WO2006115243A1 - Method of producing cancer stem cell - Google Patents

Method of producing cancer stem cell Download PDF

Info

Publication number
WO2006115243A1
WO2006115243A1 PCT/JP2006/308560 JP2006308560W WO2006115243A1 WO 2006115243 A1 WO2006115243 A1 WO 2006115243A1 JP 2006308560 W JP2006308560 W JP 2006308560W WO 2006115243 A1 WO2006115243 A1 WO 2006115243A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
tumor
cancer
cancer stem
Prior art date
Application number
PCT/JP2006/308560
Other languages
French (fr)
Japanese (ja)
Inventor
Nobuyuki Takakura
Yoshihiro Yamada
Hiroyasu Kidoya
Original Assignee
National University Corporation Kanazawa University
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 National University Corporation Kanazawa University filed Critical National University Corporation Kanazawa University
Publication of WO2006115243A1 publication Critical patent/WO2006115243A1/en

Links

Classifications

    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • C12N5/12Fused cells, e.g. hybridomas
    • C12N5/16Animal 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0695Stem cells; Progenitor cells; Precursor cells

Definitions

  • the present invention relates to a method for producing cancer stem cells.
  • Non-Patent Document 1 it is cultured in a test tube!
  • Speaking tumor cell lines do not have uniform cell growth
  • Some cells belong to a group of stem cells called SP (side populatio n) that have high drug efflux ability, and these cells differentiate into various cells It is said to have pluripotency.
  • SP side populatio n
  • tumor cells having cell surface markers that are partly different from other tumor cells among breast cancer and brain tumor cells that have developed in individuals. Has been shown to have a higher ability to engraft individuals than other tumor cells!
  • Non-patent literature l Kondo, T "et al., Proc. Natl. Acad. Sci. USA” 101 (3), pp.781-786, 2004
  • Non-Patent Document 2 AH "Iajj, M. et al” Proc. Natl. Acad. Sci., U S A "100 (7), pp.3983-39 88, 2003
  • Non-Patent Document 3 Singh, S.K., et al., Nature, 432 (7015), pp.396-401, 2004
  • An object of the present invention is to provide a method for obtaining cancer stem cells experimentally with good reproducibility.
  • the inventors of the present invention have conventionally attempted a detailed analysis of the involvement of hematopoietic cells in the molecular mechanism of angiogenesis in cancer tissues.
  • hematopoietic stem cells and hematopoietic progenitor cells invading only the mature blood cells of the myeloid and lymphocyte lineages invaded the tumor (Okamoto, R., et al. , Blood, 105 (7), pp.275 7-2763, 2005), and many of these leaked out of the blood vessels within the tumor. They found that these blood cells secrete angiogenesis-promoting factors and contribute greatly to the formation of new blood vessels formed in the tumor, that is, tumor angiogenesis.
  • a method for producing cancer stem cells comprising a step of in vitro cell fusion of tumor cells and blood cells.
  • the above method for performing the above cell fusion by co-culturing blood cells containing hematopoietic stem cells and Z or progenitor cells and tumor cells.
  • the present invention provides a cancer stem cell that can be obtained by the above method.
  • Another aspect of the present invention is a method for producing a model tumor tissue according to the present invention, wherein a mixture of cancer stem cells obtained by cell fusion of tumor cells and blood cells and the tumor cells is cultured. A method comprising the steps is provided. The present invention also provides a model tumor tissue that can be obtained by the above method.
  • a method for preparing a cancer-bearing model animal wherein a mixture of a cancer stem cell obtained by cell fusion of a tumor cell and a blood cell and the tumor cell is used as an animal.
  • a method comprising the step of administering to:
  • the present invention also provides a cancer-bearing model animal that can be obtained by the above method.
  • FIG. 1 B16 tumor cells (alone) or a mixture of B16 cells and the cancer stem cells obtained in Example 1 of Example were implanted subcutaneously in mice, and the size of the formed tumors was measured over time. It is a figure showing the measurement results.
  • FIG. 2 shows a flow cytometric analysis of a tumor formed when a B16 tumor cell labeled with a red fluorescent dye is transplanted into a mouse that expresses GFP throughout the body.
  • (A) shows the result of confirming a cell group that simultaneously expresses DS-RED and GFP formed by cell fusion in vivo in the tumor (R2), and (B) shows the cell of this R2
  • the result of having analyzed the expression of the stem cell marker (C-Kit or CD133) is shown.
  • the method of the present invention is a method for producing cancer stem cells, and includes a step of cell fusion of tumor cells and blood cells in vitro (in vitro).
  • the type of tumor cell line to be fused with blood cells is not particularly limited, and includes, for example, any cancer cells of mammals other than humans (including cancer cells such as solid cancer and blood cancer).
  • tumor cell lines such as B16 cells that are mouse melanoma cell lines
  • sublines thereof and primary tumor cells
  • the tumor cell line may be introduced with a gene for expressing a fluorescent chromoprotein in order to facilitate cell fusion and subsequent confirmation of the behavior of cells derived from tumor cells.
  • a plasmid capable of expressing a red fluorescent chromoprotein (DS-RED) can be introduced.
  • the gene transfer can be performed by a known method, and any method available to those skilled in the art, such as an electroporation method and a DEAE-DEXTRAN method, can be employed.
  • the type of blood cell is not particularly limited as long as it is a blood cell derived from an animal species that can be fused with a tumor cell.
  • blood cells derived from any blood or tissue such as peripheral blood or fetal liver may be used.
  • blood cells containing hematopoietic stem cells and Z or progenitor cells can be preferably used.
  • cells expressing CD11b containing hematopoietic stem cells as blood cells on the cell surface can be collected from the bone marrow of mammals.
  • Blood cells can be collected by a person skilled in the art, such as a method using a cell sorter, a vaning method using an antibody, a magnetic cell recovery method using magnetic beads, or a method using an automatic fluorescent cell recovery device other than EPICS. Any method can be employed.
  • blood cells are not limited to CD lib-positive cells including hematopoietic stem cells, monocytes, macrophages, and neutrophils, but are blood cells that can turn tumor cells into cancer stem cells. For example, you can use some cells.
  • blood cells may be introduced with a gene for expressing a fluorescent chromoprotein.
  • a gene for expressing a fluorescent chromoprotein it is preferable to use a gene that expresses a protein having a color tone different from that of a fluorescent chromoprotein expressed from a gene introduced into a tumor cell.
  • DS-RED red fluorescent chromoprotein
  • Blood cells that also control the bone marrow strength of mice expressing green 'fluorescent protein (GFP) or related proteins in the whole body (green mice) can be used, and fused cells can be easily identified. Is possible.
  • any means that can confirm DNA derived from tumor cells and blood cells can be adopted, for example, deletion or insertion into cell DNA.
  • Cells that have been marked with may be used, or cells with the surface labeled with a different fluorescent dye may be used for short-term culture.
  • cell fusion can be performed by simultaneously seeding and co-culturing about 10 4 tumor cells and about 10 6 blood cells, for example.
  • the medium used for the culture is not particularly limited, and can be appropriately selected by those skilled in the art.
  • a medium in which an appropriate concentration, for example, about 10% of urine serum is added to the basic medium, or a medium in which one or more appropriate cytodynamic ins are added can also be used.
  • the site force-in for example, a force that can use stem cell factor, M-CSF, etc.
  • the type and concentration are not limited to these, and those skilled in the art can appropriately select them.
