JP4653465B2 - Method for promoting natural killer cell proliferation and culture composition used therefor - Google Patents

Method for promoting natural killer cell proliferation and culture composition used therefor Download PDF

Info

Publication number
JP4653465B2
JP4653465B2 JP2004338152A JP2004338152A JP4653465B2 JP 4653465 B2 JP4653465 B2 JP 4653465B2 JP 2004338152 A JP2004338152 A JP 2004338152A JP 2004338152 A JP2004338152 A JP 2004338152A JP 4653465 B2 JP4653465 B2 JP 4653465B2
Authority
JP
Japan
Prior art keywords
cells
cell
natural killer
culture
glycosaminoglycan
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.)
Expired - Fee Related
Application number
JP2004338152A
Other languages
Japanese (ja)
Other versions
JP2006115826A (en
Inventor
泰信 小林
啓司 谷川
淳 有賀
Original Assignee
泰信 小林
啓司 谷川
淳 有賀
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 泰信 小林, 啓司 谷川, 淳 有賀 filed Critical 泰信 小林
Priority to JP2004338152A priority Critical patent/JP4653465B2/en
Publication of JP2006115826A publication Critical patent/JP2006115826A/en
Application granted granted Critical
Publication of JP4653465B2 publication Critical patent/JP4653465B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Description

この発明は、ヒト白血球中に存在するナチュラルキラー細胞(以下、NK細胞と略す場合もある)を生体外で培養する際、その増殖を促進させる方法に関するものである。  The present invention relates to a method for promoting the proliferation of natural killer cells (hereinafter sometimes abbreviated as NK cells) present in human leukocytes when cultured in vitro.

近年になって、生体の細胞性免疫機能を応用した細胞免疫療法が癌をはじめとする様々な疾患の治療や予防に適用されるようになってきた。特に癌治療の分野では、ここ十数年にわたり世界中で多くの臨床試験が実施され、今日では外科的療法、化学療法、放射線療法に次ぐ第4の治療法として認知されるようになっている。  In recent years, cellular immunotherapy that applies the cellular immune function of living bodies has been applied to the treatment and prevention of various diseases including cancer. Especially in the field of cancer treatment, many clinical trials have been conducted all over the world for the past decades, and today it is recognized as the fourth treatment after surgical therapy, chemotherapy and radiation therapy. .

癌を対象とした治療法を例にとると、細胞免疫療法は主に以下の3つの治療法に分類される。すなわちインターロイキン−2(以下IL−2と略す)などのサイトカインによって活性化されたリンパ球を治療に用いる「LAK療法」(例えば、非特許文献1参照)、抗原特異的な細胞傷害性活性を有するキラーT細胞治療に用いる「活性化リンパ球移入療法」(例えば、非特許文献2参照)、そして癌抗原に特異的な免疫反応を誘導する能力を持つ樹状細胞を応用した「樹状細胞ワクチン療法」(例えば、非特許文献3参照)である。  Taking cancer therapy as an example, cellular immunotherapy is mainly classified into the following three therapies. That is, “LAK therapy” (for example, refer to Non-Patent Document 1) using lymphocytes activated by cytokines such as interleukin-2 (hereinafter abbreviated as IL-2), antigen-specific cytotoxic activity. Applying “activated lymphocyte transfer therapy” (see, for example, Non-Patent Document 2) used for killer T cell therapy and dendritic cells capable of inducing an immune response specific to a cancer antigen “Vaccine therapy” (see, for example, Non-Patent Document 3).

このうち「LAK療法」は、体外に取り出したリンパ球分画をIL−2存在下で培養することにより、ナチュラルキラー細胞をはじめとするリンパ球の癌細胞に対する殺傷能力が顕著に高まる現象を利用する療法である。すなわち、LAK療法とは、末梢血から単核球分画(リンパ球+単球)もしくは単核球分画より単球を取り除いたリンパ球分画を調製し、その分画をIL−2を含有する培地で1〜2週間培養して抗腫瘍活性を強化させた後、この細胞を生体内に戻す療法であり、1980年代にRosenbergらにより担癌患者を対象とした臨床研究が開始されたのを端緒とし、現在でも世界中で広く実施されている(例えば、非特許文献4参照)。このように、IL−2存在下で培養され、癌細胞に対する殺傷能力が高まるように誘導された細胞をLAK細胞と呼ぶ。
Rosenberg SA,Lotze MT,Muul LM,Leitman S,Chang AE,Ettinghausen SE,Matory YL,Skibber JM,Shiloni E,Vetto JT.,N Engl J Med.1985 Dec 5;313(23):1485−92. Takayama T,Sekine T,Makuuchi M,Yamasaki S,Kosuge T,Yamamoto J,Shimada K,Sakamoto M,Hirohashi S,Ohashi Y,Kakizoe T.,Lancet.2000 Sep 2;356(9232):802−7. Nestle FO,Alijagic S,Gilliet M,Sun Y,Grabbe S,Dummer R,Burg G,Schadendorf D.,Nat Med.1998 Mar;4(3):328−32. Rosenberg SA.,Semin Oncol.1986 Jun;13(2):200−6.
Among these, “LAK therapy” utilizes a phenomenon in which the killing ability of lymphocytes including natural killer cells to cancer cells is remarkably increased by culturing the lymphocyte fraction taken out of the body in the presence of IL-2. It is a therapy to do. That is, LAK therapy refers to a mononuclear cell fraction (lymphocyte + monocyte) or a lymphocyte fraction obtained by removing monocytes from the mononuclear cell fraction from peripheral blood, and the fraction is treated with IL-2. This is a therapy in which the cells are cultured for 1 to 2 weeks to enhance the antitumor activity and then returned to the living body. In the 1980s, Rosenberg et al. Started clinical studies targeting cancer-bearing patients. It has been widely implemented all over the world even now (for example, see Non-Patent Document 4). Thus, a cell cultured in the presence of IL-2 and induced to increase the killing ability against cancer cells is referred to as a LAK cell.
Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, Matry YL, Skibber JM, Shiloni E, Vetto JT. N Engl J Med. 1985 Dec 5; 313 (23): 1485-92. Takayama T, Sekine T, Makuchi M, Yamazaki S, Kosuge T, Yamamoto J, Shimada K, Sakamoto M, Hirohashi S, Ohashi Y, Kahiko. Lancet. 2000 Sep 2; 356 (9232): 802-7. Nestle FO, Alijagic S, Gillet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadedorf D. et al. Nat Med. 1998 Mar; 4 (3): 328-32. Rosenberg SA. , Semin Oncol. 1986 Jun; 13 (2): 200-6.

しかしながら、上述した培養方法では、LAK細胞の効果の主体となるナチュラルキラー細胞が常に効率よく増殖するとは限らないことが知られている。  However, it is known that natural killer cells, which are the main effects of LAK cells, do not always efficiently proliferate in the culture method described above.

特に担癌患者ではナチュラルキラー細胞の培養が困難な症例がしばしば認められる。すなわち上記の一般的な培養方法で培養しても、ナチュラルキラー細胞の増殖がほとんど認められず、他の細胞、例えばCD56陽性、CD3陽性のT細胞の増殖が優勢となり、結果的にナチュラルキラー細胞をほとんど含まないLAK細胞が誘導されることになる。  Particularly in patients with cancer, cases in which natural killer cell culture is difficult are often observed. That is, even when cultivated by the above general culture method, almost no proliferation of natural killer cells was observed, and proliferation of other cells, for example, CD56 positive and CD3 positive T cells became dominant, resulting in natural killer cells. LAK cells that are almost free of sucrose are induced.

