JPWO2012008595A1 - Production method of anti-LR11 antibody and standard for immunological measurement - Google Patents

Production method of anti-LR11 antibody and standard for immunological measurement Download PDF

Info

Publication number
JPWO2012008595A1
JPWO2012008595A1 JP2012524618A JP2012524618A JPWO2012008595A1 JP WO2012008595 A1 JPWO2012008595 A1 JP WO2012008595A1 JP 2012524618 A JP2012524618 A JP 2012524618A JP 2012524618 A JP2012524618 A JP 2012524618A JP WO2012008595 A1 JPWO2012008595 A1 JP WO2012008595A1
Authority
JP
Japan
Prior art keywords
cancer
derived
tumor cells
soluble
urine
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.)
Pending
Application number
JP2012524618A
Other languages
Japanese (ja)
Inventor
海老沼 宏幸
宏幸 海老沼
耕平 田久保
耕平 田久保
深町 勇
勇 深町
彩舟 吉田
彩舟 吉田
英明 武城
英明 武城
知昭 中世古
知昭 中世古
齋藤 康
康 齋藤
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.)
Sekisui Medical Co Ltd
Original Assignee
Sekisui 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 Sekisui Medical Co Ltd filed Critical Sekisui Medical Co Ltd
Publication of JPWO2012008595A1 publication Critical patent/JPWO2012008595A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

抗LR11抗体を大量かつ効率的に製造するための手段及び免疫学的測定におけるLR11標準品を提供する。悪性腫瘍細胞由来又は尿由来LR11を用いることを特徴とする抗LR11抗体の製造法。A means for efficiently and efficiently producing anti-LR11 antibodies and LR11 standards in immunological measurements are provided. A method for producing an anti-LR11 antibody, comprising using malignant tumor cell-derived or urine-derived LR11.

Description

本発明は、効率的な抗LR11抗体の製造法及びLR11の免疫学的測定用標準品に関する。   The present invention relates to an efficient method for producing an anti-LR11 antibody and a standard for immunological measurement of LR11.

LR11(LDL receptor relative with 11 ligand−binding repeats、SorLA、SORL1とも称される)は、LDL受容体ファミリーに特徴的な構造を有する新規なLDL受容体類似タンパク質として同定された(特許文献1、非特許文献1)。免疫組織化学分析やin situ hybridization分析の結果、LR11は平滑筋細胞の遊走及び増殖によって形成された血管内膜肥厚部位で特異的に発現が亢進していることが報告されている(非特許文献2)。
本発明者らは、哺乳動物の血液中に可溶性LR11が存在していること、及び動脈硬化性疾患患者血液中の可溶性LR11濃度が健常者と比較して有意に高値であることを見出した(特許文献2)。本発明者らはさらに、特定の界面活性剤で試料中のLR11を処理することにより、抗LR11抗体を用いて血液中あるいは骨髄液中の可溶性LR11を簡便に測定する方法(特許文献3、非特許文献4)を開発し、様々な疾患における血液中若しくは骨髄液中の可溶性LR11の濃度を測定した結果、悪性腫瘍、特に白血病や悪性リンパ腫のような造血器腫瘍疾患で異常高値を示すことを確認し、LR11が新たな腫瘍マーカーになり得ることを見出した(特許文献4)。
LR11 (also referred to as LDL receptor relative with 11 ligand-binding repeats, SorLA, SORL1) has been identified as a novel LDL receptor-like protein having a structure characteristic of the LDL receptor family (Patent Document 1, Non-patent Document 1, Patent Document 1). As a result of immunohistochemical analysis and in situ hybridization analysis, it has been reported that LR11 is specifically expressed at an intimal thickening site formed by smooth muscle cell migration and proliferation (Non-Patent Literature). 2).
The present inventors have found that soluble LR11 is present in the blood of mammals, and that the soluble LR11 concentration in the blood of patients with arteriosclerotic disease is significantly higher than that in healthy subjects ( Patent Document 2). Furthermore, the present inventors further processed LR11 in a sample with a specific surfactant to easily measure soluble LR11 in blood or bone marrow fluid using an anti-LR11 antibody (Patent Document 3, Non-Patent Document 3). As a result of developing a patent document 4) and measuring the concentration of soluble LR11 in blood or bone marrow fluid in various diseases, it has been shown that abnormally high values are observed in malignant tumors, particularly hematopoietic tumor diseases such as leukemia and malignant lymphoma. As a result, LR11 was found to be a new tumor marker (Patent Document 4).