  • Co-culture for cell fusion can be performed, for example, at a temperature of about 37 ° C. in the presence of 5% CO for about 1 to 10 days.
  • a fusion cell of about 1 to 10%, preferably about 0.1 to several% is obtained.
  • cell fusion can be performed by co-cultivation, but polyethylene glycol can be used without using the cell fusion ability of the cells themselves.
  • Cell fusion can also be carried out by a general cell fusion method such as a fusion method using PEG or a cell fusion method using electric pulses.
  • cell fusion is also possible by mechanically injecting cell nuclei extracted from blood cells into tumors, or conversely, injecting tumor cell nuclei into blood cells.
  • the fused cells are stained yellow and can be separated using a cell sorter or the like. In this way, the cancer stem cells of the present invention can be isolated.
  • a model tumor tissue can be formed in vivo or in vitro.
  • cancer Stem cells and cancer stem cell strength Cancer cells that have been separated are considered to be in a mosaic pattern.
  • the presence of cancer stem cells promotes, for example, the growth of tumor cells, or the cancer is engrafted in the individual to form cancer tissue.
  • To produce a model tumor tissue that is closer to a real tumor tissue by mixing and culturing the fused cells that are stem cells and the tumor cells used to generate the fused cells, or transplanting the cell mixture to an animal. Is possible.
  • the model tumor tissue is obtained by mixing and culturing the fusion cells, which are cancer stem cells obtained as described above, and the tumor cells used for fusion in a ratio of, for example, about 1:10. Can be generated in vitro.
  • An appropriate medium as exemplified above can be used for the culture.
  • a tumor tissue can be formed at the transplant site. This animal can be used as a tumor model animal.
  • Examples of animals include, but are not limited to, the ability to use mice, rats, rabbits, guinea pigs, monkeys, and the like. In general, it is desirable to select an animal species that can engraft the tumor cells used for fusion.
  • the cell mixture When transplanting the above cell mixture into an animal, the cell mixture may be suspended in, for example, phosphate buffered saline (PBS), or a semi-solid medium such as collagen or matrigel, or other medium may be used.
  • PBS phosphate buffered saline
  • a semi-solid medium such as collagen or matrigel, or other medium may be used.
  • the tumor tissue formation can be evaluated by measuring the size of the tumor over time, but the cell mixture is intravenously or intraperitoneally.
  • tumors formed in the lungs, liver, etc. may be observed non-visually.
  • tumor cells fused with blood cells are transformed into highly malignant cancer cells that increase the tumor, and the phenotype of the cells It can be concluded that the cancer stem cells have been transformed. This process occurs in vivo, and tumor cells generated by transformation in vivo become cancer stem cells by cell fusion with immature blood cells such as hematopoietic stem cells, and together with surrounding tumor cells, mosaic cells It is thought to contribute to the development of cancer by forming a tumorous tissue.
  • the cancer stem cell generation mechanism and the cancer stem It becomes possible to study the formation process of cancer involving cells in detail.
  • cell fusion is performed in the presence of a test substance, and the degree of inhibition of cell fusion is confirmed, thereby screening a substance that inhibits the generation of cancer stem cells. It becomes possible to do.
  • a substance that induces apoptosis of the cancer stem cell of the present invention can be screened. Substances screened in this way are expected to be useful as anti-cancer drugs.
  • model tumor tissue or animal model tumor reproduces a malignancy closer to that of a real tumor than that of a tumor cell alone without cancer stem cells. Therefore, it is extremely useful for detailed elucidation of mechanisms such as cancer development and growth, or cancer metastasis, and is extremely useful for the development of antitumor agents effective in vivo.
  • Example 1 Cell fusion of tumor cells and blood cells in vitro
  • B16 cells which are mouse melanoma cell lines, were used as tumor cell lines to be fused with blood cells.
  • B16 melanoma cells were subcultured in normal medium supplemented with 10% urine serum in DMEM (Sigma) medium.
  • a plasmid capable of expressing red fluorescent chromoprotein (DS-RED) (Betaton's Dickinson) was introduced into B16 melanoma cells using lipofuctamine (Invitrogen).
  • DS-RED red fluorescent chromoprotein
  • Ivitrogen lipofuctamine
  • cells that express green 'fluorescent' protein (GFP) in the whole body (green mouse) bone marrow force CD lib (cell population including hematopoietic stem cells) on the cell surface are cell sorters (EPICS, Coulter) (Manufactured).
  • Example 2 Tumor formation model with cancer stem cells
  • cancer stem cells and cancer stem cells are considered to be mosaics.
  • the cancer stem cells obtained in Example 1 actually have a positive effect on the occurrence of cancer, because the cancer stem cells are thought to engraft in the individual to form cancer tissue. This was confirmed by the following experiment.
  • a mixture of B16 tumor cells 1.1 x 10 4 (B16 alone) or B16 tumor cells 1.0 x 10 4 and cancer stem cells 0.1 x 10 4 obtained in Example 1 was suspended in PBS, respectively, and C57B1 / 6 strain mice were administered and transplanted under the back skin. Tumor formation was evaluated by measuring tumor size over time. The results are shown in Fig. 1.
  • Tumor formation was not observed in individuals transplanted with 1.1 x 10 4 B16 tumor cells, whereas individuals transplanted with the same amount of B16 tumor cells mixed with 0.1 x 10 4 cancer stem cells. Significant tumor formation was observed, and it was confirmed that tumor cells including cancer stem cells changed to a cell population with high tumorigenicity.
  • Example 1 1 ⁇ 10 6 B16 tumor cells fluorescently labeled with DS-RED obtained in Example 1 (this number of cells is the number of cells that can form tumors alone) in PBS. Suspended, transplanted into C57B1 / 6 lineage green mouse, and tumor cells that simultaneously express GFP and DS-RED in the formed tumor (cells derived from the host green mouse and tumor cells fused together) We examined whether or not there exists. 14 days after transplantation of B16 tumor cells expressing DS-RED, the tumor was removed from the mouse, and the tumor tissue was minced with surgical scissors, and then disperse (Boehringer) was added to the minced tumor tissue.
  • the cells were allowed to stand at room temperature for 10 minutes, and then the cell suspension was filled into a syringe equipped with a syringe needle, and the cells were dispersed by repeating aspiration and discharge several times. These cells were analyzed by flow cytometry. As a result, as shown in FIG. 2 (A), the presence of fusion cells expressing both DS-RED and GFP was recognized. In addition, when this fused cell was stained with a monoclonal antibody against c-Kit and CD133 (both manufactured by Pharmingen) and analyzed by flow cytometry, c-Kit was almost 100% and CD133 was 50-60% was found to be expressed in the fused cells (Fig. 2 (B)). Since c-Kit and AC133 are known to be expressed in immature stem cell levels, it was confirmed that the cancer stem cell generation process obtained in Example 1 occurred in the actual tumor formation process. did it. Industrial applicability
  • cancer stem cells can be efficiently generated with high reproducibility in vitro.
  • a model tumor tissue that is closer to the actual tumor tissue can be generated in vitro, or a tumor-bearing model animal that reproduces the tumor development process in vivo can be created.

Abstract

A method of producing a cancer stem cell comprising the step of in vitro cell fusion of a tumor cell and a blood cell (for example, blood cells including a hematopoietic stem cell and/or a precursor cell) and a method of producing a cancer-bearing model animal comprising the step of administering a mixture of the cancer stem cell obtained by the cell fusion of a tumor cell and a blood cell and the tumor cell to a non-human mammal.