我々が、主に消化器癌の患者を対象として、常法にしたがってこれらの患者の末梢血リンパ球分画よりLAK細胞を誘導した結果、20例中11例では、健常人同様にナチュラルキラー細胞の増殖が旺盛なLAK細胞(ナチュラルキラー細胞の比率:平均20.4%)を誘導することができたが、20例中9例では、CD56陽性、CD3陽性のT細胞が優位となり、ナチュラルキラー細胞の増殖はほとんど認められなかった(ナチュラルキラー細胞の比率:平均2.2%)。このようなCD56陽性CD3陽性T細胞優位なLAK細胞は、ナチュラルキラー細胞優位なLAK細胞と比べて、癌細胞に対する傷害活性およびインターフェロンγやTNFαなどのサイトカイン産生能が著しく低い。したがってこのようなナチュラルキラー細胞の増殖がほとんど認められなかったLAK細胞を用いてもほとんど治療の効果が期待できない。  As a result of inducing LAK cells from peripheral blood lymphocyte fractions of these patients according to a conventional method mainly for patients with gastrointestinal cancer, in 11 cases out of 20 cases, natural killer cells as well as healthy people LAK cells (natural killer cell ratio: average 20.4%) could be induced, but in 9 out of 20 cases, CD56 positive and CD3 positive T cells were dominant, and natural killer Little cell proliferation was observed (natural killer cell ratio: average 2.2%). Such LAK cells predominantly CD56-positive CD3-positive T cells have significantly lower cytotoxic activity against cancer cells and the ability to produce cytokines such as interferon γ and TNFα than LAK cells predominantly natural killer cells. Therefore, even if LAK cells in which such natural killer cell growth is hardly observed are used, almost no therapeutic effect can be expected.

そこで、上述のようなナチュラルキラー細胞の増殖が困難な患者、つまり例えば常法通り培養しても抗腫瘍活性の低いCD56陽性CD3陽性T細胞が優位となるような患者においてもLAK療法の恩恵を享受することが可能となるような新たな細胞培養法、すなわち抗腫瘍活性の強いナチュラルキラー細胞を良好に増殖させることを可能とする新たなLAK細胞の培養方法の開発が待ち望まれている。さらに従来までの方法でナチュラルキラー細胞の増殖が良好な患者の場合でも、その増殖をさらに促進することで、LAK療法の効果もさらに高まることが期待される。  Therefore, the benefits of LAK therapy can be obtained even in patients who have difficulty in growing natural killer cells as described above, for example, patients in whom CD56-positive CD3-positive T cells with low antitumor activity are dominant even if cultured as usual. The development of a new cell culture method that can be enjoyed, that is, a new culture method of LAK cells that enables the natural killer cells with strong antitumor activity to grow well, is awaited. Further, even in the case of a patient who has a good natural killer cell proliferation by the conventional methods, it is expected that the effect of LAK therapy will be further enhanced by further promoting the proliferation.

一方、近年、リツキサンやハーセプチン等の腫瘍抗原特異的なモノクローナル抗体を主成分とする抗体医薬品が開発され治療に用いられているが、これらの医薬品の抗腫瘍作用の機序の一つがナチュラルキラー細胞による抗体依存性細胞傷害活性である事も明らかとなっている(Carson WE 他,Eur J Immunol.2001 Oct;31(10):3016−25.およびMaloney DG 他,Semin Oncol.2002 Feb;29(1 Suppl 2):2−9.)。したがって、より多くのナチュラルキラー細胞の増殖が可能となれば、LAK療法の有効性が高まることが期待されるのみでなく、上記のような抗体医薬品とLAK療法を併用することによって、その効果が相乗的に高まることが期待され、結果的に、さまざまな様相を呈する癌に対して、より有用な治療法を提供することが可能となると考えられる。  On the other hand, in recent years, antibody drugs mainly composed of monoclonal antibodies specific to tumor antigens such as Rituxan and Herceptin have been developed and used for treatment. One of the mechanisms of antitumor action of these drugs is natural killer cells. (Carson WE et al., Eur J Immunol. 2001 Oct; 31 (10): 3016-25. And Maloney DG et al., Semin Oncol. 2002 Feb; 29 (). 1 Suppl 2): 2-9.). Therefore, if more natural killer cells can be proliferated, not only the effectiveness of LAK therapy is expected to be increased, but also by using the antibody drug and LAK therapy in combination as described above, the effect is improved. It is expected to increase synergistically, and as a result, it will be possible to provide more useful treatments for cancers that exhibit various aspects.

そこで、本発明は、従来のLAK細胞の培養方法に比較して、ナチュラルキラー細胞の増殖が促進される培養方法、およびその培養方法に適する培養組成物を提供することを目的とする。  Therefore, an object of the present invention is to provide a culture method in which the growth of natural killer cells is promoted and a culture composition suitable for the culture method as compared with conventional LAK cell culture methods.

かかる問題を解決するために鋭意検討した結果、発明者らは、ナチュラルキラー細胞を含む細胞群を、ナチュラルキラー細胞が発現するCD56分子のリガンドを含む培地中で培養することによって、ナチュラルキラー細胞の増殖を選択的に促進させる事が可能であることを見出した。  As a result of diligent studies to solve such problems, the inventors have cultivated a group of cells containing natural killer cells in a medium containing a ligand of CD56 molecule expressed by natural killer cells, thereby It has been found that growth can be selectively promoted.

CD56分子は接着分子の一つである神経細胞接着分子(NCAM)のアイソフォームの一つであり、末梢血中では、ナチュラルキラー細胞や一部のT細胞のサブセットに発現することが知られていたが、これまでに、そのリガンドを加えることで、これらの細胞の増殖が促進されることはまったく知られていなかった。本発明に係る方法によれば、IL−2のみによる誘導ではナチュラルキラー細胞の増殖がほとんど認められず、CD56陽性CD3陽性T細胞等が優位となるような細胞群においても、ナチュラルキラー細胞の増殖が十分に促進される。また、もともとナチュラルキラー細胞の増殖が良好な細胞群の場合も、より多くのナチュラルキラー細胞を含有する細胞群を誘導することができる。  CD56 molecule is one of the adhesion forms of neural cell adhesion molecule (NCAM), which is one of adhesion molecules, and is known to be expressed in natural killer cells and a subset of some T cells in peripheral blood. However, it has never been known so far that the addition of the ligand promotes the growth of these cells. According to the method according to the present invention, natural killer cell proliferation is hardly observed by induction with IL-2 alone, and even in a cell group in which CD56-positive CD3-positive T cells and the like are dominant, proliferation of natural killer cells. Is fully promoted. Moreover, even in the case of a cell group in which natural killer cells proliferate originally, a cell group containing more natural killer cells can be induced.

