞)への分化誘導率を示す。図中、コントロールは無添加群の陰性対照であり、シクロスポリンAは陽性対照である。The differentiation induction rate from myocardial progenitor cells (Flk1-positive cells) to cardiomyocytes (αMHC-positive cells) by 21-2 and APT-1 at each concentration is shown. In the figure, the control is a negative control in the non-addition group, and cyclosporin A is a positive control. 各濃度のAPT-7、APT-8、APT-9およびAPT-10による心筋前駆細胞(Flk1陽性細胞)から心筋細胞(αMHC陽性細胞)への分化誘導率を示す。図中、コントロールは無添加群の陰性対照であり、シクロスポリンAは陽性対照である。The differentiation induction rate from myocardial progenitor cells (Flk1-positive cells) to cardiomyocytes (αMHC-positive cells) by each concentration of APT-7, APT-8, APT-9 and APT-10 is shown. In the figure, the control is a negative control in the non-addition group, and cyclosporin A is a positive control. Aに、Side Population(SP)細胞から心筋細胞への誘導プロトコールを示す。Bに、21-2を添加したDay21の心筋細胞のDAPIおよび抗心筋トロポニン抗体を用いた免疫染色像(写真)を示す。A shows a protocol for induction from side-population (SP) cells into cardiomyocytes. B shows immunostained images (photographs) of Day 21 cardiomyocytes supplemented with 21-2 using DAPI and anti-cardiac troponin antibody.
 本発明は、上記の式(I)式または式(II)で表される環状デプシペプチドまたはその塩を有効成分として含有する医薬組成物を提供する。さらに、当該環状デプシペプチドは、心筋前駆細胞と接触させることにより、心筋細胞を効率良く入手できることから、当該化合物と心筋前駆細胞を接触させる工程を含む心筋細胞の製造方法を提供する。 The present invention provides a pharmaceutical composition comprising the cyclic depsipeptide represented by the above formula (I) or formula (II) or a salt thereof as an active ingredient. Furthermore, since the cyclic depsipeptide can be efficiently obtained by contacting with the myocardial progenitor cell, a method for producing a cardiomyocyte including the step of contacting the compound with the myocardial progenitor cell is provided.
 本発明において、デプシペプチドとは、一つ以上のエステル結合を有するペプチドであり、環状デプシペプチドとは、少なくとも1つの環構造を有するデプシペプチドである。 In the present invention, a depsipeptide is a peptide having one or more ester bonds, and a cyclic depsipeptide is a depsipeptide having at least one ring structure.
 本発明において好ましい環状デプシペプチドは、式(I)または式(II)で表される化合物、またはその薬学上許容される塩、溶媒和物若しくはプロドラッグである。
Figure JPOXMLDOC01-appb-C000033
Figure JPOXMLDOC01-appb-C000034
(式中、R1およびR3がそれぞれH(水素原子)またはC1-C6アルキル基であり、
R2はH、C1-C6アルキル基、トリメチルシリル(TMS)基およびトリエチルシリル(TES)基からなる群から選択される置換基であり、R4はH、OH、C1-C6アルコキシ基、F、Cl、BrおよびIからなる群から選択される置換基であり、L-MはC(Me)=CH、CH=CH、CH2-CH2およびCH(Me)-CH2からなる群から選択される構造を有し、Xは、O(酸素原子)またはS(硫黄原子)であり、nは、1または2である。)
A preferred cyclic depsipeptide in the present invention is a compound represented by formula (I) or formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof.
Figure JPOXMLDOC01-appb-C000033
Figure JPOXMLDOC01-appb-C000034
Wherein R 1 and R 3 are each H (hydrogen atom) or a C1-C6 alkyl group,
R 2 is a substituent selected from the group consisting of H, C1-C6 alkyl group, a trimethylsilyl (TMS) group and triethylsilyl (TES) group, R 4 is H, OH, C1-C6 alkoxy group, F, A substituent selected from the group consisting of Cl, Br and I, and LM is selected from the group consisting of C (Me) = CH, CH = CH, CH 2 -CH 2 and CH (Me) -CH 2 X is O (oxygen atom) or S (sulfur atom), and n is 1 or 2. )
 本発明において、「C1-C6アルキル基」は、直鎖状、分枝状もしくは環状のアルキル基を意味し、例えば、メチル基、エチル基、n-プロピル基、イソプロピル基、シクロプロピル基、n-ブチル基、イソブチル基、sec-ブチル基、tert-ブチル基、シクロブチル基、n-ペンチル基、イソペンチル基、ネオペンチル基、tert-ペンチル基、シクロペエンチル基、n-ヘキシル基、2,2-ジメチルブチル基、3,3-ジメチルブチル基、2-エチルブチル基、およびシクロヘキシル基が挙げられる。
 本発明において、「C1-C6アルコキシ基」は、直鎖状、分枝状もしくは環状のアルコキシ基を意味し、例えば、メトキシ基、エトキシ基、n-プロポキシ基、イソプロポキシ基、シクロプロピルオキシ基、1-エチルプロポキシ基、2,2-ジメチルプロポキシ基、n-ブトキシ基、イソブチルオキシ基、tert-ブトキシ基、n-ペンチルオキシ基、シクロペエンチルオキシ基、n-ヘキシルオキシ基、およびシクロヘキシルオキシ基等が挙げられる。
In the present invention, the “C1-C6 alkyl group” means a linear, branched or cyclic alkyl group, for example, methyl group, ethyl group, n-propyl group, isopropyl group, cyclopropyl group, n -Butyl group, isobutyl group, sec-butyl group, tert-butyl group, cyclobutyl group, n-pentyl group, isopentyl group, neopentyl group, tert-pentyl group, cyclopeentyl group, n-hexyl group, 2,2-dimethylbutyl Groups, 3,3-dimethylbutyl group, 2-ethylbutyl group, and cyclohexyl group.
In the present invention, “C1-C6 alkoxy group” means a linear, branched or cyclic alkoxy group, for example, a methoxy group, an ethoxy group, an n-propoxy group, an isopropoxy group, a cyclopropyloxy group. 1-ethylpropoxy group, 2,2-dimethylpropoxy group, n-butoxy group, isobutyloxy group, tert-butoxy group, n-pentyloxy group, cyclopeentyloxy group, n-hexyloxy group, and cyclohexyl An oxy group etc. are mentioned.
 好ましくは、環状デプシペプチドは、表1に記載された置換基を有する式(III)から式(VIII)に記載された化合物である。 Preferably, the cyclic depsipeptide is a compound described in Formula (III) to Formula (VIII) having a substituent described in Table 1.
Figure JPOXMLDOC01-appb-T000035
Figure JPOXMLDOC01-appb-T000035
Figure JPOXMLDOC01-appb-C000036
Figure JPOXMLDOC01-appb-C000036
Figure JPOXMLDOC01-appb-C000037
Figure JPOXMLDOC01-appb-C000037
Figure JPOXMLDOC01-appb-C000038
Figure JPOXMLDOC01-appb-C000038
Figure JPOXMLDOC01-appb-C000039
Figure JPOXMLDOC01-appb-C000039
Figure JPOXMLDOC01-appb-C000040
Figure JPOXMLDOC01-appb-C000040
Figure JPOXMLDOC01-appb-C000041
Figure JPOXMLDOC01-appb-C000041
 本発明において、前記環状デプシペプチドは光学異性体、又はラセミ体を含む。本発明において、環状デプシペプチドの薬学上許容される塩としては、例えば、塩酸塩、臭化水素酸塩、リン酸塩、硫酸塩、硝酸塩等の無機酸塩、ギ酸塩、酢酸塩、プロピオン酸塩、マレイン酸塩、フマル酸塩、コハク酸塩、乳酸塩、リンゴ酸塩、酒石酸塩、クエン酸塩、アスコルビン酸塩、マロン酸塩、シュウ酸塩、グリコール酸塩、フタル酸塩、ベンゼンスルホン酸塩等の有機酸との塩、または、ナトリウム塩、カリウム塩等のアルカリ金属塩;カルシウム塩、マグネシウム塩、バリウム塩等のアルカリ土類金属塩;アルミニウム塩;アンモニウム塩;トリメチルアミン、トリエチルアミン、ピリジン、ピコリン、2,6-ルチジン、エタノールアミン、ジエタノールアミン、トリエタノールアミン、N,N-ジエチルアミン、シクロヘキシルアミン、N,N'-ジシクロヘキシルアミン、N,N'-ジベンジルエチレンジアミン、N,N-ジメチルアミノピリジン(DMAP)、1,4-ジアザビシクロ〔2.2.2〕オクタン(DABCO)、1,5-ジアザビシクロ〔4.3.0〕ノネン-5(DBN)、1,8-ジアザビシクロ〔5.4.0〕ウンデ-7-セン(DBU)等の有機塩基との塩;アルギニン、リジン、オルニチン等の塩基性アミノ酸との塩、または、アスパラギン酸、グルタミン酸等の酸性アミノ酸との塩等が挙げられる。また、これらの塩を組合せて用いることもできる。 In the present invention, the cyclic depsipeptide includes an optical isomer or a racemate. In the present invention, pharmaceutically acceptable salts of cyclic depsipeptides include, for example, inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, nitrate, formate, acetate, propionate. , Maleate, fumarate, succinate, lactate, malate, tartrate, citrate, ascorbate, malonate, oxalate, glycolate, phthalate, benzenesulfonic acid Salts with organic acids such as salts, or alkali metal salts such as sodium salts and potassium salts; alkaline earth metal salts such as calcium salts, magnesium salts and barium salts; aluminum salts; ammonium salts; trimethylamine, triethylamine, pyridine, Picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, N, N-diethylamine, cyclohexylamine N, N'-dicyclohexylamine, N, N'-dibenzylethylenediamine, N, N-dimethylaminopyridine (DMAP), 1,4-diazabicyclo [2.2.2] octane (DABCO), 1,5-diazabicyclo [ 4.3.0] Salts with organic bases such as nonene-5 (DBN) and 1,8-diazabicyclo [5.4.0] unde-7-cene (DBU); salts with basic amino acids such as arginine, lysine and ornithine Or salts with acidic amino acids such as aspartic acid and glutamic acid. These salts can also be used in combination.
 環状デプシペプチドのプロドラッグとは、生体内で加水分解されて環状デプシペプチドに変換される化合物をいい、例えば、アミノ基をアルカノイル基(アシル基)に置換した誘導体(すなわちアミド化した誘導体)、ヘミアミナールエーテル誘導体、アルコキシカルボニルオキシメチル基に置換した誘導体、N-オキシド誘導体等が挙げられる。 A cyclic depsipeptide prodrug is a compound that is hydrolyzed in vivo to be converted into a cyclic depsipeptide. For example, a derivative in which an amino group is substituted with an alkanoyl group (acyl group) (that is, an amidated derivative), hemiami Examples thereof include nal ether derivatives, derivatives substituted with alkoxycarbonyloxymethyl groups, and N-oxide derivatives.
 本発明において、環状デプシペプチドは、Doi T, et al. Chem. Asian J.6, 180, 2011、Doi T, et al. Org. Lett., 8, 531, 2006、Numajiri Y, et al. Chem Asian J, 4, 111, 2009、Zou B, et al. Org Lett, 5, 3503, 2003およびQi-Yin Chen, et al. ACS Med. Chem. Lett., 2, 861-865, 2011に記載の方法及びそれに準じた方法によって合成することができる。または、Hendrik L, et al. J. Am. Chem. Soc. 123, 5418, 2001に記載されたようにシアノバクテリアを凍結乾燥し、有機溶媒への溶解層より抽出することができる。 In the present invention, the cyclic depsipeptide is Doi T, et al. Chem. Asian J.6, 180, 2011, Doi T, et al. Org. Lett., 8, 531, 2006, Numajiri Y, et al. Chem Asian J, 4, 111, 2009, Zou B, et al. Org Lett, 5, 3503, 2003 and Qi-Yin Chen, et al. ACS Med. Chem. Lett., 2, 861-865, 2011 And can be synthesized by a method analogous thereto. Alternatively, as described in Hendrik 凍結 L, et al. J. Am. Chem. Soc. 123, 5418, and 2001, cyanobacteria can be lyophilized and extracted from a dissolved layer in an organic solvent.
 本発明はまた、環状デプシペプチドを含有する医薬組成物を提供する。本発明において対象となる疾患は、心疾患であり、例えば、心不全、虚血性心疾患または心筋症等が上げられる。 The present invention also provides a pharmaceutical composition containing a cyclic depsipeptide. The target disease in the present invention is a heart disease, and examples thereof include heart failure, ischemic heart disease or cardiomyopathy.
 本発明の環状デプシペプチドはそのまま単独で、または薬理学的に許容される担体、賦形剤、希釈剤等と混合し、適当な剤型の医薬組成物として経口的又は非経口的に投与することができる。 The cyclic depsipeptide of the present invention may be administered as it is alone or mixed with a pharmacologically acceptable carrier, excipient, diluent, etc., and administered orally or parenterally as a pharmaceutical composition of an appropriate dosage form. Can do.