Description

がん幹細胞の作成方法 技術分野  How to create cancer stem cells
[0001] 本発明はがん幹細胞の作成方法に関するものである。  [0001] The present invention relates to a method for producing cancer stem cells.
背景技術  Background art
[0002] 現代のがん研究および治療は、がんを均質な細胞集団として扱っており、がん治療 薬開発においては、ただ単純に増殖するがん細胞の細胞死を指標に治療薬の開発 力 されている。このような経緯により多くのがん治療薬や治療法が開発されてきた 力 がんを根治させるには至っていないのが現状である。一方、最近になって、がん 組織は、抗がん剤に抵抗性のがん幹細胞と、そのがん幹細胞より分ィ匕して過剰に増 殖する杭がん剤に感受性のあるがん細胞により構築されており、がん幹細胞の細胞 死を誘導しない限りがんが再発するという概念が提唱されており、非常に注目されて いる。  [0002] Modern cancer research and treatment treats cancer as a homogeneous cell population. In the development of cancer therapeutics, the development of therapeutics is based solely on the cell death of proliferating cancer cells. It is powerful. Due to these circumstances, many cancer drugs and treatments have been developed. The current situation is that the cancer has not been completely cured. On the other hand, recently, cancer tissues are sensitive to cancer stem cells that are resistant to anti-cancer drugs and pile cancer drugs that are separated from the cancer stem cells and multiply excessively. The concept of cancer recurring unless it induces cell death of cancer stem cells has been proposed and has received much attention.
[0003] 例えば、非特許文献 1によれば、試験管内で培養されて!ヽる腫瘍細胞株は均一な 細胞が増殖しているのではなぐ一部の細胞は薬剤排出能の高い SP (side populatio n)と呼ばれる幹細胞群に属し、これらの細胞は種々の細胞に分化する多分化能を有 するとされている。また、非特許文献 2及び 3では、それぞれ個体に発生した乳がん および脳腫瘍細胞の中で、他の腫瘍細胞とは一部異なる細胞表面マーカーを有す る腫瘍細胞が存在しており、これらの細胞は他の腫瘍細胞に比べ個体への生着能 力が高!ヽことが示されて!/ヽる。  [0003] For example, according to Non-Patent Document 1, it is cultured in a test tube! Speaking tumor cell lines do not have uniform cell growth Some cells belong to a group of stem cells called SP (side populatio n) that have high drug efflux ability, and these cells differentiate into various cells It is said to have pluripotency. In Non-Patent Documents 2 and 3, there are tumor cells having cell surface markers that are partly different from other tumor cells among breast cancer and brain tumor cells that have developed in individuals. Has been shown to have a higher ability to engraft individuals than other tumor cells!
[0004] これらの報告においては、腫瘍は生化学的に均質ではなぐ一部に幹細胞様の多 分化能、および移植による生着能力が高い細胞が存在しており、それががん幹細胞 であると説明されている。し力しながら、これらのがん幹細胞がいかなる分ィ匕過程を経 て他のがん細胞と異なる形質を獲得しているのかは現在のところ不明であり、また、 がん幹細胞を取得する方法にっ 、てもこれらの文献には示唆な 、し教示がな 、。が ん幹細胞を実験的に取得する方法が開発され、がん幹細胞の発生の成因が解明さ れれば、がん細胞の幹細胞化を抑制する治療法や、がん幹細胞を標的とする治療 法を開発できることが期待されるほ力、がんの悪性化や予後の診断法、あるいはがん が棲息する-ツチを解明してがんを根絶する診断や治療法への発展が期待される。 従って、がん幹細胞を実験的に再現性よく取得する方法の開発が切望されている。 非特許文献 l : Kondo, T" et al., Proc. Natl. Acad. Sci. U S A" 101(3), pp.781- 786, 2004 [0004] In these reports, tumors are not biochemically homogeneous, but some cells have stem cell-like multipotency and high engraftment potential by transplantation, which are cancer stem cells It is explained. However, it is currently unknown how these cancer stem cells have acquired different traits from other cancer cells, and how to obtain cancer stem cells. However, there is no suggestion or teaching in these documents. Once a method for experimentally obtaining cancer stem cells has been developed and the cause of cancer stem cell development has been elucidated, treatments that suppress the stem cell transformation of cancer cells and treatments that target cancer stem cells Expected to be able to develop methods, cancer malignant and prognostic methods, or cancer is inhabited-it is expected to develop into diagnosis and treatment methods that eradicate cancer by elucidating the pinch . Therefore, development of a method for obtaining cancer stem cells experimentally with good reproducibility is eagerly desired. Non-patent literature l: Kondo, T "et al., Proc. Natl. Acad. Sci. USA" 101 (3), pp.781-786, 2004
非特許文献 2 :AH"Iajj, M. et al" Proc. Natl. Acad. Sci., U S A" 100(7), pp.3983- 39 88, 2003  Non-Patent Document 2: AH "Iajj, M. et al" Proc. Natl. Acad. Sci., U S A "100 (7), pp.3983-39 88, 2003
非特許文献 3 : Singh, S.K., et al., Nature, 432(7015), pp.396- 401, 2004  Non-Patent Document 3: Singh, S.K., et al., Nature, 432 (7015), pp.396-401, 2004
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0005] 本発明の課題は、がん幹細胞を実験的に再現性よく取得する方法を提供すること にある。 [0005] An object of the present invention is to provide a method for obtaining cancer stem cells experimentally with good reproducibility.
課題を解決するための手段  Means for solving the problem
[0006] 本発明者らは、従来より、がん組織の血管新生の分子メカニズムについて造血系 細胞の関与について詳細な解析を試みてきた。その結果、腫瘍の成長中において は、骨髄球系、リンパ球系の成熟した血液細胞だけでなぐ造血幹細胞や造血前駆 細胞が腫瘍内部に多く侵入していること(Okamoto, R., et al., Blood, 105(7), pp.275 7-2763, 2005)、及びこれらの多くは腫瘍内で血管外に漏出していることを解明した。 そして、これらの血液細胞は血管形成の促進因子を分泌し、腫瘍内に形成される新 しい血管の形成、つまり腫瘍血管新生に大きく貢献することを見出した。  [0006] The inventors of the present invention have conventionally attempted a detailed analysis of the involvement of hematopoietic cells in the molecular mechanism of angiogenesis in cancer tissues. As a result, during tumor growth, hematopoietic stem cells and hematopoietic progenitor cells invading only the mature blood cells of the myeloid and lymphocyte lineages invaded the tumor (Okamoto, R., et al. , Blood, 105 (7), pp.275 7-2763, 2005), and many of these leaked out of the blood vessels within the tumor. They found that these blood cells secrete angiogenesis-promoting factors and contribute greatly to the formation of new blood vessels formed in the tumor, that is, tumor angiogenesis.