ナチュラルキラー細胞を含む細胞群としては、例えば血液から得た単核球細胞やリンパ球分画が挙げられ、これらの細胞群は当業者であれば公知の方法に従って容易に調製することが可能である。
発明の一態様として、本発明は、ナチュラルキラー細胞を含む細胞群を培養する際に、CD56分子のリガンドの一つであるグリコサミノグリカン類を培地に添加することでナチュラルキラー細胞の増殖を顕著に促進する培養方法を提供するものである。
Examples of cell groups containing natural killer cells include mononuclear cells and lymphocyte fractions obtained from blood, and those cell groups can be easily prepared according to known methods by those skilled in the art. is there.
As one aspect of the invention, when culturing a cell group containing natural killer cells, the present invention increases the growth of natural killer cells by adding glycosaminoglycans, which are one of the ligands of the CD56 molecule, to the medium. It provides a culture method that significantly promotes.

発明に用いるグリコサミノグリカンとしては、例えば、ヘパリン、ヘパラン硫酸、コンドロイチン硫酸、デルマタン硫酸などを用いることができる。これらは、いずれも硫酸基を持つ糖が規則正しく配列した多糖類であり、また、いずれもCD56分子に結合するリガンドとしての機能を有している。  As the glycosaminoglycan used in the invention, for example, heparin, heparan sulfate, chondroitin sulfate, dermatan sulfate and the like can be used. These are polysaccharides in which sugars having sulfate groups are regularly arranged, and all have a function as a ligand that binds to the CD56 molecule.

これらのグリコサミノグリカンは、一部の医薬品グレードのものを含め、試薬として容易に入手することが可能である。これらは主に動物の皮膚や軟骨などから精製されたものであるが、その由来は特定されるものではない。また上記のような天然由来のもののみならず、各々のグリコサミノグリカンの構成糖を用いて化学的に合成された人工的なグリコサミノグリカンを用いることも可能である。  These glycosaminoglycans are readily available as reagents, including some pharmaceutical grades. These are mainly purified from animal skin, cartilage, etc., but their origin is not specified. It is also possible to use artificial glycosaminoglycans that are chemically synthesized using the constituent sugars of each glycosaminoglycan as well as those naturally derived from the above.

培養時に添加するグリコサミノグリカンの濃度は、特に限定されるものではないが、低濃度であれば十分な効果が期待できず、また濃度が高すぎても逆に細胞の生育に好ましくない作用を及ぼす可能性もあるため、例えばヘパリンの場合には最終濃度で0.1ユニット/mL〜1000ユニット/mLの範囲で、好ましくは0.5ユニット/mL〜500ユニット/mL、最も好ましくは1ユニット/mL〜100ユニット/mLとする。またコンドロイチン硫酸、ヘパラン硫酸、デルマタン硫酸の場合は、最終濃度で0.1〜1000μg/mLの範囲で、好ましくは0.5μg/mL〜500μg/mL、最も好ましくは1μg/mL〜300μg/mLの範囲とする。  The concentration of glycosaminoglycan added at the time of culture is not particularly limited, but if the concentration is low, a sufficient effect cannot be expected, and if the concentration is too high, it is not preferable for cell growth. For example, in the case of heparin, the final concentration is in the range of 0.1 unit / mL to 1000 unit / mL, preferably 0.5 unit / mL to 500 unit / mL, most preferably 1 Unit / mL to 100 units / mL. In the case of chondroitin sulfate, heparan sulfate, and dermatan sulfate, the final concentration is in the range of 0.1 to 1000 μg / mL, preferably 0.5 μg / mL to 500 μg / mL, and most preferably 1 μg / mL to 300 μg / mL. Range.

またこれらのグリコサミノグリカンは一種類のみ用いても、また二種以上の混合物として用いてもかまわない。  These glycosaminoglycans may be used alone or as a mixture of two or more.

さらにCD56分子のリガンドとしては、多糖体であるグリコサミノグリカンそのもののみならず、グリコサミノグリカンがコア蛋白質に結合した形態、すなわちグリコサミノグリカンを糖鎖の構成成分とするプロテオグリカンを用いることも可能である。  Furthermore, as a ligand for the CD56 molecule, not only the glycosaminoglycan which is a polysaccharide itself but also a form in which glycosaminoglycan is bound to a core protein, that is, a proteoglycan having glycosaminoglycan as a constituent component of a sugar chain is used. Is also possible.

発明の別の態様として、本発明は、細胞群を培養する際に、白血球単核球分画もしくはリンパ球分画を、細胞表面にヘパリン、ヘパラン硫酸、コンドロイチン硫酸やデルマタン硫酸などを構成成分とするプロテオグリカンを発現する別の細胞と共培養することで、ナチュラルキラー細胞の増殖を顕著に促進する培養方法を提供するものである。  As another aspect of the invention, when culturing a cell group, the present invention comprises a leukocyte mononuclear cell fraction or lymphocyte fraction, and heparin, heparan sulfate, chondroitin sulfate, dermatan sulfate, etc. The present invention provides a culture method that significantly promotes the growth of natural killer cells by co-culturing with another cell that expresses proteoglycan.

共培養に用いることができる細胞は、各グリコサミノグリカン類に対する抗体を用いて常法によりフローサイトメーターや免疫染色により検索することが可能である。また、ヘパリチナーゼ、コンドロイチナーゼ等の酵素で細胞表面グリコサミノグリカンを分解処理することでナチュラルキラー細胞の増殖促進作用が消失または減弱するような細胞も共培養に用いることができる。このような条件を満たす細胞としては、肝臓癌細胞株のHuH2(下記に具体例を示す)、骨髄性白血病細胞株であるK562、乳癌細胞株のMCF7、また胆嚢癌細胞株であるAGなどが挙げられるが、本発明に用いることのできる細胞はこれらの細胞株に限定されるものではない。  Cells that can be used for co-culture can be searched by a flow cytometer or immunostaining by an ordinary method using antibodies against each glycosaminoglycan. In addition, cells whose cell surface glycosaminoglycan is decomposed with an enzyme such as heparitinase, chondroitinase, etc., and the growth promoting action of natural killer cells disappears or attenuates can be used for co-culture. Examples of cells that satisfy such conditions include the liver cancer cell line HuH2 (specific examples are shown below), the myeloid leukemia cell line K562, the breast cancer cell line MCF7, and the gallbladder cancer cell line AG. Examples of cells that can be used in the present invention are not limited to these cell lines.

共培養に用いる細胞株とリンパ球との混合の比率は特定されないが、好ましくは1:1から1:100の範囲で、さらに好ましくは1:5から1:20の範囲で混合して培養することができる。  Although the mixing ratio of the cell line and lymphocyte used for co-culture is not specified, it is preferably mixed in the range of 1: 1 to 1: 100, more preferably in the range of 1: 5 to 1:20. be able to.

また共培養に用いる細胞株自体が培養時に増殖するような好ましくない事態を防ぐために、事前にその分裂増殖能を抑制する処置を施すことが好ましい。この処置は、細胞株を共培養に用いる前に、マイトマイシンCとともに培養したり、あるいは放射線を照射する等、当業者が一般的に用いる手法によって行う事ができる。  In addition, in order to prevent an unfavorable situation in which the cell line itself used for co-culture is proliferated at the time of culture, it is preferable to perform a treatment for suppressing its division and proliferation ability in advance. This treatment can be performed by techniques commonly used by those skilled in the art, such as culturing with mitomycin C or irradiating with radiation before using the cell line for co-culture.