 経口投与のための組成物としては、固体または液体の剤形、具体的には錠剤(糖衣錠、フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、カプセル剤(ソフトカプセル剤を含む)、シロップ剤、乳剤、懸濁剤等が挙げられる。一方、非経口投与のための組成物としては、例えば、注射剤、坐剤等が用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤等の剤形を包含しても良い。これらの製剤は、賦形剤(例えば、乳糖、白糖、葡萄糖、マンニトール、ソルビトールのような糖誘導体;トウモロコシデンプン、バレイショデンプン、α澱粉、デキストリンのような澱粉誘導体;結晶セルロースのようなセルロース誘導体;アラビアゴム;デキストラン;プルランのような有機系賦形剤;及び、軽質無水珪酸、合成珪酸アルミニウム、珪酸カルシウム、メタ珪酸アルミン酸マグネシウムのような珪酸塩誘導体;燐酸水素カルシウムのような燐酸塩;炭酸カルシウムのような炭酸塩;硫酸カルシウムのような硫酸塩等の無機系賦形剤である)、滑沢剤(例えば、ステアリン酸、ステアリン酸カルシウム、ステアリン酸マグネシウムのようなステアリン酸金属塩;タルク;コロイドシリカ;ビーズワックス、ゲイ蝋のようなワックス類;硼酸;アジピン酸;硫酸ナトリウムのような硫酸塩;グリコール;フマル酸;安息香酸ナトリウム;DLロイシン;ラウリル硫酸ナトリウム、ラウリル硫酸マグネシウムのようなラウリル硫酸塩;無水珪酸、珪酸水和物のような珪酸類;及び、上記澱粉誘導体である)、結合剤(例えば、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルピロリドン、マクロゴール、及び、前記賦形剤と同様の化合物である)、崩壊剤(例えば、低置換度ヒドロキシプロピルセルロース、カルボキシメチルセルロース、カルボキシメチルセルロースカルシウム、内部架橋カルボキシメチルセルロースナトリウムのようなセルロース誘導体;カルボキシメチルスターチ、カルボキシメチルスターチナトリウム、架橋ポリビニルピロリドンのような化学修飾されたデンプン・セルロース類である)、乳化剤(例えば、ベントナイト、ビーガムのようなコロイド性粘土;水酸化マグネシウム、水酸化アルミニウムのような金属水酸化物;ラウリル硫酸ナトリウム、ステアリン酸カルシウムのような陰イオン界面活性剤;塩化ベンザルコニウムのような陽イオン界面活性剤;及び、ポリオキシエチレンアルキルエーテル、ポリオキシエチレンソルビタン脂肪酸エステル、ショ糖脂肪酸エステルのような非イオン界面活性剤である)、安定剤(メチルパラベン、プロピルパラベンのようなパラオキシ安息香酸エステル類;クロロブタノール、ベンジルアルコール、フェニルエチルアルコールのようなアルコール類;塩化ベンザルコニウム;フェノール、クレゾールのようなフェノール類;チメロサール;デヒドロ酢酸;及び、ソルビン酸である)、矯味矯臭剤(例えば、通常使用される、甘味料、酸味料、香料等である)、希釈剤等の添加剤を用いて周知の方法で製造される。 Compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups Agents, emulsions, suspensions and the like. On the other hand, as a composition for parenteral administration, for example, injections, suppositories and the like are used, and injections include intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, and the like. Dosage forms may be included. These formulations include excipients (eg sugar derivatives such as lactose, sucrose, sucrose, mannitol, sorbitol; starch derivatives such as corn starch, potato starch, alpha starch, dextrin; cellulose derivatives such as crystalline cellulose; Gum arabic; dextran; organic excipients such as pullulan; and silicate derivatives such as light anhydrous silicic acid, synthetic aluminum silicate, calcium silicate and magnesium metasilicate aluminate; phosphates such as calcium hydrogen phosphate; Carbonates such as calcium; inorganic excipients such as sulfates such as calcium sulfate), lubricants (eg, stearic acid metal salts such as stearic acid, calcium stearate, magnesium stearate; talc; Colloidal silica; wax like beeswax and gay wax Borax; adipic acid; sulfate such as sodium sulfate; glycol; fumaric acid; sodium benzoate; DL leucine; lauryl sulfate such as sodium lauryl sulfate and magnesium lauryl sulfate; anhydrous silicic acid, silicic acid hydrate Such as silicic acids; and the above-mentioned starch derivatives), binders (for example, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, macrogol, and compounds similar to the excipients), disintegrants ( For example, cellulose derivatives such as low substituted hydroxypropyl cellulose, carboxymethyl cellulose, carboxymethyl cellulose calcium, internally crosslinked sodium carboxymethyl cellulose; carboxymethyl starch, carboxymethyl starch sodium, crosslinked poly Chemically modified starches and celluloses such as livinylpyrrolidone), emulsifiers (for example, colloidal clays such as bentonite and bee gum; metal hydroxides such as magnesium hydroxide and aluminum hydroxide; sodium lauryl sulfate Anionic surfactants such as calcium stearate; cationic surfactants such as benzalkonium chloride; and nonionic interfaces such as polyoxyethylene alkyl ethers, polyoxyethylene sorbitan fatty acid esters, sucrose fatty acid esters Activators), stabilizers (paraoxybenzoates such as methylparaben and propylparaben; alcohols such as chlorobutanol, benzyl alcohol and phenylethyl alcohol; benzalkonium chloride; phenol, cresol Such phenols; thimerosal; dehydroacetic acid; and sorbic acid), flavoring agents (for example, commonly used sweeteners, acidulants, fragrances, etc.), well-known additives using diluents, etc. It is manufactured by the method.
 本発明における環状デプシペプチドの投与量は、患者の症状、年齢、体重等の種々の条件により変化し得る。 The dose of the cyclic depsipeptide in the present invention can vary depending on various conditions such as patient symptoms, age, weight and the like.
 その投与量は症状、年齢等により異なるが、経口投与の場合には、1回当たり下限0.1mg(好適には0.5mg)、上限1000mg(好適には500mg)を、非経口的投与の場合には、1回当たり下限0.01mg(好適には0.05mg)、上限100mg(好適には50mg)を、成人に対して1日当たり1乃至6回投与することができる。症状に応じて増量もしくは減量してもよい。 The dose varies depending on symptoms, age, etc. In the case of oral administration, the lower limit is 0.1 mg (preferably 0.5 mg) and the upper limit is 1000 mg (preferably 500 mg). Can be administered to an adult 1 to 6 times per day at a lower limit of 0.01 mg (preferably 0.05 mg) and an upper limit of 100 mg (preferably 50 mg). The dose may be increased or decreased depending on the symptoms.
 本発明は、また、環状デプシペプチドを心筋前駆細胞へ添加する工程を含む、心筋細胞の製造方法を提供する。当該工程は、in vitroであってもin vivoであっても良いが、好ましくは環状デプシペプチドを単離された心筋前駆細胞へ添加する、in vitroの方法である。
 ここで、心筋細胞とは、自己拍動の特性を有する心筋の細胞を意味する。
 また、心筋前駆細胞とは、前記心筋細胞の前駆細胞であって、培養または成熟によって心筋細胞を生じる能力を有する細胞であり、例えば、生体の心臓に含まれる心筋幹細胞および心筋前駆細胞、多能性幹細胞から分化誘導された細胞、骨髄細胞、骨格筋芽細胞または脂肪由来間葉系細胞に含まれる細胞またはこれらの細胞から分化誘導された細胞(特開2006-115771、特開2003-325169および特開2008-307205)が挙げられる。また、心筋幹細胞および心筋前駆細胞は、別段の記載がなければ特に区別されない。ここで、生体の心臓に含まれる心筋前駆細胞として、心臓に含まれる細胞のうちside populationに存在する細胞(SP細胞)が例示される(Oyama T, et al. J Cell Biol. 176, 329-341, 2007)。SP細胞とは、Hoechst33342 というDNA蛍光色素に対して高い排出能を有する細胞を意味する。心筋細胞および心筋前駆細胞は互いに混在していても良く、また、単離されていても良い。
The present invention also provides a method for producing cardiomyocytes, comprising the step of adding a cyclic depsipeptide to myocardial progenitor cells. This step may be in vitro or in vivo, but is preferably an in vitro method in which a cyclic depsipeptide is added to isolated myocardial progenitor cells.
Here, the myocardial cell means a myocardial cell having the characteristics of self-pulsation.
The myocardial progenitor cell is a progenitor cell of the cardiomyocyte and has the ability to generate a cardiomyocyte by culture or maturation. For example, a myocardial stem cell and a myocardial progenitor cell contained in a living heart Cells derived from sex stem cells, bone marrow cells, skeletal myoblasts, cells included in adipose-derived mesenchymal cells, or cells differentiated from these cells (JP 2006-115771, JP 2003-325169 and JP-A-2008-307205). Further, myocardial stem cells and myocardial progenitor cells are not particularly distinguished unless otherwise specified. Here, as a myocardial progenitor cell contained in a living heart, cells (SP cells) existing in a side population among cells contained in the heart are exemplified (Oyama T, et al. J Cell Biol. 176, 329- 341, 2007). The SP cell means a cell having a high excretion ability with respect to a DNA fluorescent dye called Hoechst33342. Cardiomyocytes and myocardial progenitor cells may be mixed with each other or isolated.
 本発明において、心筋細胞はまた、心筋マーカーである心筋トロポニン(cTNTまたはtroponin T type 2)および/またはαMHC(α myosin heavy chain)が陽性であることによって特徴づけられる。
 また、心筋前駆細胞は、特に限定されないが、Flk1/KDR、islet1、Wt1および/またはN-Cadherinが陽性であること、および/またはFACS解析においてside population(SP細胞分画)に存在することによって特徴づけられる。
In the present invention, cardiomyocytes are also characterized by being positive for myocardial markers cardiac troponin (cTNT or troponin T type 2) and / or αMHC (α myosin heavy chain).
In addition, myocardial progenitor cells are not particularly limited, but by being positive for Flk1 / KDR, islet1, Wt1 and / or N-Cadherin and / or present in the side population (SP cell fraction) in FACS analysis Characterized.
 本発明において、心筋前駆細胞は、in vitroで多能性幹細胞を分化させることによって用意されても良い。ここで、多能性幹細胞とは、生体に存在するすべての細胞に分化可能である多能性を有し、かつ、増殖能をも併せもつ幹細胞であり、それには、特に限定されないが、例えば胚性幹(ES)細胞、核移植により得られるクローン胚由来の胚性幹(ntES)細胞、精子幹細胞(「GS細胞」)、胚性生殖細胞(「EG細胞」)、人工多能性幹(iPS)細胞、培養線維芽細胞や骨髄幹細胞由来の多能性細胞(Muse細胞)などが含まれる。 In the present invention, cardiac progenitor cells may be prepared by differentiating pluripotent stem cells in vitro. Here, the pluripotent stem cell is a stem cell having pluripotency that can be differentiated into all cells existing in a living body and also having proliferative ability, and is not particularly limited, for example, Embryonic stem (ES) cells, embryonic stem (ntES) cells derived from cloned embryos obtained by nuclear transfer, sperm stem cells (“GS cells”), embryonic germ cells (“EG cells”), induced pluripotent stems (iPS) cells, cultured fibroblasts, pluripotent cells derived from bone marrow stem cells (Muse cells) and the like are included.
(A) 胚性幹細胞
 ES細胞は、ヒトやマウスなどの哺乳動物の初期胚(例えば胚盤胞)の内部細胞塊から樹立された、多能性と自己複製による増殖能を有する幹細胞である。
(A) Embryonic stem cells ES cells are stem cells established from the inner cell mass of early embryos (for example, blastocysts) of mammals such as humans and mice, and having pluripotency and proliferation ability by self-replication.
 ES細胞は、受精卵の8細胞期、桑実胚後の胚である胚盤胞の内部細胞塊に由来する胚由来の幹細胞であり、成体を構成するあらゆる細胞に分化する能力、いわゆる分化多能性と、自己複製による増殖能とを有している。ES細胞は、マウスで1981年に発見され(M.J. Evans and M.H. Kaufman (1981), Nature 292:154-156)、その後、ヒト、サルなどの霊長類でもES細胞株が樹立された (J.A. Thomson et al. (1998), Science 282:1145-1147; J.A. Thomson et al. (1995), Proc. Natl. Acad. Sci. USA, 92:7844-7848;J.A. Thomsonet al. (1996), Biol. Reprod., 55:254-259; J.A. Thomson and V.S. Marshall (1998), Curr. Top. Dev. Biol., 38:133-165)。 ES cells are embryonic stem cells derived from the inner cell mass of the blastocyst, the embryo after the morula, in the 8-cell stage of a fertilized egg, and have the ability to differentiate into any cell that constitutes an adult, so-called differentiation. And ability to proliferate by self-replication. ES cells were discovered in mice in 1981 (MJ Evans and MH Kaufman (1981), Nature 292: 154-156), and then ES cell lines were established in primates such as humans and monkeys (JA Thomson et al. (1998), Science 282: 1145-1147; JA Thomson et al. (1995), Proc. Natl. Acad. Sci. USA, 92: 7844-7848; JA Thomsonet al. (1996), Biol. Reprod. 55: 254-259; JA Thomson and VS Marshall (1998), Curr. Top. Dev. Biol., 38: 133-165).
