JP2012077006A - Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method - Google Patents

Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method Download PDF

Info

Publication number
JP2012077006A
JP2012077006A JP2010220872A JP2010220872A JP2012077006A JP 2012077006 A JP2012077006 A JP 2012077006A JP 2010220872 A JP2010220872 A JP 2010220872A JP 2010220872 A JP2010220872 A JP 2010220872A JP 2012077006 A JP2012077006 A JP 2012077006A
Authority
JP
Japan
Prior art keywords
klrg1
amino acid
positive
peptide
positive immune
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.)
Withdrawn
Application number
JP2010220872A
Other languages
Japanese (ja)
Inventor
Yoko Tokunaga
陽子 徳永
Susumu Kuno
晋 久野
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Asahi Kasei Medical Co Ltd
Original Assignee
Asahi Kasei Kuraray Medical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Asahi Kasei Kuraray Medical Co Ltd filed Critical Asahi Kasei Kuraray Medical Co Ltd
Priority to JP2010220872A priority Critical patent/JP2012077006A/en
Publication of JP2012077006A publication Critical patent/JP2012077006A/en
Withdrawn legal-status Critical Current

Links

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

PROBLEM TO BE SOLVED: To provide a new peptide as a ligand that has a low molecular weight, is inexpensive and has KLRG1 affinity and a KLRG1-positive immunocyte separation material that supports the peptide and selectively removes KLRG1-positive immunocytes in peripheral blood.SOLUTION: The KLRG1-positive immunocyte separation material includes a water-insoluble carrier to which a peptide containing an amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn and having a total number of constituent amino acid residues of ≤80 is immobilized.

Description

本発明は、KLRG1陽性免疫細胞を認識するペプチドに関する。本発明はさらに、血液、体液、細胞培養液及びこれらの処理液などのKLRG1陽性免疫細胞を含有する溶液から、KLRG1陽性免疫細胞を選択的に除去するためのKLRG1陽性免疫細胞分離材に関する。 The present invention relates to a peptide that recognizes KLRG1-positive immune cells. The present invention further relates to a KLRG1-positive immune cell separating material for selectively removing KLRG1-positive immune cells from a solution containing KLRG1-positive immune cells such as blood, body fluid, cell culture medium and treatment solutions thereof.

近年、わが国における乳癌の罹患数および死亡数の増加は著しい。乳癌はこれまでエストロゲンレセプター(ER)とプロゲステロンレセプター(PgR)の発現状況により手術以外の治療法が決められる傾向にあった。ホルモン療法、抗がん剤療法などの薬物療法が有用であり、奏功率、生存率の改善がみられている。しかし、human epidermal growth factor receptor-related 2(HER2)陽性乳癌の約50%はホルモン受容体陰性でありホルモン療法の対象外であった。最近ではHER2陽性乳癌に対するトラスツズマブを中心とする抗体療法などの分子標的療法が行われ、その有用性が示されており、今後の有力な治療法として期待されている。 In recent years, the number of morbidity and mortality of breast cancer has increased remarkably in Japan. Up to now, breast cancer has tended to be treated by a method other than surgery depending on the expression status of estrogen receptor (ER) and progesterone receptor (PgR). Drug therapy such as hormone therapy and anticancer drug therapy is useful, and the response rate and survival rate have been improved. However, about 50% of human epidermal growth factor receptor-related 2 (HER2)-positive breast cancers were hormone receptor-negative and were excluded from hormone therapy. Recently, molecular-targeted therapies such as trastuzumab-based antibody therapy for HER2-positive breast cancer have been performed, and their usefulness has been demonstrated, and they are expected as a powerful therapeutic method in the future.

乳癌細胞は種々の増殖因子とその受容体を発現して、増殖に関連するシグナル伝達系を形成している。1984年に同定された癌遺伝子HER2はその1つであり、ヒト上皮成長因子受容体human epidermal growth factor receptor(EGFR,HER)ファミリーの一員である。受容体は細胞膜貫通型の糖タンパクで、構造的な類似性からHER1、HER2、HER3、HER4が存在している(非特許文献1)。EGF受容体ファミリーは二量体形成を通じてシグナル伝達に寄与しており、リガンドが結合した受容体がもう1つの受容体と二量体を形成することによりシグナル伝達に寄与すると考えられている。 Breast cancer cells express various growth factors and their receptors to form a signal transduction system related to growth. The oncogene HER2 identified in 1984 is one of them, and is a member of the human epidermal growth factor receptor (EGFR, HER) family. The receptor is a transmembrane glycoprotein, and HER1, HER2, HER3, and HER4 are present due to structural similarity (Non-Patent Document 1). The EGF receptor family contributes to signal transduction through dimerization, and it is believed that the ligand-bound receptor contributes to signal transduction by forming a dimer with another receptor.

HER2受容体を規定するHER2/neuはproto-oncogeneの1つと考えられ、17番染色体(17q21.1)に存在する(非特許文献2)。乳癌をはじめ卵巣癌、肺癌、胃癌など種々の癌においてHER2/neu遺伝子の増幅および過剰発現が認められている。HER2陽性乳癌は、乳癌全体の20%から30%を占める。成人の正常組織ではこの遺伝子の発現は非常に弱い。その自然経過は特異であり、全身療法を施行しない場合は再発が早く、1987年にHER2の増幅が乳癌の予後不良と相関することが報告されて以降、多くの研究においてHER2陽性転移乳癌では経過が不良であることが明らかにされている。 HER2/neu, which defines the HER2 receptor, is considered to be one of proto-oncogenes and exists on chromosome 17 (17q21.1) (Non-patent document 2). Amplification and overexpression of the HER2/neu gene have been observed in various cancers such as breast cancer, ovarian cancer, lung cancer, and gastric cancer. HER2-positive breast cancer accounts for 20% to 30% of all breast cancers. Expression of this gene is very weak in normal adult tissues. Its natural history is unique, with early recurrence in the absence of systemic therapy, and in many studies since HER2 amplification was reported to correlate with poor breast cancer prognosis in HER2-positive metastatic breast cancer. Is revealed to be bad.

1986年に抗HER2モノクローナル抗体がneu形質転換細胞の悪性形質を阻害することが示され、HER2を標的とするトラスツズマブが開発されるに到った。トラスツズマブの原型となったHER2受容体の細胞外ドメインに対するマウスモノクローナル抗体mu4D5は、in vitroにおいてHER2陽性の腫瘍細胞株に対して増殖抑制作用が認められている(非特許文献3)。また、HER2陽性ヒト乳癌細胞株であるSKBR3に対するマウス脾細胞の抗体依存性細胞傷害(antibody-dependent cell cytotoxicity, ADCC)は、マウスモノクローナル抗体で増強されており、in vivoにおいてはADCC活性も抗腫瘍効果に寄与しているとの報告もある(非特許文献4)。しかし臨床応用するにあたり、マウス抗体をそのまま用いてはhuman anti-mouse antibody(HAMA)の出現が問題となる。そこで、HER2受容体の細胞外ドメインに対するマウスモノクローナル抗体の抗原結合部位である可変部のみをヒトIgG1の定常部に移植したヒト化抗体として作製されたものがトラスツズマブ(商品名:ハーセプチン(登録商標))である。 In 1986, anti-HER2 monoclonal antibody was shown to inhibit the malignant phenotype of neu-transformed cells, leading to the development of trastuzumab targeting HER2. The murine monoclonal antibody mu4D5 against the extracellular domain of the HER2 receptor, which is the prototype of trastuzumab, has been confirmed to have a growth inhibitory action against HER2-positive tumor cell lines in vitro (Non-patent Document 3). Furthermore, antibody-dependent cell cytotoxicity (ADCC) of mouse splenocytes against HER2-positive human breast cancer cell line SKBR3 is enhanced by mouse monoclonal antibody, and ADCC activity is also antitumor in vivo. There is also a report that it contributes to the effect (Non-Patent Document 4). However, for clinical application, the appearance of human anti-mouse antibodies (HAMA) becomes a problem when using mouse antibodies as they are. Therefore, trastuzumab (trade name: Herceptin (registered trademark)) was prepared as a humanized antibody in which only the variable region, which is the antigen-binding site of the mouse monoclonal antibody against the extracellular domain of HER2 receptor, was transplanted to the constant region of human IgG1. ).

HER2はPI3KやMAPKを含む様々なシグナル伝達経路ネットワークを惹起するが、トラスツズマブはHER2受容体に結合することにより、このシグナル伝達経路を抑制し、細胞周期の停止やアポトーシス、血管新生抑制などを誘導して、直接的に腫瘍細胞増殖抑制作用を示すと考えられる。また、Srcチロシンキナーゼの阻害とそれに伴うPTENの活性化、Aktの脱リン酸化も報告されている。さらに、腫瘍細胞と結合したトラスツズマブのFc部分に、NK細胞をはじめとする免疫細胞に発現するFc受容体が結合し腫瘍細胞殺傷効果を示す(ADCC活性)ことも示されている(非特許文献5から8)。これらの直接的腫瘍細胞増殖抑制作用と、ADCC活性の2つがトラスツズマブの主な作用機序と考えられている。 HER2 induces various signal transduction network including PI3K and MAPK, but trastuzumab inhibits this signal transduction pathway by binding to HER2 receptor, and induces cell cycle arrest, apoptosis, angiogenesis inhibition, etc. Then, it is considered that the tumor cell growth inhibitory effect is directly exhibited. In addition, inhibition of Src tyrosine kinase, accompanying activation of PTEN, and dephosphorylation of Akt have been reported. Furthermore, it has been shown that Fc receptors expressed in immune cells such as NK cells bind to the Fc portion of trastuzumab bound to tumor cells and exhibit a tumor cell killing effect (ADCC activity) (Non-Patent Document 1). 5 to 8). Two of these direct tumor cell growth inhibitory actions and ADCC activity are considered to be the main action mechanisms of trastuzumab.

トラスツズマブはHER2を標的としており、その治療効果はHER2タンパクの発現の程度により大きく異なる。測定法や材料などによりその陽性率は大きく異なることが知られているが、HER2蛋白の過剰発現、DNAの増幅を調べるため、免疫組織化学方法(immunohistochemical method:IHC法)とfluoresence in situ hybridization(FISH)が広く行われている。また、トラスツズマブは術後補助療法に利用すると高い効果が得られることがHERA試験をはじめとする複数の大規模臨床試験で明らかになっている。 Trastuzumab targets HER2, and its therapeutic effect greatly depends on the expression level of HER2 protein. It is known that the positive rate varies greatly depending on the measurement method and material, but in order to investigate overexpression of HER2 protein and DNA amplification, immunohistochemical method (IHC method) and fluorescence in situ hybridization( FISH) is widely practiced. In addition, trastuzumab has been shown to be highly effective when used in postoperative adjuvant therapy, as shown in several large-scale clinical trials including the HERA trial.

