JP2016093178A - Culture medium for differentiated cells derived from stem cells, production of differentiated cells from stem cells, and method for production of cell pharmaceutical composition containing differentiated cells - Google Patents

Culture medium for differentiated cells derived from stem cells, production of differentiated cells from stem cells, and method for production of cell pharmaceutical composition containing differentiated cells Download PDF

Info

Publication number
JP2016093178A
JP2016093178A JP2015220829A JP2015220829A JP2016093178A JP 2016093178 A JP2016093178 A JP 2016093178A JP 2015220829 A JP2015220829 A JP 2015220829A JP 2015220829 A JP2015220829 A JP 2015220829A JP 2016093178 A JP2016093178 A JP 2016093178A
Authority
JP
Japan
Prior art keywords
cells
cell
stem
stem cells
differentiated
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
JP2015220829A
Other languages
Japanese (ja)
Other versions
JP6744084B2 (en
Inventor
義基 中島
Yoshiki Nakashima
義基 中島
健史 大政
Takeshi Omasa
健史 大政
正義 塚原
Masayoshi Tsukahara
正義 塚原
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyowa Kirin Co Ltd
Original Assignee
Kyowa Hakko Kirin Co Ltd
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 Kyowa Hakko Kirin Co Ltd filed Critical Kyowa Hakko Kirin Co Ltd
Publication of JP2016093178A publication Critical patent/JP2016093178A/en
Application granted granted Critical
Publication of JP6744084B2 publication Critical patent/JP6744084B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

PROBLEM TO BE SOLVED: To provide a technique of preparing differentiated cells without the contamination with undifferentiated stem cells, in differentiated cells induced from stem cells; and to provide a pharmaceutical composition containing a stem cell survival inhibitor for inhibiting in vivo neoplastic transformation of a cell pharmaceutical composition containing differentiated cells derived from stem cells.SOLUTION: The invention provides a culture medium for differentiated cells derived from stem cells by which differentiated cells without contamination with undifferentiated stem cells can be obtained by compounding, into the undifferentiated stem cells, atrovastatin which has a specific survival inhibiting activity and inhibits the survival of undifferentiated stem cells without inducing the apoptosis of the differentiated cells.SELECTED DRAWING: Figure 1

Description

本発明は、幹細胞由来の分化細胞用培地、幹細胞からの分化細胞の製造及び該分化細胞を含む細胞医薬組成物の製造のための方法に関する。より詳しくは、アトルバスタチンによって幹細胞の生存を抑制して分化細胞のみを選択的に培養可能な培地等に関する。   The present invention relates to a medium for differentiated cells derived from stem cells, production of differentiated cells from stem cells, and a method for producing a cell pharmaceutical composition containing the differentiated cells. More specifically, the present invention relates to a medium or the like that can selectively culture only differentiated cells by suppressing the survival of stem cells with atorvastatin.

人工多能性幹細胞(iPS細胞)、胚性幹細胞(ES細胞)等の多分化能幹細胞を用いる再生医療技術の課題のひとつは、多分化能幹細胞を所望のタイプの細胞に分化させた後に患者の体内に移植する際に、多分化能幹細胞が未分化状態のまま残存し、分化した細胞とともに患者の体内に移植され、患者の体内で腫瘍及び癌化する危険を如何に防止するかである(非特許文献1参照)。   One of the challenges of regenerative medicine technology using pluripotent stem cells such as artificial pluripotent stem cells (iPS cells) and embryonic stem cells (ES cells) is to differentiate patients from pluripotent stem cells into desired types of cells. How to prevent the risk of pluripotent stem cells remaining in an undifferentiated state when transplanted into the body of the patient, transplanted into the patient's body together with the differentiated cells, and becoming a tumor and cancer in the patient's body (Refer nonpatent literature 1).

造腫瘍性を持つおそれのある未分化iPS細胞等の混入を評価する試験系としては、未分化多能性細胞特異的なマーカーや分化能の高い細胞に特異的なマーカー(非特許文献2参照)の発現を指標にしたフローサイトメトリーや定量的RT-PCR(qRT-PCR)が挙げられる。しかし、いずれも一定の頻度以下の未分化多能性幹細胞の混入は検出できず、最終製品を未分化多能性幹細胞培養条件に戻して培養してiPS細胞等のコロニーが出現しないことの確認などが必要である。   As a test system for evaluating contamination of undifferentiated iPS cells or the like that may have tumorigenicity, a marker specific to undifferentiated pluripotent cells or a marker specific to cells with high differentiation potential (see Non-Patent Document 2) ) Expression as an index, and quantitative RT-PCR (qRT-PCR). However, in any case, contamination of undifferentiated pluripotent stem cells below a certain frequency cannot be detected, and it is confirmed that colonies such as iPS cells do not appear when the final product is cultured after returning to undifferentiated pluripotent stem cell culture conditions. Etc. are necessary.

アトルバスタチン(Monocalcium bis{(3R,5R)−7−[2−(4−fluorophenyl)−5−(1−methylethyl)−3−phenyl−4−(phenylcarbamoyl)−1H−pyrrol−1−yl]−3,5−dihydroxyheptanoate} trihydrate、式(I)参照)は、HMG-CoA還元酵素阻害剤(スタチン)と呼ばれる薬剤の一種である。   Atorvastatin (Monocalcum bis {(3R, 5R) -7- [2- (4-fluorophenyl) -5- (1-methylethyl) -3-phenyl-4- (phenylcarbamoyl) -1H-pyrol-1-yl] -3 , 5-dihydroxyheptanoate} trihydrate (see formula (I)) is a kind of drug called HMG-CoA reductase inhibitor (statin).

スタチン薬剤は、現在、シンバスタチン(Simvastatin)、フルバスタチン(Fluvastatin)、ロバスタチン(Lovastatin)、アトルバスタチン(Atorvastatin)、ピタバスタチン(Pitavastatin)、プラバスタチン(Pravastatin)、メバスタチン(Mevastatin)及びロスバスタチン(Rosuvastatin)が知られている。スタチン薬剤は、心血管疾患についてのリスク患者における血中低密度リポタンパク質(LDL)濃度を低下させるために最も効果的な薬剤である。血中における高レベルのLDLは、血流の遮断、血管の破裂、血栓症を促進する冠動脈損傷の形成に関連している(非特許文献3参照)。   Statin drugs are currently simvastatin, fluvastatin, lovastatin, atorvastatin, pitavastatin, pravastatin, pravastatin, and mevastatin (Mevastatin). Yes. Statin drugs are the most effective drugs for reducing blood low density lipoprotein (LDL) levels in patients at risk for cardiovascular disease. High levels of LDL in the blood are associated with the formation of coronary artery damage that promotes blockage of blood flow, rupture of blood vessels, and thrombosis (see Non-Patent Document 3).

本発明に関連して、特許文献1には、iPS細胞の腫瘍化を抑制して目的の分化細胞に分化させる技術が開示されている。当該文献の実施例には、iPS細胞を骨芽細胞へ誘導し免疫不全マウスに移植する実験において、シンバスタチン、フルバスタチン又はロバスタチンを含む培地中で分化誘導を行うことにより、移植された細胞による腫瘍形成を抑制できたことが記載されている。しかし、引用文献1には、アトルバスタチンについて具体的な検討はなされておらず、またiPS細胞の心筋細胞への分化誘導に関する具体的な開示はなされていない。   In relation to the present invention, Patent Document 1 discloses a technique for suppressing iPS cell tumorigenesis and differentiating into an objective differentiated cell. In the examples of this document, in an experiment in which iPS cells are induced into osteoblasts and transplanted into immunodeficient mice, differentiation induction is performed in a medium containing simvastatin, fluvastatin, or lovastatin, so that the tumor caused by the transplanted cells. It is described that the formation could be suppressed. However, Cited Document 1 does not specifically examine atorvastatin, and does not specifically disclose the induction of iPS cell differentiation into cardiomyocytes.

国際公開第2013/100080号International Publication No. 2013/100080

J. Cell Sci.,2010,123,p.643−651J. et al. Cell Sci. 2010, 123, p. 643-651 PNAS,2013,110,51,p.20569-20574PNAS, 2013, 110, 51, p. 20569-20574 The Pharmacological Basis of Therapeutics,1996,9,p.879The Pharmacological Basis of Therapeutics, 1996, 9, p. 879

本発明は、幹細胞から誘導される分化細胞において、未分化な幹細胞の混入のない分化細胞を調製するための技術を提供することを主な目的とする。   The main object of the present invention is to provide a technique for preparing differentiated cells free from contamination of undifferentiated stem cells in differentiated cells derived from stem cells.

[1]アトルバスタチンを含む、幹細胞由来の分化細胞用培地。
[2]前記幹細胞が誘導性多能性幹細胞である[1]の培地。
[3]前記分化細胞が心筋細胞である[1]又は[2]の培地。
[4]前記幹細胞がヒト由来である[1]〜[3]のいずれかの培地。
[5]アトルバスタチンを濃度1〜20μMで含む[1]〜[4]のいずれかの培地。
[6]前記培地が無血清培地である、[1]〜[5]のいずれかの培地。
[1] A medium for differentiated cells derived from stem cells, containing atorvastatin.
[2] The medium according to [1], wherein the stem cells are inducible pluripotent stem cells.
[3] The medium according to [1] or [2], wherein the differentiated cells are cardiomyocytes.
[4] The medium according to any one of [1] to [3], wherein the stem cells are derived from human.
[5] The medium according to any one of [1] to [4], containing atorvastatin at a concentration of 1 to 20 μM.
[6] The medium according to any one of [1] to [5], wherein the medium is a serum-free medium.

[7]アトルバスタチンを有効成分とする幹細胞生存抑制剤。
[8]前記幹細胞が誘導性多能性幹細胞である[7]の幹細胞生存抑制剤。
[9]前記幹細胞がヒト由来である[7]又は[8]の幹細胞生存抑制剤。
[10]幹細胞由来の分化細胞を含む細胞医薬組成物の生体内での腫瘍化を抑制するため医薬組成物であり、[7]〜[9]のいずれかの幹細胞生存抑制剤を含む医薬組成物。
[7] A stem cell survival inhibitor comprising atorvastatin as an active ingredient.
[8] The stem cell survival inhibitor of [7], wherein the stem cells are inducible pluripotent stem cells.
[9] The stem cell survival inhibitor of [7] or [8], wherein the stem cells are derived from human.
[10] A pharmaceutical composition for suppressing tumor formation in vivo of a cell pharmaceutical composition containing differentiated cells derived from stem cells, comprising the stem cell survival inhibitor of any one of [7] to [9] object.