さらに、本発明は、本発明を実施するための培養組成物をも提供する。  Furthermore, the present invention also provides a culture composition for carrying out the present invention.

この培養組成物は、リンパ球を培養するのに一般に用いられる培地を基本組成とし、そこにナチュラルキラー細胞の増殖を促す添加物としてグリコサミノグリカン、グリコサミノグリカンを構成成分とするプロテオグリカン、および/またはグリコサミノグリカンを構成成分とするプロテオグリカンを発現する細胞を添加したものとして提供される。  This culture composition is based on a medium generally used for culturing lymphocytes, and glycosaminoglycan as an additive for promoting the growth of natural killer cells, proteoglycan containing glycosaminoglycan as a constituent, And / or a cell to which proteoglycan expressing glycosaminoglycan as a constituent is added.

リンパ球を培養するのに用いられる標準的な培地には、RPMI−1640培地、AIM−V培地、X−VIVO培地などがある。よって基本組成となる基礎培地はこれらの標準的な培地をそのまま用いることもできるが、これらの培地に限定されるものでもなく、リンパ球培養が可能である培地であればどのような培地組成であってもかまわない。そして当該発明の培養組成物は、このような基本組成に、グリコサミノグリカンとして、ヘパリン、ヘパラン硫酸、コンドロイチン硫酸、デルマタン硫酸などを添加することで構成される。グリコサミノグリカンは、最終濃度でヘパリンでは0.1ユニット/mL〜1000ユニット/mL、好ましくは0.5ユニット/mL〜500ユニット/mL、最も好ましくは1ユニット/mL〜100ユニット/mLの濃度範囲で、またコンドロイチン硫酸、ヘパラン硫酸、デルマタン硫酸では0.1μg/mL〜1000μg/mL、好ましくは0.5μg/mL〜500μg/mL、最も好ましくは1μg/mL〜300μg/mLの濃度範囲で配合された形で提供される。Standard media used to culture lymphocytes include RPMI-1640 media, AIM-V media, X-VIVO media, and the like. Therefore, these basic media can be used as they are as the basic medium, but they are not limited to these media, and any medium composition can be used as long as it is capable of lymphocyte culture. It does not matter. And the culture composition of the said invention is comprised by adding heparin, heparan sulfate, chondroitin sulfate, dermatan sulfate etc. to such a basic composition as glycosaminoglycan. Glycosaminoglycan is 0.1 units / mL to 1000 units / mL, preferably 0.5 units / mL to 500 units / mL, most preferably 1 units / mL to 100 units / mL in heparin at final concentrations. In the concentration range, and in chondroitin sulfate, heparan sulfate, and dermatan sulfate, in the concentration range of 0.1 μg / mL to 1000 μg / mL, preferably 0.5 μg / mL to 500 μg / mL, most preferably 1 μg / mL to 300 μg / mL. Provided in blended form.

本発明に係る方法によれば、ナチュラルキラー細胞を含む細胞群を、CD56分子のリガンドを含む培地で培養することにより、ナチュラルキラー細胞の増殖を選択的に促進することができる。本方法によれば、IL−2によってナチュラルキラー細胞の増殖が誘導されなかった細胞群においても、効率よくナチュラルキラー細胞を増殖させることが可能となると共に、もともとIL−2によってナチュラルキラー細胞の増殖が誘導されやすい細胞群においても、より多くのナチュラルキラー細胞を含む培養物を得ることができる。このようにして得られたナチュラルキラー細胞を高い比率で含む細胞組成物を用いれば、LAK療法の効果も高められ、また、リツキサンやハーセプチン等の抗体医薬品とLAK療法の併用により、有用な治療法を提供することも可能となると考えられる。  According to the method of the present invention, the growth of natural killer cells can be selectively promoted by culturing a cell group containing natural killer cells in a medium containing a ligand for CD56 molecule. According to this method, it becomes possible to efficiently proliferate natural killer cells even in a cell group in which natural killer cell proliferation was not induced by IL-2. Even in a group of cells that are easily induced, a culture containing more natural killer cells can be obtained. If the cell composition containing the natural killer cells thus obtained in a high ratio is used, the effect of LAK therapy can be enhanced, and a useful therapeutic method can be obtained by combining LAK therapy with antibody drugs such as Rituxan and Herceptin. It will be possible to provide

以下に具体的な例を記載して発明の効果を説明する。The effects of the invention will be described below with specific examples.

健常人由来サンプルにおける、ヘパラン硫酸およびコンドロイチン硫酸A、デルマタン硫酸によるナチュラルキラー細胞(=NK細胞)の増殖の促進。  Promotion of natural killer cell (= NK cell) proliferation by heparan sulfate, chondroitin sulfate A, and dermatan sulfate in samples from healthy individuals.

健常人の末梢血30mLより、常法に従ってリンフォセパール(免疫生物研究所社製)を用いて60,000,000個の末梢血単核球細胞を得た。その後この細胞をAIM−V培地(インビトロジェン社製)に懸濁させて150cmの培養用フラスコに播種した後に37℃のCOインキュベーターで60分間培養し、その後浮遊している細胞を回収し、33,000,000個のリンパ球分画を得た。このリンパ球分画を、牛胎児血清10%、IL−2(1,000国際単位/mL〕を含むAIM−V培地に懸濁し、24ウェルの培養プレートに1,000,000個/1mL/ウェルとなるように播種した。またヘパラン硫酸(生化学工業(株)製)、コンドロイチン硫酸A(生化学工業製)あるいはデルマタン硫酸を最終濃度で100μg/mLとなるように培地に加えた。その後37℃のCOインキュベーターで培養し、培養7日目に各ウェルに牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地を1mL加えて細胞を懸濁した後にその細胞懸濁液を2ウェルに播きかえ、また培養11日目に同様の方法で2ウェルを4ウェルに播きかえて培養した。そして培養14日目に、細胞を回収して血球計算盤を用いて細胞数を計測するとともに、フローサイトメーターを用いてNK細胞(CD3陰性、CD56陽性、CD16陽性)の比率を計測し、NK細胞数を算出した。60,000,000 peripheral blood mononuclear cells were obtained from 30 mL of peripheral blood of healthy persons using Lymphosepar (manufactured by Immunobiological Laboratories) according to a conventional method. Thereafter, the cells are suspended in AIM-V medium (manufactured by Invitrogen), seeded in a 150 cm 2 culture flask, cultured in a CO 2 incubator at 37 ° C. for 60 minutes, and then the floating cells are collected. 33,000,000 lymphocyte fractions were obtained. This lymphocyte fraction was suspended in AIM-V medium containing 10% fetal bovine serum and IL-2 (1,000 international units / mL), and 1,000,000 cells / 1 mL / mL were cultured in a 24-well culture plate. Heparan sulfate (manufactured by Seikagaku Corporation), chondroitin sulfate A (manufactured by Seikagaku Corporation) or dermatan sulfate was added to the medium to a final concentration of 100 μg / mL. Cultivate in a CO 2 incubator at 37 ° C., and on day 7 of culture, add 1 mL of AIM-V medium containing 10% fetal calf serum and IL-2 (1,000 international units / mL) to each well to suspend the cells. After that, the cell suspension was seeded in 2 wells and cultured in the same manner on day 11 of culture, with 2 wells in place of 4 wells, and on day 14 of culture, cells were collected and subjected to hemocytometer. Board As well as measuring the number of cells used, NK cells (CD3-negative, CD56-positive, CD16-positive) the ratio of measured using the flow cytometer was calculated number NK cells.