これまでの様々な検討から、トラスツズマブ投与によりHER2を阻害すると、乳癌の自然経過に大きな影響を与えることがわかっている。しかし、トラスツズマブはHER2過剰発現乳癌すべての自然経過を変更するものではないこともわかってきており、初回トラスツズマブ単剤投与に反応する症例は、HER2過剰発現乳癌の約1/3以下であるといわれている。これと同様に顕微鏡的転移癌の場合、かなりの割合の腫瘍がトラスツズマブに耐性であることが示唆される。しかし、トラスツズマブ耐性の機序については明確には解明されていない。現在のところ耐性機序として、(1)トラスツズマブの到達が不十分なため、HER2の細胞外領域の阻害が十分ではないこと、(2)HER2発現の減少、(3)下流にあるHER2機能調節因子が変化していること(p27kip1の低下、PTENの消失または不活性化など)、(4)代替経路によるシグナル伝達が生じること(insulin-like growth factor I receptor:IGF1Rの過剰発現)、(5)免疫能の低下、特にADCC活性の低下、などの可能性が示唆されている。 Various studies have shown that inhibition of HER2 by trastuzumab administration has a significant effect on the natural course of breast cancer. However, it has also been found that trastuzumab does not alter the natural course of all HER2-overexpressing breast cancers, and it is estimated that about 1/3 or less of HER2-overexpressing breast cancers respond to initial trastuzumab monotherapy. ing. Similarly, in the case of microscopic metastases, it is suggested that a significant proportion of tumors are resistant to trastuzumab. However, the mechanism of trastuzumab resistance has not been clarified. At present, the resistance mechanisms are: (1) Insufficient access of trastuzumab, so that inhibition of the extracellular region of HER2 is not sufficient, (2) reduction of HER2 expression, (3) downstream regulation of HER2 function Factor changes (p27 kip1 reduction, PTEN loss or inactivation, etc.), (4) Signal transduction by alternative pathways (insulin-like growth factor I receptor: IGF1R overexpression), ( 5) Possibility of decreased immunocompetence, especially decreased ADCC activity has been suggested.

上記(5)に掲げた免疫能と耐性機序に関する検討はあまり活発ではない。近年、トラスツズマブの主たる作用機序の1つであるADCC活性に関わる因子であるNK細胞について、その機能が明らかにされてきている。NK細胞には抑制性のレセプターが発現しており、その一つとして同定されたのがレクチン様レセプターkiller cell lectin-like receptor G1(KLRG1)である。これまでNK細胞に関して明らかにされた抑制性レセプターのリガンドのほとんどは、MHCクラスI分子にかかわるものであったが、2006年にM.Itoらにより、KLRG1のリガンドがMHCクラスI分子以外であることが報告された。 The studies on immunocompetence and resistance mechanism mentioned in (5) above are not very active. In recent years, the function of NK cells, which is a factor involved in ADCC activity, which is one of the main action mechanisms of trastuzumab, has been clarified. An inhibitory receptor is expressed in NK cells, and one of them was identified as the lectin-like receptor killer cell lectin-like receptor G1 (KLRG1). Most of the ligands of inhibitory receptors revealed so far on NK cells are related to MHC class I molecules, but in 2006, by M. Ito et al., the ligand of KLRG1 is other than MHC class I molecule. It was reported.

KLRG1は、ラット好塩基球性白血病細胞株RBL-2H3に発現する機能分子MAFA(mast cell function-associated antigen)として発見された(非特許文献9)。このKLRG1は、RBL-2H3細胞では抗KLRG1抗体でKLRG1を架橋することにより、Fcレセプター刺激による脱顆粒反応が抑制される。ラットKLRG1のcDNAクローニングから、細胞外領域にC型レクチン様の構造を持ち、細胞内領域にITIM(immunoreceptor tyrosine-based inhibitory motif)をもつII型膜貫通タンパク質のホモ二量体であることが、Pechtらにより報告されている(非特許文献10)。ヒトおよびマウスでは、KLRG1がNK細胞やT細胞の一部に発現するが(非特許文献11から13)、健常人では末梢血NK細胞の約50%に発現がみられることがわかっている。 KLRG1 was discovered as a functional molecule MAFA (mast cell function-associated antigen) expressed in the rat basophilic leukemia cell line RBL-2H3 (Non-patent document 9). This KLRG1 suppresses the degranulation reaction by Fc receptor stimulation by cross-linking KLRG1 with an anti-KLRG1 antibody in RBL-2H3 cells. From the cDNA cloning of rat KLRG1, it is found that it is a type II transmembrane protein homodimer having a C-type lectin-like structure in the extracellular region and an ITIM (immunoreceptor tyrosine-based inhibitory motif) in the intracellular region. Reported by Pecht et al. (Non-Patent Document 10). It is known that KLRG1 is expressed in a part of NK cells and T cells in humans and mice (Non-patent Documents 11 to 13), but is expressed in about 50% of peripheral blood NK cells in healthy people.

トラスツズマブ治療は、再発治療から再発予防目的の術後補助療法、術前治療へと適応が拡大されつつあり、さらなるHER2陽性癌の根治性向上への発展が期待される。適応が拡大されたことにより、トラスツズマブ投与対象例が大きく増加することが見込まれる。しかしながら、現状では、対象症例中には多数のトラスツズマブ投与無効群が存在している。 Trastuzumab treatment is expanding its application from recurrence treatment to postoperative adjuvant therapy for the purpose of preventing recurrence, and preoperative treatment, and further development to improve the curativeness of HER2-positive cancer is expected. It is expected that the number of patients treated with trastuzumab will greatly increase due to the expanded indication. However, at present, there are many trastuzumab ineffective groups among the target cases.

一方、KLRG1は、脊椎動物に遍在して発現して細胞間接着を仲介している3つの古典的カドヘリン(E-、N-、R-)と結合することが報告されている。E-、N-、R-カドヘリンによるKLRG1の連結反応はキラー細胞の細胞障害活性を調節し、カドヘリンが発現した組織への損傷を妨げることが示唆されている(特許文献14)。また、これらカドヘリンとKLRG1との結合について検討されており、E-カドヘリンについて更に、SPR測定の結果、KLRG1はE-カドヘリンのN末端の免疫グロブリン様ドメインと結合していること、KLRG1-E-カドヘリンComplexの結晶構造解析の結果、E-カドヘリンの9つのアミノ酸(3、4、5、6、7、92、93、94、95残基目)がKLRG1と相互作用しており、かつそのうちの5つのアミノ酸(4、5、7、93、95残基目)は9個の水素結合をも含んでいることが報告されている(非特許文献15)。更に、E-カドヘリンのN末端側から2から6残基目のいずれか1個のアミノ酸をアラニンに置換したE-カドヘリン変異体を用いて、KLRG1との相互作用をSPR分析、フローサイトメトリー分析、KLRG1レポーターアッセイ、および細胞障害活性測定により評価した結果、KLRG1はE-カドヘリンの2から6残基目のN末端部分を認識していることが報告されている(非特許文献16)。 On the other hand, KLRG1 has been reported to bind to three classical cadherins (E-, N-, R-) that are ubiquitously expressed in vertebrates and mediate cell-cell adhesion. It has been suggested that the KLRG1 ligation reaction by E-, N-, R-cadherin regulates the cytotoxic activity of killer cells and prevents damage to the tissues expressing cadherin (Patent Document 14). In addition, the binding between these cadherins and KLRG1 has been investigated, and further, regarding E-cadherin, as a result of SPR measurement, KLRG1 is bound to the N-terminal immunoglobulin-like domain of E-cadherin, KLRG1-E- As a result of crystal structure analysis of cadherin complex, 9 amino acids of E-cadherin (3rd, 4th, 5th, 6th, 7th, 92nd, 93rd, 94th, and 95th residues) interact with KLRG1 and It has been reported that five amino acids (residues 4, 5, 7, 93 and 95) also contain 9 hydrogen bonds (Non-Patent Document 15). Furthermore, using an E-cadherin mutant in which any one of amino acids 2 to 6 from the N-terminal side of E-cadherin was replaced with alanine, the interaction with KLRG1 was analyzed by SPR and flow cytometry. It was reported that KLRG1 recognizes the N-terminal portion of residues 2 to 6 of E-cadherin as a result of evaluation by the KLRG1 reporter assay and cytotoxic activity measurement (Non-Patent Document 16).

Pinkas-Kramarski, R., I. Arloy and Y. Yarden (1997) ErbB receptors and EGF-like ligands: cell lineage determination and oncogenesis through combinatorial signaling. J Mammary Gland Biol Neoplasia, 2:97-107.Pinkas-Kramarski, R., I. Arloy and Y. Yarden (1997) ErbB receptors and EGF-like ligands: cell lineage determination and oncogenesis through combinatorial signaling. J Mammary Gland Biol Neoplasia, 2:97-107. Schechter, A.L., D.F. Stern, L. Vaidyanathan, S.J. Decker, J.A. Drebin, M.I. Greene and R.A. Weinberg (1984) The neu oncogene: an erb-B-related gene encoding a 185,000-Mr tumour antigen. Nature, 312:513-516.Schechter, AL, DF Stern, L. Vaidyanathan, SJ Decker, JA Drebin, MI Greene and RA Weinberg (1984) The neu oncogene: an erb-B-related gene encoding a 185,000-Mr tumour antigen. Nature, 312:513- 516. Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM, Ullrich A (1989) p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol, 9(3):1165-72.Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM, Ullrich A (1989) p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol, 9(3):1165- 72. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Lang I, Nitz U, Iwata H, Thomssen C, Lohrisch C, Suter TM, Ruschoff J, Suto T, Greatorex V, Ward C, Straehle C, McFadden E, Dolci MS, Gelber RD; Herceptin Adjuvant (HERA) Trial Study Team (2005) Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med, 353(16):1659-72Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Lang I, Nitz U, Iwata H, Thomssen C, Lohrisch C, Suter TM, Ruschoff J, Suto T, Greatorex V, Ward C, Straehle C, McFadden E, Dolci MS, Gelber RD; Herceptin Adjuvant (HERA) Trial Study Team (2005) Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer.N Engl J Med, 353(16):1659-72 Clynes RA, Towers TL, Presta LG, Ravetch JV (2000) Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med, 6(4):443-6.Clynes RA, Towers TL, Presta LG, Ravetch JV (2000) Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med, 6(4):443-6. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK (2002) Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature, 416(6878):279-80Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK (2002) Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature, 416(6878):279-80 Gennari R, Menard S, Fagnoni F, Ponchio L, Scelsi M, Tagliabue E, Castiglioni F, Villani L, Magalotti C, Gibelli N, Oliviero B, Ballardini B, Da Prada G, Zambelli A, Costa A (2004) Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res., 10(17):5650-5Gennari R, Menard S, Fagnoni F, Ponchio L, Scelsi M, Tagliabue E, Castiglioni F, Villani L, Magalotti C, Gibelli N, Oliviero B, Ballardini B, Da Prada G, Zambelli A, Costa A (2004) Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res., 10(17):5650-5 Barok, et al., (2007) Trastuzumab causes antibody-dependent cellular cytotoxicity-mediated growth inhibition of submacroscopic JIMT-1 breast cancer xenografts despite intrinsic drug resistance. Molecular Cancer Therapeutics, 6:2065-2072.Barok, et al., (2007) Trastuzumab causes antibody-dependent cellular cytotoxicity-mediated growth inhibition of submacroscopic JIMT-1 breast cancer xenografts despite intrinsic drug resistance. Molecular Cancer Therapeutics, 6:2065-2072. Ortega Soto E, Pecht I (1988) A monoclonal antibody that inhibits secretion from rat basophilic leukemia cells and binds to a novel membrane component. J Immunol, 141(12):4324-32.Ortega Soto E, Pecht I (1988) A monoclonal antibody that inhibits secretion from rat basophilic leukemia cells and binds to a novel membrane component.J Immunol, 141(12):4324-32. Abramson J, Xu R, Pecht I (2002) An unusual inhibitory receptor--the mast cell function-associated antigen (MAFA). Mol Immunol, 38(16-18):1307-13.Abramson J, Xu R, Pecht I (2002) An unusual inhibitory receptor--the mast cell function-associated antigen (MAFA). Mol Immunol, 38(16-18):1307-13. Hanke T, Corral L, Vance RE, Raulet DH (1998) 2F1 antigen, the mouse homolog of the rat "mast cell function-associated antigen", is a lectin-like type II transmembrane receptor expressed by natural killer cells. Eur J Immunol., 28(12):4409-17Hanke T, Corral L, Vance RE, Raulet DH (1998) 2F1 antigen, the mouse homolog of the rat "mast cell function-associated antigen", is a lectin-like type II transmembrane receptor expressed by natural killer cells.Eur J Immunol ., 28(12):4409-17 Butcher S, Arney KL, Cook GP (1998) MAFA-L, an ITIM-containing receptor encoded by the human NK cell gene complex and expressed by basophils and NK cells. Eur J Immunol., 28(11):3755-62Butcher S, Arney KL, Cook GP (1998) MAFA-L, an ITIM-containing receptor encoded by the human NK cell gene complex and expressed by basophils and NK cells. Eur J Immunol., 28(11):3755-62 Blaser C, Kaufmann M, Pircher H (1998) Virus-activated CD8 T cells and lymphokine-activated NK cells express the mast cell function-associated antigen, an inhibitory C-type lectin. J Immunol., 161(12):6451-4Blaser C, Kaufmann M, Pircher H (1998) Virus-activated CD8 T cells and lymphokine-activated NK cells express the mast cell function-associated antigen, an inhibitory C-type lectin. J Immunol., 161(12):6451- Four Masayuki Ito, Takuma Maruyama, Naotoshi Saito, Satoru Koganei, Kazuo Yamamoto, Naoki Matsumoto (2006) Killer Cell lectin-like receptor G1 bind three members of the classical cadherin family to inhibit NK cell cytotoxicity. J Exp Med., 203(2):289-95Masayuki Ito, Takuma Maruyama, Naotoshi Saito, Satoru Koganei, Kazuo Yamamoto, Naoki Matsumoto (2006) Killer Cell lectin-like receptor G1 bind three members of the classical cadherin family to inhibit NK cell cytotoxicity.J Exp Med., 203(2) :289-95 Li Y, Hofmann M, Wang Q, Teng L, Chlewicki LK, Pircher H, Mariuzza RA. (2009)Structure of natural killer cell receptor KLRG1 bound to E-cadherin reveals basis for MHC-independent missing self recognition. Immunity.,17;31(1):35-46Li Y, Hofmann M, Wang Q, Teng L, Chlewicki LK, Pircher H, Mariuzza RA. (2009) Structure of natural killer cell receptor KLRG1 bound to E-cadherin reveals basis for MHC-independent missing self recognition. Immunity., 17 31(1):35-46 Nakamura S, Kuroki K, Ohki I, Sasaki K, Kajikawa M, Maruyama T, Ito M, Kameda Y, Ikura M, Yamamoto K, Matsumoto N, Maenaka K.(2009) Molecular basis for E-cadherin recognition by killer cell lectin-like receptor G1 (KLRG1). J Biol Chem., 2;284(40):27327-35Nakamura S, Kuroki K, Ohki I, Sasaki K, Kajikawa M, Maruyama T, Ito M, Kameda Y, Ikura M, Yamamoto K, Matsumoto N, Maenaka K. (2009) Molecular basis for E-cadherin recognition by killer cell lectin -like receptor G1 (KLRG1). J Biol Chem., 2;284(40):27327-35.