[11]幹細胞から分化細胞を製造する方法であって、分化誘導後の細胞をアトルバスタチンにより処理する工程を含む方法。
[12]前記幹細胞が誘導性多能性幹細胞である[11]の方法。
[13]前記分化細胞が心筋細胞である[11]又は[12]の方法
[14]前記幹細胞がヒト由来である[11]〜[13]のいずれかの方法。
[15]幹細胞を培養する工程と、幹細胞を分化誘導する工程と、をさらに含む[11]〜[14]のいずれかの方法。
[11] A method for producing a differentiated cell from a stem cell, comprising a step of treating the cell after differentiation induction with atorvastatin.
[12] The method of [11], wherein the stem cell is an inducible pluripotent stem cell.
[13] The method according to [11] or [12], wherein the differentiated cells are cardiomyocytes [14] The method according to any one of [11] to [13], wherein the stem cells are derived from human.
[15] The method according to any one of [11] to [14], further comprising a step of culturing stem cells and a step of inducing differentiation of stem cells.

[16]幹細胞由来の分化細胞を含む細胞医薬組成物を製造する方法であって、前記幹細胞を分化誘導する工程と、分化誘導後の細胞をアトルバスタチンにより処理する工程と、を含む方法。
[17]前記幹細胞が誘導性多能性幹細胞である[16]の方法。
[18]前記分化細胞が心筋細胞である[16]又は[17]の方法
[19]前記幹細胞がヒト由来である[16]〜[18]のいずれかの方法。
[20]幹細胞を培養する工程、をさらに含む[16]〜[19]のいずれかの方法。
[16] A method for producing a cell pharmaceutical composition comprising differentiated cells derived from stem cells, the method comprising the step of inducing differentiation of the stem cells and the step of treating the cells after differentiation induction with atorvastatin.
[17] The method according to [16], wherein the stem cell is an inducible pluripotent stem cell.
[18] The method according to [16] or [17], wherein the differentiated cells are cardiomyocytes [19] The method according to any one of [16] to [18], wherein the stem cells are derived from human.
[20] The method according to any one of [16] to [19], further comprising a step of culturing stem cells.

[21]幹細胞及び分化細胞を含む細胞混合物から分化細胞のみを分離する方法であって、
前記細胞混合物をアトルバスタチンにより処理する手順を含む方法。
[22]前記幹細胞が誘導性多能性幹細胞である[21]の方法。
[23]前記分化細胞が心筋細胞である[21]又は[22]の方法
[24]前記幹細胞及び前記分化細胞がヒト由来である[21]〜[23]のいずれかの方法。
[25]幹細胞を培養する工程と、幹細胞を分化誘導する工程と、をさらに含む[21]〜[24]のいずれかの方法。
[21] A method for separating only differentiated cells from a cell mixture containing stem cells and differentiated cells,
Treating the cell mixture with atorvastatin.
[22] The method of [21], wherein the stem cell is an inducible pluripotent stem cell.
[23] The method according to [21] or [22], wherein the differentiated cell is a cardiomyocyte. [24] The method according to any one of [21] to [23], wherein the stem cell and the differentiated cell are derived from human.
[25] The method according to any one of [21] to [24], further comprising a step of culturing a stem cell and a step of inducing differentiation of the stem cell.

[26][1]の培地、[7]の幹細胞生存抑制剤、[10]の医薬組成物の製造のためのアトルバスタチンの使用。
[27]幹細胞由来の分化細胞を含む細胞医薬組成物の製造のためのアトルバスタチンの使用。
[28]幹細胞由来の分化細胞を含む細胞医薬組成物の生体内での腫瘍化を抑制するためのアトルバスタチンの使用。
[26] Use of atorvastatin for the production of the medium of [1], the stem cell survival inhibitor of [7], and the pharmaceutical composition of [10].
[27] Use of atorvastatin for the production of a cell pharmaceutical composition containing differentiated cells derived from stem cells.
[28] Use of atorvastatin for suppressing in vivo tumorigenesis of a cell pharmaceutical composition containing differentiated cells derived from stem cells.

本発明により、幹細胞から誘導される分化細胞において、未分化な幹細胞の混入のない分化細胞を調製するための技術が提供される。   The present invention provides a technique for preparing differentiated cells free from contamination of undifferentiated stem cells in differentiated cells derived from stem cells.

培養ヒトiPS細胞に対するスタチン類の生存抑制活性を評価した結果を示すグラフである(実施例1)。It is a graph which shows the result of having evaluated the survival inhibitory activity of statins with respect to a cultured human iPS cell (Example 1). 培養ヒトiPS細胞に対するアトルバスタチンの生存抑制活性を評価した結果を示すグラフである(実施例1)。It is a graph which shows the result of having evaluated the survival inhibitory activity of atorvastatin with respect to a cultured human iPS cell (Example 1). 培養ヒトiPS細胞に対するアトルバスタチンの生存抑制活性を評価した結果を示すグラフである(実施例1)。It is a graph which shows the result of having evaluated the survival inhibitory activity of atorvastatin with respect to a cultured human iPS cell (Example 1). 培養心筋細胞に対するアトルバスタチンの生存抑制活性(細胞死誘導活性)を評価した結果を示すグラフである(実施例2)。It is a graph which shows the result of having evaluated the survival inhibitory activity (cell death induction activity) of the atorvastatin with respect to a cultured cardiomyocyte (Example 2).

以下、本発明を実施するための好適な形態について図面を参照しながら説明する。なお、以下に説明する実施形態は、本発明の代表的な実施形態の一例を示したものであり、これにより本発明の範囲が狭く解釈されることはない。   DESCRIPTION OF EXEMPLARY EMBODIMENTS Hereinafter, preferred embodiments for carrying out the invention will be described with reference to the drawings. In addition, embodiment described below shows an example of typical embodiment of this invention, and, thereby, the range of this invention is not interpreted narrowly.

1.分化細胞用培地
本発明に係る分化細胞用培地は、幹細胞から分化細胞を誘導するために用いられるものであり、アトルバスタチンを含むことを特徴とする。本発明において、アトルバスタチンは、未分化な幹細胞に特異的な生存抑制活性を有し、分化細胞(特に心筋細胞)の細胞死を誘発することなく、未分化幹細胞の生存を抑制することが明らかとなった。
1. Medium for differentiated cells The medium for differentiated cells according to the present invention is used for inducing differentiated cells from stem cells, and is characterized by containing atorvastatin. In the present invention, it is apparent that atorvastatin has a survival inhibitory activity specific to undifferentiated stem cells and suppresses the survival of undifferentiated stem cells without inducing cell death of differentiated cells (particularly cardiomyocytes). became.

本発明に係る培地において、アトルバスタチンの濃度は、未分化幹細胞に対して生存抑制活性を示し、かつ分化細胞の細胞死を誘発することがない濃度である限りにおいて、特に限定されない。このような濃度は、実施例記載の方法及び従来公知の方法を用いて当業者が適宜設定可能である。アトルバスタチンの濃度は、例えば0.01〜2000μM、好ましくは0.1〜200μM、より好ましくは1〜20μMとされる。   In the medium according to the present invention, the concentration of atorvastatin is not particularly limited as long as it is a concentration that exhibits a survival inhibitory activity against undifferentiated stem cells and does not induce cell death of differentiated cells. Such a concentration can be appropriately set by those skilled in the art using the methods described in the Examples and conventionally known methods. The concentration of atorvastatin is, for example, 0.01 to 2000 μM, preferably 0.1 to 200 μM, more preferably 1 to 20 μM.

本発明に係る培地は、幹細胞の培養や幹細胞からの分化細胞の誘導のために従来用いられている培地(基礎培地)に、アトルバスタチンを上記濃度で添加することにより調製できる。このような培地としては、例えば、以下を挙げることができる。   The medium according to the present invention can be prepared by adding atorvastatin at the above concentration to a medium conventionally used for culturing stem cells or inducing differentiated cells from stem cells (basal medium). Examples of such a medium include the following.

[基礎培地]
RPMI-1640培地、EagleのMEM培地、ダルベッコ改変MEM培地、Glasgow’s MEM培地、α-MEM培地、199培地、IMDM培地、DMEM培地Hybridoma Serum free培地、Chemically Defined Hybridoma Serum Free培地、Ham’s Medium F-12、Ham’s Medium F-10、Ham’s Medium F12K、ATCC-CRCM30、DM-160、DM-201、BME、Fischer、McCoy’s 5A、Leibovitz’s L-15、RITC80-7、MCDB105、MCDB107、MCDB131、MCDB153、MCDB201、NCTC109、NCTC135、Waymouth’s MB752/1、CMRL-1066、Williams’ medium E、Brinster’s BMOC-3 Medium、E8 Medium、E8 medium(以上サーモフィッシャーサイエンティフィック)、ReproFF2(リプロセル社)、EX-CELL 302培地(SAFC社)またはEX-CELL-CD-CHO(SAFC社)及びこれらの混合物。
[Basic medium]
RPMI-1640 medium, Eagle's MEM medium, Dulbecco's modified MEM medium, Glasgow's MEM medium, α-MEM medium, 199 medium, IMDM medium, DMEM medium Hybridoma Serum free medium, Chemically defined HybridSumFarm medium's medium F-12, Ham's Medium F-10, Ham's Medium F12K, ATCC-CRCM30, DM-160, DM-201, BME, Fischer, McCoy's 5A, Leibovitz's L-15, RITC80-7, MCDB105, MCDB107, MCDB131, MCDB153, MCDB201, NCTC109, NCTC135, Waymout's MB752 / 1, CMRL-1 66, Williams' medium E, Brinster's BMOC-3 Medium, E8 Medium, E8 medium (above Thermo Fisher Scientific), ReproFF2 (Reprocell), EX-CELL 302 medium (SAFC) or EX-CELL-CD -CHO (SAFC) and mixtures thereof.

本発明に係る培地は、これらの培地にアトルバスタチンを予め添加あるいは細胞培養中に添加することによって調製できる。   The medium according to the present invention can be prepared by adding atorvastatin to these mediums in advance or during cell culture.