 ES細胞は、対象動物の受精卵の胚盤胞から内部細胞塊を取出し、内部細胞塊を線維芽細胞のフィーダー上で培養することによって樹立することができる。また、継代培養による細胞の維持は、白血病抑制因子(leukemia inhibitory factor (LIF))、塩基性線維芽細胞成長因子(basic fibroblast growth factor (bFGF))などの物質を添加した培養液を用いて行うことができる。ヒトおよびサルのES細胞の樹立と維持の方法については、例えばUSP5,843,780; Thomson JA, et al. (1995), Proc Natl. Acad. Sci. U S A. 92:7844-7848; Thomson JA, et al. (1998), Science. 282:1145-1147; H. Suemori et al. (2006), Biochem. Biophys. Res. Commun., 345:926-932; M. Ueno et al. (2006), Proc. Natl. Acad. Sci. USA, 103:9554-9559; H. Suemori et al. (2001), Dev. Dyn., 222:273-279;H. Kawasaki et al. (2002), Proc. Natl. Acad. Sci. USA, 99:1580-1585;Klimanskaya I, et al. (2006), Nature. 444:481-485などに記載されている。 ES cells can be established by taking an inner cell mass from a blastocyst of a fertilized egg of a target animal and culturing the inner cell mass on a fibroblast feeder. In addition, maintenance of cells by subculture is performed using a culture solution to which substances such as leukemia inhibitory factor (LIF) and basic fibroblast growth factor (basic fibroblast growth factor (bFGF)) are added. It can be carried out. For methods of establishing and maintaining human and monkey ES cells, see, for example, USP 5,843,780; Thomson JA, et al. (1995), Proc Natl. Acad. Sci. U S A. 92: 7844-7848; Thomson JA, et al. (1998), Science. 282: 1145-1147; H. Suemori et al. (2006), Biochem. Biophys. Res. Commun., 345: 926-932; M. Ueno et al. (2006), Proc. Natl. Acad. Sci. USA, 103: 9554-9559; H. Suemori et al. (2001), Dev. Dyn., 222: 273-279; H. Kawasaki et al. (2002), Proc. Natl Acad. Sci. USA, ; 99: 1580-1585; Klimanskaya I, et al. (2006), Nature. 444: 481-485.
 ES細胞作製のための培養液として、例えば0.1mM 2-メルカプトエタノール、0.1mM 非必須アミノ酸、2mM L-グルタミン酸、20% KSRおよび4ng/ml bFGFを補充したDMEM/F-12培養液を使用し、37℃、2% CO2/98% 空気の湿潤雰囲気下でヒトES細胞を維持することができる(O. Fumitaka et al. (2008), Nat. Biotechnol., 26:215-224)。また、ES細胞は、3~4日おきに継代する必要があり、このとき、継代は、例えば1mM CaCl2および20% KSRを含有するPBS中の0.25% トリプシンおよび0.1mg/mlコラゲナーゼIVを用いて行うことができる。 For example, DMEM / F-12 culture medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM non-essential amino acid, 2 mM L-glutamic acid, 20% KSR and 4 ng / ml bFGF is used as the culture medium for ES cell production. Human ES cells can be maintained in a humid atmosphere of 37 ° C., 2% CO 2 /98% air (O. Fumitaka et al. (2008), Nat. Biotechnol., 26: 215-224). ES cells also need to be passaged every 3-4 days, where passage is eg 0.25% trypsin and 0.1 mg / ml collagenase IV in PBS containing 1 mM CaCl 2 and 20% KSR. Can be used.
 ES細胞の選択は、一般に、アルカリホスファターゼ、Oct-3/4、Nanogなどの遺伝子マーカーの発現を指標にしてReal-Time PCR法で行うことができる。特に、ヒトES細胞の選択では、OCT-3/4、NANOG、ECADなどの遺伝子マーカーの発現を指標とすることができる(E. Kroon et al. (2008), Nat. Biotechnol., 26:443-452)。 ES cells can be generally selected by Real-Time PCR using the expression of gene markers such as alkaline phosphatase, Oct-3 / 4, Nanog as an index. In particular, in the selection of human ES cells, the expression of gene markers such as OCT-3 / 4, NANOG, and ECAD can be used as an index (E. Kroon et al. (2008), Nat. Biotechnol., 26: 443). -452).
 ヒトES細胞株は、例えばWA01(H1)およびWA09(H9)は、WiCell Reserch Instituteから、KhES-1、KhES-2およびKhES-3は、京都大学再生医科学研究所(京都、日本)から入手可能である。 Human ES cell lines, for example, WA01 (H1) and WA09 (H9) are obtained from the WiCell Research Institute, and KhES-1, KhES-2 and KhES-3 are obtained from the Institute of Regenerative Medicine (Kyoto, Japan), Kyoto University Is possible.
(B) 精子幹細胞
 精子幹細胞は、精巣由来の多能性幹細胞であり、精子形成のための起源となる細胞である。この細胞は、ES細胞と同様に、種々の系列の細胞に分化誘導可能であり、例えばマウス胚盤胞に移植するとキメラマウスを作出できるなどの性質をもつ(M. Kanatsu-Shinohara et al. (2003) Biol. Reprod., 69:612-616; K. Shinohara et al. (2004), Cell, 119:1001-1012)。神経膠細胞系由来神経栄養因子(glial cell line-derived neurotrophic factor (GDNF))を含む培養液で自己複製可能であるし、またES細胞と同様の培養条件下で継代を繰り返すことによって、精子幹細胞を得ることができる(竹林正則ら(2008),実験医学,26巻,5号(増刊),41~46頁,羊土社(東京、日本))。
(B) Sperm stem cells Sperm stem cells are testis-derived pluripotent stem cells that are the origin of spermatogenesis. Like ES cells, these cells can be induced to differentiate into various types of cells, and have characteristics such as the ability to create chimeric mice when transplanted into mouse blastocysts (M. Kanatsu-Shinohara et al. ( 2003) Biol. Reprod., 69: 612-616; K. Shinohara et al. (2004), Cell, 119: 1001-1012). It is capable of self-replication in a culture medium containing glial cell line-derived neurotrophic factor (GDNF), and by repeating subculture under the same culture conditions as ES cells, Stem cells can be obtained (Masatake Takebayashi et al. (2008), Experimental Medicine, Vol. 26, No. 5 (extra number), 41-46, Yodosha (Tokyo, Japan)).
(C) 胚性生殖細胞
 胚性生殖細胞は、胎生期の始原生殖細胞から樹立される、ES細胞と同様な多能性をもつ細胞であり、LIF、bFGF、幹細胞因子(stem cell factor)などの物質の存在下で始原生殖細胞を培養することによって樹立しうる(Y. Matsui et al. (1992), Cell, 70:841-847; J.L. Resnick et al. (1992), Nature, 359:550-551)。
(C) Embryonic germ cells Embryonic germ cells are cells that are established from embryonic primordial germ cells and have the same pluripotency as ES cells, such as LIF, bFGF, stem cell factor, etc. It can be established by culturing primordial germ cells in the presence of these substances (Y. Matsui et al. (1992), Cell, 70: 841-847; JL Resnick et al. (1992), Nature, 359: 550 -551).
(D) 人工多能性幹細胞
 人工多能性幹(iPS)細胞は、特定の初期化因子を、DNA又はタンパク質の形態で体細胞に導入することによって作製することができる、ES細胞とほぼ同等の特性、例えば分化多能性と自己複製による増殖能、を有する体細胞由来の人工の幹細胞である(K. Takahashi and S. Yamanaka (2006) Cell, 126:663-676; K. Takahashi et al. (2007), Cell, 131:861-872; J. Yu et al. (2007), Science, 318:1917-1920; Nakagawa, M.ら,Nat. Biotechnol. 26:101-106 (2008);国際公開WO 2007/069666)。初期化因子は、ES細胞に特異的に発現している遺伝子、その遺伝子産物もしくはnon-cording RNAまたはES細胞の未分化維持に重要な役割を果たす遺伝子、その遺伝子産物もしくはnon-coding RNA、あるいは低分子化合物によって構成されてもよい。初期化因子に含まれる遺伝子として、例えば、Oct3/4、Sox2、Sox1、Sox3、Sox15、Sox17、Klf4、Klf2、c-Myc、N-Myc、L-Myc、Nanog、Lin28、Fbx15、ERas、ECAT15-2、Tcl1、beta-catenin、Lin28b、Sall1、Sall4、Esrrb、Nr5a2、Tbx3またはGlis1等が例示され、これらの初期化因子は、単独で用いても良く、組み合わせて用いても良い。初期化因子の組み合わせとしては、WO2007/069666、WO2008/118820、WO2009/007852、WO2009/032194、WO2009/058413、WO2009/057831、WO2009/075119、WO2009/079007、WO2009/091659、WO2009/101084、WO2009/101407、WO2009/102983、WO2009/114949、WO2009/117439、WO2009/126250、WO2009/126251、WO2009/126655、WO2009/157593、WO2010/009015、WO2010/033906、WO2010/033920、WO2010/042800、WO2010/050626、WO 2010/056831、WO2010/068955、WO2010/098419、WO2010/102267、WO 2010/111409、WO 2010/111422、WO2010/115050、WO2010/124290、WO2010/147395、WO2010/147612、Huangfu D, et al. (2008), Nat. Biotechnol., 26: 795-797、Shi Y, et al. (2008), Cell Stem Cell, 2: 525-528、Eminli S, et al. (2008), Stem Cells. 26:2467-2474、Huangfu D, et al. (2008), Nat Biotechnol. 26:1269-1275、Shi Y, et al. (2008), Cell Stem Cell, 3, 568-574、Zhao Y, et al. (2008), Cell Stem Cell, 3:475-479、Marson A, (2008), Cell Stem Cell, 3, 132-135、Feng B, et al. (2009), Nat Cell Biol. 11:197-203、R.L. Judson et al., (2009), Nat. Biotech., 27:459-461、Lyssiotis CA, et al. (2009), Proc Natl Acad Sci U S A. 106:8912-8917、Kim JB, et al. (2009), Nature. 461:649-643、Ichida JK, et al. (2009), Cell Stem Cell. 5:491-503、Heng JC, et al. (2010), Cell Stem Cell. 6:167-74、Han J, et al. (2010), Nature. 463:1096-100、MaliP, et al. (2010), Stem Cells. 28:713-720、Maekawa M, et al. (2011), Nature. 474:225-9.に記載の組み合わせが例示される。
(D) Artificial pluripotent stem cells Artificial pluripotent stem (iPS) cells can be produced by introducing specific reprogramming factors into somatic cells in the form of DNA or protein, which is almost equivalent to ES cells It is an artificial stem cell derived from a somatic cell having the characteristics of, for example, differentiation pluripotency and proliferation ability by self-replication (K. Takahashi and S. Yamanaka (2006) Cell, 126: 663-676; K. Takahashi et al (2007), Cell, 131: 861-872; J. Yu et al. (2007), Science, 318: 1917-1920; Nakagawa, M. et al., Nat. Biotechnol. 26: 101-106 (2008); International publication WO 2007/069666). The reprogramming factor is a gene specifically expressed in ES cells, its gene product or non-cording RNA, a gene that plays an important role in maintaining undifferentiation of ES cells, its gene product or non-coding RNA, or It may be constituted by a low molecular compound. Examples of genes included in the reprogramming factor include Oct3 / 4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15 -2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3 or Glis1 etc. are exemplified, and these reprogramming factors may be used alone or in combination. As combinations of reprogramming factors, WO2007 / 069666, WO2008 / 118820, WO2009 / 007852, WO2009 / 032194, WO2009 / 058413, WO2009 / 057831, WO2009 / 075119, WO2009 / 079007, WO2009 / 091659, WO2009 / 101084, WO2009 / 101407, WO2009 / 102983, WO2009 / 114949, WO2009 / 117439, WO2009 / 126250, WO2009 / 126251, WO2009 / 126655, WO2009 / 157593, WO2010 / 009015, WO2010 / 033906, WO2010 / 033920, WO2010 / 042800, WO2010 / 050626, WO 2010/056831, WO2010 / 068955, WO2010 / 098419, WO2010 / 102267, WO 2010/111409, WO 2010/111422, WO2010 / 115050, WO2010 / 124290, WO2010 / 147395, WO2010 / 147612, Huangfu D, et al. 2008), Nat. Biotechnol., 26: 795-797, Shi Y, et al. (2008), Cell Stem Cell, 2: 525-528, Eminli S, et al. (2008), Stem Cells. 26: 2467 -2474, Huangfu D, et al. (2008), Nat Biotechnol. 26: 1269-1275, Shi Y, et al. (2008), Cell Stem Cell, 3, 568-574, Zhao Y, et al. (2008 ), Cell Stem Cell, 3: 475-479, Marson A, (2008), Cell Stem Cell, 3, 132-135, Feng B, et al. (2009), Nat Cell Biol. 11: 197-203, RL Judson et al., (2009), Nat. Biotech., 27: 459-461, Lyssiotis CA, et al. (2009), Proc Natl Acad Sci US A. 106: 8912-8917, Kim JB, et al. 2009), Nature. 461: 649-643, Ichida JK, et al. (2009), Cell Stem Cell. 5: 491-503, Heng JC, et al. (2010), Cell Stem Cell. 6: 167-74 , Han J, et al. (2010), Nature. 463: 1096-100, MaliP, et al. (2010), Stem Cells. 28: 713-720, Maekawa M, et al. (2011), Nature. : The combination described in 225-9.