本発明は、低分子量で安価な、KLRG1に親和性を有するリガンドとしての新規なペプチド、並びに上記ペプチドを担持した、末梢血中のKLRG1陽性免疫細胞を選択的に除去するためのKLRG1陽性免疫細胞分離材を提供することを解決すべき課題とした。 The present invention relates to a novel peptide having a low molecular weight and low cost as a ligand having an affinity for KLRG1, and a KLRG1 positive immune cell carrying the above peptide for selectively removing KLRG1 positive immune cells in peripheral blood. Providing a separating material was a problem to be solved.

本発明者らは上記課題を解決するために鋭意検討した結果、E−カドヘリンのN末端の2から7残基目のアミノ酸配列を含む、1から8残基目のアミノ酸配列からなるペプチドでは、KLRG1との結合能力は発現できないが、C末端側を更に12残基目まで伸ばしたペプチドは、KLRG1との結合能力を持ち、また、このペプチドを水不溶性担体の表面に固定することで、末梢血からKLRG1陽性免疫細胞を選択的に除去することができるという事実を見出し、本発明を完成するに至った。 As a result of intensive studies to solve the above-mentioned problems, the present inventors have found that in a peptide consisting of an amino acid sequence of 1 to 8 residues, which contains the amino acid sequence of 2 to 7 residues at the N-terminus of E-cadherin, Although the ability to bind to KLRG1 cannot be expressed, a peptide in which the C-terminal side is further extended to the 12th residue has the ability to bind to KLRG1, and by immobilizing this peptide on the surface of a water-insoluble carrier, The present inventors have completed the present invention by finding the fact that KLRG1-positive immune cells can be selectively removed from blood.

即ち、本発明の態様は以下を含む。
(1) Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下のペプチドが固定されている水不溶性担体からなる、KLRG1陽性免疫細胞分離材。
(2) 配列番号1のアミノ酸配列を含むペプチドは、そのC末端側に水不溶性担体に固定するための活性アミノ酸残基が付加されている、(1)に記載のKLRG1陽性免疫細胞分離材。
(3) 前記活性アミノ酸残基がLysである、(2)に記載のKLRG1陽性免疫細胞分離材。
(4) 配列番号1のペプチドは、スペーサーを介して前記水不溶性担体に固定されている、(1)に記載のKLRG1陽性免疫細胞分離材。
(5) 前記スペーサーは、前記水不溶性担体との結合末端側に活性アミノ酸残基が付加されている、(4)に記載のKLRG1陽性免疫細胞分離材。
(6) 前記活性アミノ酸残基がLysである、(4)または(5)に記載のKLRG1陽性免疫細胞分離材。
(7) 前記水不溶性担体が織布もしくは不織布である、(1)〜(6)のいずれか1項に記載のKLRG1陽性免疫細胞分離材。
(8) 前記構成アミノ酸残基の総数が50個以下である、(1)〜(7)のいずれか1項に記載のKLRG1陽性免疫細胞分離材。
(9) 前記構成アミノ酸残基の総数が40個以下である、(1)〜(7)のいずれか1項に記載のKLRG1陽性免疫細胞分離材。
(10) 前記構成アミノ酸残基の総数が30個以下である、(1)〜(7)のいずれか1項に記載のKLRG1陽性免疫細胞分離材。
(11) Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列からなるペプチドがスペーサーを介して前記水不溶性担体に固定されている、KLRG1陽性免疫細胞分離材。
(12) Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下である、KLRG1陽性免疫細胞親和性ペプチド。
(13) 配列番号1のアミノ酸配列のC末端側にLysを有する、(12)に記載のKLRG1陽性免疫細胞親和性ペプチド。
(14) Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下であるペプチドを含む、KLRG1陽性免疫細胞分離剤。
(15) (1)〜(11)のいずれか1項に記載のKLRG1陽性免疫細胞分離材にKLRG1陽性免疫細胞を含有する溶液を接触させることを含む、KLRG1陽性免疫細胞を分離する方法。
That is, aspects of the invention include the following.
(1) A peptide containing the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) and having a total number of constituent amino acid residues of 80 or less A KLRG1-positive immune cell separating material comprising a water-insoluble carrier to which is immobilized.
(2) The KLRG1-positive immune cell separating material according to (1), wherein the peptide containing the amino acid sequence of SEQ ID NO: 1 has an active amino acid residue for immobilizing on a water-insoluble carrier added to its C-terminal side.
(3) The KLRG1-positive immune cell separating material according to (2), wherein the active amino acid residue is Lys.
(4) The KLRG1-positive immune cell separating material according to (1), wherein the peptide of SEQ ID NO: 1 is immobilized on the water-insoluble carrier via a spacer.
(5) The KLRG1-positive immune cell separating material according to (4), wherein the spacer has an active amino acid residue added to the binding terminal side with the water-insoluble carrier.
(6) The KLRG1-positive immune cell separating material according to (4) or (5), wherein the active amino acid residue is Lys.
(7) The KLRG1-positive immune cell separating material according to any one of (1) to (6), wherein the water-insoluble carrier is a woven or non-woven fabric.
(8) The KLRG1-positive immune cell separating material according to any one of (1) to (7), wherein the total number of the constituent amino acid residues is 50 or less.
(9) The KLRG1-positive immune cell separating material according to any one of (1) to (7), wherein the total number of the constituent amino acid residues is 40 or less.
(10) The KLRG1-positive immune cell separating material according to any one of (1) to (7), wherein the total number of the constituent amino acid residues is 30 or less.
(11) A peptide consisting of the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) is immobilized on the water-insoluble carrier via a spacer. A KLRG1-positive immune cell separating material.
(12) The amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) is included, and the total number of constituent amino acid residues is 80 or less. , KLRG1-positive immune cell affinity peptide.
(13) The KLRG1-positive immune cell affinity peptide according to (12), which has Lys at the C-terminal side of the amino acid sequence of SEQ ID NO: 1.
(14) The amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) is included, and the total number of constituent amino acid residues is 80 or less. A KLRG1-positive immune cell separating agent comprising a peptide.
(15) A method for separating KLRG1-positive immune cells, which comprises contacting a solution containing KLRG1-positive immune cells with the KLRG1-positive immune cell separating material according to any one of (1) to (11).

本発明の分離材を用いれば、例えば、血液、体液、細胞培養液、及びこれらの処理液中から、KLRG1陽性免疫細胞を選択的に除去することができる。したがって、上皮性癌細胞で発現する癌特異的膜抗原に対する抗体を投与しても従来は抗癌効果が発揮されなかった患者においても、有効な抗癌効果を得ることが期待できる。 If the separating material of the present invention is used, for example, KLRG1-positive immune cells can be selectively removed from blood, body fluid, cell culture fluid, and treatment liquids thereof. Therefore, it can be expected that an effective anti-cancer effect can be obtained even in a patient who has not conventionally exhibited an anti-cancer effect even when an antibody against a cancer-specific membrane antigen expressed in epithelial cancer cells is administered.

また、本発明のペプチドを用いれば、KLRG1陽性免疫細胞中のKLRG1とKLRG1に対するリガンド(例えば、E−カドヘリン)との結合を阻害し、KLRG1を介した抑制性シグナルを遮断することができる。したがって、上皮性癌細胞で発現する癌特異的膜抗原に対する、抗体依存性細胞障害活性を有する抗体と、前記癌特異的膜抗原が陽性であり、かつKLRG1に対するリガンドが陽性である上皮性癌細胞と単核球を含む細胞集団とを接触させても、従来は上皮性癌細胞に対する細胞障害活性が発揮されなかった場合においても、十分な細胞障害活性を得ることが期待できる。 Further, by using the peptide of the present invention, it is possible to inhibit the binding between KLRG1 and a ligand for KLRG1 (eg, E-cadherin) in KLRG1-positive immune cells, and block the inhibitory signal mediated by KLRG1. Therefore, an antibody having antibody-dependent cytotoxic activity against a cancer-specific membrane antigen expressed in epithelial cancer cells, and an epithelial cancer cell positive for the cancer-specific membrane antigen and positive for a ligand for KLRG1 It is expected that sufficient cytotoxic activity will be obtained even when the cell population containing mononuclear cells is brought into contact with the cell population in the past, even when the cytotoxic activity against epithelial cancer cells was not exerted.

以下、本発明を詳細に説明する。なお、本発明は、以下の実施の形態に限定されるものではなく、その要旨の範囲内で種々変形して実施することができる。 Hereinafter, the present invention will be described in detail. The present invention is not limited to the following embodiments, and can be variously modified and implemented within the scope of the gist.

本発明のKLRG1陽性免疫細胞分離材は、血液、体液、細胞培養液、及びこれらを処理して得られた液などのKLRG1陽性免疫細胞を含有する溶液から、KLRG1陽性免疫細胞を選択的に除去することができる分離材であって、Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下のペプチドが水不溶性担体に固定されていることを特徴とする。 The KLRG1-positive immune cell separating material of the present invention selectively removes KLRG1-positive immune cells from a solution containing KLRG1-positive immune cells such as blood, body fluid, cell culture solution, and a solution obtained by treating these. A separation material capable of being, which comprises the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1), It is characterized in that peptides having a total number of 80 or less are immobilized on a water-insoluble carrier.