LR11の遺伝子配列は解明されている(特許文献1、非特許文献1)ものの、全長LR11は250kDaという大きな分子量を有しているため、組換え全長LR11タンパク質の大量発現には成功していない。また、LR11を分泌する培養細胞、LR11遺伝子を安定的に発現するLDL受容体欠損CHO細胞、全長LR11遺伝子を導入したCOS−7細胞も報告されている(非特許文献5)が、LR11の発現量が非常に僅かであるか、遺伝子工学的な処理が必要であった。このように、抗体を作製するための免疫原の供給源は限られているか、LR11産生細胞とするための加工が必要であった。   Although the gene sequence of LR11 has been elucidated (Patent Document 1, Non-Patent Document 1), since full-length LR11 has a large molecular weight of 250 kDa, large-scale expression of recombinant full-length LR11 protein has not been successful. In addition, cultured cells that secrete LR11, LDL receptor-deficient CHO cells that stably express the LR11 gene, and COS-7 cells into which the full-length LR11 gene has been introduced have been reported (Non-Patent Document 5). The amount was very small or required genetic engineering treatment. Thus, the source of the immunogen for producing the antibody is limited, or processing for obtaining an LR11 producing cell is necessary.

抗LR11抗体はポリクローナル抗体、モノクローナル抗体ともすでに市販されているが、上記した事情より、免疫原としてLR11の部分アミノ酸配列を有する合成ペプチドを使用して作製されている。そのため、当該市販抗体は、全長LR11など生物試料中のLR11に対する反応性が非常に弱いものであるか又はLR11を固相に固定化した場合にしか反応しないものであるなど、実用的に十分な性能を有する抗体とは言い難いものであった。   Although anti-LR11 antibodies are already commercially available as polyclonal antibodies and monoclonal antibodies, due to the circumstances described above, they are prepared using a synthetic peptide having a partial amino acid sequence of LR11 as an immunogen. For this reason, the commercially available antibody is practically sufficient, for example, it is very weak in reactivity to LR11 in a biological sample such as full-length LR11 or reacts only when LR11 is immobilized on a solid phase. It was hard to say that the antibody has performance.

また、血液中の可溶性LR11を、抗体を作製するための免疫原の供給源とするためには、大量の採血が必要であり供血者への負担を考慮すると非現実的である。   Moreover, in order to use soluble LR11 in blood as a source of an immunogen for producing an antibody, a large amount of blood is required, which is unrealistic considering the burden on the donor.

上記した各事情は、LR11の免疫学的測定用標準品を作製する際の標品LR11の供給源として考えた場合においても同様である。
以上のように、抗LR11抗体を作製する際の免疫原の供給源、あるいは免疫学的測定用標準品を作製する際の標品LR11の供給源として、侵襲性が低い手段で大量かつ簡便にLR11を収集することができ、さらに生物試料中の存在状態を維持した状態でLR11を収集できる供給源が求められていたのである。
The above-mentioned circumstances are the same even when considered as a supply source of the standard LR11 when producing a standard for immunological measurement of LR11.
As described above, as a source of an immunogen for producing an anti-LR11 antibody or a source of a standard LR11 for producing a standard for immunological measurement, it can be easily produced in a large amount by means of low invasiveness. There was a need for a source that can collect LR11 and that can collect LR11 while maintaining the presence in the biological sample.

特開平9−163988号公報Japanese Patent Laid-Open No. 9-163988 WO2008/155891WO2008 / 155891 WO2009/116268WO2009 / 116268 特許2009−285492号公報Japanese Patent No. 2009-285492

J.Biol.Chem.1996;271,24761−24768J. et al. Biol. Chem. 1996; 271, 24761-24768. Arterioscler.Thromb.Vasc.Biol.1999;19,2687−2695Arterioscler. Thromb. Vasc. Biol. 1999; 19, 2687-2695. J.Clin.Invest.2008;118,2733−2746J. et al. Clin. Invest. 2008; 118, 2733-2746 Clin.Chem.2009;55,1801−1808Clin. Chem. 2009; 55, 1801-1808 Circ.Res.2004;94,752−758Circ. Res. 2004; 94, 752-758.