その結果を表1に示した。培養開始前の時点、すなわち末梢血リンパ球分画中に8.5%であったNK細胞が、培地中に最終濃度で100μg/mLのヘパラン硫酸、コンドロイチン硫酸Aあるいはデルマタン硫酸を加えて2週間培養することで、各々48.2%、24.2%、49.2%となり、またNK細胞数も培養開始時の各々72.5倍、23.5倍、112.7倍となった。
一方、比較例として、グリコサミノグリカンを加えない条件で2週間培養した場合は、NK細胞の比率は17・0%までしか増加せず、細胞数も16.5倍に留まった。

Figure 0004653465
The results are shown in Table 1. NK cells that had been 8.5% in the peripheral blood lymphocyte fraction at the time prior to the start of culture were added 100 μg / mL heparan sulfate, chondroitin sulfate A or dermatan sulfate in the medium for 2 weeks. By culturing, the numbers were 48.2%, 24.2%, and 49.2%, respectively, and the NK cell numbers were 72.5 times, 23.5 times, and 112.7 times, respectively, at the start of the culture.
On the other hand, as a comparative example, when cultured for 2 weeks under the condition where glycosaminoglycan was not added, the ratio of NK cells increased only to 17.0%, and the number of cells remained at 16.5 times.
Figure 0004653465

健常人由来サンプルにおけるヘパリンによるNK細胞の増殖の促進。Promotion of proliferation of NK cells by heparin in samples from healthy individuals.

健常人の末梢血20mLより、常法に従ってリンフォセパールを用いて血単核球細胞を調製し、その後この細胞をAIM−V培地に懸濁して75cmの培養用フラスコに播種した後に37℃のCOインキュベーターで60分間培養し、その後浮遊している細胞を回収し、13,000,000個のリンパ球分画を得た。このリンパ球分画を、牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地に懸濁し、24ウェルの培養プレートに1,000,000個/1mL/ウェルとなるように播種した。またヘパリン(持田製薬製)を最終濃度で5ユニット/mLとなるように加えた。その後37℃のCOインキュベーターで培養し、培養7日目に各ウェルに牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地を1mL加えて細胞を懸濁した後にその細胞懸濁液を2ウェルに播きかえ、また培養11日目に同様の方法で2ウェルを4ウェルに播きかえて培養した。そして培養14日目に、細胞を回収して血球計算盤を用いて細胞数を計測するとともに、フローサイトメーターを用いてNK細胞(CD3陰性、CD56陽性、CD16陽性)の比率を計測し、NK細胞数を算出した。Blood mononuclear cells were prepared from 20 mL of normal human peripheral blood using Lymphosepar according to a conventional method, and the cells were then suspended in AIM-V medium and seeded in a 75 cm 2 culture flask. The cells were cultured in a 2 incubator for 60 minutes, and then the floating cells were collected to obtain 13,000,000 lymphocyte fractions. This lymphocyte fraction was suspended in AIM-V medium containing 10% fetal bovine serum and IL-2 (1,000 international units / mL), and 1,000,000 cells / 1 mL / mL were cultured in a 24-well culture plate. It seed | inoculated so that it might become a well. Heparin (Mochida Pharmaceutical) was added to a final concentration of 5 units / mL. Thereafter, the cells were cultured in a CO 2 incubator at 37 ° C., and on the seventh day of the culture, 1 mL of AIM-V medium containing 10% fetal calf serum and IL-2 (1,000 international units / mL) was added to each well, and the cells were suspended. After turbidity, the cell suspension was seeded in 2 wells, and on the 11th day of culture, 2 wells were seeded in 4 wells and cultured. On the 14th day of culture, the cells were collected and the number of cells was counted using a hemocytometer, and the ratio of NK cells (CD3 negative, CD56 positive, CD16 positive) was measured using a flow cytometer. Cell number was calculated.

その結果を表2に示した。培養開始前の時点、すなわち末梢血リンパ球分画中に4.1%であったNK細胞が、培地中に最終濃度で5ユニット/mLのヘパリンを加えて2週間培養することで、11.6%となり、NK細胞数も培養開始時の21.6倍となった。
一方、比較例として、ヘパリンを加えない条件で2週間培養したところ、NK細胞の比率は5.5%までしか増加せず、細胞数も10.2倍に留まった。

Figure 0004653465
The results are shown in Table 2. NK cells that were 4.1% in the time before the start of culture, that is, peripheral blood lymphocyte fraction, were cultured for 2 weeks by adding 5 units / mL heparin at a final concentration in the medium. The number of NK cells was 21.6 times that at the start of culture.
On the other hand, as a comparative example, when cultured for 2 weeks without adding heparin, the ratio of NK cells increased only to 5.5%, and the number of cells remained 10.2 times.
Figure 0004653465

担癌患者由来サンプルにおけるヘパリンによるNK細胞の選択的な増殖の促進。Promotion of selective proliferation of NK cells by heparin in samples from patients with cancer.

肝臓癌の患者末梢血より調製したリンパ球分画を、24ウェルの培養プレートに、牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地に懸濁し、1,000,000個/1mL/ウェルとなるように播種した。またヘパリン(持田製薬(株)製)を最終濃度で5ユニットmL(5単位/mL)となるように加えた。その後37℃のCOインキュベーターで培養し、培養7日目に各ウェルに牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地を1mL加えて細胞を懸濁した後にその細胞懸濁液を2ウェルに播きかえ、また培養11日目に同様の方法で2ウェルを4ウェルに播きかえて培養した。そして培養14日目に、細胞を回収して血球計算盤を用いて細胞数を計測するとともに、フローサイトメーターを用いてNK細胞(CD3陰性、CD56陽性、CD16陽性)ならびにCD56陽性T細胞(CD3陽性、CD56陽性、CD16陰性)の比率を計測し、各々の細胞数を算出した。The lymphocyte fraction prepared from the peripheral blood of a patient with liver cancer was suspended in an AIM-V medium containing fetal bovine serum 10%, IL-2 (1,000 international units / mL) in a 24-well culture plate, It seed | inoculated so that it might become 1,000,000 piece / 1mL / well. Heparin (manufactured by Mochida Pharmaceutical Co., Ltd.) was added to a final concentration of 5 units mL (5 units / mL). Thereafter, the cells were cultured in a CO 2 incubator at 37 ° C., and on the seventh day of the culture, 1 mL of AIM-V medium containing 10% fetal calf serum and IL-2 (1,000 international units / mL) was added to each well, and the cells were suspended. After turbidity, the cell suspension was seeded in 2 wells, and on the 11th day of culture, 2 wells were seeded in 4 wells and cultured. On the 14th day of culture, the cells were collected and counted using a hemocytometer, and NK cells (CD3 negative, CD56 positive, CD16 positive) and CD56 positive T cells (CD3) were measured using a flow cytometer. (Positive, CD56 positive, CD16 negative) were measured, and the number of each cell was calculated.