本発明の分離材に用いるペプチドの配列番号1は、E−カドヘリンのN末端1から12残基目のアミノ酸配列である。KLRG1陽性免疫細胞を含有する溶液から、KLRG1陽性免疫細胞を選択的に除去することができる分離材において、本発明者らは水不溶性担体表面上のKLRG1と結合するリガンドとして、E−カドヘリンに着目し、E−カドヘリンタンパクの一部をペプチドとして切り出して設計することを検討した。本発明者らは、先行文献(非特許文献15、16)からKLRG1との相互作用においてE−カドヘリンのN末端部分の2から7残基目、および92から95残基目のアミノ酸配列が重要であると推測し、検討を進めたところ、N末端部分の2から7残基目を含む、1から8残基目のアミノ酸配列からなるペプチドを水不溶性担体に固定した場合は、KLRG1陽性免疫細胞との結合能力はなく、KLRG1陽性免疫細胞を含有する溶液からKLRG1陽性免疫細胞を選択的に除去することはできないが、ペプチドのアミノ酸配列を更にC末端側に4アミノ酸伸ばした、E−カドヘリンN末端部分1から12残基目の配列番号1を含むペプチドを水不溶性担体に固定した場合は、N末端部分の92から95残基目のアミノ酸配列を含むペプチドがなくても、KLRG1陽性免疫細胞との結合能力を発現し、KLRG1陽性免疫細胞を含有する溶液からKLRG1陽性免疫細胞を選択的に除去することができることを見出し、本発明に至った。 SEQ ID NO: 1 of the peptide used in the separation material of the present invention is the amino acid sequence of the 12th residue from the N-terminal of E-cadherin. In a separation material capable of selectively removing KLRG1-positive immune cells from a solution containing KLRG1-positive immune cells, the present inventors have focused on E-cadherin as a ligand that binds to KLRG1 on the surface of a water-insoluble carrier. Then, it was examined to cut out a part of E-cadherin protein as a peptide for designing. The present inventors have found that the amino acid sequences at the 2nd to 7th residues and at the 92nd to 95th residues of the N-terminal portion of E-cadherin are important in the interaction with KLRG1 from the previous literature (Non-Patent Documents 15 and 16). As a result of further studies, it was confirmed that when a peptide consisting of the amino acid sequence of the 1st to 8th residues, including the 2nd to 7th residues of the N-terminal part, was immobilized on a water-insoluble carrier, KLRG1-positive immunity It has no ability to bind to cells and cannot selectively remove KLRG1-positive immune cells from a solution containing KLRG1-positive immune cells, but the amino acid sequence of the peptide is further extended by 4 amino acids to the C-terminal side, and E-cadherin When a peptide containing SEQ ID NO: 1 at the N-terminal portion 1 to 12 residues was immobilized on a water-insoluble carrier, KLRG1-positive immunity was obtained even without a peptide containing an amino acid sequence at the N-terminal portion 92 to 95 residues. The present inventors have found that KLRG1-positive immune cells can be selectively removed from a solution that expresses a cell-binding ability and that contains KLRG1-positive immune cells, and completed the present invention.

E−カドヘリンのN末端部分1から8残基目のアミノ酸配列からなるペプチドの場合は、ペプチドは水不溶性担体上でKLRG1と結合し捕捉するための立体構造が保てないため、結合能力を得ることができないが、E−カドヘリンのN末端部分1から12残基目のアミノ酸配列である12個のアミノ酸(配列番号1)を含むペプチドの場合は、水不溶性担体上で、KLRG1と相互作用できる立体構造をとることができ、結合、捕捉能力を発現できると推定している。 In the case of a peptide consisting of the amino acid sequence of the 1st to 8th residues of the N-terminal portion of E-cadherin, the peptide cannot maintain a three-dimensional structure for binding and capturing KLRG1 on a water-insoluble carrier, and thus has a binding ability. However, in the case of a peptide containing 12 amino acids (SEQ ID NO: 1) which is the amino acid sequence of residues 1 to 12 of the N-terminal portion of E-cadherin, it can interact with KLRG1 on a water-insoluble carrier. It is presumed that it can have a three-dimensional structure and can exhibit binding and capturing ability.

また本発明によれば、水不溶性担体表面上のペプチドのアミノ酸数は12個以上80個以下である。水不溶性担体表面上において、配列番号1はその配列のC末端側およびN末端側に更にアミノ酸を延長してもよく、アミノ酸数が増えても配列番号1の立体構造は維持されるが、アミノ酸数が増えるに従い、KLRG1陽性免疫細胞以外の細胞を非特異的に吸着しやすくなるため、アミノ酸数は12個以上80個以下が好ましい。また合成方法やコストから12個以上50個以下がより好ましく、12個以上40個以下がより好ましく、12個以上30個以下がさらに好ましく、12個以上20個以下が特に好ましく、12個以上15個以下が最も好ましい。配列番号1のアミノ酸配列のN末端側に延長されるアミノ酸の種類は特に限定されず、任意のアミノ酸を延長することができる。配列番号1のアミノ酸配列のC末端側に延長されるアミノ酸の種類も特に限定されず、任意のアミノ酸を延長してもよいし、あるいはE−カドヘリンのN末端側の13残基目以降のアミノ酸配列を、配列番号1のアミノ酸配列のC末端側に延長してもよい。 Further, according to the present invention, the number of amino acids of the peptide on the surface of the water-insoluble carrier is 12 or more and 80 or less. On the surface of the water-insoluble carrier, SEQ ID NO: 1 may further extend amino acids to the C-terminal side and N-terminal side of the sequence, and the three-dimensional structure of SEQ ID NO: 1 is maintained even if the number of amino acids is increased. As the number increases, cells other than the KLRG1-positive immune cells are more likely to be nonspecifically adsorbed, so the number of amino acids is preferably 12 or more and 80 or less. Moreover, 12 or more and 50 or less are more preferable, 12 or more and 40 or less are more preferable, 12 or more and 30 or less are more preferable, 12 or more and 20 or less are especially preferable, 12 or more and 15 from the synthetic method or cost. The number of pieces or less is most preferable. The type of amino acid extended to the N-terminal side of the amino acid sequence of SEQ ID NO: 1 is not particularly limited, and any amino acid can be extended. The type of amino acid extended to the C-terminal side of the amino acid sequence of SEQ ID NO: 1 is not particularly limited, and any amino acid may be extended, or the amino acid at the 13th residue and after on the N-terminal side of E-cadherin. The sequence may be extended C-terminal to the amino acid sequence of SEQ ID NO:1.

また、ペプチドの末端はフリーでもよいし、例えばN末端はアセチル化、C末端はアミド化によりブロッキングしてもよいし、また水不溶性担体への固定などを考慮して、N末端またはC末端に官能基を導入してもよい。 In addition, the terminal of the peptide may be free, for example, the N-terminal may be blocked by acetylation and the C-terminal may be blocked by amidation, and in consideration of immobilization on a water-insoluble carrier, etc. A functional group may be introduced.

本発明の分離材において、配列番号1のアミノ酸配列を含むペプチドは、そのC末端側に水不溶性担体に固定するための活性アミノ酸残基を付加するとより好ましい。活性アミノ酸残基とは、側鎖に水不溶性担体と結合するための活性基を持つアミノ酸であり、例えば、リシン、アルギニン、システイン、トリプトファン、ヒスチジン、アスパラギン、アスパラギン酸、グルタミン、グルタミン酸、セリン、トレオニン、チロシンが挙げられ、水不溶性担体との固定のしやすさから、好ましくはリシン、システインであり、より好ましくはリシンである。 In the separating material of the present invention, the peptide containing the amino acid sequence of SEQ ID NO: 1 is more preferably added with an active amino acid residue for immobilizing it on a water-insoluble carrier at its C-terminal side. The active amino acid residue is an amino acid having an active group for binding to a water-insoluble carrier in its side chain, for example, lysine, arginine, cysteine, tryptophan, histidine, asparagine, aspartic acid, glutamine, glutamic acid, serine, threonine. , Tyrosine, and lysine and cysteine are preferable and lysine is more preferable because they are easily fixed to a water-insoluble carrier.

本発明の分離材において、配列番号1のペプチドはスペーサーを介して水不溶性担体に固定されてもよい。本明細書中でいうスペーサーとは、配列番号1のアミノ酸配列を含むペプチドと水不溶性担体との間に位置し、該ペプチドを水不溶性担体に間隔を空けて固定できる任意の長さの分子を意味する。スペーサーは、スペーサーの流動性や排除体積効果により、細胞と分離材との非特異的な相互作用を防げる。スペーサーの長さとしては、スペーサーのないものから、その中に含まれる原子数で2000までが好ましい結果を与える。あまり長すぎるスペーサーは熱安定性及び耐放射線性が悪く、溶出物の原因となる。より好ましくはスペーサーのないものから、その中に含まれる原子数で1000以下であり、さらに好ましくは500以下であり、特に好ましくは200以下である。スペーサーは、水不溶性担体の表面上に導入してもよいし、ペプチドに導入してもよい。その場合、スペーサーの導入位置は配列番号1のアミノ酸配列を中断しない位置とする。配列番号1よりもC末端側がより好ましい。スペーサーの具体的材料としては、これらに限定はされないが、ポリエチレングリコールやなどの高分子材料やポリペプチドなどが挙げられる。また、スペーサーにおいては、水不溶性担体との結合末端に、水不溶性担体への固定などを考慮して官能基を導入してもよいし、活性アミノ酸残基が付加されていることが好ましい。スペーサーに付加することができる活性アミノ酸残基としては、本明細書中上記したものが挙げられる。 In the separation material of the present invention, the peptide of SEQ ID NO: 1 may be immobilized on a water-insoluble carrier via a spacer. The term “spacer” as used herein refers to a molecule that is located between a peptide containing the amino acid sequence of SEQ ID NO: 1 and a water-insoluble carrier and that can fix the peptide to the water-insoluble carrier with a space between them. means. The spacer prevents non-specific interaction between cells and the separation material due to the fluidity of the spacer and the excluded volume effect. With respect to the length of the spacer, a preferable result is obtained from one having no spacer to 2000 in terms of the number of atoms contained therein. Spacers that are too long have poor thermal stability and radiation resistance, and cause elution. The number of atoms contained therein is more preferably 1,000 or less, still more preferably 500 or less, and particularly preferably 200 or less, from the one having no spacer. The spacer may be introduced on the surface of the water-insoluble carrier or may be introduced on the peptide. In that case, the position at which the spacer is introduced is a position where the amino acid sequence of SEQ ID NO: 1 is not interrupted. The C-terminal side is more preferable than SEQ ID NO: 1. Specific materials for the spacer include, but are not limited to, polymeric materials such as polyethylene glycol and the like, polypeptides, and the like. In addition, in the spacer, a functional group may be introduced at the binding end with the water-insoluble carrier in consideration of immobilization on the water-insoluble carrier, or an active amino acid residue is preferably added. Active amino acid residues that can be added to the spacer include those mentioned herein above.