本発明の課題は、抗LR11抗体を大量かつ効率的に製造するための手段、特に生物試料中のLR11に反応する抗LR11抗体を製造するための手段、及び免疫学的測定におけるLR11の標準品を提供することにある。   The object of the present invention is to provide a means for efficiently and efficiently producing anti-LR11 antibodies, particularly a means for producing anti-LR11 antibodies that react with LR11 in a biological sample, and a standard product of LR11 in immunological measurement. Is to provide.

本発明者らは、LR11の新たな供給源について検討した結果、悪性腫瘍細胞の表面にLR11が発現すること及び悪性腫瘍細胞の培養上清中にLR11が分泌されること、また、ヒトの尿中にも可溶性LR11が存在していることを見出した。さらに悪性腫瘍細胞由来及びヒト尿由来のLR11を免疫原や抗体の特異性評価用抗原として使用することにより、全長LR11など生物試料中のLR11に反応する抗体の作製に成功した。また、これらの悪性腫瘍細胞由来及びヒト尿由来LR11は、LR11の免疫学的測定用標準品としても有用であることを見出した。本発明はかかる知見に基づいて完成したものである。   As a result of studying a new source of LR11, the present inventors have found that LR11 is expressed on the surface of malignant tumor cells, that LR11 is secreted into the culture supernatant of malignant tumor cells, and human urine. It was found that soluble LR11 was also present therein. Furthermore, by using LR11 derived from malignant tumor cells and human urine as an antigen for evaluating the specificity of an immunogen or antibody, an antibody that reacts with LR11 in a biological sample such as full-length LR11 was successfully produced. Further, it was found that these malignant tumor cell-derived and human urine-derived LR11 are also useful as a standard for immunological measurement of LR11. The present invention has been completed based on such findings.

すなわち、本発明は、悪性腫瘍細胞由来又は尿由来のLR11を用いることを特徴とする抗LR11抗体の製造法を提供するものである。
また、本発明は、悪性腫瘍細胞由来又は尿由来のLR11を含有するLR11の免疫学的測定用標準品を提供するものである。
なお、本明細書において、「LR11」とは、特にことわらない限り抗LR11抗体との反応性を有するタンパク質を指す。すなわち、「全長LR11」や「可溶性LR11」として公知のタンパク質やそれらが部分的に断片化されたり、修飾されたものをいう。
That is, this invention provides the manufacturing method of the anti- LR11 antibody characterized by using LR11 derived from a malignant tumor cell or urine.
The present invention also provides a standard for immunological measurement of LR11 containing LR11 derived from malignant tumor cells or urine.
In the present specification, “LR11” refers to a protein having reactivity with an anti-LR11 antibody unless otherwise specified. That is, it refers to proteins known as “full length LR11” or “soluble LR11” and those partially fragmented or modified.

本発明の方法によれば、LR11が細胞表面に発現している悪性腫瘍細胞自体、若しくは前記悪性腫瘍細胞の培養上清中に分泌される可溶性LR11、又はヒトの尿中に存在している可溶性LR11を、免疫原や特異性評価用抗原として用いることにより、工業的に有利にLR11に対する特異抗体の作製が可能となる。
また、悪性腫瘍細胞由来又は尿由来のLR11を含有する標準品を用いることで、正確にLR11を免疫学的測定することができる。
According to the method of the present invention, malignant tumor cells expressing LR11 on the cell surface itself, soluble LR11 secreted in the culture supernatant of the malignant tumor cells, or soluble present in human urine By using LR11 as an immunogen or an antigen for specificity evaluation, it becomes possible to produce a specific antibody against LR11 in an industrially advantageous manner.
Moreover, LR11 can be accurately measured immunologically by using a standard product containing LR11 derived from malignant tumor cells or urine.