その結果を表3に示した。培養開始前の時点、すなわち末梢血リンパ球分画中に5.0%であったNK細胞が、培地中に最終濃度で5ユニット/mLのヘパリンを加えて2週間培養することで、27.9%となり、またNK細胞数も培養開始時の30.0倍となった。
比較例として、ヘパリンを加えない条件で2週間培養したところ、NK細胞の比率は16.5%までしか増加せず、細胞数も16.6倍に留まった。
一方、CD56陽性T細胞は、末梢血リンパ球分画中に11.0%であり、ヘパリンを加えて培養した場合は32.5%(細胞数15.7倍)となり、ヘパリンを加えずに培養した場合は約44.7%(細胞数20.5倍)であった。
以上の結果から、ヘパリンはNK細胞の増殖を促進する作用を有するが、CD56陽性T細胞の増殖には逆に抑制的に機能する作用を有することが明らかとなった。

Figure 0004653465
The results are shown in Table 3. NK cells, which were 5.0% in the peripheral blood lymphocyte fraction before the start of culture, were cultured for 2 weeks by adding 5 units / mL heparin at a final concentration in the medium. The number of NK cells was 30.0 times that at the start of culture.
As a comparative example, when cultured for 2 weeks under the condition where heparin was not added, the ratio of NK cells increased only to 16.5%, and the number of cells remained at 16.6 times.
On the other hand, CD56-positive T cells account for 11.0% in the peripheral blood lymphocyte fraction, and 32.5% (15.7 times the number of cells) when cultured with heparin, without adding heparin. When cultured, it was about 44.7% (20.5 times the number of cells).
From the above results, it was clarified that heparin has an action of promoting the proliferation of NK cells, but has an action of functioning suppressively on the proliferation of CD56-positive T cells.
Figure 0004653465

IL−2によりCD56陽性T細胞が優位となる担癌患者由来サンプルにおける、コンドロイチン硫酸Aおよびデルマタン硫酸によるNK細胞の選択的な増殖の促進。Promotion of selective proliferation of NK cells by chondroitin sulfate A and dermatan sulfate in a sample derived from a cancer-bearing patient in which CD56-positive T cells are predominant by IL-2.

胃癌患者の末梢血より調製したリンパ球分画を、24ウェルの培養プレートに、牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地に懸濁し、1,000,000個/1mL/ウェルとなるように播種した。またコンドロイチン硫酸Aあるいはデルマタン硫酸を最終濃度で100μg/mLとなるように培地に加えた。その後37℃のCOインキュベーターで培養し、培養7日目に各ウェルに牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地を1mL加えて細胞を懸濁した後にその細胞懸濁液を2ウェルに播きかえ、また培養11日目に同様の方法で2ウェルを4ウェルに播きかえて培養した。そして培養14日目に、細胞を回収して血球計算盤を用いて細胞数を計測するとともに、フローサイトメーターを用いてNK細胞(CD3陰性、CD56陽性、CD16陽性)の比率ならびにCD3陽性、CD56陽性細胞の比率を計測し、NK細胞数およびCD56陽性T細胞数を算出した。Lymphocyte fractions prepared from peripheral blood of gastric cancer patients are suspended in AIM-V medium containing fetal bovine serum 10%, IL-2 (1,000 international units / mL) in a 24-well culture plate. , 000,000 cells / 1 mL / well. In addition, chondroitin sulfate A or dermatan sulfate was added to the medium so as to have a final concentration of 100 μg / mL. Thereafter, the cells were cultured in a CO 2 incubator at 37 ° C., and on the seventh day of the culture, 1 mL of AIM-V medium containing 10% fetal calf serum and IL-2 (1,000 international units / mL) was added to each well, and the cells were suspended. After turbidity, the cell suspension was seeded in 2 wells, and on the 11th day of culture, 2 wells were seeded in 4 wells and cultured. On the 14th day of culture, the cells were collected and the number of cells was counted using a hemocytometer, and the ratio of NK cells (CD3 negative, CD56 positive, CD16 positive) and CD3 positive, CD56 were measured using a flow cytometer. The ratio of positive cells was measured, and the number of NK cells and the number of CD56 positive T cells were calculated.

その結果を表4に示した。培養開始前の時点、すなわち末梢血リンパ球分画中に1.3%であったNK細胞は、グリコサミノグリカンを加えない条件で2週間培養したところ、細胞数は4.5倍に増加したものの、その比率は0.5%に低下した。しかしながら培地中に最終濃度で100μg/mLのコンドロイチン硫酸Aもしくはデルマタン硫酸を加えて2週間培養すると、NK細胞の比率は各々2.4%、2.3%となり、またNK細胞数も各々培養開始時の26.1倍および26.3倍となり、NK細胞の増殖が顕著に促進されることが明らかとなった。
一方、末梢血リンパ球分画中に1.2%であったCD56陽性T細胞数は、グリコサミノグリカンを加えない条件で2週間培養することで、細胞数は420.8倍に増加し、その比率は約42.2%となった。一方、100μg/mLのコンドロイチン硫酸Aもしくはデルマタン硫酸存在下で2週間培養した場合も、CD56陽性T細胞の比率は各々36.9%および39.8%となり、また細胞数も各々培養開始時の458.3倍および521.8倍であり、CD56陽性T細胞数の増殖はグリコサミノグリカンを加えない場合とほぼ同程度であった。
以上の結果から、コンドロイチン硫酸Aもしくはデルマタン硫酸の添加によってNK細胞の増殖が選択的に促進されることが確認された。

Figure 0004653465
The results are shown in Table 4. NK cells, which were 1.3% in the peripheral blood lymphocyte fraction at the time before the start of culture, were cultured for 2 weeks under the condition where glycosaminoglycan was not added, the number of cells increased 4.5 times. However, the ratio fell to 0.5%. However, when 100 μg / mL of chondroitin sulfate A or dermatan sulfate was added to the medium at the final concentration and cultured for 2 weeks, the ratio of NK cells was 2.4% and 2.3%, respectively, and the number of NK cells was also started. It was revealed that the proliferation of NK cells was significantly promoted by 26.1 times and 26.3 times the time.
On the other hand, the number of CD56-positive T cells, which was 1.2% in the peripheral blood lymphocyte fraction, increased to 420.8 times by culturing for 2 weeks under the condition where glycosaminoglycan was not added. The ratio was about 42.2%. On the other hand, when cultured for 2 weeks in the presence of 100 μg / mL of chondroitin sulfate A or dermatan sulfate, the ratios of CD56-positive T cells were 36.9% and 39.8%, respectively, and the number of cells was also the same as at the beginning of the culture. 458.3 times and 521.8 times, and the proliferation of CD56 positive T cells was almost the same as when no glycosaminoglycan was added.
From the above results, it was confirmed that the addition of chondroitin sulfate A or dermatan sulfate selectively promotes the growth of NK cells.
Figure 0004653465

健常人由来サンプルにおけるグリコサミノグリカン発現細胞との共培養によるNK細胞の選択的な増殖の促進。Promotion of selective proliferation of NK cells by co-culture with glycosaminoglycan-expressing cells in samples from healthy individuals.