 上記初期化因子には、ヒストンデアセチラーゼ(HDAC)阻害剤[例えば、バルプロ酸 (VPA)、トリコスタチンA、酪酸ナトリウム、MC 1293、M344等の低分子阻害剤、HDACに対するsiRNAおよびshRNA(例、HDAC1 siRNA Smartpool( (Millipore)、HuSH 29mer shRNA Constructs against HDAC1 (OriGene)等)等の核酸性発現阻害剤など]、MEK阻害剤(例えば、PD184352、PD98059、U0126、SL327およびPD0325901)、Glycogen synthase kinase-3阻害剤(例えば、BioおよびCHIR99021)、DNAメチルトランスフェラーゼ阻害剤(例えば、5-azacytidine)、ヒストンメチルトランスフェラーゼ阻害剤(例えば、BIX-01294 等の低分子阻害剤、Suv39hl、Suv39h2、SetDBlおよびG9aに対するsiRNAおよびshRNA等の核酸性発現阻害剤など)、L-channel calcium agonist (例えばBayk8644)、酪酸、TGFβ阻害剤またはALK5阻害剤(例えば、LY364947、SB431542、616453およびA-83-01)、p53阻害剤(例えばp53に対するsiRNAおよびshRNA)、ARID3A阻害剤(例えば、ARID3Aに対するsiRNAおよびshRNA)、miR-291-3p、miR-294、miR-295およびmir-302などのmiRNA、Wnt Signaling(例えばsoluble Wnt3a)、神経ペプチドY、プロスタグランジン類(例えば、プロスタグランジンE2およびプロスタグランジンJ2)、hTERT、SV40LT、UTF1、IRX6、GLISl、PITX2、DMRTBl等の樹立効率を高めることを目的として用いられる因子も含まれており、本明細書においては、これらの樹立効率の改善目的にて用いられた因子についても初期化因子と別段の区別をしないものとする。 The reprogramming factors include histone deacetylase (HDAC) inhibitors [for example, small molecule inhibitors such as valproate (VPA), trichostatin A, sodium butyrate, MC 1293, M344, siRNA and shRNA against HDAC (eg , Nucleic acid expression inhibitors such as HDAC1 siRNA Smartpool ((Millipore), HuSH 29mer shRNA Constructs against HDAC1 (OriGene), etc.)], MEK inhibitors (eg PD184352, PD98059, U0126, SL327 and PD0325901), Glycogen synthase kin -3 inhibitors (eg, Bio and CHIR99021), DNA methyltransferase inhibitors (eg, 5-azacytidine), histone methyltransferase inhibitors (eg, small molecule inhibitors such as BIX-01294, Suv39hl, Suv39h2, SetDBl and G9a Nucleic acid expression inhibitors such as siRNA and shRNA), L-channelLcalcium agonist (eg Bayk8644), butyric acid, TGFβ inhibitor or ALK5 inhibitor (eg LY364947, SB4315) 42, 616453 and A-83-01), p53 inhibitors (eg siRNA and shRNA against p53), ARID3A inhibitors (eg siRNA and shRNA against ARID3A), miR-291-3p, miR-294, miR-295 and miRNA such as mir-302, Wnt Signaling (eg soluble Wnt3a), neuropeptide Y, prostaglandins (eg prostaglandin E2 and prostaglandin J2), hTERT, SV40LT, UTF1, IRX6, GLISL, PITX2, DMRTBl Factors used for the purpose of improving establishment efficiency such as the above are also included. In this specification, factors used for the purpose of improving establishment efficiency are not distinguished from initialization factors. And
 初期化因子は、タンパク質の形態の場合、例えばリポフェクション、細胞膜透過性ペプチド(例えば、HIV由来のTATおよびポリアルギニン)との融合、マイクロインジェクションなどの手法によって体細胞内に導入してもよい。 In the case of a protein form, the reprogramming factor may be introduced into a somatic cell by a technique such as lipofection, fusion with a cell membrane-permeable peptide (for example, HIV-derived TAT and polyarginine), or microinjection.
 一方、DNAの形態の場合、例えば、ウイルス、プラスミド、人工染色体などのベクター、リポフェクション、リポソーム、マイクロインジェクションなどの手法によって体細胞内に導入することができる。ウイルスベクターとしては、レトロウイルスベクター、レンチウイルスベクター(以上、Cell, 126, pp.663-676, 2006; Cell, 131, pp.861-872, 2007; Science, 318, pp.1917-1920, 2007)、アデノウイルスベクター(Science, 322, 945-949, 2008)、アデノ随伴ウイルスベクター、センダイウイルスベクター(WO 2010/008054)などが例示される。また、人工染色体ベクターとしては、例えばヒト人工染色体(HAC)、酵母人工染色体(YAC)、細菌人工染色体(BAC、PAC)などが含まれる。プラスミドとしては、哺乳動物細胞用プラスミドを使用しうる(Science, 322:949-953, 2008)。ベクターには、核初期化物質が発現可能なように、プロモーター、エンハンサー、リボゾーム結合配列、ターミネーター、ポリアデニル化サイトなどの制御配列を含むことができるし、さらに、必要に応じて、薬剤耐性遺伝子(例えばカナマイシン耐性遺伝子、アンピシリン耐性遺伝子、ピューロマイシン耐性遺伝子など)、チミジンキナーゼ遺伝子、ジフテリアトキシン遺伝子などの選択マーカー配列、緑色蛍光タンパク質(GFP)、βグルクロニダーゼ(GUS)、FLAGなどのレポーター遺伝子配列などを含むことができる。また、上記ベクターには、体細胞への導入後、初期化因子をコードする遺伝子もしくはプロモーターとそれに結合する初期化因子をコードする遺伝子を共に切除するために、それらの前後にLoxP配列を有してもよい。 On the other hand, in the case of DNA, it can be introduced into somatic cells by techniques such as vectors such as viruses, plasmids, artificial chromosomes, lipofection, liposomes, and microinjection. Virus vectors include retrovirus vectors, lentivirus vectors (cell, 126, pp.663-676, 2006; Cell, 131, pp.861-872, 2007; Science, 318, pp.1917-1920, 2007 ), Adenovirus vectors (Science, 322, 945-949, 2008), adeno-associated virus vectors, Sendai virus vectors (WO 2010/008054) and the like. Examples of artificial chromosome vectors include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), and bacterial artificial chromosomes (BAC, PAC). As a plasmid, a plasmid for mammalian cells can be used (Science, 322: 949-953, 2008). The vector can contain regulatory sequences such as a promoter, enhancer, ribosome binding sequence, terminator, polyadenylation site, etc. so that a nuclear reprogramming substance can be expressed. Selective marker sequences such as kanamycin resistance gene, ampicillin resistance gene, puromycin resistance gene, thymidine kinase gene, diphtheria toxin gene, reporter gene sequences such as green fluorescent protein (GFP), β-glucuronidase (GUS), FLAG, etc. Can be included. In addition, the above vector has a LoxP sequence before and after the introduction of the gene into a somatic cell in order to excise the gene or promoter encoding the reprogramming factor and the gene encoding the reprogramming factor that binds to it. May be.
 また、RNAの形態の場合、例えばリポフェクション、マイクロインジェクションなどの手法によって体細胞内に導入しても良く、分解を抑制するため、5-メチルシチジンおよびpseudouridine (TriLink Biotechnologies)を取り込ませたRNAを用いても良い(Warren L, (2010) Cell Stem Cell. 7:618-630)。 In the case of RNA, it may be introduced into somatic cells by techniques such as lipofection and microinjection, and in order to suppress degradation, RNA incorporating 5-methylcytidine and pseudouridine® (TriLink® Biotechnologies) is used. Yes (Warren L, (2010) Cell Stem Cell. 7: 618-630).
 iPS細胞誘導のための培養液としては、例えば、10~15%FBSを含有するDMEM、DMEM/F12又はDME培養液(これらの培養液にはさらに、LIF、penicillin/streptomycin、puromycin、L-グルタミン、非必須アミノ酸類、β-メルカプトエタノールなどを適宜含むことができる。)または市販の培養液[例えば、マウスES細胞培養用培養液(TX-WES培養液、トロンボX社)、霊長類ES細胞培養用培養液(霊長類ES/iPS細胞用培養液、リプロセル社)、無血清培地(mTeSR、Stemcell Technology社)]などが含まれる。 Examples of the culture medium for inducing iPS cells include DMEM, DMEM / F12 or DME culture medium containing 10 to 15% FBS (these culture media include LIF, penicillin / streptomycin, puromycin, L-glutamine). , Non-essential amino acids, β-mercaptoethanol, etc.) or commercially available culture media (eg, culture media for mouse ES cell culture (TX-WES culture solution, Thrombo X), primate ES cells) Culture medium for culture (primate ES / iPS cell culture medium, Reprocell), serum-free medium (mTeSR, Stemcell Technology).
 培養法の例としては、たとえば、37℃、5%CO2存在下にて、10%FBS含有DMEM又はDMEM/F12培養液上で体細胞と初期化因子とを接触させ約4~7日間培養し、その後、細胞をフィーダー細胞(たとえば、マイトマイシンC処理STO細胞、SNL細胞等)上にまきなおし、体細胞と初期化因子の接触から約10日後からbFGF含有霊長類ES細胞培養用培養液で培養し、該接触から約30~約45日又はそれ以上ののちにiPS様コロニーを生じさせることができる。 As an example of the culture method, for example, a somatic cell is brought into contact with a reprogramming factor on a DMEM or DMEM / F12 medium containing 10% FBS at 37 ° C. in the presence of 5% CO 2 for about 4 to 7 days. Then, re-spread the cells on feeder cells (for example, mitomycin C-treated STO cells, SNL cells, etc.), and use bFGF-containing primate ES cell culture medium about 10 days after contact between the somatic cells and the reprogramming factor. Culturing and generating iPS-like colonies about 30 to about 45 days or more after the contact.
 あるいは、37℃、5% CO2存在下にて、フィーダー細胞(たとえば、マイトマイシンC処理STO細胞、SNL細胞等)上で10%FBS含有DMEM培養液(これにはさらに、LIF、ペニシリン/ストレプトマイシン、ピューロマイシン、L-グルタミン、非必須アミノ酸類、β-メルカプトエタノールなどを適宜含むことができる。)で培養し、約25~約30日又はそれ以上ののちにES様コロニーを生じさせることができる。望ましくは、フィーダー細胞の代わりに、初期化される体細胞そのものを用いる(Takahashi K, et al. (2009), PLoS One. 4:e8067またはWO2010/137746)、もしくは細胞外基質(例えば、Laminin-5(WO2009/123349)およびマトリゲル(BD社))を用いる方法が例示される。 Alternatively, 10% FBS-containing DMEM medium (including LIF, penicillin / streptomycin, etc.) on feeder cells (eg, mitomycin C-treated STO cells, SNL cells, etc.) in the presence of 5% CO 2 at 37 ° C. Can be suitably included with puromycin, L-glutamine, non-essential amino acids, β-mercaptoethanol, etc.) and can form ES-like colonies after about 25 to about 30 days or more . Desirably, instead of feeder cells, somatic cells to be reprogrammed themselves are used (Takahashi K, et al. (2009), PLoS One. 4: e8067 or WO2010 / 137746), or extracellular matrix (eg, Laminin- 5 (WO2009 / 123349) and Matrigel (BD)) are exemplified.