[0007] また、腫瘍内への血液細胞の侵入を抑制すると腫瘍の増大が生じてこないことから 、本発明者らは、血液細胞には血管形成を促進する以外に腫瘍の悪性化に関与す る機序が存在するとの仮説に基づいて、これらの血液細胞と腫瘍細胞との相互関係 を解析した。その結果、造血幹細胞 Z前駆細胞を含有する血液細胞と腫瘍細胞との 共培養により、腫瘍細胞と血液細胞とが細胞融合すること、及び血液細胞と細胞融 合した上記の腫瘍細胞は、融合前には発現して 、な力つた幹細胞の表面抗原であ る CD133 (AC133)や c-kitの発現が誘導されていることを見出した。また、血液細胞と 細胞融合した少量の腫瘍細胞及び血液細胞と細胞融合して ゝな 、少量の腫瘍細胞 (個体に移植しても腫瘍を形成することのできな 、腫瘍細胞である)を混合してマウス に移植することにより、非常に大きな悪性度の高い腫瘍形成が誘導されることを見出 した。これは、がん糸且織の形成において通常のがん細胞とがん幹細胞とが混在した モザイク状態においてがんの成長が促進すること実験的に再現したものと考えられる 。本発明は上記の知見を基にして完成された。 [0007] Further, since suppression of blood cell invasion into a tumor does not cause an increase in tumor, the present inventors are involved in the malignant transformation of tumors in addition to promoting blood vessel formation in blood cells. Based on the hypothesis that a mechanism exists, we analyzed the correlation between these blood cells and tumor cells. As a result, by co-culturing blood cells containing hematopoietic stem cell Z progenitor cells and tumor cells, the tumor cells and blood cells fuse, and the above-mentioned tumor cells fused with blood cells are not fused. It was found that the expression of CD133 (AC133) and c-kit, which are surface antigens of stem cells with strong expression, was induced. In addition, a small amount of tumor cells fused with blood cells and a small amount of tumor cells fused with blood cells. It was found that mixing tumor cells (which cannot form tumors even when transplanted into individuals) and transplanting them into mice induces very high malignant tumor formation. . This is considered to have been experimentally reproduced that the growth of cancer is promoted in a mosaic state in which normal cancer cells and cancer stem cells are mixed in the formation of cancer thread and tissue. The present invention has been completed based on the above findings.
[0008] すなわち、本発明により、がん幹細胞の作成方法であって、腫瘍細胞と血液細胞と をインビトロで細胞融合する工程を含む方法が提供される。上記の発明の好ま ヽ態 様によれば、造血幹細胞及び Z又は前駆細胞を含有する血液細胞と腫瘍細胞との 共培養により上記細胞融合を行なう上記方法が提供される。また、本発明により、上 記の方法により得ることができるがん幹細胞が提供される。 [0008] That is, according to the present invention, there is provided a method for producing cancer stem cells, comprising a step of in vitro cell fusion of tumor cells and blood cells. According to a preferred embodiment of the present invention, there is provided the above method for performing the above cell fusion by co-culturing blood cells containing hematopoietic stem cells and Z or progenitor cells and tumor cells. In addition, the present invention provides a cancer stem cell that can be obtained by the above method.
別の観点カゝらは、本発明により、モデル腫瘍組織の作成方法であって、腫瘍細胞と 血液細胞とを細胞融合することにより得られるがん幹細胞と上記腫瘍細胞との混合 物を培養する工程を含む方法が提供される。また、本発明により、上記の方法により 得ることができるモデル腫瘍組織が提供される。  Another aspect of the present invention is a method for producing a model tumor tissue according to the present invention, wherein a mixture of cancer stem cells obtained by cell fusion of tumor cells and blood cells and the tumor cells is cultured. A method comprising the steps is provided. The present invention also provides a model tumor tissue that can be obtained by the above method.
さらに別の観点からは、本発明により、担がんモデル動物の作成方法であって、腫 瘍細胞と血液細胞とを細胞融合することにより得られるがん幹細胞と上記腫瘍細胞と の混合物を動物に投与する工程を含む方法が提供される。また、本発明により、上 記の方法により得ることができる担がんモデル動物が提供される。  From another point of view, according to the present invention, there is provided a method for preparing a cancer-bearing model animal, wherein a mixture of a cancer stem cell obtained by cell fusion of a tumor cell and a blood cell and the tumor cell is used as an animal. There is provided a method comprising the step of administering to: The present invention also provides a cancer-bearing model animal that can be obtained by the above method.
図面の簡単な説明  Brief Description of Drawings
[0009] [図 1]B16腫瘍細胞(単独)又は B16細胞と実施例の例 1で得たがん幹細胞との混合 物をマウスの皮下に移植し、形成された腫瘍のサイズを経時的に測定した結果を示 した図である。  [0009] [Fig. 1] B16 tumor cells (alone) or a mixture of B16 cells and the cancer stem cells obtained in Example 1 of Example were implanted subcutaneously in mice, and the size of the formed tumors was measured over time. It is a figure showing the measurement results.
[図 2]赤色の蛍光色素でラベルされた B16腫瘍細胞を GFPを全身に発現するマウスに 移植した際に形成された腫瘍のフローサイトメトリー解析を示した図である。図中、 (A) は腫瘍中に生体内で細胞融合により形成された DS-RED及び GFPを同時に発現する 細胞群が確認された結果を示し(R2)、(B)はこの R2の細胞について幹細胞マーカー (C- Kit又は CD 133)の発現を解析した結果を示す。  FIG. 2 shows a flow cytometric analysis of a tumor formed when a B16 tumor cell labeled with a red fluorescent dye is transplanted into a mouse that expresses GFP throughout the body. In the figure, (A) shows the result of confirming a cell group that simultaneously expresses DS-RED and GFP formed by cell fusion in vivo in the tumor (R2), and (B) shows the cell of this R2 The result of having analyzed the expression of the stem cell marker (C-Kit or CD133) is shown.
発明を実施するための最良の形態 [0010] 本発明の方法は、がん幹細胞の作成方法であって、腫瘍細胞と血液細胞とをイン ビトロ(生体外)で細胞融合する工程を含むことを特徴として 、る。 BEST MODE FOR CARRYING OUT THE INVENTION [0010] The method of the present invention is a method for producing cancer stem cells, and includes a step of cell fusion of tumor cells and blood cells in vitro (in vitro).
血液細胞と融合させる腫瘍細胞株の種類は特に限定されず、例えば、ヒトゃヒト以 外の哺乳類動物の任意のがん細胞(固形癌及び血液癌などのがん細胞を包含する The type of tumor cell line to be fused with blood cells is not particularly limited, and includes, for example, any cancer cells of mammals other than humans (including cancer cells such as solid cancer and blood cancer).
)を用いることができる。例えば、腫瘍細胞株 (マウスメラノーマ細胞株である B16細胞 など)、そのサブラインやプライマリー腫瘍細胞などを用いることができる。腫瘍細胞 株は、細胞融合やその後の腫瘍細胞由来の細胞の挙動の確認を容易にするため、 蛍光色素蛋白を発現するための遺伝子を導入しておいてもよい。このような目的のた めには、例えば、赤色の蛍光色素蛋白(DS-RED)を発現できるプラスミドを導入する ことができる。遺伝子導入は公知の手法により行なうことが可能であり、例えば、エレ タトロポレーシヨン法、 DEAE-DEXTRAN法などの当業者に利用可能な任意の方法を 採用することができる。 ) Can be used. For example, tumor cell lines (such as B16 cells that are mouse melanoma cell lines), sublines thereof, and primary tumor cells can be used. The tumor cell line may be introduced with a gene for expressing a fluorescent chromoprotein in order to facilitate cell fusion and subsequent confirmation of the behavior of cells derived from tumor cells. For this purpose, for example, a plasmid capable of expressing a red fluorescent chromoprotein (DS-RED) can be introduced. The gene transfer can be performed by a known method, and any method available to those skilled in the art, such as an electroporation method and a DEAE-DEXTRAN method, can be employed.