健常人の末梢血より調製したリンパ球分画を、24ウェルの培養プレートに、牛胎児血清10%、IL−2(1,000国際単位/mL)を含むAIM−V培地に懸濁し、2,000,000個/2mL/ウェルとなるように播種した。また、予め最終濃度で50μg/mlのマイトマイシンC存在下で30分培養してその分裂増殖能を停止させたHuH2細胞株(肝臓癌細胞株)を、200,000個/2mL/ウェルとなるように播種した(リンパ球分画細胞数:HuH2細胞数=10:1)。また一部のHuH2細胞は、事前にリン酸緩衝生理食塩水中にて最終濃度で10mU/mLのヘパリチナーゼI(生化学工業製)、で37℃、4時間酵素処理を行うことで細胞表面のグリコサミノグリカンを消化し、酵素処理を行わなかった細胞を用いた場合と比較した。The lymphocyte fraction prepared from the peripheral blood of a healthy person is suspended in an AIM-V medium containing fetal bovine serum 10% and IL-2 (1,000 international units / mL) in a 24-well culture plate. , 000,000 pieces / 2 mL / well. In addition, a HuH2 cell line (liver cancer cell line) that has been cultured for 30 minutes in the presence of mitomycin C at a final concentration of 30 μm in advance to stop its division and proliferation ability to become 200,000 cells / 2 mL / well. (Number of lymphocyte fractional cells: number of HuH2 cells = 10: 1). In addition, some HuH2 cells were previously subjected to enzyme treatment at 37 ° C. for 4 hours in phosphate buffered saline with 10 mU / mL heparitinase I (manufactured by Seikagaku Corporation) at a final concentration of glycoproteins on the cell surface. It was compared with the case of using cells that had been digested with saminoglycan and not subjected to enzyme treatment.

その結果を表5に示した。培養開始前の時点、すなわち末梢血リンパ球分画中に6.8%であったNK細胞は、IL−2存在下で7日間培養したところ、NK細胞の比率は10.4%、細胞数は1.9倍に増加したが、HuH2細胞と共培養することでNK細胞の比率は14.5%、細胞数は培養開始時の3.7倍となり、NK細胞の増殖が顕著に促進されることが明らかとなった。一方、事前にヘパリチナーゼIで前処理を行なったHuH2細胞と共培養すると、NK細胞の比率は11.9%となり、またNK細胞数も培養開始時の2.5倍となり、ヘパリチナーゼIで細胞表面のグリコサミノグリカンを消化したHuH2細胞では、無処置のHuH2細胞と比較してNK細胞の増殖を促進する作用の顕著な低下が認められた。
以上の結果から、HuH2細胞との共培養によりNK細胞の増殖が選択的に促進されること、そしてそのNK細胞の増殖促進作用がHuH2細胞の発現するグリコサミノグリカンを介した作用であることが確認された。

Figure 0004653465
The results are shown in Table 5. NK cells that had been 6.8% in the peripheral blood lymphocyte fraction before the start of culture were cultured in the presence of IL-2 for 7 days. The NK cell ratio was 10.4%, the number of cells. Increased by 1.9 times, but by co-culture with HuH2 cells, the ratio of NK cells was 14.5%, the number of cells was 3.7 times that at the start of culture, and the proliferation of NK cells was significantly promoted. It became clear. On the other hand, when co-cultured with HuH2 cells pretreated with heparitinase I in advance, the ratio of NK cells was 11.9%, and the number of NK cells was 2.5 times that at the start of the culture. In the HuH2 cells digested with the glycosaminoglycan, a significant decrease in the effect of promoting the proliferation of NK cells was observed as compared with the untreated HuH2 cells.
From the above results, the proliferation of NK cells is selectively promoted by co-culture with HuH2 cells, and the proliferation promoting action of NK cells is an action via glycosaminoglycan expressed by HuH2 cells. Was confirmed.
Figure 0004653465

発明の産業上の利用分野Industrial use of the invention

本発明は、ヒト白血球中に存在するナチュラルキラー細胞を生体外で培養して増殖させる方法に関するものである。本発明に記載された方法で培養されたナチュラルキラー細胞は、癌やウイルス性疾患等の治療および/または予防のための細胞免疫療法に用いることができる。  The present invention relates to a method for culturing and expanding natural killer cells present in human leukocytes in vitro. Natural killer cells cultured by the method described in the present invention can be used for cell immunotherapy for the treatment and / or prevention of cancer, viral diseases and the like.

Claims (10)

ナチュラルキラー細胞の増殖を促進させる方法であって、
ナチュラルキラー細胞を含む細胞群を、以下(a)〜(c)の少なくとも一つを含む培地中で培養する工程を含む、方法。
(a)ヘパリン、コンドロイチン硫酸、ヘパラン硫酸、デルマタン硫酸からなる群より選択される一以上の物質を含むグリコサミノグリカン。
(b)前記(a)のグリコサミノグリカンを構成成分とするプロテオグリカン。
(c)前記(a)のグリコサミノグリカン、及び、前記(a)のグリコサミノグリカンを構成成分とするプロテオグリカン、の少なくも一つを発現する細胞。
A method of promoting the growth of natural killer cells,
A method comprising culturing a cell group containing natural killer cells in a medium containing at least one of the following (a) to (c) :
(A) A glycosaminoglycan containing one or more substances selected from the group consisting of heparin, chondroitin sulfate, heparan sulfate, and dermatan sulfate.
(B) A proteoglycan comprising the glycosaminoglycan of (a) as a constituent component.
(C) A cell that expresses at least one of the glycosaminoglycan of (a) and the proteoglycan having the glycosaminoglycan of (a) as a constituent component.
前記細胞群が、血液から得た単核球細胞またはリンパ球分画である、請求項1に記載の方法。   The method according to claim 1, wherein the cell group is a mononuclear cell or lymphocyte fraction obtained from blood. 前記培地が、さらにインターロイキン2を含む、請求項1または2に記載の方法。   The method according to claim 1 or 2, wherein the medium further contains interleukin-2. 前記(c)の細胞が、ヘパリチナーゼ処理またはコンドロイチナーゼ処理によって、そのナチュラルキラー細胞の増殖促進能が低下する細胞である、請求項1から3のいずれか1項に記載の方法。 The method according to any one of claims 1 to 3, wherein the cell of (c) is a cell whose proliferation promoting ability of natural killer cells is reduced by heparitinase treatment or chondroitinase treatment. 前記細胞群が、癌患者由来のものである、請求項1から4のいずれか1項に記載の方法。The method according to any one of claims 1 to 4, wherein the cell group is derived from a cancer patient. 以下(a)〜(c)の少なくとも一つを含むナチュラルキラー細胞の増殖を促進させる培養組成物:
(a)ヘパリン、コンドロイチン硫酸、ヘパラン硫酸、デルマタン硫酸からなる群より選択される一以上の物質を含むグリコサミノグリカン。
(b)前記(a)のグリコサミノグリカンを構成成分とするプロテオグリカン。
(c)前記(a)のグリコサミノグリカン、及び、前記(a)のグリコサミノグリカンを構成成分とするプロテオグリカン、の少なくも一つを発現する細胞。
A culture composition for promoting the growth of natural killer cells comprising at least one of the following (a) to (c):
(A) A glycosaminoglycan containing one or more substances selected from the group consisting of heparin, chondroitin sulfate, heparan sulfate, and dermatan sulfate .
(B) A proteoglycan comprising the glycosaminoglycan of (a) as a constituent component.
(C) A cell that expresses at least one of the glycosaminoglycan of (a) and the proteoglycan having the glycosaminoglycan of (a) as a constituent component .
前記ナチュラルキラー細胞が、血液から得た単核球細胞またはリンパ球分画である細胞群に含まれるものである、請求項6に記載の培養組成物。The culture composition according to claim 6, wherein the natural killer cell is contained in a cell group that is a mononuclear cell or lymphocyte fraction obtained from blood. さらにインターロイキン2を含む、請求項6または7に記載の培養組成物。The culture composition according to claim 6 or 7, further comprising interleukin-2. 前記(c)の細胞が、ヘパリチナーゼ処理またはコンドロイチナーゼ処理によって、そのナチュラルキラー細胞の増殖促進能が低下する細胞である、請求項6から8のいずれか1項に記載の培養組成物。The culture composition according to any one of claims 6 to 8, wherein the cell (c) is a cell whose natural killer cell proliferation promoting ability is reduced by heparitinase treatment or chondroitinase treatment. 前記ナチュラルキラー細胞が、癌患者由来のものである、請求項6から9のいずれか1項に記載の培養組成物。The culture composition according to any one of claims 6 to 9, wherein the natural killer cell is derived from a cancer patient.
JP2004338152A 2004-10-25 2004-10-25 Method for promoting natural killer cell proliferation and culture composition used therefor Expired - Fee Related JP4653465B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2004338152A JP4653465B2 (en) 2004-10-25 2004-10-25 Method for promoting natural killer cell proliferation and culture composition used therefor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2004338152A JP4653465B2 (en) 2004-10-25 2004-10-25 Method for promoting natural killer cell proliferation and culture composition used therefor