 この他にも、血清を含有しない培地を用いて培養する方法も例示される(Sun N, et al. (2009), Proc Natl Acad Sci U S A. 106:15720-15725)。さらに、樹立効率を上げるため、低酸素条件(0.1%以上、15%以下の酸素濃度)によりiPS細胞を樹立しても良い(Yoshida Y, et al. (2009), Cell Stem Cell. 5:237-241またはWO2010/013845)。 In addition to this, a method of culturing using a medium not containing serum is also exemplified (Sun N, et al. (2009), Proc Natl Acad Sci U S A. 106: 15720-15725). Furthermore, in order to increase the establishment efficiency, iPS cells may be established under hypoxic conditions (oxygen concentration of 0.1% or more and 15% or less) (Yoshida Y, et al. (2009), Cell Stem Cell. 5: 237 -241 or WO2010 / 013845).
 上記培養の間には、培養開始2日目以降から毎日1回新鮮な培養液と培養液交換を行う。また、核初期化に使用する体細胞の細胞数は、限定されないが、培養ディッシュ100cm2あたり約5×103~約5×106細胞の範囲である。 During the culture, the culture medium is exchanged with a fresh culture medium once a day from the second day onward. The number of somatic cells used for nuclear reprogramming is not limited, but ranges from about 5 × 10 3 to about 5 × 10 6 cells per 100 cm 2 of culture dish.
 iPS細胞は、形成したコロニーの形状により選択することが可能である。一方、体細胞が初期化された場合に発現する遺伝子(例えば、Oct3/4、Nanog)と連動して発現する薬剤耐性遺伝子をマーカー遺伝子として導入した場合は、対応する薬剤を含む培養液(選択培養液)で培養を行うことにより樹立したiPS細胞を選択することができる。また、マーカー遺伝子が蛍光タンパク質遺伝子の場合は蛍光顕微鏡で観察することによって、発光酵素遺伝子の場合は発光基質を加えることによって、また発色酵素遺伝子の場合は発色基質を加えることによって、iPS細胞を選択することができる。 IPS cells can be selected according to the shape of the formed colonies. On the other hand, when a drug resistance gene that is expressed in conjunction with a gene that is expressed when somatic cells are initialized (for example, Oct3 / 4, Nanog) is introduced as a marker gene, a culture solution containing the corresponding drug (selection The established iPS cells can be selected by culturing with the culture medium. In addition, if the marker gene is a fluorescent protein gene, iPS cells are selected by observing with a fluorescence microscope, in the case of a luminescent enzyme gene, by adding a luminescent substrate, and in the case of a chromogenic enzyme gene, by adding a chromogenic substrate can do.
 本明細書中で使用する「体細胞」なる用語は、卵子、卵母細胞、ES細胞などの生殖系列細胞または分化全能性細胞を除くあらゆる動物細胞(好ましくは、ヒトを含む哺乳動物細胞)をいう。体細胞には、非限定的に、胎児(仔)の体細胞、新生児(仔)の体細胞、および成熟した健全なもしくは疾患性の体細胞のいずれも包含されるし、また、初代培養細胞、継代細胞、および株化細胞のいずれも包含される。具体的には、体細胞は、例えば(1)神経幹細胞、造血幹細胞、間葉系幹細胞、歯髄幹細胞等の組織幹細胞(体性幹細胞)、(2)組織前駆細胞、(3)リンパ球、上皮細胞、内皮細胞、筋肉細胞、線維芽細胞(皮膚細胞等)、毛細胞、肝細胞、胃粘膜細胞、腸細胞、脾細胞、膵細胞(膵外分泌細胞等)、脳細胞、肺細胞、腎細胞および脂肪細胞等の分化した細胞などが例示される。 As used herein, the term “somatic cell” refers to any animal cell (preferably, a mammalian cell including a human) except a germ line cell such as an egg, oocyte, ES cell, or totipotent cell. Say. Somatic cells include, but are not limited to, fetal (pup) somatic cells, neonatal (pup) somatic cells, and mature healthy or diseased somatic cells. , Passage cells, and established cell lines. Specifically, somatic cells include, for example, (1) neural stem cells, hematopoietic stem cells, mesenchymal stem cells, tissue stem cells such as dental pulp stem cells (somatic stem cells), (2) tissue progenitor cells, (3) lymphocytes, epithelium Cells, endothelial cells, muscle cells, fibroblasts (skin cells, etc.), hair cells, hepatocytes, gastric mucosal cells, enterocytes, spleen cells, pancreatic cells (exocrine pancreas cells, etc.), brain cells, lung cells, kidney cells Examples thereof include differentiated cells such as fat cells.
 また、iPS細胞を移植用細胞の材料として用いる場合、拒絶反応が起こらないという観点から、移植先の個体のHLA遺伝子型が同一もしくは実質的に同一である体細胞を用いることが望ましい。ここで、「実質的に同一」とは、移植した細胞に対して免疫抑制剤により免疫反応が抑制できる程度にHLA遺伝子型が一致していることであり、例えば、HLA-A、HLA-BおよびHLA-DRの3遺伝子座あるいはHLA-Cを加えた4遺伝子座が一致するHLA型を有する体細胞である。 In addition, when iPS cells are used as a material for transplantation cells, it is desirable to use somatic cells having the same or substantially the same HLA genotype as the transplant destination individual from the viewpoint that rejection does not occur. Here, “substantially the same” means that the HLA genotype matches the transplanted cells to such an extent that an immune response can be suppressed by an immunosuppressive agent. For example, HLA-A, HLA-B And somatic cells having an HLA type in which 3 loci of HLA-DR or 4 loci plus HLA-C are matched.
(E) 核移植により得られたクローン胚由来のES細胞
 nt ES細胞は、核移植技術によって作製されたクローン胚由来のES細胞であり、受精卵由来のES細胞とほぼ同じ特性を有している (T. Wakayama et al. (2001), Science, 292:740-743; S. Wakayama et al. (2005), Biol. Reprod., 72:932-936; J. Byrne et al. (2007), Nature, 450:497-502)。すなわち、未受精卵の核を体細胞の核と置換することによって得られたクローン胚由来の胚盤胞の内部細胞塊から樹立されたES細胞がnt ES(nuclear transfer ES)細胞である。nt ES細胞の作製のためには、核移植技術(J.B. Cibelli et al. (1998), Nature Biotechnol., 16:642-646)とES細胞作製技術(上記)との組み合わせが利用される(若山清香ら(2008),実験医学,26巻,5号(増刊), 47~52頁)。核移植においては、哺乳動物の除核した未受精卵に、体細胞の核を注入し、数時間培養することで初期化することができる。
(E) Cloned embryo-derived ES cells obtained by nuclear transfer nt ES cells are cloned embryo-derived ES cells produced by nuclear transfer technology and have almost the same characteristics as ES cells derived from fertilized eggs (T. Wakayama et al. (2001), Science, 292: 740-743; S. Wakayama et al. (2005), Biol. Reprod., 72: 932-936; J. Byrne et al. (2007) , Nature, 450: 497-502). That is, an ES cell established from an inner cell mass of a clonal embryo-derived blastocyst obtained by replacing the nucleus of an unfertilized egg with the nucleus of a somatic cell is an nt ES (nuclear transfer ES) cell. For the production of nt ES cells, a combination of nuclear transfer technology (JB Cibelli et al. (1998), Nature Biotechnol., 16: 642-646) and ES cell production technology (above) is used (Wakayama). Seika et al. (2008), Experimental Medicine, Vol. 26, No. 5 (extra number), 47-52). Nuclear transfer can be initialized by injecting a somatic cell nucleus into a mammal's enucleated unfertilized egg and culturing for several hours.
(F) Multilineage-differentiating Stress Enduring cells(Muse細胞)
 Muse細胞は、WO2011/007900に記載された方法にて製造された多能性幹細胞であり、詳細には、線維芽細胞または骨髄間質細胞を長時間トリプシン処理、好ましくは8時間または16時間トリプシン処理した後、浮遊培養することで得られる多能性を有した細胞であり、SSEA-3およびCD105が陽性である。
(F) Multilineage-differentiating Stress Enduring cells (Muse cells)
Muse cells are pluripotent stem cells produced by the method described in WO2011 / 007900. Specifically, fibroblasts or bone marrow stromal cells are treated with trypsin for a long time, preferably 8 or 16 hours. It is a pluripotent cell obtained by suspension culture after treatment, and is positive for SSEA-3 and CD105.
 本発明では、多能性幹細胞から心筋前駆細胞への分化誘導方法は特に限定されないが、例えば、以下の方法を用いることができる。 In the present invention, the method for inducing differentiation from pluripotent stem cells to myocardial progenitor cells is not particularly limited. For example, the following method can be used.
 多能性幹細胞を任意の方法で分離し、浮遊培養もしくはコーティング処理された培養皿を用いて接着培養または/およびフィーダー細胞との共培養を行ってもよい。また、浮遊培養および接着培養を組み合わせて行っても良い。ここで、分離の方法としては、力学的、プロテアーゼ活性とコラゲナーゼ活性を有する分離溶液(例えば、Accutase(TM)およびAccumax(TM)が挙げられる)またはコラゲナーゼ活性のみを有する分離液を用いても良い。ここで、浮遊培養においては、培養皿の表面が、細胞との接着性を向上させる目的で人工的に処理(例えば、細胞外マトリックス等によるコーティング処理)されていないもの、もしくは、人工的に接着を抑制する処理(例えば、ポリヒドロキシエチルメタクリル酸(poly-HEMA)によるコーティング処理)したものを使用できる。接着培養においては、例えば、マトリゲル(BD)、I型コラーゲン、IV型コラーゲン、ゼラチン、ラミニン、ヘパラン硫酸プロテオグリカン、またはエンタクチン、およびこれらの組み合わせを用いてコーティング処理された培養皿を使用できる。また、共培養に用いられる細胞は、OP9細胞(Nishikawa, S.I. et al, Development 125, 1747-1757 (1998))またはEND-2細胞(Mummery C, et al, Circulation. 107:2733-40 (2003))が例示される。 Pluripotent stem cells may be separated by any method, and adhesion culture or / and co-culture with feeder cells may be performed using a culture dish subjected to suspension culture or coating treatment. Further, suspension culture and adhesion culture may be performed in combination. Here, as a separation method, a separation solution having mechanical, protease activity and collagenase activity (for example, Accutase (TM) and Accumax (TM)) or a separation solution having only collagenase activity may be used. . Here, in suspension culture, the surface of the culture dish is not artificially treated (for example, coated with an extracellular matrix or the like) for the purpose of improving adhesion to cells, or artificially adhered. Can be used (for example, coating treatment with polyhydroxyethyl methacrylic acid (poly-HEMA)). In the adhesion culture, for example, a culture dish coated with matrigel (BD), type I collagen, type IV collagen, gelatin, laminin, heparan sulfate proteoglycan, or entactin, and combinations thereof can be used. The cells used for co-culture are OP9 cells (Nishikawa, SI et al, Development 125, 1747-1757 (1998)) or END-2 cells (Mummery C, et al, Circulation. 107: 2733-40 (2003 )) Is exemplified.
 本工程における培地は、動物細胞の培養に用いられる培地を基礎培地として調製することができる。基礎培地としては、例えばIMDM培地、Medium 199培地、Eagle's Minimum Essential Medium (EMEM)培地、αMEM培地、Doulbecco's modified Eagle's Medium (DMEM)培地、Ham's F12培地、RPMI 1640培地、Fischer's培地、およびこれらの混合培地などが包含される。好ましくは、αMEMまたはDMEMである。培地には、血清が含有されていてもよいし、あるいは無血清でもよい。必要に応じて、例えば、アルブミン、トランスフェリン、Knockout Serum Replacement(KSR)(ES細胞培養時のFBSの血清代替物)、脂肪酸、インスリン、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3'-チオールグリセロール、ITS-サプリメントなどの1つ以上の血清代替物を含んでもよいし、B27-サプリメント、N2-サプリメント、脂質、アミノ酸、L-グルタミン、Glutamax(Invitrogen)、非必須アミノ酸、ビタミン、サイトカイン、Wntシグナル阻害剤、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類などの1つ以上の物質も含有しうる。サイトカインとして、アクチビンAおよびBMP4が例示される。Wntシグナル阻害剤として、XAV939(Shih-Min A. Huang, et al, Nature 461, 614-620, 2009)、ビタミンA(レチノイン酸),リチウム,フラボノイド、Dickkopf1(Dkk1)、インスリン様増殖因子結合タンパク質(IGFBP)(WO2009/131166)、βカテニンに対するsiRNA等が例示される。 The medium in this step can be prepared using a medium used for animal cell culture as a basal medium. Examples of the basal medium include IMDM medium, Medium 、 199 medium, Eagle's'Minimum Essential Medium (EMEM) medium, αMEM medium, Doulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium Etc. are included. ΑMEM or DMEM is preferable. The medium may contain serum or may be serum-free. If necessary, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (serum substitute for FBS during ES cell culture), fatty acid, insulin, collagen precursor, trace element, 2-mercaptoethanol, 3'-thiol May contain one or more serum replacements such as glycerol, ITS-supplements, B27-supplements, N2-supplements, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), non-essential amino acids, vitamins, cytokines, Wnt It may also contain one or more substances such as signal inhibitors, antibiotics, antioxidants, pyruvate, buffers, inorganic salts. Examples of cytokines include activin A and BMP4. As Wnt signal inhibitors, XAV939 (Shih-Min A. Huang, et al, Nature 461, 614-620, 2009), vitamin A (retinoic acid), lithium, flavonoid, Dickkopf1 (Dkk1), insulin-like growth factor binding protein Examples include (IGFBP) (WO2009 / 131166), siRNA for β-catenin, and the like.