[0011] 血液細胞は、腫瘍細胞と融合可能な動物種由来の血液細胞であれば、その種類 は特に限定されない。例えば、末梢血や胎児肝臓など、任意の血液や組織由来の 血液細胞であってもよい。例えば、造血幹細胞及び Z又は前駆細胞を含有する血液 細胞を好ましく用いることができる。例えば、血液細胞として造血幹細胞を含む CD11 bを細胞表面に発現する細胞を哺乳類動物の骨髄力 採取することができる。血液 細胞の採取には、例えば、セルソーターを用いる方法、抗体によるバニング法、マグ ネティックビーズを用いる磁気による細胞回収法、 EPICS以外の自動蛍光細胞回収 装置を用いる方法など、当業者に利用可能な任意の方法を採用することができる。も つとも、血液細胞としては、造血幹細胞や単球、マクロファージ及び好中球を含む CD lib陽性細胞に限定されることはなぐ腫瘍細胞をがん幹細胞化させることのできる血 液細胞であるならば 、かなる細胞を用いてもょ 、。  [0011] The type of blood cell is not particularly limited as long as it is a blood cell derived from an animal species that can be fused with a tumor cell. For example, blood cells derived from any blood or tissue such as peripheral blood or fetal liver may be used. For example, blood cells containing hematopoietic stem cells and Z or progenitor cells can be preferably used. For example, cells expressing CD11b containing hematopoietic stem cells as blood cells on the cell surface can be collected from the bone marrow of mammals. Blood cells can be collected by a person skilled in the art, such as a method using a cell sorter, a vaning method using an antibody, a magnetic cell recovery method using magnetic beads, or a method using an automatic fluorescent cell recovery device other than EPICS. Any method can be employed. In any case, blood cells are not limited to CD lib-positive cells including hematopoietic stem cells, monocytes, macrophages, and neutrophils, but are blood cells that can turn tumor cells into cancer stem cells. For example, you can use some cells.
[0012] 血液細胞には、細胞融合やその後の血液細胞由来の細胞の挙動の確認を容易に するため、蛍光色素蛋白を発現するための遺伝子を導入しておいてもよい。このよう な目的のためには、腫瘍細胞に導入した遺伝子から発現される蛍光色素蛋白と異な る色調の蛋白質を発現する遺伝子を用いることが好ましい。例えば、腫瘍細胞に赤 色の蛍光色素蛋白(DS-RED)を発現できるプラスミドを導入した場合には、血液細胞 としてグリーン'フルオレセント ·プロテイン (GFP)又はその類縁蛋白質を全身に発現 するマウス (green mouse)の骨髄力も調節した血液細胞を用いることができ、融合さ れた細胞の同定を容易に行なうことが可能になる。もっとも、上記の目的のためには、 腫瘍細胞及び血液細胞由来の DNAを確認することができる手段であれば任意の手 段を採用することが可能であり、例えば、細胞の DNAに欠損や挿入によるマーキング を行なった細胞を用いたり、短期の培養の際には細胞表面を異なる蛍光色素で標識 した細胞を用いてもよい。 [0012] In order to facilitate confirmation of cell fusion and subsequent behavior of cells derived from blood cells, blood cells may be introduced with a gene for expressing a fluorescent chromoprotein. For this purpose, it is preferable to use a gene that expresses a protein having a color tone different from that of a fluorescent chromoprotein expressed from a gene introduced into a tumor cell. For example, when a plasmid capable of expressing red fluorescent chromoprotein (DS-RED) is introduced into tumor cells, blood cells Blood cells that also control the bone marrow strength of mice expressing green 'fluorescent protein (GFP) or related proteins in the whole body (green mice) can be used, and fused cells can be easily identified. Is possible. However, for the above purpose, any means that can confirm DNA derived from tumor cells and blood cells can be adopted, for example, deletion or insertion into cell DNA. Cells that have been marked with may be used, or cells with the surface labeled with a different fluorescent dye may be used for short-term culture.
[0013] 細胞の融合は、例えば、例えば 104個程度の腫瘍細胞と例えば 106個程度の血液細 胞とを同時に播種して共培養することにより行なうことができる。培養に用いる培地は 特に限定されず、当業者が適宜選択可能である力 例えば、 DMEMなどのほか、 RP ΜΙ、 α ΜΕΜ、 F12培地などを用いることができる。培養に際しては、基本培地に適宜 の濃度、例えば 10%程度のゥシ血清を添加した培地や、適宜のサイト力インなどを 1 種又は 2種以上添加した培地を用いることもできる。サイト力インとしては、例えば、 ste m cell factorや M-CSFなどを用いることができる力 これらに限定されることはなぐ 種類や濃度は当業者が適宜選択可能である。細胞融合のための共培養は、例えば 、 37°C程度の温度で 5% CO存在下に 1〜10日程度行なうことができ、全細胞中の 0.0 [0013] For example, cell fusion can be performed by simultaneously seeding and co-culturing about 10 4 tumor cells and about 10 6 blood cells, for example. The medium used for the culture is not particularly limited, and can be appropriately selected by those skilled in the art. For example, in addition to DMEM, RPΜΙ, αΜΕΜ, F12 medium and the like can be used. In culturing, a medium in which an appropriate concentration, for example, about 10% of urine serum is added to the basic medium, or a medium in which one or more appropriate cytodynamic ins are added can also be used. As the site force-in, for example, a force that can use stem cell factor, M-CSF, etc. The type and concentration are not limited to these, and those skilled in the art can appropriately select them. Co-culture for cell fusion can be performed, for example, at a temperature of about 37 ° C. in the presence of 5% CO for about 1 to 10 days.
2  2
1〜10%程度、好ましくは 0.1〜数 %程度の融合細胞が得られる。上記のように、腫瘍細 胞及び Z又は血液細胞自体が細胞融合能を有する場合には、共培養により細胞融 合を行なうことができるが、細胞自体の細胞融合能を用いずに、ポリエチレングリコー ル (PEG)を用いた融合法や電気パルスによる細胞融合法などの一般的な細胞融合 方法により細胞融合を行なうこともできる。さらに、血液細胞から取り出した細胞核を 腫瘍に機械的に注入する、あるいは逆に腫瘍細胞の核を血液細胞に注入することに よっても細胞融合が可能である。腫瘍細胞を赤色蛍光色素、血液細胞を緑色蛍光色 素で標識した場合には、融合細胞は黄色に染色されるので、セルソーターなどを用 いて分離することが可能である。このようにして、本発明のがん幹細胞を単離すること ができる。  A fusion cell of about 1 to 10%, preferably about 0.1 to several% is obtained. As described above, when tumor cells and Z or blood cells themselves have cell fusion ability, cell fusion can be performed by co-cultivation, but polyethylene glycol can be used without using the cell fusion ability of the cells themselves. Cell fusion can also be carried out by a general cell fusion method such as a fusion method using PEG or a cell fusion method using electric pulses. Furthermore, cell fusion is also possible by mechanically injecting cell nuclei extracted from blood cells into tumors, or conversely, injecting tumor cell nuclei into blood cells. When tumor cells are labeled with a red fluorescent dye and blood cells are labeled with a green fluorescent dye, the fused cells are stained yellow and can be separated using a cell sorter or the like. In this way, the cancer stem cells of the present invention can be isolated.