また、培地には、必要に応じて細胞の生存又は増殖に必要な生理活性物質及び栄養因子などを添加できる。これらの添加物は、培地に予め添加されていてもよく、細胞培養中に添加されてもよい。培養中に添加する方法は、1溶液または2種以上の混合溶液などいかなる形態によってでもよく、連続的または断続的な添加であってよい。   In addition, physiologically active substances and nutrient factors necessary for cell survival or growth can be added to the medium as necessary. These additives may be added in advance to the medium, or may be added during cell culture. The method to be added during the culture may be in any form such as one solution or a mixed solution of two or more kinds, and may be continuous or intermittent addition.

生理活性物質としては、インシュリン、IGF−1、トランスフェリン、アルブミンまたは補酵素Q10などが挙げられる。
栄養因子としては、糖、アミノ酸、ビタミン、加水分解物または脂質などが挙げられる。
糖としては、グルコース、マンノースまたはフルクトースなどが挙げられ、1種または2種以上を組み合わせて用いられる。
アミノ酸としては、L−アラニン、L−アルギニン、L−アスパラギン、L−アスパラギン酸、L−システイン、L−グルタミン酸、L−グルタミン、グリシン、L−ヒスチジン、L−イソロイシン、L−ロイシン、L−リジン、L−メチオニン、L−フェニルアラニン、L−プロリン、L−セリン、L−スレオニン、L−トリプトファン、L−チロシンまたはL−バリンなどが挙げられ、1種または2種以上を組み合わせて用いられる。
ビタミンとしては、d−ビオチン、D−パントテン酸、コリン、葉酸、myo−イノシトール、ナイアシンアミド、ピロドキサール、リボフラビン、チアミン、シアノコバラミンまたはDL−α―トコフェロールなどが挙げられ、1種または2種以上を組み合わせて用いられる。
加水分解物としては、大豆、小麦、米、えんどう豆、とうもろこし、綿実、酵母抽出物などを加水分解したものが挙げられる。
脂質としては、コレステロール、リノール酸またはリノレイン酸などが挙げられる。
A physiologically active substance, insulin, IGF-1, transferrin, and the like albumin or coenzyme Q 10.
Nutritional factors include sugars, amino acids, vitamins, hydrolysates or lipids.
Examples of the sugar include glucose, mannose, and fructose, and one kind or a combination of two or more kinds is used.
As amino acids, L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine , L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine and the like, and one kind or a combination of two or more kinds is used.
Examples of vitamins include d-biotin, D-pantothenic acid, choline, folic acid, myo-inositol, niacinamide, pyrodoxal, riboflavin, thiamine, cyanocobalamin or DL-α-tocopherol, and one or a combination of two or more Used.
Examples of the hydrolyzate include hydrolyzed soybeans, wheat, rice, peas, corn, cottonseed, yeast extract and the like.
Examples of lipids include cholesterol, linoleic acid, and linolenic acid.

さらに、培地には、カナマイシン、ストレプトマイシン、ペニシリンまたはハイグロマイシンなどの抗生物質を必要に応じて添加してもよい。シアル酸等の酸性物質を培地に添加する場合には、培地のpHを細胞の成育に適した中性域であるpH5〜9、好ましくはpH6〜8に調整することが望ましい。   Further, an antibiotic such as kanamycin, streptomycin, penicillin or hygromycin may be added to the medium as necessary. When an acidic substance such as sialic acid is added to the medium, it is desirable to adjust the pH of the medium to pH 5-9, preferably pH 6-8, which is a neutral range suitable for cell growth.

本発明に係る培地は、血清含培地であっても無血清培地であってもよい。異種動物由来成分の混入防止の観点からは血清を含有しないか、培養される幹細胞と同種動物由来の血清が用いられることが好ましい。ここで、無血清培地とは、無調整又は未精製の血清を含まない培地を意味する。無血清培地は、精製された血液由来成分や動物組織由来成分(例えば、増殖因子)を含有していてもよい。   The medium according to the present invention may be a serum-containing medium or a serum-free medium. From the viewpoint of preventing contamination with heterogeneous animal-derived components, it is preferable not to contain serum or to use serum derived from the same species as the cultured stem cells. Here, the serum-free medium means a medium containing no unadjusted or unpurified serum. The serum-free medium may contain a purified blood-derived component or animal tissue-derived component (for example, a growth factor).

本発明に係る培地は、血清と同様に、血清代替物についてもこれを含んでいても含んでいなくともよい。血清代替物としては、例えば、アルブミン、脂質リッチアルブミン及び組換えアルブミン等のアルブミン代替物、植物デンプン、デキストラン、タンパク質加水分解物、トランスフェリン又は他の鉄輸送体、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’-チオグリセロールあるいはこれらの均等物などが挙げられ得る。血清代替物の具体例として、例えば、国際公開第98/30679号記載の方法により調製されるものや、市販のknockout Serum Replacement(KSR社)、Chemically-defined Lipid concentrated(Life Technologies社)及びGlutamax(Life Technologies社)などが挙げられる。   The medium according to the present invention may or may not contain a serum substitute as well as serum. Serum substitutes include, for example, albumin substitutes such as albumin, lipid-rich albumin and recombinant albumin, plant starch, dextran, protein hydrolysates, transferrin or other iron transporters, fatty acids, insulin, collagen precursors, trace amounts An element, 2-mercaptoethanol, 3′-thioglycerol, or an equivalent thereof may be used. Specific examples of serum substitutes include, for example, those prepared by the method described in WO 98/30679, commercially available knockout Serum Replacement (KSR), Chemically-defined Lipid concentrated (Life Technologies max) and Gluta Life Technologies).

[幹細胞]
本発明が対象とする「幹細胞」は、自己複成能及び分化増殖能を有する未熟な細胞をいい、分化能力に応じて、多能性幹細胞(pluripotent stem cell)、複能性幹細胞(multipotent stem cell)、単能性幹細胞(unipotent stem cell)等が含まれる。「幹細胞」は、一般に、未分化状態を保持したまま増殖できる「自己再生能」と、三胚葉系列すべてに分化できる「分化多能性」とを有する未分化細胞と定義されている。
多能性幹細胞とは、生体を構成する全ての組織や細胞へ分化し得る能力を有する細胞を意味する。
複能性幹細胞とは、全ての種類ではないが、複数種の組織や細胞へ分化し得る能力を有する細胞を意味する。
単能性幹細胞とは、特定の組織や細胞へ分化し得る能力を有する細胞を意味する。
[Stem cells]
The “stem cell” targeted by the present invention refers to an immature cell having self-replication ability and differentiation / proliferation ability, and depending on the differentiation ability, a pluripotent stem cell, a multipotent stem cell (multipotent stem cell). cell), unipotent stem cells, and the like. A “stem cell” is generally defined as an undifferentiated cell having “self-renewal ability” capable of proliferating while maintaining an undifferentiated state and “differentiation pluripotency” capable of differentiating into all three germ layers.
A pluripotent stem cell means a cell having an ability to differentiate into all tissues and cells constituting a living body.
A multipotent stem cell means a cell having the ability to differentiate into multiple types of tissues and cells, although not all types.
A unipotent stem cell means a cell having the ability to differentiate into a specific tissue or cell.

幹細胞の由来種も特に限定されず、例えば、ラット、マウス、ハムスター、モルモット等のげっ歯類、ウサギ等のウサギ目、ブタ、ウシ、ヤギ、ヒツジ等の有蹄目、イヌ、ネコ等のネコ目、ヒト、サル、アカゲザル、マーモセット、オランウータン、チンパンジーなどの霊長類などの細胞であってよい。   The stem cell origin species is not particularly limited, and examples thereof include rodents such as rats, mice, hamsters, and guinea pigs, rabbit eyes such as rabbits, ungulates such as pigs, cows, goats, and sheep, and cats such as dogs and cats. The cells may be eyes, humans, monkeys, rhesus monkeys, marmosets, orangutans, chimpanzees and other primates.

幹細胞の具体例としては、筋芽細胞、血管内皮細胞、骨芽細胞、脂肪細胞、筋細胞、心筋細胞、軟骨細胞等へ分化する間葉系幹細胞、ニューロンやグリア細胞へ分化する神経幹細胞、白血球、赤血球、血小板、肥満細胞、樹状細胞等へ分化する造血幹細胞又は骨髄幹細胞、スフェロイド状態から胚様体(EB体)と呼ばれる擬似的な胚の形成を経て様々な組織への分化・誘導のステップに進むことが知られている胚性幹細胞(Embryonic stem cell:ES細胞)や誘導性多能性幹細胞(induced pluripotent cell:iPS細胞)、始原生殖細胞に由来する胚性生殖(EG)細胞、精巣組織からのGS細胞の樹立培養過程で単離されるmultipotent germline stem(mGS)細胞、骨髄から単離されるmultipotent adult progenitor cell(MAPC)等の多能性幹細胞などが挙げられる。   Specific examples of stem cells include myoblasts, vascular endothelial cells, osteoblasts, adipocytes, myocytes, cardiomyocytes, mesenchymal stem cells that differentiate into chondrocytes, neural stem cells that differentiate into neurons and glial cells, leukocytes Hematopoietic stem cells or bone marrow stem cells that differentiate into red blood cells, platelets, mast cells, dendritic cells, etc., and differentiation / induction into various tissues through formation of pseudo embryos called embryoid bodies (EB bodies) from the spheroid state Embryonic stem cells (ES cells), inducible pluripotent cells (iPS cells), embryonic germ cells (EG) cells derived from primordial germ cells, known to proceed to the steps, Multipotent germline stem (mGS) isolated in the process of establishing GS cells from testis tissue Cells, such as multipotent adult progenitor cell (MAPC) pluripotent stem cells, such as isolated from the bone marrow and the like.

多能性幹細胞としては、特に、上述のES細胞またはiPS細胞を挙げることができる。体細胞の核を核移植することによって作製された初期胚を培養することによって樹立した幹細胞も、多能性幹細胞としてまた好ましい(Nature,1997,385,p.810、Science,1998,280,p.1256、Nature Biotechnology,1999,17,p.456、Nature,1998,394,p.369、Nature Genetics,1999,22,p.127、Proc.Natl.Acad.Sci.USA,1999,96,p.14984、Nature Genetics,2000,24,p.109)。   Examples of pluripotent stem cells include the aforementioned ES cells or iPS cells. Stem cells established by culturing early embryos produced by nuclear transfer of somatic cell nuclei are also preferred as pluripotent stem cells (Nature, 1997, 385, p. 810, Science, 1998, 280, p. 1256, Nature Biotechnology, 1999, 17, p.456, Nature, 1998, 394, p.369, Nature Genetics, 1999, 22, p.127, Proc. Natl. Acad. Sci. USA, 1999, 96, p. 14984, Nature Genetics, 2000, 24, p. 109).