各種造血器腫瘍細胞株におけるLR11の発現を、細胞溶解液及び細胞培養液を用いて、ウェスタンブロット法で検出した図である。It is the figure which detected the expression of LR11 in various hematopoietic tumor cell lines by Western blotting using a cell lysis solution and a cell culture solution. 各種造血器腫瘍細胞株の培養液中の可溶性LR11をELISAで測定した結果を表すグラフである。It is a graph showing the result of having measured soluble LR11 in the culture solution of various hematopoietic tumor cell lines by ELISA. 各種造血器腫瘍細胞株における細胞表面でのLR11の発現を、フローサイトメトリーで解析した結果を示した図である。It is the figure which showed the result of having analyzed the expression of LR11 on the cell surface in various hematopoietic tumor cell lines by flow cytometry. 各種上皮性悪性腫瘍細胞株における細胞表面でのLR11発現を、膜画分溶解液を用いて、ウェスタンブロット法で検出した図である。It is the figure which detected the LR11 expression on the cell surface in various epithelial malignant tumor cell lines by the western blotting using the membrane fraction solution. 各種上皮性悪性腫瘍細胞株の培養液中の可溶性LR11をELISAで測定した結果を表すグラフである。It is a graph showing the result of having measured soluble LR11 in the culture solution of various epithelial malignant tumor cell lines by ELISA. ヒト尿由来の可溶性LR11、CCRF−SB細胞株由来の可溶性LR11、ヒト血清由来可溶性LR11、COLO201細胞株由来の可溶性LR11を、ウェスタンブロット法で比較した図である。It is the figure which compared soluble LR11 derived from human urine, soluble LR11 derived from CCRF-SB cell line, soluble LR11 derived from human serum, and soluble LR11 derived from COLO201 cell line by Western blotting. NB−4細胞株を腹腔内免疫したマウスの血清中の抗LR11抗体価を測定した結果を表すグラフである。It is a graph showing the result of having measured the anti- LR11 antibody titer in the serum of the mouse | mouth which immunized intraperitoneally with NB-4 cell strain.

本発明において抗LR11抗体の作製又はLR11の免疫学的測定用標準品に用いられるLR11は、悪性腫瘍細胞由来又は尿由来である。   In the present invention, LR11 used for preparation of an anti-LR11 antibody or a standard for immunological measurement of LR11 is derived from malignant tumor cells or urine.

本発明者らの知見によれば、悪性腫瘍細胞表面にはLR11が顕著に発現しており、また悪性腫瘍細胞の培養液中には高濃度の可溶性LR11が分泌されている。従って、細胞表面にLR11を発現している悪性腫瘍細胞を直接免疫原として用いることや、当該細胞の培養液から可溶性LR11を採取して、免疫原、抗LR11抗体の特異性評価、あるいは免疫学的測定用標準品として使用することができる。   According to the knowledge of the present inventors, LR11 is remarkably expressed on the surface of malignant tumor cells, and a high concentration of soluble LR11 is secreted in the culture solution of malignant tumor cells. Therefore, malignant tumor cells expressing LR11 on the cell surface can be used directly as an immunogen, or soluble LR11 can be collected from the culture medium of the cells to evaluate the specificity of the immunogen or anti-LR11 antibody, or immunology It can be used as a standard for standard measurement.

本発明で使用できる悪性腫瘍細胞は、例えば、ATCC(American Type Culture Collection)やヒューマンサイエンス研究資源バンク(Health Science Research Resources Bank)等に登録されている細胞株で、LR11が細胞表面に発現しているものであれば特に制限されること無く使用することができる。悪性腫瘍の種類としては、造血器腫瘍または上皮性腫瘍が挙げられ、造血器腫瘍が好ましい。   Examples of malignant tumor cells that can be used in the present invention are cell lines registered in the ATCC (American Type Culture Collection), the Human Science Research Resource Bank, etc., and LR11 is expressed on the cell surface. If it is, it can be used without particular limitation. Examples of the type of malignant tumor include hematopoietic tumor or epithelial tumor, and hematopoietic tumor is preferable.

造血器腫瘍の例として、白血病及び悪性リンパ腫が挙げられる。白血病には急性白血病及び慢性白血病が含まれ、悪性リンパ腫には非ホジキンリンパ腫が含まれる。このうち、急性白血病及び悪性リンパ腫が好ましい。さらに具体的には、造血器腫瘍細胞のうち、急性骨髄性白血病由来株のHL−60、ML−2、NB−4、急性リンパ性白血病由来株のNALL−1、MOLT−4、CCRF−SB、悪性リンパ腫由来株のDaudi、U937などが好ましく、特に、HL−60、ML−2、NB−4、MOLT−4、CCRF−SB、U937が好ましい。また、前記株化された細胞以外に、白血病患者および悪性リンパ腫患者由来の腫瘍細胞の細胞溶解液または細胞培養液から精製等して使用することもできる。   Examples of hematopoietic tumors include leukemia and malignant lymphoma. Leukemia includes acute leukemia and chronic leukemia, and malignant lymphoma includes non-Hodgkin lymphoma. Of these, acute leukemia and malignant lymphoma are preferred. More specifically, among hematopoietic tumor cells, HL-60, ML-2, and NB-4 of acute myeloid leukemia-derived strains, NALL-1, MOLT-4, CCRF-SB of acute lymphocytic leukemia-derived strains Malignant lymphoma-derived strains such as Daudi and U937 are preferable, and HL-60, ML-2, NB-4, MOLT-4, CCRF-SB and U937 are particularly preferable. In addition to the established cells, the cells can also be purified from a cell lysate or cell culture solution of tumor cells derived from leukemia patients and malignant lymphoma patients.