本発明の分離材において、配列番号1のペプチドがスペーサーを介して水不溶性担体に固定されている場合の具体的態様としては、スペーサーがペプチドに導入されている場合、スペーサーが水不溶性担体に導入されている場合、あるいはスペーサーがペプチドと水不溶性担体の両方に導入されている場合などが挙げられる。即ち、ペプチドとスペーサーとの結合体を水不溶性担体に固定化してもよいし、ペプチドを、スペーサーを有する水不溶性担体に固定化してもよいし、ペプチドとスペーサーとの結合体を、スペーサーを有する水不溶性担体に固定化してもよい。この際、ペプチドを水不溶性担体に固定するための官能基(活性アミノ酸残基など)は、ペプチド、スペーサー又は水不溶性担体の何れか1以上に適宜付加することができる。 In the separation material of the present invention, the peptide of SEQ ID NO: 1 is immobilized on the water-insoluble carrier via a spacer. Specific examples of the embodiment include the case where the spacer is introduced into the peptide and the spacer is introduced into the water-insoluble carrier. Or a spacer is introduced into both the peptide and the water-insoluble carrier. That is, the peptide-spacer conjugate may be immobilized on a water-insoluble carrier, the peptide may be immobilized on a water-insoluble carrier having a spacer, or the peptide-spacer conjugate may have a spacer. It may be immobilized on a water-insoluble carrier. At this time, a functional group (an active amino acid residue or the like) for fixing the peptide to the water-insoluble carrier can be appropriately added to any one or more of the peptide, the spacer and the water-insoluble carrier.

本発明で用いることのできる水不溶性担体の形状としては、液層でKLRG1陽性免疫細胞を含有する溶液との接触を良好に行うために、接触頻度の面より表面積が大きいことが好ましい。例を挙げると、不織布、繊維状、綿状、糸状、束状、簾状、織布等の繊維構造体、平膜、スポンジ等の高分子多孔質体、あるいは粒子(磁気粒子など)等の構造が挙げられる。特に血液細胞の吸着性、分離材としての取り扱い性からみて、織布、不織布が好ましく、中でも細胞との多点的な接触が可能である点で不織布が最も好ましい。 The shape of the water-insoluble carrier that can be used in the present invention is preferably a surface area larger than the contact frequency in order to make good contact with a solution containing KLRG1-positive immune cells in the liquid layer. Examples include fibrous structures such as non-woven fabrics, fibrous, cotton-like, yarn-like, bundle-like, blind-like and woven fabrics, flat membranes, polymeric porous bodies such as sponges, and particles (such as magnetic particles). The structure is mentioned. In particular, woven fabrics and non-woven fabrics are preferable from the viewpoint of adsorbability of blood cells and handleability as a separating material, and among them, non-woven fabrics are most preferable because they can be contacted with cells at multiple points.

水不溶性担体の形状が不織布等の繊維状担体を用いる場合、繊維径が細胞吸着能力に寄与するため、有効な平均繊維径のものを用いることが重要である。本発明の平均繊維径の測定は走査型電子顕微鏡で繊維状基材の表面を撮影し、目視により撮影面上に分散している糸の直径をランダムに100個以上測定して求める。繊維径が大きすぎる場合にはKLRG1陽性免疫細胞の吸着量及び吸着速度が低下するし、小さすぎる場合には、KLRG1陽性免疫細胞以外のリンパ球、単球、顆粒球、血小板等の非特異吸着をおこしやすい点から、平均繊維径0.5μm以上30μm以下が好ましい。更に好ましくは1μm以上20μm以下である。最も好ましくは2μm以上10μm以下である。また、フィラメントは、モノフィラメントでもマルチフィラメントでも構わないし、多孔質フィラメントでも異型フィラメントでも構わない。 When a fibrous carrier such as a non-woven fabric is used as the shape of the water-insoluble carrier, the fiber diameter contributes to the cell adsorbing ability, and therefore it is important to use an effective average fiber diameter. The average fiber diameter of the present invention is determined by photographing the surface of the fibrous substrate with a scanning electron microscope and visually measuring 100 or more randomly distributed diameters of the yarns dispersed on the photographed surface. If the fiber diameter is too large, the adsorption amount and adsorption rate of KLRG1-positive immune cells will decrease, and if it is too small, non-specific adsorption of lymphocytes other than KLRG1-positive immune cells, monocytes, granulocytes, platelets, etc. The average fiber diameter is preferably 0.5 μm or more and 30 μm or less from the viewpoint of easily causing the above. More preferably, it is 1 μm or more and 20 μm or less. Most preferably, it is 2 μm or more and 10 μm or less. Further, the filament may be a monofilament or a multifilament, and may be a porous filament or an atypical filament.

水不溶性担体の形状が粒状の場合、球状、多角球状等いかなる形状であっても有用に用いられる。また表面は平滑であっても、凹凸があってもKLRG1陽性免疫細胞を選択的に除去できる表面であれば有用に用いられる。また、粒径は、50μm以上10mm以下が好ましい。粒径は50μm未満であると粒子の流出防止が困難になる傾向にあるので好ましくない。粒径が10mmより大きい場合十分な表面積が得られなくなる傾向にあるので好ましくない。粒径は、好ましくは、80μm以上8mm以下、最も好ましくは100μm以上6mm以下である。 When the water-insoluble carrier has a granular shape, it can be effectively used in any shape such as spherical or polygonal. Further, any surface can be usefully used as long as it has a smooth surface or unevenness and can selectively remove KLRG1-positive immune cells. The particle size is preferably 50 μm or more and 10 mm or less. If the particle size is less than 50 μm, it is difficult to prevent the particles from flowing out, which is not preferable. If the particle size is larger than 10 mm, a sufficient surface area tends not to be obtained, which is not preferable. The particle size is preferably 80 μm or more and 8 mm or less, and most preferably 100 μm or more and 6 mm or less.

本発明で用いることのできる水不溶性担体の材質としては、ポリエチレンテレフタレート、ポリブチレンテレフタレート及びポリオキシエチレンテレフタレート等のポリエステル、ポリアクリルニトリル、ナイロン6、ナイロン6,6等のポリアミド、芳香族ポリアミド、ポリスチレン及びその誘導体、ポリエチレン、ポリプロピレン、ポリブテン等のポリオレフィン、メチルメタクリレート、エチルメタクリレート等のメタクリル酸エステル誘導体を重合して得られる高分子化合物、メチルアクリレート、エチルアクリレート等のアクリル酸エステル誘導体を重合して得られる高分子化合物、ポリトリフルオロクロルエチレン、ポリビニルホルマール、ポリスルホン、ポリウレタン、ポリビニルアセタール、ポリカーボネート等の合成高分子、セルロース及び/又はその誘導体等の再生繊維などが挙げられる。これらを単独で用いてもよいし、上記材質をブレンド、アロイ化して用いてもよい。好ましくは、繊維の成形性や活性基の導入しやすさの点より、ポリエチレン、ポリプロピレン等のポリオレフィン、スチレンブタジエン共重合体等のポリスチレン等が挙げられる。特に、溶出物の少ない点でポリエチレン、ポリプロピレンが推奨される。また、これらに親水性を付与する目的で表面をコーティング、放射線グラフト等の方法により親水性高分子材料で修飾した材料も有用に用いられる。 Examples of the material of the water-insoluble carrier that can be used in the present invention include polyesters such as polyethylene terephthalate, polybutylene terephthalate and polyoxyethylene terephthalate, polyamides such as polyacrylonitrile, nylon 6, nylon 6,6, aromatic polyamide, polystyrene. And its derivatives, polyolefins such as polyethylene, polypropylene and polybutene, polymer compounds obtained by polymerizing methacrylic acid ester derivatives such as methyl methacrylate and ethyl methacrylate, and acrylic acid ester derivatives such as methyl acrylate and ethyl acrylate. Examples of the polymer compound include synthetic polymers such as polytrifluorochloroethylene, polyvinyl formal, polysulfone, polyurethane, polyvinyl acetal, and polycarbonate, and regenerated fibers such as cellulose and/or its derivatives. These may be used alone, or may be used by blending or alloying the above materials. Preferable examples include polyolefin such as polyethylene and polypropylene, polystyrene such as styrene-butadiene copolymer, and the like, from the viewpoint of moldability of fibers and easy introduction of active groups. In particular, polyethylene and polypropylene are recommended in terms of less eluate. Further, a material whose surface is modified with a hydrophilic polymer material by a method such as coating or radiation grafting for the purpose of imparting hydrophilicity to these is also useful.

本発明のペプチドを水不溶性担体の表面に存在させる方法は、共有結合、イオン結合、物理的吸着、包埋あるいは担体表面への沈殿不溶化などあらゆる公知の方法を用いることができるが、リガンドの溶出性よりみて、共有結合により固定、不溶化して用いることが好ましい。そのため通常固定化酵素、アフィニティークロマトフラフィーで用いられる公知の担体への活性基導入法が良好に用いられる。例を挙げると、カルボキシル基を有する担体を用い、このカルボキシル基をN−ヒドロキシコハク酸イミドと反応させることによって、スクシンイミドオキシカルボニル基に変換し、これにペプチドをアミノ基の部分で反応させる方法(活性エステル法)、アミノ基またはカルボキシル基を有する担体を用い、ジシクロヘキシルカルボジイミドなどの縮合試薬の存在下で、担体のアミノ基またはカルボキシル基とペプチドのカルボキシル基またはアミノ基を縮合反応させて結合させる方法(縮合法)、担体とペプチドとをグルタルアルデヒドなどの2個以上の官能基を有する化合物を用いて架橋させて結合させる方法(担体架橋法)、水酸基を有する担体を用い、臭化シアンなどのハロゲン化シアンを担体に作用させ、ペプチドやタンパク質のアミノ基の部分で反応させて結合させる方法、チオール基を有する担体を用い、ペプチドやタンパク質のチオール基の部分で反応させる方法、およびエピクロロヒドリンなどのエポキシドを担体に作用させ、ペプチドのアミノ基の部分や水酸基の部分で反応させて結合させる方法、N−ヒドロキシメチルハロアセトアミド等を用いて担体表面にα−アセトアミノハロゲン基を導入しペプチドやタンパクのチオール基の部分で反応させる方法、γ線や電子線を照射してラジカルを発生させた水不溶性担体にメタクリル酸グリシジルなどのビニル化合物をグラフト重合してエポキシ基を導入しペプチドやタンパクのアミノ基やチオール基の部分で反応させる方法等が挙げられるが、これに限定されるものではない。また水不溶性担体上に直接ペプチドを合成する方法として、水不溶性担体上に保護基を外す条件においても切断されない結合性の基を付加し、上記水不溶性担体上でペプチドを下記のペプチドの固相合成法で合成し、保護基が付加されている全ての官能基から保護基を除去することにより得る方法が挙げられる。 As the method for allowing the peptide of the present invention to be present on the surface of the water-insoluble carrier, any known method such as covalent bond, ionic bond, physical adsorption, embedding, or precipitation insolubilization on the carrier surface can be used, but ligand elution From the viewpoint of properties, it is preferable to use by fixing and insolubilizing by covalent bond. Therefore, a well-known method of introducing an active group into a carrier, which is usually used for immobilized enzymes and affinity chromatography, is favorably used. For example, a method of using a carrier having a carboxyl group and converting the carboxyl group to N-hydroxysuccinimide to convert to a succinimideoxycarbonyl group, and reacting the peptide at the amino group portion ( Active ester method), a method of using a carrier having an amino group or a carboxyl group, and subjecting the amino group or the carboxyl group of the carrier to the carboxyl group or the amino group of the peptide in the presence of a condensation reagent such as dicyclohexylcarbodiimide to bond them. (Condensation method), a method in which a carrier and a peptide are crosslinked and bonded using a compound having two or more functional groups such as glutaraldehyde (carrier crosslinking method), a carrier having a hydroxyl group is used, and cyan bromide or the like is used. A method in which cyanogen halide is allowed to act on a carrier to react and bond at the amino group portion of a peptide or protein, a method of using a carrier having a thiol group and a reaction at the thiol group portion of a peptide or protein, and epichlorohydride A method in which an epoxide such as phosphorus is allowed to act on the carrier to react by binding at the amino group portion or hydroxyl group portion of the peptide, and an α-acetaminohalogen group is introduced onto the carrier surface using N-hydroxymethylhaloacetamide or the like. Method of reacting at the thiol group of peptide or protein, grafting a vinyl compound such as glycidyl methacrylate to a water-insoluble carrier that has generated radicals by irradiation with γ-rays or electron beams and introducing an epoxy group into the peptide or peptide Examples thereof include a method of reacting at the amino group or thiol group of the protein, but the method is not limited thereto. As a method for directly synthesizing a peptide on a water-insoluble carrier, a binding group that is not cleaved even under the condition of removing a protective group is added to the water-insoluble carrier, and the peptide is immobilized on the water-insoluble carrier as a solid phase of the following peptide. Examples include a method of synthesizing by a synthetic method and removing the protective group from all the functional groups to which the protective group is added.