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及び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 are obtained from, for example, WA01 (H1) and WA09 (H9) from the WiCell Research Institute, and KhES-1, KhES-2 and KhES-3 from the Institute of Regenerative Medicine, Kyoto University (Kyoto, Japan). Is possible.

iPS細胞としては、例えば、皮膚細胞等の体細胞に複数の遺伝子(初期化因子)を導入して得られる、ES細胞同様の多分化能を獲得した細胞が挙げられる。例えばOct3/4遺伝子、Klf4遺伝子、C-Myc遺伝子及びSox2遺伝子を導入することによって得られるiPS細胞、Oct3/4遺伝子、Klf4遺伝子及びSox2遺伝子を導入することによって得られるiPS細胞(Nature Biotechnology,2008,26,101-106)等が挙げられる。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、Nat. Biotechnol., 2008,26,p.795−797、Cell Stem Cell, 2008,2,p.525−528、Stem Cells. 2008,26,p.2467−2474、Nat Biotechnol. 2008,26,p.1269−1275、Cell Stem Cell, 2008,3, p.568−574、Cell Stem Cell, 2008,3,p.475−479、Cell Stem Cell, 2008,3, p.132−135、Nat Cell Biol.,2009,11,p.197−203、Nat. Biotech., 2009,27,p.459−461、Proc Natl Acad Sci U S A.,2009,106,p.8912−8917、Nature,2009,461,p.643−649、Cell Stem Cell,2009, 5,p.491−503、Cell Stem Cell,2010,6,p.167−74、Nature,2010,463,p.1096−100、Stem Cells,2010,28,p.713−720、Nature,2011,474,p.225−9に記載の組み合わせが例示される。iPS細胞は、所定の機関(理研バイオリソースセンター、京都大学)より入手可能である。   Examples of iPS cells include cells obtained by introducing a plurality of genes (reprogramming factors) into somatic cells such as skin cells and having acquired pluripotency similar to ES cells. For example, iPS cells obtained by introducing Oct3 / 4 gene, Klf4 gene, C-Myc gene and Sox2 gene, iPS cells obtained by introducing Oct3 / 4 gene, Klf4 gene and Sox2 gene (Nature Biotechnology, 2008) , 26, 101-106). 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, and ECAT15. -2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3, or Glis1, etc. These initialization factors may be used alone or in combination. Combinations of reprogramming factors include WO2007 / 069666, WO2008 / 118820, WO2009 / 007852, WO2009 / 032194, WO2009 / 058413, WO2009 / 057831, WO2009 / 0775119, 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 / 045 626, WO 2010/056831, WO 2010/068955, WO 2010/098419, WO 2010/102267, WO 2010/111409, WO 2010/111422, WO 2010/115050, WO 2010/124290, WO 2010/147395, WO 2010/147612, Nat. Biotechnol. 2008, 26, p. 795-797, Cell Stem Cell, 2008, 2, p. 525-528, Stem Cells. 2008, 26, p. 2467-2474, Nat Biotechnol. 2008, 26, p. 1269-1275, Cell Stem Cell, 2008, 3, p. 568-574, Cell Stem Cell, 2008, 3, p. 475-479, Cell Stem Cell, 2008, 3, p. 132-135, Nat Cell Biol., 2009, 11, p. 197-203, Nat. Biotech., 2009, 27, p. 459-461, Proc Natl Acad Sci USA, 2009, 106, p. 8912-8917, Nature, 2009, 461, p. 643-649, Cell Stem Cell, 2009, 5, p. 491-503, Cell Stem Cell, 2010, 6, p. 167-74, Nature, 2010, 463, p. 1096-100, Stem Cells, 2010, 28, p. 713-720, Nature, 2011, 474, p. The combination of 225-9 is illustrated. iPS cells are available from predetermined institutions (RIKEN BioResource Center, Kyoto University).

複能性幹細胞としては、特に、間葉系幹細胞、造血系幹細胞、神経系幹細胞、骨髄幹細胞及び生殖幹細胞等の体性幹細胞等が挙げられる。複能性幹細胞は、好ましくは間葉系幹細胞、より好ましくは骨髄間葉系幹細胞である。なお、間葉系幹細胞とは、骨芽細胞、軟骨芽細胞及び脂肪芽細胞等の間葉系の細胞全て又はいくつかへの分化が可能な幹細胞又はその前駆細胞の集団を広義に意味する。   Examples of multipotent stem cells include somatic stem cells such as mesenchymal stem cells, hematopoietic stem cells, neural stem cells, bone marrow stem cells, and reproductive stem cells. The multipotent stem cell is preferably a mesenchymal stem cell, more preferably a bone marrow mesenchymal stem cell. Note that the mesenchymal stem cell broadly means a group of stem cells or progenitor cells that can differentiate into all or some mesenchymal cells such as osteoblasts, chondroblasts, and lipoblasts.

本発明に係る培地は、いずれの幹細胞の培養にも好適に使用することができるが、好ましくは間葉系幹細胞、ES細胞又はiPS細胞の培養に、より好ましくはiPS細胞の培養に、特に好ましくはヒトiPS細胞の培養に使用することができる。ヒトiPS細胞としてより具体的には253G1株(理研セルバンクNo. HPS0002)、201B7株(理研セルバンクNo. HPS0063)、409B2株(理研セルバンクNo. HPS0076)、454E2株(理研セルバンクNo. HPS0077)、HiPS−RIKEN−1A株(理研セルバンクNo. HPS0003)、HiPS−RIKEN−2A株(理研セルバンク No. HPS0009)、HiPS−RIKEN−12A株(理研セルバンク No. HPS0029)、Nips−B2株(理研セルバンクNo. HPS0223)などを挙げることができる。   The medium according to the present invention can be suitably used for culturing any stem cell, but is preferably used for culturing mesenchymal stem cells, ES cells or iPS cells, more preferably for culturing iPS cells. Can be used to culture human iPS cells. More specifically, as human iPS cells, 253G1 strain (RIKEN Cell Bank No. HPS0002), 201B7 strain (RIKEN Cell Bank No. HPS0063), 409B2 strain (RIKEN Cell Bank No. HPS0076), 454E2 strain (RIKEN Cell Bank No. HPS0077), HiPS -RIKEN-1A strain (RIKEN Cell Bank No. HPS0003), HiPS-RIKEN-2A strain (RIKEN Cell Bank No. HPS0009), HiPS-RIKEN-12A strain (RIKEN Cell Bank No. HPS0029), and Nippon-B2 strain (RIKEN Cell Bank No. HPS0223).

[幹細胞生存抑制剤等]
上述の通り、アトルバスタチンは、未分化な幹細胞に特異的な生存抑制活性を有し、分化細胞(特に心筋細胞)の細胞死を誘発することなく、未分化幹細胞の生存を抑制する。このため、アトルバスタチンは、in vitroでの細胞培養において幹細胞生存抑制剤として利用でき、さらにin vivoにおいても幹細胞生存抑制剤として、幹細胞由来の分化細胞を含む細胞医薬組成物の生体内での腫瘍化を抑制するため利用できる可能性がある。
[Stem cell survival inhibitor, etc.]
As described above, atorvastatin has a survival inhibitory activity specific to undifferentiated stem cells, and suppresses the survival of undifferentiated stem cells without inducing cell death of differentiated cells (particularly cardiomyocytes). For this reason, atorvastatin can be used as a stem cell survival inhibitor in cell culture in vitro, and also in vivo as a stem cell survival inhibitor in vivo, a cell pharmaceutical composition containing stem cell-derived differentiated cells. May be used to suppress

2.分化細胞の製造方法
本発明に係る分化細胞の製造方法は、分化誘導後の細胞をアトルバスタチンにより処理する工程を含むことを特徴とする。より具体的には、本発明に係る分化細胞の製造方法は、上述の本発明に係る分化細胞用培地中で分化誘導後の細胞を培養する手順を含むことを特徴とする。ただし、本発明に係る分化細胞の製造方法は、分化誘導前の幹細胞がアトルバスタチンにより処理されることを排除する趣旨ではない。すなわち、本発明に係る分化細胞の製造方法では、少なくとも分化誘導後の細胞(分化細胞に加えて未分化状態を維持した幹細胞を含み得る)がアトルバスタチンにより処理されるものであり、加えて分化誘導前の幹細胞もがアトルバスタチンにより処理されてもよい。
2. Method for Producing Differentiated Cell The method for producing a differentiated cell according to the present invention includes a step of treating the cell after differentiation induction with atorvastatin. More specifically, the method for producing differentiated cells according to the present invention is characterized by including a procedure for culturing cells after differentiation induction in the above-described medium for differentiated cells according to the present invention. However, the method for producing differentiated cells according to the present invention is not intended to exclude that stem cells before differentiation induction are treated with atorvastatin. That is, in the method for producing differentiated cells according to the present invention, at least cells after differentiation induction (which may include stem cells that have maintained an undifferentiated state in addition to differentiated cells) are treated with atorvastatin, in addition to differentiation induction. Previous stem cells may also be treated with atorvastatin.

アトルバスタチンは、未分化な幹細胞に特異的な生存抑制活性を有し、分化細胞の細胞死を誘発することなく、未分化幹細胞の生存を抑制する。このため、本発明に係る分化細胞の製造方法によれば、未分化幹細胞の混入がない(あるいは極めて少ない)分化細胞を得ることができる。   Atorvastatin has a survival inhibitory activity specific to undifferentiated stem cells, and suppresses the survival of undifferentiated stem cells without inducing cell death of differentiated cells. For this reason, according to the method for producing a differentiated cell according to the present invention, it is possible to obtain a differentiated cell free from (or very little) of undifferentiated stem cells.