上皮性悪性腫瘍の例として、胃癌、肝臓癌、膵臓癌、肺癌、前立腺癌、膀胱癌、食道癌、乳癌、子宮頸癌、卵巣癌、結腸癌、大腸癌、腎臓癌、胆嚢癌、神経腫瘍(グリオーマ)、黒色細胞腫(メラノーマ)が含まれる。特に、上皮性悪性腫瘍のうち、胃癌由来株のMKN1、GCIY、肝臓癌由来株のHLE、HepG2、HuH−7、JHH−6、膵臓癌由来株のAsPC−1、BxPC−3、肺癌由来株のOka−C−1、前立腺癌由来株のPC−3、膀胱癌由来株のKMBC−2、食道癌由来株のT.Tn、乳癌由来株のYMB−1、子宮頸癌由来株のHeLa、卵巣癌由来株のOVISE、結腸癌・大腸癌由来株のCOLO201、腎臓癌由来株のCaki−1、胆嚢癌由来株のOCUG−1、神経腫瘍由来株のKINGS−1、黒色細胞腫由来株のG−361などが好ましい。また、前記株化された細胞以外に、上皮性悪性腫瘍患者由来の腫瘍細胞の細胞溶解液または細胞培養液から精製等して使用することもできる。   Examples of epithelial malignant tumors include stomach cancer, liver cancer, pancreatic cancer, lung cancer, prostate cancer, bladder cancer, esophageal cancer, breast cancer, cervical cancer, ovarian cancer, colon cancer, colon cancer, kidney cancer, gallbladder cancer, nerve tumor (Glioma), melanoma (melanoma). In particular, among epithelial malignant tumors, gastric cancer-derived strains MKN1, GCIY, liver cancer-derived strains HLE, HepG2, HuH-7, JHH-6, pancreatic cancer-derived strains AsPC-1, BxPC-3, lung cancer-derived strains Oka-C-1, prostate cancer-derived strain PC-3, bladder cancer-derived strain KMBC-2, esophageal cancer-derived strain T. Tn, breast cancer-derived strain YMB-1, cervical cancer-derived strain HeLa, ovarian cancer-derived strain OVISE, colon cancer / colon cancer-derived strain COLO201, kidney cancer-derived strain Caki-1, and gallbladder cancer-derived strain OCUG -1, neuronal tumor-derived strain KINGS-1, melanoma-derived strain G-361, and the like are preferable. In addition to the established cells, the cells can also be used after purification from a cell lysate or cell culture solution of tumor cells derived from patients with epithelial malignant tumors.

また、本発明者らは、尿中にも可溶性LR11が高濃度で存在することを見出した。尿としては、ヒト尿が特に好ましい。尿は、一般に夾雑タンパク質が少なく、また侵襲性が実質的に存在せず、かつ、大量に収集することが容易であるため、本発明におけるLR11の供給源として好適である。   The present inventors have also found that soluble LR11 is present in urine at a high concentration. As urine, human urine is particularly preferable. Urine is generally suitable as a supply source of LR11 in the present invention because it has few contaminating proteins, is substantially non-invasive, and can be easily collected in large quantities.