[0014] 上記のようにして得られたがん幹細胞を用いて、モデル腫瘍組織をインビボ又はィ ンビトロで形成させることができる。先に説明したとおり、腫瘍形成に際しては、がん 幹細胞とがん幹細胞力 分ィ匕したがん細胞がモザイク状となっていると考えられる。 がん幹細胞が存在することによって、例えば腫瘍細胞の増殖が促進され、あるいは がんが個体内で生着してがん組織を形成すると考えられることから、上記のようにして 得られたがん幹細胞である融合細胞と上記融合細胞の生成に用いた腫瘍細胞とを 混合して培養し、あるいは上記細胞混合物を動物に移植することにより、より現実の 腫瘍組織に近いモデル腫瘍組織を生成することが可能になる。 [0014] By using the cancer stem cells obtained as described above, a model tumor tissue can be formed in vivo or in vitro. As explained earlier, during tumor formation, cancer Stem cells and cancer stem cell strength Cancer cells that have been separated are considered to be in a mosaic pattern. The presence of cancer stem cells promotes, for example, the growth of tumor cells, or the cancer is engrafted in the individual to form cancer tissue. To produce a model tumor tissue that is closer to a real tumor tissue by mixing and culturing the fused cells that are stem cells and the tumor cells used to generate the fused cells, or transplanting the cell mixture to an animal. Is possible.
[0015] 例えば、上記のようにして得られたがん幹細胞である融合細胞と融合に用いた腫瘍 細胞とを、例えば 1:10程度の割合で混合して培養することによりモデル腫瘍組織をィ ンビトロで生成することができる。培養には上記に例示したような適宜の培地を用いる ことができる。また、例えば上記の割合でがん幹細胞である融合細胞と融合に用いた 腫瘍細胞とを含む細胞混合物をヒト以外の哺乳類動物に移植することにより、移植部 位に腫瘍組織を形成することができ、この動物を腫瘍モデル動物として用いることが できる。動物としては、例えば、マウス、ラット、ゥサギ、モルモット、サルなどを用いるこ とができる力 これらに限定されることはない。一般的には、融合に用いた腫瘍細胞 が生着可能な動物種を選択することが望ましい。上記細胞混合物を動物に移植する に際しては、細胞混合物を例えばリン酸緩衝生理食塩水 (PBS)に懸濁してもよぐある いはコラーゲンやマトリゲルなどの半固形培地やその他の培地を用いてもょ 、。腫瘍 組織の形成に関する評価は、例えば、皮下に細胞混合物を移植した場合には、経時 的に腫瘍のサイズを肉眼的に計測することにより行なうことができるが、静脈内又は 腹腔内に細胞混合物を移植した場合には、肺や肝臓等で形成される腫瘍を非肉眼 的に観察すればよい。 [0015] For example, the model tumor tissue is obtained by mixing and culturing the fusion cells, which are cancer stem cells obtained as described above, and the tumor cells used for fusion in a ratio of, for example, about 1:10. Can be generated in vitro. An appropriate medium as exemplified above can be used for the culture. In addition, for example, by transplanting a cell mixture containing a fusion cell that is a cancer stem cell and the tumor cell used for fusion at the above-mentioned ratio into a mammal other than a human, a tumor tissue can be formed at the transplant site. This animal can be used as a tumor model animal. Examples of animals include, but are not limited to, the ability to use mice, rats, rabbits, guinea pigs, monkeys, and the like. In general, it is desirable to select an animal species that can engraft the tumor cells used for fusion. When transplanting the above cell mixture into an animal, the cell mixture may be suspended in, for example, phosphate buffered saline (PBS), or a semi-solid medium such as collagen or matrigel, or other medium may be used. Oh ,. For example, when the cell mixture is implanted subcutaneously, the tumor tissue formation can be evaluated by measuring the size of the tumor over time, but the cell mixture is intravenously or intraperitoneally. When transplanted, tumors formed in the lungs, liver, etc. may be observed non-visually.
[0016] V、かなる特定の理論に拘泥するわけではな 、が、血液細胞と細胞融合した腫瘍細 胞は腫瘍を増大させる悪性度の高いがん細胞に変化し、またその細胞の表現型から がん幹細胞に形質転換したものと結論できる。このような過程は生体内においても生 じており、生体内において形質転換により生じた腫瘍細胞は造血幹細胞など未熟な 血液細胞と細胞融合することによりがん幹細胞となり、周囲の腫瘍細胞とともにモザィ ク状の腫瘍組織を形成してがんの発生に寄与するものと考えられる。  [0016] V, not bound by any particular theory, but tumor cells fused with blood cells are transformed into highly malignant cancer cells that increase the tumor, and the phenotype of the cells It can be concluded that the cancer stem cells have been transformed. This process occurs in vivo, and tumor cells generated by transformation in vivo become cancer stem cells by cell fusion with immature blood cells such as hematopoietic stem cells, and together with surrounding tumor cells, mosaic cells It is thought to contribute to the development of cancer by forming a tumorous tissue.
[0017] 本発明のがん幹細胞を用いることにより、がん幹細胞の発生メカニズム及びがん幹 細胞が関与するがんの形成過程を詳細に研究することが可能になる。また、本発明 のがん幹細胞の生成方法において、被検物質の存在下で細胞融合を行い、細胞融 合の阻害程度を確認することにより、がん幹細胞の生成を阻害する物質をスクリー二 ングすることが可能になる。また、本発明のがん幹細胞のアポトーシスを誘導する物 質をスクリーニングすることもできる。このようにしてスクリーニングされた物質は、抗悪 性腫瘍剤として有用であることが期待される。さら〖こ、本発明により提供されるモデル 腫瘍組織又はモデル腫瘍動物は、がん幹細胞が存在しな ヽ腫瘍細胞単独の場合に 比べて、より現実の腫瘍に近い悪性度を再現していることから、がんの発生や成長、 又はがんの転移などのメカニズムの詳細な解明に極めて有用であり、インビボで有効 な抗腫瘍剤の開発に極めて有用である。 [0017] By using the cancer stem cells of the present invention, the cancer stem cell generation mechanism and the cancer stem It becomes possible to study the formation process of cancer involving cells in detail. In the cancer stem cell generation method of the present invention, cell fusion is performed in the presence of a test substance, and the degree of inhibition of cell fusion is confirmed, thereby screening a substance that inhibits the generation of cancer stem cells. It becomes possible to do. In addition, a substance that induces apoptosis of the cancer stem cell of the present invention can be screened. Substances screened in this way are expected to be useful as anti-cancer drugs. Furthermore, the model tumor tissue or animal model tumor provided by the present invention reproduces a malignancy closer to that of a real tumor than that of a tumor cell alone without cancer stem cells. Therefore, it is extremely useful for detailed elucidation of mechanisms such as cancer development and growth, or cancer metastasis, and is extremely useful for the development of antitumor agents effective in vivo.
実施例  Example
[0018] 例 1:試験管内における腫瘍細胞と血液細胞の細胞融合  [0018] Example 1: Cell fusion of tumor cells and blood cells in vitro
血液細胞と融合させる腫瘍細胞株としてはマウスメラノーマ細胞株である B16細胞 を用いた。 B16メラノーマ細胞は、 DMEM (シグマ社製)培地に 10%のゥシ血清を添カロ した通常の培地で継代した。 B16メラノーマ細胞に赤色の蛍光色素蛋白(DS-RED)を 発現できるプラスミド (ベタトン'ディッキンソン社製)をリポフエクタミン (インビトロジェン 社製)により遺伝子導入した。別途、グリーン 'フルオレセント 'プロテイン (GFP)を全身 に発現するマウス(green mouse)の骨髄力 CD libを細胞表面に発現する細胞(造 血幹細胞を含む細胞集団)をセルソーター(EPICS,コールター社製)を用いて採取し た。  B16 cells, which are mouse melanoma cell lines, were used as tumor cell lines to be fused with blood cells. B16 melanoma cells were subcultured in normal medium supplemented with 10% urine serum in DMEM (Sigma) medium. A plasmid capable of expressing red fluorescent chromoprotein (DS-RED) (Betaton's Dickinson) was introduced into B16 melanoma cells using lipofuctamine (Invitrogen). Separately, cells that express green 'fluorescent' protein (GFP) in the whole body (green mouse) bone marrow force CD lib (cell population including hematopoietic stem cells) on the cell surface are cell sorters (EPICS, Coulter) (Manufactured).