また、本発明に係る分化細胞の製造方法と同様のアトルバスタチン処理工程に、幹細胞及び分化細胞を含む細胞混合物を供すれば、分化細胞の細胞死を誘発することなく、未分化幹細胞の生存を抑制することができるので、細胞混合物から未分化幹細胞の混入がない(あるいは極めて少ない)分化細胞のみを分離することもできる(分化細胞の分離方法)。   In addition, if a cell mixture containing stem cells and differentiated cells is used in the atorvastatin treatment step similar to the method for producing differentiated cells according to the present invention, the survival of undifferentiated stem cells is suppressed without inducing cell death of the differentiated cells. Therefore, it is possible to separate only differentiated cells that are free from (or very little) undifferentiated stem cells from the cell mixture (method for separating differentiated cells).

本発明に係る分化細胞の製造方法は、特に幹細胞から心筋細胞を製造するために好適に用いられる。実施例において後述するように、フルバスタチン等のスタチンでは、幹細胞から分化誘導した心筋細胞に対して細胞死誘導活性が発現する場合がある。このため、本発明に係る分化細胞の製造方法は、幹細胞から心筋細胞を製造する場合に特に好適となる。   The method for producing differentiated cells according to the present invention is suitably used particularly for producing cardiomyocytes from stem cells. As will be described later in Examples, statins such as fluvastatin may exhibit cell death-inducing activity against cardiomyocytes differentiated from stem cells. For this reason, the method for producing differentiated cells according to the present invention is particularly suitable for producing cardiomyocytes from stem cells.

本発明に係る分化細胞の製造方法は、上記のアトルバスタチンによる処理工程に加えて、幹細胞を培養する工程と、幹細胞を分化誘導する工程と、をさらに含んでいてもよい。これらの工程において、細胞の培養及び幹細胞の分化誘導は、従来公知の手法に従って行えばよく、アトルバスタチンの存在下又は非存在下で行うことができる。   The method for producing differentiated cells according to the present invention may further include a step of culturing stem cells and a step of inducing differentiation of stem cells, in addition to the treatment step with atorvastatin. In these steps, cell culture and stem cell differentiation induction may be performed according to a conventionally known technique, and can be performed in the presence or absence of atorvastatin.

細胞培養に用いられる培養器は、特に限定されないが、フラスコ、ディッシュ、シャーレ、マイクロウエルプレート、マイクロスライド、チャンバースライド、チューブ、トレイ、培養バック又はタンクなどの培養槽などが挙げられ得る。これらの培養器の基材も、特に限定されず、ガラスや、ポリプロピレイン及びポリスチレンなどの各種プラスチック、ステンレスなどの金属又はそれらの組み合わせが挙げられる。   The incubator used for cell culture is not particularly limited, and examples thereof include flasks, dishes, petri dishes, microwell plates, microslides, chamber slides, tubes, trays, culture vessels such as culture bags or tanks. The substrate of these incubators is not particularly limited, and examples thereof include glass, various plastics such as polypropylene and polystyrene, metals such as stainless steel, or combinations thereof.

培養器は、細胞接着性であっても細胞非接着性であってもよく、目的に応じて適宜選ばれる。細胞接着性の培養器は、培養器の表面と細胞との接着性を向上させる目的で、細胞外マトリックス(ECM)等の任意の細胞支持用基質でコーティングされたものであり得る。細胞支持用基質は、幹細胞又はフィーダー細胞(用いられる場合)の接着を目的とする任意の物質であり得る。このような細胞支持用基質としては、コラーゲン、ゼラチン、ポリ-L-リジン、ポリ-D-リジン、ラミニン(または、ラミニンの一部構造体)、及びフィブロネクチン並びにそれらの混合物、例えばマトリゲル、並びに溶解細胞膜調製物が挙げられる(Lancet,2005,365,9471,p.1636-1641参照)。   The incubator may be cell-adhesive or non-cell-adhesive, and is appropriately selected according to the purpose. The cell-adhesive incubator can be coated with any cell-supporting substrate such as an extracellular matrix (ECM) for the purpose of improving the adhesion between the surface of the incubator and the cells. The cell support substrate can be any substance intended to adhere to stem cells or feeder cells (if used). Such cell support substrates include collagen, gelatin, poly-L-lysine, poly-D-lysine, laminin (or a partial structure of laminin), and fibronectin and mixtures thereof such as matrigel and lysis Cell membrane preparations are mentioned (see Lancet, 2005, 365, 9471, p. 1636-1641).

培養される幹細胞は、分散細胞又は非分散細胞であり得る。分散細胞とは、細胞分散を促進するために処理された細胞をいう。分散細胞としては、数個(典型的に2〜50、2〜20、又は2〜10個)の細胞からなる小さな細胞塊を形成している細胞が挙げられる。分散細胞は、浮遊(懸濁)細胞、又は接着細胞であり得る。   The cultured stem cells can be dispersed cells or non-dispersed cells. Dispersed cells refer to cells that have been treated to promote cell dispersion. Examples of the dispersed cells include cells forming a small cell mass composed of several cells (typically 2 to 50, 2 to 20, or 2 to 10 cells). The dispersed cells can be floating (suspension) cells or adherent cells.

細胞の培養密度は、細胞の生存及び増殖を促進する効果を達成し得るような密度である限り特に限定されない。好ましくは1.0×101〜1.0×107細胞/ml、より好ましくは1.0×102〜1.0×107細胞/ml、さらにより好ましくは1.0×103〜1.0×107細胞/ml、最も好ましくは3.0×104〜1.0×107細胞/mlである。 The culture density of the cells is not particularly limited as long as the density can achieve the effect of promoting cell survival and proliferation. Preferably 1.0 × 10 1 to 1.0 × 10 7 cells / ml, more preferably 1.0 × 10 2 to 1.0 × 10 7 cells / ml, even more preferably 1.0 × 10 3 to 1.0 × 10 7 cells / ml, most preferably 3.0 × 10 4 to 1.0 × 10 7 cells / ml.

温度、CO2濃度、酸素濃度及びpHなどの培養条件は、動物組織に由来する細胞の培養に従来用いられている技術に基づいて適宜設定できる。例えば、培養温度は、特に限定されるものではないが30〜40℃、好ましくは37℃であり得る。CO2濃度は、1〜10%、好ましくは2〜5%であり得る。酸素濃度は、10〜25%であり得る。 Culture conditions such as temperature, CO 2 concentration, oxygen concentration and pH can be appropriately set based on techniques conventionally used for culturing cells derived from animal tissues. For example, the culture temperature is not particularly limited, but may be 30 to 40 ° C, preferably 37 ° C. CO 2 concentration is 1-10%, preferably be a 2-5%. The oxygen concentration can be 10-25%.

幹細胞の接着培養を行う場合、フィーダー細胞の存在下で培養してもよい。フィーダー細胞には、胎児線維芽細胞等のストローマ細胞を用いることができる(例えば、Manipulating the Mouse Embryo A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, 1994、Gene Targeting, A Practical Approach, IRL Press at Oxford University Press, 1993;Proc. Natl. Acad. Sci. USA,1981,78,12,p.7634−7638、Nature,1981,292,5819,p.154−156、J. Virol.,1969,4,5,p.549−553、Science,1996,272,5262,p.722−724、J. Cell. Physiol.,1982,112,1,p.89−95、国際公開第01/088100号、同第2005/080554号参照)。   When performing adherent culture of stem cells, the cells may be cultured in the presence of feeder cells. As the feeder cells, stromal cells such as fetal fibroblasts can be used (for example, Manipulating the Mouse Embryo A Laboratory Manual, Second Edition, Cold Spring Harbor Pt, 1994, Geskin A Laboratory Press, 1994). Oxford University Press, 1993; Proc. Natl. Acad. Sci. USA, 1981, 78, 12, p.7634-7638, Nature, 1981, 292, 5819, p.154-156, J. Virol., 1969, 4 , 5, p.549-553, Sc ence, 1996,272,5262, p.722-724, J. Cell. Physiol., 1982,112,1, p.89-95, International Publication No. WO 01/088100, see Nos. No. 2005/080554).

幹細胞の浮遊培養の態様としては、担体上での浮遊培養(J. Biotechnology,2007,132,2,p.227−236)又はメチルセルロースなどの高分子ポリマーを用いた浮遊培養(Stem Cell Reports,2014,2,5,p.734−745)などが挙げられる。幹細胞の浮遊培養との用語は、培地中において、培養器又はフィーダー細胞(用いられる場合)に対して非接着性の条件下で幹細胞を培養することをいう。幹細胞の浮遊培養としては、幹細胞の分散培養及び幹細胞の凝集浮遊培養が挙げられる。幹細胞の分散培養との用語は、懸濁された幹細胞を培養することをいい、数個(例、2〜20個)の幹細胞からなる小さな細胞塊の分散培養が挙げられる。分散培養を継続した場合、培養された分散細胞がより大きな幹細胞塊を形成し、その後凝集浮遊培養が実行され得る。このような凝集浮遊培養としては、胚様体培養法(Curr. Opin. Cell Biol.,1995,7,6,p.862-869参照)、SFEB法(Nature Neuroscience,2005,8,3,p.288-296、国際公開第2005/123902号)、メッシュフィルターを用いて機械的処理により細胞株を継代させるスフェア培養法(Stem Cell Reports,2014,2,5,p.734−745)が挙げられる。   Examples of the suspension culture of stem cells include suspension culture on a carrier (J. Biotechnology, 2007, 132, 2, p. 227-236) or suspension culture using a polymer such as methyl cellulose (Stem Cell Reports, 2014). 2, 5, p. 734-745). The term stem cell suspension culture refers to culturing stem cells in a medium under conditions that are non-adherent to the incubator or feeder cells (if used). Stem cell suspension culture includes stem cell dispersion culture and stem cell aggregation suspension culture. The term stem cell dispersion culture refers to culturing suspended stem cells, and examples include dispersion culture of small cell clusters composed of several (eg, 2 to 20) stem cells. When the dispersion culture is continued, the cultured dispersion cells form a larger stem cell mass, and then aggregated suspension culture can be performed. Examples of such agglutination suspension culture include embryoid body culture method (see Curr. Opin. Cell Biol., 1995, 7, 6, p. 862-869), SFEB method (Nature Neuroscience, 2005, 8, 3, p. 288-296, International Publication No. 2005/123902), a sphere culture method (Stem Cell Reports, 2014, 2, 5, p. 734-745) in which a cell line is passaged by mechanical treatment using a mesh filter. Can be mentioned.