水不溶性担体へのペプチドの固定量は、特に限定されないが、ペプチドの機能を有効に発現するためには、水不溶性担体の表面積当たり0.01nmol/m2以上100μmol/m2以下が好ましく、より好ましくは0.1nmol/m2以上10μmol/m2以下であり、更に好ましくは1.0nmol/m2以上1.0μmol/m2以下である。 The amount of the peptide immobilized on the water-insoluble carrier is not particularly limited, but in order to effectively express the function of the peptide, it is preferably 0.01 nmol/m 2 or more and 100 μmol/m 2 or less per surface area of the water-insoluble carrier. preferably at 0.1 nmol / m 2 or more 10 .mu.mol / m 2 or less, further preferably 1.0 nmol / m 2 or more 1.0 [mu] mol / m 2 or less.

本発明において、KLRG1陽性免疫細胞の種類は特に限定されないが、KLRG1陽性NK細胞、KLRG1陽性T細胞などが挙げられる。 In the present invention, the type of KLRG1-positive immune cells is not particularly limited, and examples thereof include KLRG1-positive NK cells and KLRG1-positive T cells.

本発明において、KLRG1陽性免疫細胞を含有する溶液が血液である場合、血液の抗凝固目的で抗凝固剤を血液中に加えることができる。抗凝固剤を例示すると、抗凝固活性を有する化合物であれば、特に限定されないが、ヘパリン、低分子ヘパリン、メシル酸ナファモスタット、メシル酸ガベキセート、アルガトロバン、クエン酸ナトリウム等が好適例として挙げられ、好ましくはヘパリンあるいはクエン酸ナトリウムが良好に用いられる。 In the present invention, when the solution containing KLRG1-positive immune cells is blood, an anticoagulant can be added to blood for the purpose of anticoagulating blood. Examples of the anticoagulant include, but are not particularly limited to, compounds having anticoagulant activity, heparin, low molecular weight heparin, nafamostat mesylate, gabexate mesylate, argatroban, sodium citrate and the like are preferred examples. Heparin or sodium citrate is preferably used.

ペプチドの合成は、ペプチド合成において通常用いられる方法、例えば、固相法または液相法により行われるが、固相法により行う方が、操作が簡便であるため好ましい。合成したペプチドは、さらに精製することにより、純度の高いものを得ることができる。ペプチドの精製は有機化合物の精製に一般的に用いられている方法が使用できる。特にカラムクロマトグラフィーによる精製は、効率良く精製を行えるので好ましい。また、スペーサーの導入は公知の合成法で行うことができ、例えば固相法によるペプチド合成においてアミノ基とカルボキシル基を有するスペーサー分子をアミノ酸と同様に導入してもよいし、液相法で導入してもよい。 Peptide synthesis is performed by a method usually used in peptide synthesis, for example, a solid phase method or a liquid phase method, but the solid phase method is preferable because the operation is simple. By further purifying the synthesized peptide, a highly pure peptide can be obtained. For purification of the peptide, a method generally used for purification of organic compounds can be used. In particular, purification by column chromatography is preferable because efficient purification can be performed. The spacer can be introduced by a known synthesis method. For example, in peptide synthesis by the solid phase method, a spacer molecule having an amino group and a carboxyl group may be introduced in the same manner as an amino acid, or by a liquid phase method. You may.

本発明の分離材は、以上述べてきたように、血液、体液、細胞培養液、及びこれらの処理液などのKLRG1陽性免疫細胞を含有する溶液から、抗体やタンパクの様に高価なリガンドを必要としないにもかかわらず選択的にKLRG1陽性免疫細胞を分離することができることから、少なくとも入口と出口を有する容器に充填して細胞選択除去を目的とする体外循環用KLRG1陽性免疫細胞分離器として有効に用いることができる。 As described above, the separation material of the present invention requires expensive ligands such as antibodies and proteins from solutions containing KLRG1-positive immune cells such as blood, body fluids, cell culture fluids, and treatment solutions thereof. Since it is possible to selectively separate KLRG1-positive immune cells, it is effective as a KLRG1-positive immune cell separator for extracorporeal circulation for the purpose of selective cell removal by filling a container having at least an inlet and an outlet. Can be used for.

本発明のKLRG1陽性免疫細胞分離材は、例えば、上皮性癌細胞で発現する癌特異的膜抗原が陽性であり、かつKLRG1に対するリガンドが陽性である上皮性癌の生体の末梢血中のKLRG1陽性免疫細胞を前記生体外で選択的に減ずる工程(a)と、該生体に、前記上皮性癌細胞で発現する前記癌特異的膜抗原に対する抗体であって抗体依存性細胞障害活性を有する抗体を含有する癌治療剤を投与する工程(b)とを含む、前記上皮性癌の生体を処置する方法において、上記工程(a)において用いることができる。上記工程(a)においては、好ましくは、生体の末梢血を体外循環して、KLRG1陰性免疫細胞よりKLRG1陽性免疫細胞に対して高い親和性を有する細胞分離器である本発明のKLRG1陽性免疫細胞分離材に通し、KLRG1陽性免疫細胞を選択的に減ずることができる。上記の上皮性癌細胞で発現する癌特異的膜抗原は、好ましくはHER2であり、上皮性癌細胞で発現する癌特異的膜抗原に対する抗体は、好ましくはトラスツズマブである。 The KLRG1-positive immune cell separating material of the present invention is, for example, KLRG1-positive in the peripheral blood of a living body of epithelial cancer in which the cancer-specific membrane antigen expressed in epithelial cancer cells is positive and the ligand for KLRG1 is positive. A step (a) of selectively reducing immune cells in vitro, and an antibody against the cancer-specific membrane antigen expressed in the epithelial cancer cells, the antibody having antibody-dependent cytotoxic activity in the organism; The method for treating a living body of epithelial cancer, which comprises the step (b) of administering the contained cancer therapeutic agent, can be used in the step (a). In the step (a), preferably, a KLRG1-positive immune cell of the present invention, which is a cell separator that extracorporeally circulates peripheral blood of a living body and has a higher affinity for a KLRG1-positive immune cell than a KLRG1-negative immune cell. KLRG1-positive immune cells can be selectively reduced through the separating material. The cancer-specific membrane antigen expressed in the epithelial cancer cells is preferably HER2, and the antibody against the cancer-specific membrane antigen expressed in the epithelial cancer cells is preferably trastuzumab.

その他、別の態様として、患者体内から取り出した免疫細胞を細胞培養処理により増殖させ、該免疫細胞を本発明の分離材と接触させ、KLRG1陽性免疫細胞を選択的に除去する。その後、KLRG1陽性免疫細胞が選択的に除去された被処理液を前記患者に再び戻すことができる。 In another embodiment, immune cells taken out from the patient's body are grown by cell culture treatment, and the immune cells are brought into contact with the separating material of the present invention to selectively remove KLRG1-positive immune cells. Then, the liquid to be treated from which the KLRG1-positive immune cells have been selectively removed can be returned to the patient again.

また本発明のペプチドはKLRG1陽性免疫細胞に親和性を有することから、例えば、FITCなどの蛍光物質を導入し、KLRG1陽性免疫細胞を標識して、フローサイトメーターを用いたKLRG1陽性免疫細胞の検出、分析用に用いたり、またはマイクロプレートに固定して、抗体やタンパクに代わるELISA法としてKLRG1またはKLRG1陽性免疫細胞の検出、分析用に有効に用いることができる。また、これらペプチドをKLRG1陽性免疫細胞が関与している疾患に対する医薬品及び医薬品のキャリヤー物質としても有効に用いることが期待できる。例えば、KLRG1陽性免疫細胞中のKLRG1とKLRG1に対するリガンド(例えば、E−カドヘリン)との結合を阻害し、KLRG1を介した抑制性シグナルを遮断することができる。したがって、上皮性癌細胞で発現する癌特異的膜抗原に対する、抗体依存性細胞障害活性を有する抗体と、前記癌特異的膜抗原が陽性であり、かつKLRG1に対するリガンドが陽性である上皮性癌細胞と単核球を含む細胞集団とを接触させても、従来は上皮性癌細胞に対する細胞障害活性が発揮されなかった場合においても、十分な細胞障害活性を得ることが期待できる。 In addition, since the peptide of the present invention has an affinity for KLRG1-positive immune cells, for example, a fluorescent substance such as FITC is introduced, KLRG1-positive immune cells are labeled, and KLRG1-positive immune cells are detected using a flow cytometer. It can be effectively used for detection and analysis of KLRG1 or KLRG1-positive immune cells as an ELISA method instead of an antibody or protein after being used for analysis or immobilized on a microplate. In addition, it can be expected that these peptides can be effectively used as drugs and carrier substances for drugs against diseases in which KLRG1-positive immune cells are involved. For example, the binding of KLRG1 in a KLRG1-positive immune cell to a ligand for KLRG1 (eg, E-cadherin) can be inhibited, and the inhibitory signal mediated by KLRG1 can be blocked. Therefore, an antibody having antibody-dependent cytotoxic activity against a cancer-specific membrane antigen expressed in epithelial cancer cells, and an epithelial cancer cell positive for the cancer-specific membrane antigen and positive for a ligand for KLRG1 It is expected that sufficient cytotoxic activity will be obtained even when the cell population containing mononuclear cells is brought into contact with the cell population in the past, even when the cytotoxic activity against epithelial cancer cells was not exerted.

以下の実施例により、本発明を更に詳細に説明するが、本発明はこれらによって限定されるものではない。 The present invention will be described in more detail with reference to the following examples, but the present invention is not limited thereto.

<水不溶性担体の作製>
ポリプロピレン製不織布(以下PP不織布と略す、TAPYRUS社製 P080HW−00F、目付80g/m2、平均繊維径5.1μm)0.158m2(面積)を脱酸素剤とともに酸素低透過性袋に封入し十分に酸素を除いた後、−78℃にて100kGyのγ線を照射した。グリシジルメタクリレート(GMA)50mLを450mLのメタノールに溶解し、40℃にて60分間窒素を通気して脱酸素処理を行った。耐圧ガラス容器に上記の不織布をすばやく入れ、減圧後、上記の溶液を引き込み40℃にて15分反応させた。反応後、取り出した不織布をジメチルホルムアミド及びメタノールにより洗浄し、35℃にて1晩真空乾燥することで、GMAがグラフト重合されたPP不織布(以下、GMA不織布と略す)を得た。γ線照射前からの重量増加より算出されるグラフト率は99%であった。ここでグラフト率(%)は以下の式で示される値である。
グラフト率(%)=[(グラフト後不織布重量−グラフト前不織布重量)/(グラフト前不織布重量)]×100
<Preparation of water-insoluble carrier>
Polypropylene non-woven fabric (hereinafter abbreviated as PP non-woven fabric, P080HW-00F manufactured by TAPYRUS Co., Ltd., weight 80 g/m 2 , average fiber diameter 5.1 μm) 0.158 m 2 (area) is enclosed in a low oxygen permeability bag together with an oxygen scavenger. After sufficiently removing oxygen, it was irradiated with 100 kGy γ-rays at −78° C. 50 mL of glycidyl methacrylate (GMA) was dissolved in 450 mL of methanol, and nitrogen was passed through at 40° C. for 60 minutes for deoxidation treatment. The above-mentioned non-woven fabric was quickly put in a pressure-resistant glass container, and after the pressure was reduced, the above-mentioned solution was drawn in and reacted at 40°C for 15 minutes. After the reaction, the unwoven fabric taken out was washed with dimethylformamide and methanol and vacuum dried at 35° C. overnight to obtain a PP nonwoven fabric in which GMA was graft-polymerized (hereinafter, abbreviated as GMA nonwoven fabric). The graft ratio calculated from the increase in weight before irradiation with γ rays was 99%. Here, the graft ratio (%) is a value represented by the following formula.
Graft ratio (%)=[(weight of nonwoven fabric after grafting-weight of nonwoven fabric before grafting)/(weight of nonwoven fabric before grafting)]×100

次に、上記GMA不織布の10gを、エタノールに浸漬し、続いて注射用水にて洗浄することで親水化処理した後、25wt%アンモニア水の200mlに浸漬し、25℃で4時間静置した。反応後、取り出した不織布を注射用水にて洗浄水が中性となるまで洗浄を行い、35℃にて1晩真空乾燥することで、アミノ化されたGMA不織布(以下、GMA(NH3)不織布と略す)を得た。 Next, 10 g of the GMA non-woven fabric was immersed in ethanol and then washed with water for injection to make it hydrophilic, then immersed in 200 ml of 25 wt% ammonia water, and allowed to stand at 25° C. for 4 hours. After the reaction, the non-woven fabric taken out was washed with water for injection until the washing water became neutral, and vacuum dried overnight at 35° C. to obtain an aminated GMA non-woven fabric (hereinafter, referred to as GMA(NH3) non-woven fabric). Abbreviated).