[0019] 104個の B16腫瘍細胞と 106個の CDllb陽性血液細胞とを 12 weU培養皿に同時に播 種し、 10%のゥシ血清を含む DMEM基本培地に stem cell factor (ギブコネ土製)及び M -CSF (ギブコネ土製)をそれぞれ最終濃度 20ng/ml及び lOng/mlとなるように添加した培 地を用いて共培養した。 37°Cで 5% CO 下に 1〜10日間共培養したところ、赤と緑の [0019] 10 4 B16 tumor cells and 10 6 CDllb-positive blood cells were seeded simultaneously in a 12 weU culture dish and stem cell factor (Gibconnet) made in DMEM basal medium containing 10% urine serum And M-CSF (manufactured by Gibconnet) were co-cultured using a medium to which final concentrations of 20 ng / ml and lOng / ml were added, respectively. When co-cultured at 37 ° C under 5% CO for 1-10 days, red and green
2  2
蛍光蛋白を同時に発現することにより黄色に染色された融合細胞の存在が認められ 、その割合は全細胞中 0.1-1.0%であった。赤と緑の蛍光を同時に発生する融合細 胞 (DS-RED陽性及び GFP陽性の融合細胞)をセルソーター(EPICS)を用いて分離し 、本発明のがん幹細胞を得た。 [0020] 例 2:がん幹細胞による腫瘍形成モデル The presence of fused cells stained yellow by co-expressing fluorescent protein was observed, and the ratio was 0.1-1.0% of all cells. Fusion cells (DS-RED positive and GFP positive fusion cells) that generate red and green fluorescence simultaneously were separated using a cell sorter (EPICS) to obtain the cancer stem cells of the present invention. [0020] Example 2: Tumor formation model with cancer stem cells
がん幹細胞による腫瘍形成の現場では、がん幹細胞とがん幹細胞力 分ィ匕したが ん細胞がモザイクとなっていると考えられる。がん幹細胞が存在することによってがん が個体内で生着してがん組織を形成すると考えられることから、例 1で得られたがん 幹細胞が、実際にがんの発生にプラスに働くことを以下の実験により確認した。 B16 腫瘍細胞 1.1 X 104個 (B16単独)、又は B16腫瘍細胞 1.0 X 104個と例 1で得たがん幹細 胞 0.1 X 104個との混合物をそれぞれ PBSに懸濁して、 C57B1/6系統マウスの背部皮 下に投与移植した。腫瘍形成の評価は、経時的に腫瘍のサイズを計測することにより 行なった。その結果を図 1に示す。 B16腫瘍細胞 1.1 X 104個を移植した個体では腫瘍 の形成が認められなかったのに対して、 0.1 X 104のがん幹細胞を混合した同量の B1 6腫瘍細胞を移植した個体には顕著な腫瘍形成が認められ、がん幹細胞を含む腫瘍 細胞が腫瘍形成能の高い細胞集団に変化することが確認された。 In the field of tumor formation by cancer stem cells, cancer stem cells and cancer stem cells are considered to be mosaics. The cancer stem cells obtained in Example 1 actually have a positive effect on the occurrence of cancer, because the cancer stem cells are thought to engraft in the individual to form cancer tissue. This was confirmed by the following experiment. A mixture of B16 tumor cells 1.1 x 10 4 (B16 alone) or B16 tumor cells 1.0 x 10 4 and cancer stem cells 0.1 x 10 4 obtained in Example 1 was suspended in PBS, respectively, and C57B1 / 6 strain mice were administered and transplanted under the back skin. Tumor formation was evaluated by measuring tumor size over time. The results are shown in Fig. 1. Tumor formation was not observed in individuals transplanted with 1.1 x 10 4 B16 tumor cells, whereas individuals transplanted with the same amount of B16 tumor cells mixed with 0.1 x 10 4 cancer stem cells. Significant tumor formation was observed, and it was confirmed that tumor cells including cancer stem cells changed to a cell population with high tumorigenicity.
[0021] また、例 1で得た DS-REDで蛍光標識した B16腫瘍細胞 1 X 106個(この細胞数はこ の腫瘍細胞が単独で腫瘍形成が可能な細胞数である)を PBSに懸濁し、 C57B1/6系 統の green mouseに移植し、形成された腫瘍中に GFPと DS-REDを同時に発現する腫 瘍細胞(宿主の green mouse由来の細胞と腫瘍細胞とが融合した細胞)が存在するか 否かを検討した。 DS- REDを発現する B16腫瘍細胞を移植後 14日目に腫瘍をマウス より取り出し、腫瘍組織を手術用ハサミで細切した後に、細切された腫瘍組織にディ スパーゼ(ベーリンガー社製)を添加して 10分間室温で放置し、その後、細胞懸濁液 を注射針をつけた注射器に充填し、数回の吸引と排出を繰り返して細胞を分散させ た。これらの細胞をフローサイトメトリー法により解析した。この結果、図 2(A)に示すよ うに、 DS-REDおよび GFPを同時に発現する融合細胞の存在が認められた。また、こ の融合細胞に対して c-Kitおよび CD133に対するモノクローナル抗体(いずれもファ 一ミンジェン社製)で染色してフローサイトメトリー法で解析すると、 c-Kitに関してはほ ぼ 100%、 CD133に関して 50-60%が融合細胞に発現していることが認められた(図 2( B) )。 c-Kitや AC133は未熟な幹細胞レベルの細胞に発現することが知られているこ とから、例 1で得られたがん幹細胞の生成過程が実際の腫瘍形成過程において生じ ていることが確認できた。 産業上の利用可能性 [0021] In addition, 1 × 10 6 B16 tumor cells fluorescently labeled with DS-RED obtained in Example 1 (this number of cells is the number of cells that can form tumors alone) in PBS. Suspended, transplanted into C57B1 / 6 lineage green mouse, and tumor cells that simultaneously express GFP and DS-RED in the formed tumor (cells derived from the host green mouse and tumor cells fused together) We examined whether or not there exists. 14 days after transplantation of B16 tumor cells expressing DS-RED, the tumor was removed from the mouse, and the tumor tissue was minced with surgical scissors, and then disperse (Boehringer) was added to the minced tumor tissue. The cells were allowed to stand at room temperature for 10 minutes, and then the cell suspension was filled into a syringe equipped with a syringe needle, and the cells were dispersed by repeating aspiration and discharge several times. These cells were analyzed by flow cytometry. As a result, as shown in FIG. 2 (A), the presence of fusion cells expressing both DS-RED and GFP was recognized. In addition, when this fused cell was stained with a monoclonal antibody against c-Kit and CD133 (both manufactured by Pharmingen) and analyzed by flow cytometry, c-Kit was almost 100% and CD133 was 50-60% was found to be expressed in the fused cells (Fig. 2 (B)). Since c-Kit and AC133 are known to be expressed in immature stem cell levels, it was confirmed that the cancer stem cell generation process obtained in Example 1 occurred in the actual tumor formation process. did it. Industrial applicability
本発明により、がん幹細胞を効率的に再現性よくインビトロで作成することができる 。このがん幹細胞を用いて、実際の腫瘍組織により近いモデル腫瘍組織をインビトロ で生成することができ、あるいはインビボで腫瘍の発生過程を再現した担がんモデル 動物を作成することができる。  According to the present invention, cancer stem cells can be efficiently generated with high reproducibility in vitro. Using these cancer stem cells, a model tumor tissue that is closer to the actual tumor tissue can be generated in vitro, or a tumor-bearing model animal that reproduces the tumor development process in vivo can be created.