 培養温度は、以下に限定されないが、約30~40℃、好ましくは約37℃であり、CO2含有空気の雰囲気下で培養が行われ、CO2濃度は、好ましくは約2~5%である。培養時間は、Flk1/KDRが発現するために必要な日数であり、例えば4日以上である。 The culture temperature is not limited to the following, but is about 30 to 40 ° C., preferably about 37 ° C. The culture is performed in an atmosphere of CO 2 -containing air, and the CO 2 concentration is preferably about 2 to 5%. is there. The culture time is the number of days required for Flk1 / KDR to be expressed, for example, 4 days or longer.
 本発明において、心筋細胞を製造するための環状デプシペプチドを心筋前駆細胞へ添加する工程では、上記多能性幹細胞から心筋前駆細胞への分化誘導方法と同様の条件下において実施され得る。環状デプシペプチドを添加する期間は、cTNTまたはαMHCが発現するために必要な日数であり、例えば6日以上である。環状デプシペプチドを添加する濃度は用いる前駆細胞と環状デプシペプチドの種類に応じて適宜選択されるが、好ましくは0.25nM~25nMである。 In the present invention, the step of adding a cyclic depsipeptide for producing cardiomyocytes to myocardial progenitor cells can be performed under the same conditions as in the differentiation induction method from pluripotent stem cells to myocardial progenitor cells. The period during which the cyclic depsipeptide is added is the number of days required for cTNT or αMHC to be expressed, for example, 6 days or longer. The concentration at which cyclic depsipeptide is added is appropriately selected according to the type of progenitor cells and cyclic depsipeptide to be used, but is preferably 0.25 nM to 25 nM.
[実施例]
 以下に実施例を挙げて本発明をより具体的に説明するが、本発明がこれらに限定されないことは言うまでもない。
[Example]
Hereinafter, the present invention will be described more specifically with reference to examples, but it goes without saying that the present invention is not limited thereto.
 マウスα-ミオシン重鎖(MHC)プロモーターにより駆動されるEGFP遺伝子を有するEMG7マウスES細胞から分化誘導し、純化されたFlk1陽性細胞を心筋前駆細胞とし、環状デプシペプチドを添加して培養することにより、心筋細胞へ分化誘導した(Yamashita, J.K. et al, FASEB J. 19, 1534-1536 (2005))。このFlk1陽性細胞をOP9間質細胞上へ播種すると同時に、シクロスポリンA、21-2(式(III))(Luesch, Bioorg Med Chem, 2002)およびAPT-1(式(IV))(Luesch, J Am Chem Soc, 2001)を添加した。同様に、対照として、薬剤の無添加による培養を行った。OP9細胞上へ播種した後、6日後に細胞を回収して、FACSによりEGFP陽性細胞数をカウントした。ここで、回収した全細胞数に対するEGFP陽性細胞数を算出し、対照である無添加群に対するこの値の比率(Fold increase)を用いて各薬剤について評価した。その結果を図1および表2に示す。21-2は、2 nMから4nMの濃度範囲において顕著な活性が確認できた。また、APT-1は、3 nMから4.5nMの濃度範囲において顕著な活性が確認できた。これらの活性は、シクロスポリンAより高かった。
Figure JPOXMLDOC01-appb-C000042
Figure JPOXMLDOC01-appb-C000043
By inducing differentiation from an EMG7 mouse ES cell having an EGFP gene driven by a mouse α-myosin heavy chain (MHC) promoter and using purified Flk1-positive cells as myocardial progenitor cells, adding cyclic depsipeptide and culturing, Differentiation into cardiomyocytes was induced (Yamashita, JK et al, FASEB J. 19, 1534-1536 (2005)). At the same time as seeding these Flk1-positive cells onto OP9 stromal cells, cyclosporin A, 21-2 (formula (III)) (Luesch, Bioorg Med Chem, 2002) and APT-1 (formula (IV)) (Luesch, J Am Chem Soc, 2001) was added. Similarly, as a control, culturing without addition of a drug was performed. After seeding on OP9 cells, the cells were collected 6 days later, and the number of EGFP positive cells was counted by FACS. Here, the number of EGFP positive cells relative to the total number of collected cells was calculated, and each drug was evaluated using the ratio (Fold increase) of this value with respect to the control group without addition. The results are shown in FIG. 21-2 was able to confirm remarkable activity in the concentration range of 2 nM to 4 nM. In addition, APT-1 was confirmed to have significant activity in the concentration range of 3 nM to 4.5 nM. These activities were higher than cyclosporin A.
Figure JPOXMLDOC01-appb-C000042
Figure JPOXMLDOC01-appb-C000043
Figure JPOXMLDOC01-appb-T000044
Figure JPOXMLDOC01-appb-T000044
 実施例1と同様の方法を用いて、APT-7(式(V))、APT-8(式(VI))(Numajiri Y, et al, Chem. Asian J. 2009, 4, 111 - 125)、APT-9(式(VII))(Numajiri Y, et al, Chem. Asian J. 2009, 4, 111 - 125)およびAPT-10(式(VIII))(Doi T, et al, Chem. Asian J. 2011, 6, 180 - 188)についても評価を行った。その結果を図2および表3に示す。APT-7、APT-8、APT-9およびAPT-10は、それぞれ、10nM、7.5 nM、7.5nM から20nMおよび1 nM から1.5nMの濃度範囲において顕著な活性が確認できた。これらの活性は、シクロスポリンAより高かった。
Figure JPOXMLDOC01-appb-C000045
Figure JPOXMLDOC01-appb-C000046
Figure JPOXMLDOC01-appb-C000047
Figure JPOXMLDOC01-appb-C000048
Using the same method as in Example 1, APT-7 (formula (V)), APT-8 (formula (VI)) (Numajiri Y, et al, Chem. Asian J. 2009, 4, 111-125) , APT-9 (formula (VII)) (Numajiri Y, et al, Chem. Asian J. 2009, 4, 111-125) and APT-10 (formula (VIII)) (Doi T, et al, Chem. Asian J. 2011, 6, 180-188) was also evaluated. The results are shown in FIG. APT-7, APT-8, APT-9 and APT-10 were able to confirm remarkable activity in the concentration ranges of 10 nM, 7.5 nM, 7.5 nM to 20 nM and 1 nM to 1.5 nM, respectively. These activities were higher than cyclosporin A.
Figure JPOXMLDOC01-appb-C000045
Figure JPOXMLDOC01-appb-C000046
Figure JPOXMLDOC01-appb-C000047
Figure JPOXMLDOC01-appb-C000048
Figure JPOXMLDOC01-appb-T000049
Figure JPOXMLDOC01-appb-T000049
 心筋細胞中のside population細胞(SP細胞)は、Oyamaらの方法を用いて単離した(Oyama T, et al. J Cell Biol. 176, 329-341, 2007)。詳細には、新生児ラットより抽出した心筋細胞を1.0×106 cells/mlで3%FBS含有PBS 中に懸濁させ、1μg/ml Hoechst 33342を添加し、37℃で60分間暗所にてインキュベート後、フローサイトメーターを用いてSP細胞として単離した。SP細胞は、350nmのUVレーザーでHoechst 33342を励起させ、450nm(Hoechst blue)と675nm(Hoechst red)において特異的位置に検出される細胞として分離した(Goodell MA, et al. J Exp Med. 183:1797-1806, 1996)。この時、Propidium iodideで染色される細胞を死細胞として除去した。得られたSP細胞は、10%FBSを含有するIMDM中で培養し、24時間後に21-2を添加して72時間培養を続けた。さらに、21-2の非添加の条件で培養を継続し、SP細胞単離から3週間後にcTNT抗体染色により細胞を観察した(図3A)。その結果を図3Bに示す。サルコメア構造を持った心筋細胞が誘導され、一部拍動することが確認された。以上より、21-2は、これまで報告されてきた心筋SP細胞から心筋細胞への分化誘導が可能であったオキシトシンやトリコスタチンAと少なくとも同等の機能を有していると推定される。 Side population cells (SP cells) in cardiomyocytes were isolated using the method of Oyama et al. (Oyama T, et al. J Cell Biol. 176, 329-341, 2007). Specifically, cardiomyocytes extracted from newborn rats were suspended at 1.0 × 10 6 cells / ml in PBS containing 3% FBS, 1 μg / ml Hoechst 33342 was added, and incubated at 37 ° C. for 60 minutes in the dark. Thereafter, the cells were isolated as SP cells using a flow cytometer. SP cells were excited as Hoechst 33342 with a 350 nm UV laser and separated as cells detected at specific locations at 450 nm (Hoechst blue) and 675 nm (Hoechst red) (Goodell MA, et al. J Exp Med. 183 : 1797-1806, 1996). At this time, cells stained with Propidium iodide were removed as dead cells. The obtained SP cells were cultured in IMDM containing 10% FBS, and after 21 hours, 21-2 was added and cultivation was continued for 72 hours. Furthermore, the culture was continued under the condition of no addition of 21-2, and the cells were observed by cTNT antibody staining 3 weeks after SP cell isolation (FIG. 3A). The result is shown in FIG. 3B. It was confirmed that cardiomyocytes with sarcomere structure were induced and partly pulsated. Based on the above, it is presumed that 21-2 has at least the same function as oxytocin and trichostatin A, which have been reported so far, capable of inducing differentiation from myocardial SP cells to cardiomyocytes.
 以上の結果から、ここで示した環状デプシペプチドには心筋前駆細胞からの心筋細胞の分化誘導能を有することが確認された。 From the above results, it was confirmed that the cyclic depsipeptide shown here has the ability to induce cardiomyocyte differentiation from myocardial progenitor cells.
 本発明を好ましい態様を強調して説明してきたが、好ましい態様が変更され得ることは当業者にとって自明であろう。本発明は、本発明が本明細書に詳細に記載された以外の方法で実施され得ることを意図する。したがって、本発明は添付の「請求の範囲」の精神及び範囲に包含されるすべての変更を含むものである。
 ここで述べられた特許及び特許出願明細書を含む全ての刊行物に記載された内容は、ここに引用されたことによって、その全てが明示されたと同程度に本明細書に組み込まれるものである。
While the invention has been described with emphasis on preferred embodiments, it will be apparent to those skilled in the art that the preferred embodiments can be modified. The present invention contemplates that the present invention may be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications encompassed within the spirit and scope of the appended claims.
The contents of all publications, including the patents and patent application specifications mentioned herein, are hereby incorporated by reference herein to the same extent as if all were expressly cited. .
 本発明によれば、心筋前駆細胞からの心筋細胞誘導効率を顕著に向上させることができるので、心疾患の治療または多能性幹細胞を用いた細胞移植治療への応用に特に有用である。 According to the present invention, the efficiency of inducing cardiomyocytes from myocardial progenitor cells can be remarkably improved, which is particularly useful for the treatment of heart diseases or cell transplantation using pluripotent stem cells.