幹細胞の分化誘導は、例えば心筋細胞の分化誘導プロセスでは、培地(例えば、STEMdiff APEL Medium、STEMCELL社)に0.5ng/ml BMP-4を添加し、1日後、培地を10ng/ml BMP-4、10ng/ml Activin A、5ng/ml bFGFを添加した物に交換し、4日目後、培地を10ng/ml VEGF、150ng/ml Dkk1を添加した物に交換し、8日目後、培地を10ng/ml VEGF、150ng/ml Dkk1、10ng/ml bFGFを添加した物に交換することで自律的な拍動を伴う心筋細胞を確認できる。   For stem cell differentiation induction, for example, in the cardiomyocyte differentiation induction process, 0.5 ng / ml BMP-4 is added to a medium (for example, STEMdiff APEL Medium, STEMCELL), and one day later, the medium is 10 ng / ml BMP-4. The medium was replaced with 10 ng / ml Activin A, 5 ng / ml bFGF, and after 4 days, the medium was replaced with 10 ng / ml VEGF, 150 ng / ml Dkk1, and after 8 days, the medium was replaced. Cardiomyocytes with autonomous pulsation can be confirmed by exchanging them with those added with 10 ng / ml VEGF, 150 ng / ml Dkk1, 10 ng / ml bFGF.

また、例えば、軟骨細胞の分化誘導プロセスでは、培地(90%αMEM培地、10%牛胎児血清(FBS)、2mM L-グルタミン、0.1μMのデキサメタゾン)中で、間葉系幹細胞を培養することによって行うことができる。さらに、例えばレチノイン酸などの分化誘導剤を培地に添加することにより、幹細胞を神経系細胞などに分化させることが可能となる。分化誘導剤には、BMP阻害剤、Wnt阻害剤、Nodal阻害剤、レチノイン酸なども用いることができる。血小板分化誘導プロセスの過程で必要となる巨核球の形成には、血清含有培地(20%FCS)において誘導された胚葉体を経て、トロンボポエチン(TPO)、インターロイキン3(IL3)、インターロイキン6(IL6)及び幹細胞因子(SCF)等の因子が使用され得る。   Further, for example, in the chondrocyte differentiation induction process, mesenchymal stem cells are cultured in a medium (90% αMEM medium, 10% fetal bovine serum (FBS), 2 mM L-glutamine, 0.1 μM dexamethasone). Can be done by. Furthermore, for example, by adding a differentiation inducer such as retinoic acid to the medium, it becomes possible to differentiate the stem cells into nervous system cells. BMP inhibitors, Wnt inhibitors, Nodal inhibitors, retinoic acid and the like can also be used as differentiation inducers. For the formation of megakaryocytes required in the process of inducing platelet differentiation, thrombopoietin (TPO), interleukin 3 (IL3), interleukin 6 (through the embryoid body induced in a serum-containing medium (20% FCS) Factors such as IL6) and stem cell factor (SCF) may be used.

3.細胞医薬組成物の製造方法
本発明に係る、幹細胞由来の分化細胞を含む細胞医薬組成物の製造方法は、幹細胞を分化誘導する工程と、分化誘導後の細胞をアトルバスタチンにより処理する工程と、を含むことを特徴とする。この製造方法は、さらに、幹細胞を培養する工程を含むことができる。
3. Method for Producing Cell Pharmaceutical Composition According to the present invention, a method for producing a cell pharmaceutical composition comprising a stem cell-derived differentiated cell comprises a step of inducing differentiation of a stem cell, and a step of treating the cell after differentiation induction with atorvastatin. It is characterized by including. This production method can further include a step of culturing stem cells.

分化誘導工程及びアトルバスタチンによる処理工程は、上記の分化細胞の製造方法と同様の手順によって行うことができる。本発明に係る細胞医薬組成物の製造方法においても、分化誘導前の幹細胞がアトルバスタチンにより処理されることは排除されないものとする。   The differentiation induction step and the treatment step with atorvastatin can be performed by the same procedure as the above-described method for producing differentiated cells. In the method for producing a cell pharmaceutical composition according to the present invention, it is not excluded that stem cells before differentiation induction are treated with atorvastatin.

アトルバスタチンは、未分化な幹細胞に特異的な生存抑制活性を有し、分化細胞の細胞死を誘発することなく、未分化幹細胞の生存を抑制する。このため、本発明に係る細胞医薬組成物の製造方法によれば、分化細胞を含み、未分化幹細胞の混入がない(あるいは極めて少ない)細胞医薬組成物を得ることができる。   Atorvastatin has a survival inhibitory activity specific to undifferentiated stem cells, and suppresses the survival of undifferentiated stem cells without inducing cell death of differentiated cells. Therefore, according to the method for producing a cell pharmaceutical composition according to the present invention, a cell pharmaceutical composition containing differentiated cells and free from (or very little) undifferentiated stem cells can be obtained.

細胞医薬組成物は、分散された分化細胞、所定形状の細胞塊を形成した分化細胞集団、あるいは組織構造や小器官を形成した分化細胞集団であり得る。細胞医薬組成物は、再生医療用の細胞ソースのために利用され得る。本発明に係る製造方法により得られる細胞医薬組成物は、未分化幹細胞の混入がない(あるいは極めて少ない)ため、移植後に再生内で腫瘍化又は癌化するおそれがなく、優れた安全性が期待できる。例えば、本発明に係る細胞医薬組成物により得られる心筋細胞を含む細胞医薬組成物は、シート状に形成されてて、心疾患の治療のため好適に心臓へ移植され得るものである。   The cell pharmaceutical composition may be a dispersed differentiated cell, a differentiated cell population forming a cell cluster of a predetermined shape, or a differentiated cell population forming a tissue structure or organelle. The cell pharmaceutical composition can be utilized for cell sources for regenerative medicine. The cell pharmaceutical composition obtained by the production method according to the present invention has no (or very little) contamination of undifferentiated stem cells, so there is no risk of tumor formation or cancer within regeneration after transplantation, and excellent safety is expected. it can. For example, a cell pharmaceutical composition containing cardiomyocytes obtained by the cell pharmaceutical composition according to the present invention is formed into a sheet and can be suitably transplanted to the heart for the treatment of heart disease.

[実施例1:ヒトiPS細胞維持培養におけるアトルバスタチンによる細胞生存抑制]
培養ヒトiPS細胞に対するアトルバスタチンの生存抑制活性を評価した。
[Example 1: Inhibition of cell survival by atorvastatin in human iPS cell maintenance culture]
The inhibitory activity of atorvastatin on cultured human iPS cells was evaluated.

(方法)
ヒトiPS細胞は、京都大学iPS細胞研究所山中伸弥教授が樹立したヒト人工多能性幹細胞(201B7)を、理化学研究所セルバンク(No.HPS0063)より入手し使用した。ヒト多能性幹細胞培養の実践プロトコール(第2版)(理化学研究所 発生・再生科学総合研究センター 幹細胞研究支援・開発室作成 http://www.cdb.riken.jp/hsct/protocol.html)に従い、細胞のフィーダー層としてマウス胎児線維芽細胞(マイトマイシン処理で不活化、MEF)を蒔いたプラスチック培養皿の上で未分化ヒトiPS細胞を培養した。
(Method)
For human iPS cells, human induced pluripotent stem cells (201B7) established by Professor Shinya Yamanaka of Kyoto University iPS Cell Laboratory were obtained from RIKEN Cell Bank (No. HPS0063) and used. Practical protocol for human pluripotent stem cell culture (2nd edition) (Created by RIKEN Center for Developmental and Regenerative Sciences, Stem Cell Research Support and Development Office: http://www.cdb.riken.jp/hsct/protocol.html) Then, undifferentiated human iPS cells were cultured on plastic culture dishes in which mouse fetal fibroblasts (inactivated by mitomycin treatment, MEF) were seeded as a feeder layer of the cells.

培養液(維持培地)には、D-MEMF12(Sigma D6421)に最終濃度20% KSR(Life Technologies)、最終濃度1% NON-ESSENTIAL AMINO ACID(×100)(非必須アミノ酸;SIGMA D7145)、2mM L-グルタミン酸及び、80μM 2-メルカプトエタノールを添加したものを用いた。培養は、37℃、5% CO2下で行った。3〜4日毎に継代を行った。解離液(リン酸バッファー緩衝生理学的食塩水に0.25%トリプシン、1mg/mlコラゲナーゼIV液、1mM CaCl2を添加したもの;全てLife Technologies)を用いて、iPS細胞をフィーダー層から解離し、ピペッティングで小細胞塊(細胞数が約50-100個程度の細胞集団)に分散した後、前日にMEFを播種し形成させたフィーダー層の上に蒔いた。 In the culture medium (maintenance medium), D-MEMF12 (Sigma D6421), final concentration 20% KSR (Life Technologies), final concentration 1% NON-ESSENTIAL AMINO ACID (× 100) (non-essential amino acid; SIGMA D7145), 2 mM What added L-glutamic acid and 80 micromol 2-mercaptoethanol was used. The culture was performed at 37 ° C. and 5% CO 2 . Passaging was performed every 3-4 days. Dissociate the iPS cells from the feeder layer using a dissociation solution (phosphate buffer buffered physiological saline supplemented with 0.25% trypsin, 1 mg / ml collagenase IV solution, 1 mM CaCl 2 ; all Life Technologies) After dispersing into small cell masses (a cell population of about 50-100 cells) by pipetting, they were seeded on the feeder layer formed by seeding MEF the day before.

上記のように培養したヒトiPS細胞を、フィーダー細胞から小細胞塊として解離し、さらに混入するフィーダー細胞を除去するために細胞接着性の培養プレート(0.1% ゼラチンコート)の底に吸着させ、培養液(アッセイ培地)で37℃、1時間培養した(iPS細胞塊はプレートに吸着しないが、混入するフィーダー細胞は強く吸着する)。iPS細胞塊をピペッティング操作により小細胞塊へ細かく砕いた後、24ウェル培養プレートを用いて、Growth Factor Reduced BD Matrigel (BD)上に、1×105個/0.65cm2/培地液量1.0mlで播種した。スタチンを含むアッセイ培地で数日間培養後に、形成されたアルカリフォスファターゼ染色陽性(ALP+)のコロニー数を未分化細胞として計測し、スタチンを含まないコントロール群と比較した。アッセイ培地はEssential8(サーモフィシャーサイエンティフィック、A1517001)を用いた。 The human iPS cells cultured as described above are dissociated from the feeder cells as small cell clusters and adsorbed to the bottom of a cell-adhesive culture plate (0.1% gelatin coat) in order to remove the contaminating feeder cells. The cells were cultured in a culture solution (assay medium) at 37 ° C. for 1 hour (iPS cell mass was not adsorbed on the plate, but contaminated feeder cells were strongly adsorbed). After crushing the iPS cell mass into small cell masses by pipetting, 1 × 10 5 cells / 0.65 cm 2 / medium volume on a Growth Factor Reduced BD Matrigel (BD) using a 24-well culture plate Seeded at 1.0 ml. After culturing for several days in an assay medium containing statins, the number of formed alkaline phosphatase staining positive (ALP +) colonies was counted as undifferentiated cells and compared with a control group containing no statins. The assay medium used was Essential 8 (Thermo Fisher Scientific, A151701).