次に、GMA(NH3)不織布1.0gを、ポリエチレングリコールジグリシジルエーテル(ナガセケムテックス(株)製、デナコールEX−821、n=4、以下EX−821と略す)12mlに浸漬し、反応容器全体をゆるやかに振とうしながら25℃で22時間保った。その後、取り出した不織布を注射用水で10回洗浄を行い、40℃にて1晩真空乾燥することで、GMA(NH3)不織布にEX−821を導入した不織布(以下、EX−821不織布と略す)を得た。 Next, 1.0 g of GMA (NH3) non-woven fabric was immersed in 12 ml of polyethylene glycol diglycidyl ether (Nagase Chemtex Co., Ltd., Denacol EX-821, n=4, hereinafter abbreviated as EX-821), and a reaction vessel. The whole was kept at 25° C. for 22 hours with gentle shaking. After that, the taken out non-woven fabric is washed 10 times with water for injection and vacuum dried at 40° C. overnight to form a non-woven fabric in which EX-821 is introduced into the GMA(NH3) non-woven fabric (hereinafter, abbreviated as EX-821 non-woven fabric) Got

<ペプチドの作製>
表1に記載のペプチドをFmoc固相法により合成した。p-alkoxybenzyl alcohol樹脂を担体としてペプチド鎖を伸長した。N末端にビオチンを修飾したペプチド(ペプチド1、7)は、ペプチド鎖伸長後、N末端のFmocを脱保護した後、D(+)-ビオチンをペプチド鎖の伸長と同様にN末端に結合した。リシンの側鎖にビオチンを修飾したペプチド(ペプチド2、8)は、Fmoc-Lys(biotin)-OHを用いてペプチド伸長した。N末端をアセチル化したペプチド(ペプチド3、4,5、6、9、10)は、ペプチド鎖伸長後、N末端のFmocを脱保護した後、無水酢酸と反応させ、アセチル化した。N末端未修飾のペプチド(ペプチド2、8)はN末端のFmocを脱保護した。ペプチド6は、スペーサーとしてFmoc−(保護)アミノ酸の代わりに8-(Fmoc-amino)-3,6-dioxaoctanoic acid(Peptides International,Inc.社製、Fmoc−mini−PEG)を用い、繰り返し3回伸長し導入した。次に、脱樹脂、脱保護を行い粗ペプチドを得た後、逆相高速液体クロマトグラフィーで精製を行い、得られた精製ペプチドはMALDI TOF−MSにより分子量を確認し、同定した。
<Preparation of peptide>
The peptides listed in Table 1 were synthesized by the Fmoc solid phase method. The peptide chain was extended using p-alkoxybenzyl alcohol resin as a carrier. For peptides modified with biotin at the N-terminus (peptides 1 and 7), after extension of the peptide chain, deprotection of Fmoc at the N-terminus was followed by attachment of D(+)-biotin to the N-terminus in the same manner as the extension of the peptide chain. .. The peptides in which the side chain of lysine was modified with biotin (peptides 2 and 8) were peptide-extended using Fmoc-Lys(biotin)-OH. The N-terminal acetylated peptides (peptides 3, 4, 5, 6, 9, and 10) were acetylated by reacting with acetic anhydride after deprotecting the N-terminal Fmoc after peptide chain extension. Unmodified N-terminal peptides (peptides 2, 8) deprotected N-terminal Fmoc. Peptide 6 was repeatedly used three times using 8-(Fmoc-amino)-3,6-dioxaoctanoic acid (Peptides International, Inc., Fmoc-mini-PEG) instead of Fmoc-(protected) amino acid as a spacer. It was extended and introduced. Next, after deresinating and deprotecting to obtain a crude peptide, purification was carried out by reverse phase high performance liquid chromatography, and the obtained purified peptide was identified by confirming its molecular weight by MALDI TOF-MS.

Figure 2012077006
Figure 2012077006

<細胞分離材の作製>
上記で得られたEX−821不織布を直径0.68cmの円に切断したもの8枚を、エタノールに浸漬し、続いてカルシウム、マグネシウムを含まないリン酸緩衝液(以下、PBS(−)と略す)にて洗浄することで親水化処理した。また表1に記載のペプチドをPBS(−)に溶解し200nmol/mLのペプチド溶液を調製した。親水化処理した8枚のEX−821不織布をペプチド溶液0.8mlに浸漬し、37℃で40時間静置し、ペプチドを固定した不織布(以下、ペプチド固定不織布と略す)を得た。次にペプチド固定不織布をPBS(−)で洗浄後、0.2%ポリオキシエチレンソルビタンモノラウレート/PBS(−)溶液0.8mlに浸漬し、常温で2.5時間静置した後、PBS(−)で洗浄し、細胞分離材を得た。
<Preparation of cell separation material>
Eight pieces obtained by cutting the EX-821 non-woven fabric obtained above into circles having a diameter of 0.68 cm were immersed in ethanol, and subsequently, a phosphate buffer solution containing no calcium or magnesium (hereinafter abbreviated as PBS(−)). ), it was made hydrophilic. Further, the peptides shown in Table 1 were dissolved in PBS(-) to prepare a 200 nmol/mL peptide solution. Eight hydrophilic EX-821 non-woven fabrics were immersed in 0.8 ml of the peptide solution and allowed to stand at 37° C. for 40 hours to obtain a non-woven fabric on which peptides were immobilized (hereinafter abbreviated as peptide-immobilized non-woven fabric). Next, the peptide-immobilized nonwoven fabric is washed with PBS(-), immersed in 0.8 ml of 0.2% polyoxyethylene sorbitan monolaurate/PBS(-) solution, and allowed to stand at room temperature for 2.5 hours, and then PBS. The cells were washed with (-) to obtain a cell separation material.

<細胞分離器の作製>
入口と出口を有する容積約1mlの容器に、上記細胞分離材4枚と充填液としてPBS(−)とを充填して、細胞分離器を得た。
<Preparation of cell separator>
A container having an inlet and an outlet and having a volume of about 1 ml was filled with the above-mentioned four cell separation materials and PBS(-) as a filling solution to obtain a cell separator.

実施例1〜6、比較例1〜4
<KLRG1陽性NK細胞の選択的除去性能評価>
細胞分離器のカラム入口からシリンジポンプを用いて生理食塩液4mlを流速1.0ml/minで通液し、プライミングした。続いて、抗凝固剤としてACD−A(acid citrate dextrose solution−A)液を添加したヒト新鮮血液2.0ml(血液:ACD−A液=8:1)を、細胞分離器のカラム入口から流速0.2ml/minで通液し、カラム出口から処理後の血液を回収した。
Examples 1-6, Comparative Examples 1-4
<Evaluation of selective removal performance of KLRG1-positive NK cells>
4 ml of physiological saline was passed through the column inlet of the cell separator using a syringe pump at a flow rate of 1.0 ml/min for priming. Subsequently, 2.0 ml of human fresh blood (blood:ACD-A solution=8:1) to which ACD-A (acid citrate dextrose solution-A) solution was added as an anticoagulant was flowed from the column inlet of the cell separator. The liquid was passed at 0.2 ml/min, and the treated blood was collected from the column outlet.

細胞分離器で処理した前後の血液中のリンパ球数をミクロセルカウンター(シスメックス(株)社製、XT−1800i)を用いて測定した。またそれらの血液を各々PBS(−)で1:1に希釈したものをFicoll−Paque PLUS(GE Healthcare社製)の上に重層し、400×gで30分間遠心して単核球浮遊液を採取した。この単核球浮遊液を、直接または間接的に蛍光標識した抗ヒトCD3抗体、抗ヒトCD56抗体、抗ヒトKLRG1抗体を用いて染色し、フローサイトメーター(BECTON DICKINSON社製、FACS CantoII)を用いてリンパ球中のNK細胞数、NK細胞のKLRG1陽性率を測定した。 The number of lymphocytes in blood before and after the treatment with the cell separator was measured using a microcell counter (XT-1800i manufactured by Sysmex Corporation). In addition, each of these bloods was diluted 1:1 with PBS(-) and overlaid on Ficoll-Paque PLUS (manufactured by GE Healthcare), and centrifuged at 400 xg for 30 minutes to collect a mononuclear cell suspension. did. This mononuclear cell suspension was directly or indirectly fluorescently labeled with anti-human CD3 antibody, anti-human CD56 antibody, anti-human KLRG1 antibody, and a flow cytometer (BECTON DICKINSON, FACS CantoII) was used. The number of NK cells in lymphocytes and the KLRG1 positive rate of NK cells were measured.

細胞分離器で処理した前後の血液中のNK細胞数、NK細胞のKLRG1陽性率から、KLRG1陽性NK細胞除去率とKLRG1陰性NK細胞除去率を算出し、その比(=KLRG1陽性NK細胞除去率/KLRG1陰性NK細胞除去率)からKLRG1陽性NK細胞の選択的除去性能を評価した。結果を表2に示す。 The KLRG1-positive NK cell removal rate and the KLRG1-negative NK cell removal rate were calculated from the number of NK cells in the blood before and after the treatment with a cell separator and the KLRG1-positive rate of the NK cells, and the ratio (=KLRG1-positive NK cell removal rate) was calculated. /KLKL1-negative NK cell removal rate), the selective removal performance of KLRG1-positive NK cells was evaluated. The results are shown in Table 2.

参考例1
水不溶性担体をEX−821不織布の代わりにGMA不織布を用い、またペプチドの代わりにリコンビナントヒトE−カドヘリン(R&D Systems Inc.社製、Recombinant Human E−Cadherin/Fc Chimera)を水不溶性担体に固定した以外は実施例1と同様に、細胞分離材のKLRG1陽性NK細胞の選択的除去性能を評価した。結果を表2に示す。
Reference example 1
As the water-insoluble carrier, a GMA nonwoven fabric was used instead of the EX-821 nonwoven fabric, and instead of the peptide, recombinant human E-cadherin (Recombinant Human E-Cadherin/Fc Chimera, manufactured by R&D Systems Inc.) was immobilized on the water-insoluble carrier. As in Example 1 except for the above, the ability of the cell separating material to selectively remove KLRG1-positive NK cells was evaluated. The results are shown in Table 2.