Claims (9)

  1.  以下の式(I)または式(II)(式中、R1およびR3はそれぞれHまたはC1-C6アルキル基であり、R2はH、C1-C6アルキル基、トリメチルシリル基およびトリエチルシリル基からなる群から選択される置換基であり、R4はH、OH、C1-C6アルコキシ基、F、Cl、BrおよびIからなる群から選択される置換基であり、L-MはC(Me)=CH、CH=CH、CH2-CH2およびCH(Me)-CH2からなる群から選択される構造を有し、Xは、OまたはSであり、nは、1または2である)で表される環状デプシペプチドまたはその薬学上許容される塩、溶媒和物もしくはプロドラッグを有効成分として含有する医薬組成物。
    Figure JPOXMLDOC01-appb-C000001
    Figure JPOXMLDOC01-appb-C000002
    The following formula (I) or formula (II) (wherein R 1 and R 3 are each H or C1-C6 alkyl group, R 2 is H, C1-C6 alkyl group, trimethylsilyl group and triethylsilyl group) R 4 is a substituent selected from the group consisting of H, OH, C1-C6 alkoxy group, F, Cl, Br and I, and LM is C (Me) = Having a structure selected from the group consisting of CH, CH═CH, CH 2 —CH 2 and CH (Me) —CH 2 , X is O or S, and n is 1 or 2. A pharmaceutical composition comprising the represented cyclic depsipeptide or a pharmaceutically acceptable salt, solvate or prodrug thereof as an active ingredient.
    Figure JPOXMLDOC01-appb-C000001
    Figure JPOXMLDOC01-appb-C000002
  2.  R1がイソプロピル基またはt-ブチル基であり、R2がHであり、R3がメチル基であり、R4がメトキシ基であり、L-MがC(Me) =CHであり、XがOまたはSであり、nが1である、請求項1に記載の医薬組成物。 R 1 is an isopropyl group or a t-butyl group, R 2 is H, R 3 is a methyl group, R 4 is a methoxy group, LM is C (Me) = CH, and X is O Or the pharmaceutical composition of Claim 1 which is S and n is 1.
  3.  環状デプシペプチドが、次の式(III)から式(VIII)で表される化合物から成る群より選択される、請求項1に記載の医薬組成物;
    Figure JPOXMLDOC01-appb-C000003
    Figure JPOXMLDOC01-appb-C000004
    Figure JPOXMLDOC01-appb-C000005
    Figure JPOXMLDOC01-appb-C000006
    Figure JPOXMLDOC01-appb-C000007
    Figure JPOXMLDOC01-appb-C000008
    The pharmaceutical composition according to claim 1, wherein the cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII):
    Figure JPOXMLDOC01-appb-C000003
    Figure JPOXMLDOC01-appb-C000004
    Figure JPOXMLDOC01-appb-C000005
    Figure JPOXMLDOC01-appb-C000006
    Figure JPOXMLDOC01-appb-C000007
    Figure JPOXMLDOC01-appb-C000008
  4.  心不全、虚血性心疾患または心筋症から選択される心疾患の治療に用いられる請求項1~3のいずれか一項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 3, which is used for the treatment of a heart disease selected from heart failure, ischemic heart disease or cardiomyopathy.
  5.  以下の式(I)または式(II)(式中、R1およびR3はそれぞれHまたはC1-C6アルキル基であり、R2はH、C1-C6アルキル基、トリメチルシリル基およびトリエチルシリル基からなる群から選択される置換基であり、R4はH、OH、C1-C6アルコキシ基、F、Cl、BrおよびIからなる群から選択される置換基であり、L-MはC(Me)=CH、CH=CH、CH2-CH2およびCH(Me)-CH2からなる群から選択される構造を有し、Xは、OまたはSであり、nは、1または2である)で表される環状デプシペプチドを心筋前駆細胞へ添加する工程を含む、心筋細胞の製造方法。
    Figure JPOXMLDOC01-appb-C000009
    Figure JPOXMLDOC01-appb-C000010
    The following formula (I) or formula (II) (wherein R 1 and R 3 are each H or C1-C6 alkyl group, R 2 is H, C1-C6 alkyl group, trimethylsilyl group and triethylsilyl group) R 4 is a substituent selected from the group consisting of H, OH, C1-C6 alkoxy group, F, Cl, Br and I, and LM is C (Me) = Having a structure selected from the group consisting of CH, CH═CH, CH 2 —CH 2 and CH (Me) —CH 2 , X is O or S, and n is 1 or 2. A method for producing cardiomyocytes, comprising a step of adding the expressed cyclic depsipeptide to myocardial progenitor cells.
    Figure JPOXMLDOC01-appb-C000009
    Figure JPOXMLDOC01-appb-C000010
  6.  R1がイソプロピル基またはt-ブチル基であり、R2がHであり、R3がメチル基であり、R4がメトキシ基であり、L-MがC(Me)=CHであり、XがOまたはSであり、nが1である、請求項5に記載の方法。 R 1 is an isopropyl group or a t-butyl group, R 2 is H, R 3 is a methyl group, R 4 is a methoxy group, LM is C (Me) = CH, and X is O 6. The method of claim 5, wherein S is n and n is 1.
  7.  環状デプシペプチドが、次の式(III)から式(VIII)で表される化合物から成る群より選択される、請求項5に記載の方法。
    Figure JPOXMLDOC01-appb-C000011
    Figure JPOXMLDOC01-appb-C000012
    Figure JPOXMLDOC01-appb-C000013
    Figure JPOXMLDOC01-appb-C000014
    Figure JPOXMLDOC01-appb-C000015
    Figure JPOXMLDOC01-appb-C000016
    The method according to claim 5, wherein the cyclic depsipeptide is selected from the group consisting of compounds represented by the following formulas (III) to (VIII).
    Figure JPOXMLDOC01-appb-C000011
    Figure JPOXMLDOC01-appb-C000012
    Figure JPOXMLDOC01-appb-C000013
    Figure JPOXMLDOC01-appb-C000014
    Figure JPOXMLDOC01-appb-C000015
    Figure JPOXMLDOC01-appb-C000016
  8.  前記心筋前駆細胞が、side population細胞(SP細胞)を含む生体内の心筋前駆細胞、または多能性幹細胞から誘導された心筋前駆細胞である、請求項5~7のいずれか一項に記載の方法。 The myocardial progenitor cell according to any one of claims 5 to 7, wherein the myocardial progenitor cell is an in vivo myocardial progenitor cell including side-population cells (SP cells) or a myocardial progenitor cell derived from a pluripotent stem cell. Method.
  9.  前記多能性幹細胞から誘導された心筋前駆細胞が、Flk1 (KDR)陽性の細胞である、請求項8に記載の方法。 The method according to claim 8, wherein the myocardial progenitor cell derived from the pluripotent stem cell is a Flk1 (KDR) positive cell.
PCT/JP2013/077279 2012-09-03 2013-10-07 Novel use method for cyclic depsipeptide WO2014042290A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2012193433A JP6024880B2 (en) 2012-09-03 2012-09-03 Novel usage of cyclic depsipeptide
JP2012-193433 2012-09-03