Claims

請求の範囲 The scope of the claims
[1] がん幹細胞の作成方法であって、腫瘍細胞と血液細胞とをインビトロで細胞融合する 工程を含む方法。  [1] A method for producing cancer stem cells, comprising the step of in vitro cell fusion of tumor cells and blood cells.
[2] 造血幹細胞及び Z又は前駆細胞を含有する血液細胞と腫瘍細胞との共培養により 上記細胞融合を行なう請求項 1に記載の方法。  [2] The method according to [1], wherein the cell fusion is performed by co-culturing blood cells containing hematopoietic stem cells and Z or progenitor cells with tumor cells.
[3] 請求項 1に記載の方法により得ることができるがん幹細胞。 [3] A cancer stem cell obtainable by the method according to claim 1.
[4] モデル腫瘍組織の作成方法であって、腫瘍細胞と血液細胞とを細胞融合すること〖こ より得られるがん幹細胞及び上記腫瘍細胞の混合物を培養する工程を含む方法。  [4] A method for producing a model tumor tissue, comprising a step of culturing a cancer stem cell obtained by cell fusion of tumor cells and blood cells and a mixture of the tumor cells.
[5] 担がんモデル動物の作成方法であって、腫瘍細胞と血液細胞とを細胞融合すること により得られるがん幹細胞及び上記腫瘍細胞の混合物をヒト以外の哺乳類動物に投 与する工程を含む方法。  [5] A method for preparing a cancer-bearing model animal, comprising a step of administering a cancer stem cell obtained by cell fusion of tumor cells and blood cells and a mixture of the tumor cells to a mammal other than a human. Including methods.
[6] 請求項 5に記載の方法により得ることができる担がんモデル動物。  [6] A cancer-bearing model animal obtainable by the method according to claim 5.
PCT/JP2006/308560 2005-04-25 2006-04-24 Method of producing cancer stem cell WO2006115243A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005-126266 2005-04-25
JP2005126266A JP4752051B2 (en) 2005-04-25 2005-04-25 How to make cancer stem cells

Publications (1)

Publication Number Publication Date
WO2006115243A1 true WO2006115243A1 (en) 2006-11-02

Family

ID=37214857

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/308560 WO2006115243A1 (en) 2005-04-25 2006-04-24 Method of producing cancer stem cell

Country Status (2)

Country Link
JP (1) JP4752051B2 (en)
WO (1) WO2006115243A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009142271A1 (en) * 2008-05-21 2009-11-26 財団法人新産業創造研究機構 Cancer stem cell having high level of sld5 expression therein
CN113957052A (en) * 2020-07-21 2022-01-21 中南大学湘雅医院 Culture method and culture kit for primary acro-melanoma cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5894385A (en) * 1981-11-30 1983-06-04 Asahi Chem Ind Co Ltd Cell fusion and reagent for it

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5894385A (en) * 1981-11-30 1983-06-04 Asahi Chem Ind Co Ltd Cell fusion and reagent for it

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
AL-HAJJ M. ET AL.: "Prospective identification of tumorigenic breast cancer cell", PROC. NATL. ACAD. SCI. USA, vol. 100, no. 7, 2003, pages 3983 - 3988, XP002297672 *
BERGERS G. AND BENJAMIN L.E.: "TUMORIGENESIS AND THE ANGIOGENIC SWITCH", NAT. REV. CANCER, vol. 3, no. 6, 2003, pages 401 - 410, XP003001237 *
DUELLI D. AND LAZEBNIK Y.: "Cell fusion: A hidden enemy?", CANCER CELL, vol. 3, no. 5, May 2003 (2003-05-01), pages 445 - 448, XP003001238 *
RUBIO D. ET AL.: "Spontaneous Human Adult Stem Cell Transformation", CANCER RES., vol. 65, no. 8, 15 April 2005 (2005-04-15), pages 3035 - 3039, XP003001236 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009142271A1 (en) * 2008-05-21 2009-11-26 財団法人新産業創造研究機構 Cancer stem cell having high level of sld5 expression therein
JP5058336B2 (en) * 2008-05-21 2012-10-24 ジーン・ステム株式会社 Cancer stem cells that highly express SLD5
CN113957052A (en) * 2020-07-21 2022-01-21 中南大学湘雅医院 Culture method and culture kit for primary acro-melanoma cells
CN113957052B (en) * 2020-07-21 2023-08-01 中南大学湘雅医院 Culture method and culture kit for primary acromelama cells

Also Published As

Publication number Publication date
JP4752051B2 (en) 2011-08-17
JP2006296364A (en) 2006-11-02

Similar Documents

Publication Publication Date Title
EP3158056B1 (en) Single cell-derived organoids
Garraway et al. Human prostate sphere‐forming cells represent a subset of basal epithelial cells capable of glandular regeneration in vivo
Maier et al. Explant outgrowth, propagation and characterization of human pericytes
US11261426B2 (en) Pluripotent stem cell that can be isolated from body tissue
KR20070047850A (en) Hematopoietic differentiation of human embryonic stem cells
CN112080472A (en) Method for culturing human lung cancer organoid 3D model special for biomedical function research
TW200411059A (en) Dedifferentiated, programmable stem cells of monocytic origin, and their production and use
EP3645704A1 (en) Single lung cell-derived organoids
Bonde et al. Recent advances in hematopoietic stem cell biology
US11834680B2 (en) Single kidney cell-derived organoids
Schubbert et al. Methods for PTEN in stem cells and cancer stem cells
JP7148402B2 (en) Method for inducing differentiation of pluripotent stem cells in vitro
CN109152799A (en) Pancreatic stem cells and application thereof
CN102712897B (en) Heart tissue derived cell
WO2006117237A2 (en) Regeneration system, its production and use
WO2006115243A1 (en) Method of producing cancer stem cell
Yuzhakova et al. Highly invasive fluorescent/bioluminescent patient-derived orthotopic model of glioblastoma in mice
WO2021221179A1 (en) Establishment of mouse model using human pancreatic cancer organoid
CN107460170B (en) Establishment and application of human pituitary adenoma cell line
WO2003042375A1 (en) Stem cells originating in salivary duet epithelial cells and use thereof
US20080153160A1 (en) Human malignant cystosarcoma phyllodes derived mouse cell line and its applications
CN114317398B (en) Gli1 and EpCAM gene co-labeled hepatic progenitor cell population and application thereof
KR102660814B1 (en) Methods for improving hematopoietic transplants
JP2008148693A (en) Method for isolation of stem cell
Nanduri et al. Retrograde intra-ductal salivary gland stem cell transplantation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06732271

Country of ref document: EP

Kind code of ref document: A1