(結果1)
スタチン類を20.0μM含むアッセイ培地で48時間培養後、コロニー(ALP+)数を測定した。アトルバスタチン添加群は、コントロール群に比べて、コロニー数は有意に低下した。他のスタチン類(フルバスタチン、ロバスタチン、メバスタチン、シンバスタチン)については、未分化幹細胞に対する明らかな生存抑制活性は認められなかった(図1)。
(Result 1)
After culturing in an assay medium containing 20.0 μM statins for 48 hours, the number of colonies (ALP +) was measured. The number of colonies in the atorvastatin added group was significantly lower than that in the control group. For other statins (fluvastatin, lovastatin, mevastatin, simvastatin), no obvious survival inhibitory activity against undifferentiated stem cells was observed (FIG. 1).

(結果2)
アトルバスタチン10.0μM含むアッセイ培地で24時間培養後、コロニー(ALP+)数を測定した。アトルバスタチン添加群では、コロニー数はコントロール群に比べ60%以下と有意に低下し、未分化幹細胞の生存抑制が認められた(図2)。
(Result 2)
After culturing in an assay medium containing 10.0 μM atorvastatin for 24 hours, the number of colonies (ALP +) was measured. In the atorvastatin-added group, the number of colonies was significantly reduced to 60% or less compared to the control group, and suppression of survival of undifferentiated stem cells was observed (FIG. 2).

(結果3)
アトルバスタチン1μMもしくは10μMを含むアッセイ培地で96時間培養後、コロニー(ALP+)数を測定した。アトルバスタチン添加群では、コントロール群に比べ、コロニー数は有意に低下し、未分化幹細胞の生存抑制が認められた(図3)。
(Result 3)
After culturing in an assay medium containing 1 μM or 10 μM atorvastatin for 96 hours, the number of colonies (ALP +) was measured. In the atorvastatin addition group, the number of colonies was significantly reduced as compared to the control group, and the survival of undifferentiated stem cells was suppressed (FIG. 3).

[実施例2:分化心筋細胞におけるアトルバスタチンによる細胞生存抑制]
iPS細胞から分化させた分化心筋細胞に対するアトルバスタチンの生存抑制活性(細胞死誘導活性)を評価した。
[Example 2: Inhibition of cell survival by atorvastatin in differentiated cardiomyocytes]
The survival inhibitory activity (cell death inducing activity) of atorvastatin against differentiated cardiomyocytes differentiated from iPS cells was evaluated.

(方法)
ヒトiPS細胞をフィーダー細胞から小細胞塊として解離し、さらに混入するフィーダー細胞を除去するために細胞接着性の培養プレート(0.1% ゼラチンコート)の底に吸着させ、アッセイ培地で37℃、1時間培養した(iPS細胞塊はプレートに吸着しないが、混入するフィーダー細胞は強く吸着する)。iPS細胞塊をピペッティング操作により小細胞塊へ細かく砕いた後、48ウェル培養プレートを用いて、Growth Factor Reduced BD Matrigel(BD)上に、高密度(1×106個/0.65cm2/培地容量0.5ml)で播種した。
(Method)
Human iPS cells were dissociated from feeder cells as small cell clumps and adsorbed to the bottom of cell-adhesive culture plates (0.1% gelatin coated) to remove further contaminated feeder cells, and 37 ° C. in assay medium. The cells were cultured for 1 hour (iPS cell mass was not adsorbed on the plate, but contaminated feeder cells were strongly adsorbed). After crushing the iPS cell mass into small cell masses by pipetting, using a 48-well culture plate, the density is increased (1 × 10 6 cells / 0.65 cm 2 /0.6) on the Growth Factor Reduced BD Matrigel (BD). Seeded in a medium volume of 0.5 ml).

心筋への分化誘導は、PSdif−Cardio Cardiomyocyte Differentiation Kit(Stem RD)を用い、キット付属のプロトコールに従って行った。分化誘導培地(PSdif−Cardio(登録商標)A、及びB、及びC)中で6日間培養後に、心筋培養培地(CardioGro(登録商標))に培地交換した。分化誘導された心筋細胞の拍動を顕微鏡下に確認し、コントロール群、及び実験群(アトルバスタチン10.0μM又はフルバスタチン10.0μMを心筋培養培地へ添加)とし、24時間後にLive/Dead Cell Staining Kit II(PromoKine)を用いて死細胞の割合を、蛍光顕微鏡を用いて測定した。   Differentiation into the myocardium was performed using PSdif-Cardio Cardiomyocyte Differentiation Kit (Stem RD) according to the protocol attached to the kit. After culturing in a differentiation induction medium (PSdif-Cardio (registered trademark) A, B, and C) for 6 days, the medium was changed to a myocardial culture medium (CardioGro (registered trademark)). The pulsation of the cardiomyocytes induced to differentiate was confirmed under a microscope, and were set as a control group and an experimental group (adding atorvastatin 10.0 μM or fluvastatin 10.0 μM to the myocardial culture medium), and after 24 hours, Live / Dead Cell Staining The ratio of dead cells was measured using a fluorescence microscope using Kit II (PromoKine).

(結果)
iPS細胞から分化誘導した心筋細胞に対して、フルバスタチンは、有意な細胞死誘導活性を示した(図4)。一方、アトルバスタチンでは死細胞の割合はコントロール群と同程度であり、分化細胞の生存抑制及び細胞死の誘導は認められなかった。
(result)
Fluvastatin showed a significant cell death-inducing activity against cardiomyocytes induced to differentiate from iPS cells (FIG. 4). On the other hand, in atorvastatin, the proportion of dead cells was the same as that in the control group, and differentiation suppression of differentiated cells and induction of cell death were not observed.

以上の結果より、アトルバスタチンは、分化細胞の細胞死を誘発することなく、未分化幹細胞の生存のみを著しく抑制することが明らかとなった。これらの結果は、アトルバスタチンが、未分化幹細胞に特異的な生存抑制活性を有することを示す。   From the above results, it was revealed that atorvastatin significantly suppresses only the survival of undifferentiated stem cells without inducing cell death of differentiated cells. These results indicate that atorvastatin has a survival inhibitory activity specific to undifferentiated stem cells.

Claims (28)

アトルバスタチンを含む、幹細胞由来の分化細胞用培地。   A medium for differentiated cells derived from stem cells, containing atorvastatin. 前記幹細胞が誘導性多能性幹細胞である請求項1記載の培地。   The medium according to claim 1, wherein the stem cells are inducible pluripotent stem cells. 前記分化細胞が心筋細胞である請求項1又は2に記載の培地。   The medium according to claim 1 or 2, wherein the differentiated cells are cardiomyocytes. 前記幹細胞がヒト由来である請求項1〜3のいずれか一項に記載の培地。   The medium according to any one of claims 1 to 3, wherein the stem cell is derived from a human. アトルバスタチンを濃度1〜20μMで含む請求項1〜4のいずれか一項に記載の培地。   The medium according to any one of claims 1 to 4, comprising atorvastatin at a concentration of 1 to 20 µM. 前記培地が無血清培地である、請求項1〜5のいずれか一項に記載の培地。   The medium according to any one of claims 1 to 5, wherein the medium is a serum-free medium. アトルバスタチンを有効成分とする幹細胞生存抑制剤。   A stem cell survival inhibitor containing atorvastatin as an active ingredient. 前記幹細胞が誘導性多能性幹細胞である請求項7記載の幹細胞生存抑制剤。   The stem cell survival inhibitor according to claim 7, wherein the stem cells are inducible pluripotent stem cells. 前記幹細胞がヒト由来である請求項7又は8に記載の幹細胞生存抑制剤。   The stem cell survival inhibitor according to claim 7 or 8, wherein the stem cell is derived from a human. 幹細胞由来の分化細胞を含む細胞医薬組成物の生体内での腫瘍化を抑制するため医薬組成物であり、請求項7〜9のいずれか一項に記載の幹細胞生存抑制剤を含む医薬組成物。   A pharmaceutical composition containing a stem cell survival inhibitor according to any one of claims 7 to 9, which is a pharmaceutical composition for suppressing tumor formation in vivo of a cellular pharmaceutical composition comprising differentiated cells derived from stem cells. . 幹細胞から分化細胞を製造する方法であって、分化誘導後の細胞をアトルバスタチンにより処理する工程を含む方法。   A method for producing a differentiated cell from a stem cell, comprising a step of treating the cell after differentiation induction with atorvastatin. 前記幹細胞が誘導性多能性幹細胞である請求項11記載の方法。   The method according to claim 11, wherein the stem cell is an inducible pluripotent stem cell. 前記分化細胞が心筋細胞である請求項11又は12記載の方法   The method according to claim 11 or 12, wherein the differentiated cells are cardiomyocytes. 前記幹細胞がヒト由来である請求項11〜13のいずれか一項に記載の方法。   The method according to any one of claims 11 to 13, wherein the stem cell is derived from a human. 幹細胞を培養する工程と、幹細胞を分化誘導する工程と、をさらに含む請求項11〜14のいずれか一項に記載の方法。   The method according to any one of claims 11 to 14, further comprising a step of culturing the stem cell and a step of inducing differentiation of the stem cell. 幹細胞由来の分化細胞を含む細胞医薬組成物を製造する方法であって、前記幹細胞を分化誘導する工程と、分化誘導後の細胞をアトルバスタチンにより処理する工程と、を含む方法。   A method for producing a cell pharmaceutical composition comprising a stem cell-derived differentiated cell, the method comprising a step of inducing differentiation of the stem cell, and a step of treating the cell after differentiation induction with atorvastatin. 前記幹細胞が誘導性多能性幹細胞である請求項16記載の方法。   The method according to claim 16, wherein the stem cell is an inducible pluripotent stem cell. 前記分化細胞が心筋細胞である請求項16又は17記載の方法   The method according to claim 16 or 17, wherein the differentiated cells are cardiomyocytes. 前記幹細胞がヒト由来である請求項16〜18のいずれか一項に記載の方法。   The method according to any one of claims 16 to 18, wherein the stem cell is derived from a human. 幹細胞を培養する工程、をさらに含む請求項16〜19のいずれか一項に記載の方法。   The method according to any one of claims 16 to 19, further comprising culturing stem cells. 幹細胞及び分化細胞を含む細胞混合物から分化細胞のみを分離する方法であって、前記細胞混合物をアトルバスタチンにより処理する手順を含む方法。   A method for separating only differentiated cells from a cell mixture containing stem cells and differentiated cells, comprising a step of treating the cell mixture with atorvastatin. 前記幹細胞が誘導性多能性幹細胞である請求項21記載の方法。   The method according to claim 21, wherein the stem cells are inducible pluripotent stem cells. 前記分化細胞が心筋細胞である請求項21又は22記載の方法   The method according to claim 21 or 22, wherein the differentiated cells are cardiomyocytes. 前記幹細胞及び前記分化細胞がヒト由来である請求項21〜23のいずれか一項に記載の方法。   The method according to any one of claims 21 to 23, wherein the stem cells and the differentiated cells are derived from a human. 幹細胞を培養する工程と、幹細胞を分化誘導する工程と、をさらに含む請求項21〜24のいずれか一項に記載の方法。   The method according to any one of claims 21 to 24, further comprising a step of culturing the stem cell and a step of inducing differentiation of the stem cell. 請求項1記載の培地、請求項7記載の幹細胞生存抑制剤、請求項10記載の医薬組成物の製造のためのアトルバスタチンの使用。   Use of atorvastatin for the production of the medium according to claim 1, the stem cell survival inhibitor according to claim 7, and the pharmaceutical composition according to claim 10. 幹細胞由来の分化細胞を含む細胞医薬組成物の製造のためのアトルバスタチンの使用。   Use of atorvastatin for the production of a cell pharmaceutical composition comprising differentiated cells derived from stem cells. 幹細胞由来の分化細胞を含む細胞医薬組成物の生体内での腫瘍化を抑制するためのアトルバスタチンの使用。   Use of atorvastatin for suppressing in vivo tumorigenesis of a cell pharmaceutical composition containing differentiated cells derived from stem cells.
JP2015220829A 2014-11-11 2015-11-11 Stem cell-derived medium for differentiated cells, method for producing differentiated cells from stem cells, and method for producing cell pharmaceutical composition containing the differentiated cells Active JP6744084B2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2014228786 2014-11-11
JP2014228786 2014-11-11