Publications (2)

Publication Number Publication Date
JP2006115826A JP2006115826A (en) 2006-05-11
JP4653465B2 true JP4653465B2 (en) 2011-03-16

Family

ID=36534293

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2004338152A Expired - Fee Related JP4653465B2 (en) 2004-10-25 2004-10-25 Method for promoting natural killer cell proliferation and culture composition used therefor

Country Status (1)

Country Link
JP (1) JP4653465B2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19836339B4 (en) 1998-08-11 2011-12-22 N.V. Nutricia carbohydrate mix
EP1597978A1 (en) 2004-05-17 2005-11-23 Nutricia N.V. Synergism of GOS and polyfructose
US8252769B2 (en) 2004-06-22 2012-08-28 N. V. Nutricia Intestinal barrier integrity
WO2009096772A1 (en) * 2008-02-01 2009-08-06 N.V. Nutricia Composition for stimulating natural killer cell activity
KR20120093002A (en) 2011-02-14 2012-08-22 (주)에이티젠 Method for diagnosing cancer and diagnosis kit using measurement of nk cell activity
JP5856025B2 (en) * 2012-08-02 2016-02-09 阿部 博幸 Methods for obtaining monocytes or NK cells

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6176420A (en) * 1984-09-10 1986-04-18 ピー、エフ、メデイカマン Membrane polysaccharide and manufacture
JP2003530826A (en) * 1999-11-17 2003-10-21 ユニバーシティー オブ ロチェスター Human ex vivo immune system
WO2004061090A1 (en) * 2002-12-27 2004-07-22 Sanwa Shurui Co., Ltd. Composition capable of activating natural killer cells and process for producing the same, composition containing natural killer cells activated by the former composition and process for producing the same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6176420A (en) * 1984-09-10 1986-04-18 ピー、エフ、メデイカマン Membrane polysaccharide and manufacture
JP2003530826A (en) * 1999-11-17 2003-10-21 ユニバーシティー オブ ロチェスター Human ex vivo immune system
WO2004061090A1 (en) * 2002-12-27 2004-07-22 Sanwa Shurui Co., Ltd. Composition capable of activating natural killer cells and process for producing the same, composition containing natural killer cells activated by the former composition and process for producing the same

Also Published As

Publication number Publication date
JP2006115826A (en) 2006-05-11

Similar Documents

Publication Publication Date Title
Lupo et al. Natural killer cells as allogeneic effectors in adoptive cancer immunotherapy
Lee et al. Haploidentical natural killer cells infused before allogeneic stem cell transplantation for myeloid malignancies: a phase I trial
CN102597223B (en) Process for production of natural killer cells
TWI612137B (en) Method for producing composition containing immune cells and composition for treating cancer
Alexia et al. Polyoxidonium® activates cytotoxic lymphocyte responses through dendritic cell maturation: clinical effects in breast cancer
Song et al. IL‐12/IL‐18‐preactivated donor NK cells enhance GVL effects and mitigate GvHD after allogeneic hematopoietic stem cell transplantation
JP7268039B2 (en) Methods of producing natural killer cells and compositions for cancer therapy
US20130157364A1 (en) Medium composition for culturing self-activated lymphocytes and method for culturing self-activated lymphocytes using same
WO2012099093A1 (en) Process for production of nk-cell-enriched blood preparation
US20220333078A1 (en) Method for ex vivo expansion CD34+HSPCs into NK cells using an aryl hydrocarbon receptor antagonist
Suck et al. Emerging natural killer cell immunotherapies: large-scale ex vivo production of highly potent anticancer effectors
KR20090127973A (en) A method for cultivating self activated lymphocyte
EA038848B1 (en) Method for proliferating natural killer cells and compositions for proliferating natural killer cells
Torelli et al. Expansion of natural killer cells with lytic activity against autologous blasts from adult and pediatric acute lymphoid leukemia patients in complete hematologic remission
JP4275680B2 (en) Culture methods for lymphocyte activity / proliferation
JP2017012010A (en) Method of growing natural killer cells and composition for growing natural killer cells
WO2013167136A1 (en) Improving adoptive cell therapy with interferon gamma
JP4653465B2 (en) Method for promoting natural killer cell proliferation and culture composition used therefor
CN109439627B (en) Method and composition for polarizing and amplifying CD4+ T cells and application of composition in curing tumors expressing specific antigens
CN114058580A (en) Method for in vitro proliferation of natural killer cells and natural killer T cells
JP6782503B1 (en) A method for producing a cell composition for treating cancer, a cell composition for treating cancer produced thereby, and a method for treating cancer using the cell composition for treating cancer.
CN110747167B (en) Preparation method and application of hemizygous BAK cell
Lee et al. Tumor necrosis factor-α enhances IL-15-induced natural killer cell differentiation
Pan et al. Natural Killer Cells at the Forefront of Cancer Immunotherapy with Immune Potency, Genetic Engineering, and Nanotechnology
JP4106488B2 (en) Use of stem cells and CD6-depleted stem cells for induction of immune tolerance against allografts and / or treatment of leukemia

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20071018

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20100907

A521 Written amendment

Free format text: JAPANESE INTERMEDIATE CODE: A821

Effective date: 20101021

RD02 Notification of acceptance of power of attorney

Free format text: JAPANESE INTERMEDIATE CODE: A7422

Effective date: 20101021

A521 Written amendment

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20101025

A521 Written amendment

Free format text: JAPANESE INTERMEDIATE CODE: A821

Effective date: 20101021

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: 20101207

A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20101217

R150 Certificate of patent or registration of utility model

Ref document number: 4653465

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150

Free format text: JAPANESE INTERMEDIATE CODE: R150

FPAY Renewal fee payment (event date is renewal date of database)

Free format text: PAYMENT UNTIL: 20131224

Year of fee payment: 3

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250

LAPS Cancellation because of no payment of annual fees