Figure 2012077006
Figure 2012077006

実施例7及び8、比較例5及び6
<KLRG1陽性リンパ球およびKLRG1陰性リンパ球へのペプチド結合性能評価>
抗凝固剤としてACD−A液を添加したヒト新鮮血液から、上記方法を用いて単核球浮遊液を作製した。またFcレセプターブロッキング剤(MBL社製、ClearBack(Human Fc receptor blocking reagent))を用いて単核球浮遊液をFcレセプターのブロッキング処理をした。
Examples 7 and 8, Comparative Examples 5 and 6
<Evaluation of peptide binding ability to KLRG1-positive lymphocytes and KLRG1-negative lymphocytes>
A mononuclear cell suspension was prepared from the fresh human blood to which the ACD-A solution was added as an anticoagulant by using the above method. The mononuclear cell suspension was subjected to Fc receptor blocking treatment using an Fc receptor blocking agent (ClearBack (Human Fc receptor blocking reagent) manufactured by MBL).

単核球浮遊液を1次抗体としてマウス抗ヒトKLRG1ポリクローナル抗体(Abnova Coporation社製、KLRG1 purified MaxPab mouse polyclonal antibody(B01P))と2次抗体としてFITC標識ヤギ抗マウスIgG(γ鎖)抗体(Southern Biotech社製)を用いて染色し、KLRG1陽性リンパ球とKLRG1陰性リンパ球を識別した。続いて表3のbiotin標識ペプチドと反応後、PE Streptavidin(BD Biosciences社製)で標識し、フローサイトメーターを用いてKLRG1陽性リンパ球へのペプチド陽性率およびKLRG1陰性リンパ球へのペプチド陽性率を評価した。結果を表3に示す。 A mouse anti-human KLRG1 polyclonal antibody (manufactured by Abnova Corporation, KLRG1 purified MaxPab mouse polyclonal antibody (B01P)) using a mononuclear cell suspension as a primary antibody, and a FITC-labeled goat anti-mouse IgG (γC) antibody as a secondary antibody. (Manufactured by Biotech) was used to distinguish between KLRG1-positive lymphocytes and KLRG1-negative lymphocytes. Then, after reacting with the biotin-labeled peptide of Table 3, it was labeled with PE Streptavidin (manufactured by BD Biosciences), and the peptide-positive rate to KLRG1-positive lymphocytes and the peptide-positive rate to KLRG1-negative lymphocytes were measured using a flow cytometer. evaluated. The results are shown in Table 3.

なお、本評価系を確認するために、biotin標識ペプチドの代わりにヒトKLRG1に対して交差反応するハムスター抗マウスKLRG1モノクローナル抗体(eBioscience社製、Biotin anti−mouse KLRG1/MAFA)を使用し、同様にKLRG1陽性リンパ球およびKLRG1陰性リンパ球へのKLRG1モノクローナル抗体結合性能を評価した。結果、KLRG1陽性リンパ球へのKLRG1モノクローナル抗体の陽性率は96.5%、KLRG1陰性リンパ球へのKLRG1モノクローナル抗体の陽性率は4.2%であり、本評価系を用いることでKLRG1陽性リンパ球およびKLRG1陰性リンパ球へのペプチド結合性能が評価できることを確認した。 In order to confirm the present evaluation system, a hamster anti-mouse KLRG1 monoclonal antibody (manufactured by eBioscience, Biotin anti-mouse KLRG1/MAFA) that cross-reacts with human KLRG1 was used instead of the biotin-labeled peptide, and the same manner was used. The ability of KLRG1 monoclonal antibody to bind to KLRG1-positive lymphocytes and KLRG1-negative lymphocytes was evaluated. As a result, the positive rate of KLRG1 monoclonal antibody to KLRG1-positive lymphocytes was 96.5%, and the positive rate of KLRG1 monoclonal antibody to KLRG1-negative lymphocytes was 4.2%. It was confirmed that the peptide binding ability to spheres and KLRG1-negative lymphocytes can be evaluated.

Figure 2012077006
Figure 2012077006

Claims (15)

Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下のペプチドが固定されている水不溶性担体からなる、KLRG1陽性免疫細胞分離材。 A peptide containing the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) and having a total number of constituent amino acid residues of 80 or less is immobilized. A material for separating KLRG1-positive immune cells, which comprises a water-insoluble carrier. 配列番号1のアミノ酸配列を含むペプチドは、そのC末端側に水不溶性担体に固定するための活性アミノ酸残基が付加されている、請求項1に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to claim 1, wherein an active amino acid residue for fixing to a water-insoluble carrier is added to the C-terminal side of the peptide containing the amino acid sequence of SEQ ID NO: 1. 前記活性アミノ酸残基がLysである、請求項2に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to claim 2, wherein the active amino acid residue is Lys. 配列番号1のペプチドは、スペーサーを介して前記水不溶性担体に固定されている、請求項1に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to claim 1, wherein the peptide of SEQ ID NO: 1 is immobilized on the water-insoluble carrier via a spacer. 前記スペーサーは、前記水不溶性担体との結合末端側に活性アミノ酸残基が付加されている、請求項4に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to claim 4, wherein the spacer has an active amino acid residue added to the binding terminal side with the water-insoluble carrier. 前記活性アミノ酸残基がLysである、請求項4または5に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to claim 4 or 5, wherein the active amino acid residue is Lys. 前記水不溶性担体が織布もしくは不織布である、請求項1〜6のいずれか1項に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to any one of claims 1 to 6, wherein the water-insoluble carrier is a woven fabric or a non-woven fabric. 前記構成アミノ酸残基の総数が50個以下である、請求項1〜7のいずれか1項に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to any one of claims 1 to 7, wherein the total number of the constituent amino acid residues is 50 or less. 前記構成アミノ酸残基の総数が40個以下である、請求項1〜7のいずれか1項に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to any one of claims 1 to 7, wherein the total number of the constituent amino acid residues is 40 or less. 前記構成アミノ酸残基の総数が30個以下である、請求項1〜7のいずれか1項に記載のKLRG1陽性免疫細胞分離材。 The KLRG1-positive immune cell separating material according to any one of claims 1 to 7, wherein the total number of the constituent amino acid residues is 30 or less. Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列からなるペプチドがスペーサーを介して前記水不溶性担体に固定されている、KLRG1陽性免疫細胞分離材。 KLRG1 in which a peptide consisting of the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) is immobilized on the water-insoluble carrier via a spacer. Positive immune cell separation material. Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下である、KLRG1陽性免疫細胞親和性ペプチド。 KLRG1-positive, containing the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1), and having a total number of constituent amino acid residues of 80 or less Immune cell affinity peptide. 配列番号1のアミノ酸配列のC末端側にLysを有する、請求項12に記載のKLRG1陽性免疫細胞親和性ペプチド。 The KLRG1-positive immune cell affinity peptide according to claim 12, which has Lys at the C-terminal side of the amino acid sequence of SEQ ID NO: 1. Asp−Trp−Val−Ile−Pro−Pro−Ile−Ser−Cys−Pro−Glu−Asn(配列番号1)のアミノ酸配列を含み、かつ構成アミノ酸残基の総数が80個以下であるペプチドを含む、KLRG1陽性免疫細胞分離剤。 Includes a peptide containing the amino acid sequence of Asp-Trp-Val-Ile-Pro-Pro-Ile-Ser-Cys-Pro-Glu-Asn (SEQ ID NO: 1) and having a total number of constituent amino acid residues of 80 or less. , A KLRG1-positive immune cell separating agent. 請求項1〜11のいずれか1項に記載のKLRG1陽性免疫細胞分離材にKLRG1陽性免疫細胞を含有する溶液を接触させることを含む、KLRG1陽性免疫細胞を分離する方法。 A method for separating KLRG1-positive immune cells, which comprises contacting a solution containing KLRG1-positive immune cells with the KLRG1-positive immune cell separating material according to any one of claims 1 to 11.
JP2010220872A 2010-09-30 2010-09-30 Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method Withdrawn JP2012077006A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2010220872A JP2012077006A (en) 2010-09-30 2010-09-30 Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2010220872A JP2012077006A (en) 2010-09-30 2010-09-30 Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method

Publications (1)

Publication Number Publication Date
JP2012077006A true JP2012077006A (en) 2012-04-19

Family

ID=46237664

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2010220872A Withdrawn JP2012077006A (en) 2010-09-30 2010-09-30 Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method

Country Status (1)

Country Link
JP (1) JP2012077006A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016501526A (en) * 2012-12-06 2016-01-21 エンリヴェックス セラピューティクス リミテッド A therapeutic apoptotic cell preparation, its production method and its use

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016501526A (en) * 2012-12-06 2016-01-21 エンリヴェックス セラピューティクス リミテッド A therapeutic apoptotic cell preparation, its production method and its use
US10077426B2 (en) 2012-12-06 2018-09-18 Enlivex Therapeutics Ltd Therapeutic apoptotic cell preparations, method for producing same and uses thereof
JP2019047813A (en) * 2012-12-06 2019-03-28 エンリヴェックス セラピューティクス リミテッド Therapeutic apoptotic cell preparations, method for producing the same and uses thereof
US10927343B2 (en) 2012-12-06 2021-02-23 Enlivex Therapeutics Ltd Therapeutic apoptotic cell preparations, method for producing same and uses thereof

Similar Documents

Publication Publication Date Title
Jayaraman et al. CAR-T design: Elements and their synergistic function
Hou et al. Navigating CAR-T cells through the solid-tumour microenvironment
AU2014337195B2 (en) Chimeric receptor that triggers antibody-dependent cell cytotoxicity against multiple tumors
US20190276540A1 (en) Compositions and methods for tcr reprogramming using fusion proteins
Shin et al. Enhancement of the tumor penetration of monoclonal antibody by fusion of a neuropilin-targeting peptide improves the antitumor efficacy
US20180154064A1 (en) Methods and devices for removal of immunosuppressive ligands
EP4321171A2 (en) Sirp alpha-antibody fusion proteins
JP2023052073A (en) Cell surface conjugates and related cell compositions and methods
AU2019206573B2 (en) Immune cells expressing a chimeric antigen receptor
WO2021027867A1 (en) Chimeric antigen receptor, construction method therefor and application thereof
EP3518943A1 (en) Methods of adoptive cell therapy
Quazi An overview of CAR T cell mediated B cell maturation antigen therapy
US10106601B2 (en) Inhibition of PLGF to treat philadelphia chromosome positive leukemia
CN111944053B (en) anti-BCMA CAR and expression vector and application thereof
KR101364428B1 (en) Cancer therapeutic agent comprising antibody against cancer-specific membrane antigen
JP2023519970A (en) Enhanced antigen-negative cell death in antigen-targeted immunotherapy
JP2012077006A (en) Klrg1-positive immunocyte-affinity peptide, klrg1-positive immunocyte separation material and klrg1-positive immunocyte separation method
CN116814664A (en) Preparation and application of CEA chimeric antigen receptor T cells for expanding tumor recognition epitope
Shin et al. Characterization of monoclonal antibodies against human leukocyte common antigen (CD45)
CN112048021A (en) ROR 2-targeted chimeric antigen receptor, expression gene, expression vector, T cell and application thereof
CN113498418A (en) Chimeric Antigen Receptors (CARs) and their use in medicine
JP5781300B2 (en) Cancer therapeutic agent containing antibody
JP2015092865A (en) Humanized anti-cd20 chimeric antigen receptor
CN116063569B (en) EPHA2 chimeric antigen receptor and uses thereof
WO2023072279A1 (en) Sirpa mutant and application thereof

Legal Events

Date Code Title Description
A711 Notification of change in applicant

Free format text: JAPANESE INTERMEDIATE CODE: A712

Effective date: 20120419

A300 Application deemed to be withdrawn because no request for examination was validly filed

Free format text: JAPANESE INTERMEDIATE CODE: A300

Effective date: 20131203