Publications (2)

Publication Number Publication Date
JP2016093178A true JP2016093178A (en) 2016-05-26
JP6744084B2 JP6744084B2 (en) 2020-08-19

Family

ID=56069643

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2015220829A Active JP6744084B2 (en) 2014-11-11 2015-11-11 Stem cell-derived medium for differentiated cells, method for producing differentiated cells from stem cells, and method for producing cell pharmaceutical composition containing the differentiated cells

Country Status (1)

Country Link
JP (1) JP6744084B2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018074457A1 (en) * 2016-10-17 2018-04-26 学校法人慶應義塾 Undifferentiated stem cell-removing agent, and method for removing undifferentiated stem cells
WO2018117127A1 (en) * 2016-12-20 2018-06-28 大日本住友製薬株式会社 Agent for removing undifferentiated ips cells
WO2018225705A1 (en) 2017-06-05 2018-12-13 テルモ株式会社 Method for producing cell culture
WO2020067438A1 (en) 2018-09-27 2020-04-02 国立大学法人大阪大学 Sheeting method for pluripotent stem cell-derived cells
CN112481202A (en) * 2020-11-30 2021-03-12 深圳博雅感知药业有限公司 Method for serum-free separation culture of umbilical cord mesenchymal stem cells by using platelet lysate
CN114891729A (en) * 2022-04-15 2022-08-12 北京全式金生物技术股份有限公司 Application of folic acid in promoting differentiation of human pluripotent stem cells to cardiac muscle cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070190651A1 (en) * 2006-02-10 2007-08-16 Board Of Regents Of The University Of Texas System Myogenic Development and Protection of Stem Cells Against Inflammation and Apoptosis By Statins and Isoprenoid Pathway Inhibitors
WO2013100080A1 (en) * 2011-12-27 2013-07-04 国立大学法人大阪大学 Method for inducing differentiation enabling tumorigenesis of ips cells to be suppressed

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070190651A1 (en) * 2006-02-10 2007-08-16 Board Of Regents Of The University Of Texas System Myogenic Development and Protection of Stem Cells Against Inflammation and Apoptosis By Statins and Isoprenoid Pathway Inhibitors
WO2013100080A1 (en) * 2011-12-27 2013-07-04 国立大学法人大阪大学 Method for inducing differentiation enabling tumorigenesis of ips cells to be suppressed

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LEE MI-HEE ET AL.: "Simvastatin suppresses self-renewal of mouse embryonic stem cells by inhibiting RhoA geranylgeranyla", STEM CELLS, vol. 25 (2007), JPN6019043205, pages 1654 - 1663, ISSN: 0004270028 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018074457A1 (en) * 2016-10-17 2018-04-26 学校法人慶應義塾 Undifferentiated stem cell-removing agent, and method for removing undifferentiated stem cells
JPWO2018074457A1 (en) * 2016-10-17 2019-07-18 学校法人慶應義塾 An undifferentiated stem cell removing agent and an undifferentiated stem cell removing method
WO2018117127A1 (en) * 2016-12-20 2018-06-28 大日本住友製薬株式会社 Agent for removing undifferentiated ips cells
JPWO2018117127A1 (en) * 2016-12-20 2019-10-24 大日本住友製薬株式会社 Remover of undifferentiated iPS cells
JP7123809B2 (en) 2016-12-20 2022-08-23 住友ファーマ株式会社 Removal agent for undifferentiated iPS cells
US11434472B2 (en) 2016-12-20 2022-09-06 Sumitomo Pharma Co., Ltd. Agent for removing undifferentiated iPS cells
WO2018225705A1 (en) 2017-06-05 2018-12-13 テルモ株式会社 Method for producing cell culture
WO2020067438A1 (en) 2018-09-27 2020-04-02 国立大学法人大阪大学 Sheeting method for pluripotent stem cell-derived cells
CN112481202A (en) * 2020-11-30 2021-03-12 深圳博雅感知药业有限公司 Method for serum-free separation culture of umbilical cord mesenchymal stem cells by using platelet lysate
CN112481202B (en) * 2020-11-30 2022-11-15 深圳博雅感知药业有限公司 Method for serum-free separation culture of umbilical cord mesenchymal stem cells by using platelet lysate
CN114891729A (en) * 2022-04-15 2022-08-12 北京全式金生物技术股份有限公司 Application of folic acid in promoting differentiation of human pluripotent stem cells to cardiac muscle cells
CN114891729B (en) * 2022-04-15 2024-04-05 北京全式金生物技术股份有限公司 Application of folic acid in promoting differentiation of human pluripotent stem cells into myocardial cells

Also Published As

Publication number Publication date
JP6744084B2 (en) 2020-08-19

Similar Documents

Publication Publication Date Title
JP6744084B2 (en) Stem cell-derived medium for differentiated cells, method for producing differentiated cells from stem cells, and method for producing cell pharmaceutical composition containing the differentiated cells
JP6986016B2 (en) Purification of stem cell-derived retinal pigment epithelium using MACS
US10828335B2 (en) Production of midbrain dopaminergic neurons and methods for the use thereof
US11492594B2 (en) Method for producing engineered heart muscle (EHM)
JP5700301B2 (en) Method for inducing differentiation of neural crest cells from pluripotent stem cells
JP2019115351A (en) Method for producing ciliary body margin-like structure
CA3007107A1 (en) Methods of differentiating retinal cells
US20180251732A1 (en) Novel method for chondrogenic induction
KR20170132281A (en) Neural stem cell proliferation medium containing human serum albumin
JP6981403B2 (en) Medium for neural stem cells that enhances neural differentiation potential
WO2017082296A1 (en) Cartilaginous tissue mass, method for producing same, and culture medium for inducing cartilaginous tissue mass from stem cells
WO2015182140A1 (en) Culture medium and culturing method for anchorage-dependent cells, cell composition including stem cells and/or differentiated cells derived from stem cells, and production method for cell composition
WO2017082294A1 (en) Stem cell culture medium, proliferation promoter, and culturing method, cell composition comprising stem cells, and method for producing same
JP7217542B2 (en) Method for removing contamination such as undifferentiated iPS cells that may have tumorigenicity using a differentiation control compound
WO2023037544A1 (en) Method for producing pluripotent stem cells
WO2017082295A1 (en) Cartilaginous tissue mass, method for producing same, and culture medium for inducing cartilaginous tissue mass from stem cells
WO2016190288A1 (en) Medium for culturing stem cells, growth stimulator, and culture method
WO2024024742A1 (en) In vitro culturing method for inducing ovarian follicles from fetal ovarian cells of primate
Novosadova et al. Fibroblast-like cells as an effective feeder for the cultivation and derivation of new lines of human induced pluripotent stem cells
EP4384600A1 (en) Muscle cells differentiated from pluripotent cells, methods of producing same and use thereof

Legal Events

Date Code Title Description
A711 Notification of change in applicant

Free format text: JAPANESE INTERMEDIATE CODE: A711

Effective date: 20181005

A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20181015

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A821

Effective date: 20181005

RD02 Notification of acceptance of power of attorney

Free format text: JAPANESE INTERMEDIATE CODE: A7422

Effective date: 20190620

RD04 Notification of resignation of power of attorney

Free format text: JAPANESE INTERMEDIATE CODE: A7424

Effective date: 20190703

A711 Notification of change in applicant

Free format text: JAPANESE INTERMEDIATE CODE: A712

Effective date: 20190816

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20191112

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20200110

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20200602

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20200701

TRDD Decision of grant or rejection written
A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20200714

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20200730

R150 Certificate of patent or registration of utility model

Ref document number: 6744084

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250