Publications (1)

Publication Number Publication Date
WO2014042290A1 true WO2014042290A1 (en) 2014-03-20

Family

ID=50278388

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2013/077279 WO2014042290A1 (en) 2012-09-03 2013-10-07 Novel use method for cyclic depsipeptide

Country Status (2)

Country Link
JP (1) JP6024880B2 (en)
WO (1) WO2014042290A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06135992A (en) * 1991-01-24 1994-05-17 Merck & Co Inc New endoserine receptor antagonist isolated from microbispora
JPH11171897A (en) * 1997-05-29 1999-06-29 Meiji Milk Prod Co Ltd Cyclic depsipeptide and medicine containing the same as active ingedient
WO2009032338A1 (en) * 2007-09-09 2009-03-12 University Of Florida Research Foundation Apratoxin therapeutic agents: mechanism and methods of treatment
JP2009520683A (en) * 2005-10-27 2009-05-28 リード ビリオン リミテッド Pharmaceutical compositions and methods for regeneration of muscle fibers in the treatment of muscle injury
WO2010065563A2 (en) * 2008-12-01 2010-06-10 University Of Florida Research Foundation, Inc. Apratoxin therapeutic agents: mechanism and methods of treatment

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06135992A (en) * 1991-01-24 1994-05-17 Merck & Co Inc New endoserine receptor antagonist isolated from microbispora
JPH11171897A (en) * 1997-05-29 1999-06-29 Meiji Milk Prod Co Ltd Cyclic depsipeptide and medicine containing the same as active ingedient
JP2009520683A (en) * 2005-10-27 2009-05-28 リード ビリオン リミテッド Pharmaceutical compositions and methods for regeneration of muscle fibers in the treatment of muscle injury
WO2009032338A1 (en) * 2007-09-09 2009-03-12 University Of Florida Research Foundation Apratoxin therapeutic agents: mechanism and methods of treatment
WO2010065563A2 (en) * 2008-12-01 2010-06-10 University Of Florida Research Foundation, Inc. Apratoxin therapeutic agents: mechanism and methods of treatment

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DAWEI MA ET AL. ET AL.: "Total Synthesis of the Cyclodepsipeptide Apratoxin A and Its Analogues and Assessment of Their Biological Activities, Chemistry", A EUROPEAN JOURNAL, vol. 12, no. 29, 2006, pages 7615 - 7626 *

Also Published As

Publication number Publication date
JP2014047193A (en) 2014-03-17
JP6024880B2 (en) 2016-11-16

Similar Documents

Publication Publication Date Title
JP6461787B2 (en) Method for inducing alveolar epithelial progenitor cells
JP7166631B2 (en) Novel chondrocyte induction method
JP6429280B2 (en) Efficient cardiomyocyte induction method
JP6738572B2 (en) Method for inducing differentiation of alveolar epithelial cells
JP6143027B2 (en) How to induce astrocytes
JP6694215B2 (en) New chondrocyte induction method
JP6373253B2 (en) New cardiomyocyte marker
JP5995247B2 (en) Method for producing dendritic cells from pluripotent stem cells
JP6025067B2 (en) New cardiomyocyte marker
WO2016114354A1 (en) Method for selecting skeletal muscle progenitor cell
JP6730709B2 (en) Method for inducing differentiation of airway epithelial cells
WO2021085462A1 (en) Method for producing hemogenic endothelial cell and/or hematopoietic progenitor cell from pluripotent stem cell
JP6024880B2 (en) Novel usage of cyclic depsipeptide
JP7429294B2 (en) Skeletal muscle progenitor cells and methods for purifying the same, compositions for treating myogenic diseases, and methods for producing cell groups containing skeletal muscle progenitor cells
WO2021015086A1 (en) Method for producing mature myotube cells from skeletal muscle stem cells
WO2017073794A1 (en) Method for producing three-dimensional myocardial tissue from pluripotent stem cells
JP7072756B2 (en) Method for inducing differentiation of pluripotent stem cells into mesoderm progenitor cells and blood vascular progenitor cells
JP2020115771A (en) Method for producing cartilage tissue from pluripotent stem cell

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: 13836947

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13836947

Country of ref document: EP

Kind code of ref document: A1