前記のように、免疫原として細胞表面にLR11を発現している悪性腫瘍細胞を直接用いることができる。一方、免疫原、抗LR11抗体の特異性評価用および免疫学的測定用標準品としては、悪性腫瘍細胞の溶解液若しくは培養液、又は尿から採取されたLR11を用いるのが好ましい。
悪性腫瘍細胞の溶解液若しくは培養液、又は尿からLR11を採取するには、例えば、RAP(受容体関連タンパク質)等のLR11親和性物質を担持させた不溶性担体に尿中の可溶性LR11を吸着させ、適当な緩衝液を用いて洗浄した後、前記不溶性担体に吸着した可溶性LR11を溶出させればよい。
As described above, malignant tumor cells expressing LR11 on the cell surface can be used directly as an immunogen. On the other hand, it is preferable to use LR11 collected from a lysate or culture solution of malignant tumor cells, or urine as a standard for immunogen, anti-LR11 antibody specificity evaluation and immunological measurement.
In order to collect LR11 from a lysate or culture solution of malignant tumor cells or urine, for example, soluble LR11 in urine is adsorbed on an insoluble carrier carrying an LR11 affinity substance such as RAP (receptor-related protein). After washing with an appropriate buffer, soluble LR11 adsorbed on the insoluble carrier may be eluted.

これらのLR11は、さらに、イオン交換クロマトグラフィー法、疎水クロマトグラフィー法、ゲルろ過法及び特異抗体によるアフィニティ法などにより精製して用いてもよい。   These LR11s may be further purified and used by ion exchange chromatography, hydrophobic chromatography, gel filtration, specific antibody affinity, and the like.

当該悪性腫瘍細胞由来又はヒト尿由来のLR11は、多量かつ安定して製造できるので、LR11の免疫学的測定用標準品として有用であり、また抗LR11抗体製造時の免疫原として、あるいは抗LR11抗体の特異性評価用として有用である。
抗LR11抗体作製時の免疫原と特異性評価には、由来の異なるLR11を組み合わせて用いてもよく、例えば、細胞表面にLR11が発現している悪性腫瘍細胞を免疫原に使用する場合は、抗LR11抗体の特異性評価では、尿由来の可溶性LR11を用いること、更に尿由来可溶性LR11を免疫原に使用する場合は、抗LR11抗体の特異性評価では、悪性腫瘍細胞から分泌された可溶性LR11を用いるか、若しくは細胞表面にLR11が発現している悪性腫瘍細胞そのものを用いることが考えられる。また、免疫原にLR11の部分アミノ酸配列を有する合成ペプチドを用いた場合においても、特異性評価用に悪性腫瘍細胞由来又はヒト尿由来のLR11を用いることによって、全長LR11など生物試料中のLR11に対する反応性が強い抗体を得ることができる。上記したように、本発明における免疫原あるいは標準品の供給源は、生物試料の形態としても、存在するLR11の形態としても、いわゆる天然の状態に極めて近いものであるため、合成ペプチドであったり遺伝子組み換えであったりした従来の供給源に対して、LR11の性状の保存の観点から特に有利であるといえる。
Since the LR11 derived from the malignant tumor cell or human urine can be produced in a large amount and stably, it is useful as a standard for immunological measurement of LR11, and is also useful as an immunogen when producing anti-LR11 antibody, or as anti-LR11. This is useful for evaluating the specificity of antibodies.
For immunogen and specificity evaluation at the time of anti-LR11 antibody production, LR11 of different origins may be used in combination. For example, when malignant tumor cells expressing LR11 on the cell surface are used as the immunogen, In the evaluation of the specificity of the anti-LR11 antibody, the soluble LR11 derived from urine is used, and when the soluble LR11 derived from urine is used as an immunogen, the specificity of the anti-LR11 antibody is evaluated by the soluble LR11 secreted from malignant tumor cells. It is conceivable to use malignant tumor cells themselves in which LR11 is expressed on the cell surface. Further, even when a synthetic peptide having a partial amino acid sequence of LR11 is used as an immunogen, by using LR11 derived from malignant tumor cells or human urine for specificity evaluation, LR11 in a biological sample such as full-length LR11 can be used. A highly reactive antibody can be obtained. As described above, the source of the immunogen or the standard product in the present invention is very close to the so-called natural state, both in the form of a biological sample and in the form of existing LR11. It can be said that it is particularly advantageous from the viewpoint of preserving the properties of LR11 with respect to conventional sources such as genetic recombination.

前記LR11は、モノクローナル抗体、ポリクローナル抗体のいずれに対しても免疫原ともなりうる。当該抗体は周知の方法にて作製することができる。例えば、ポリクローナル抗体の作製には、免疫する動物としてマウス、ラット、ハムスター、ウサギ、ヤギ、ヒツジ、ニワトリなどが用いられる。抗LR11ポリクローナル抗体を含有する抗血清は、前記LR11を動物の皮下、皮内、腹腔などに一回又は複数回投与し得ることができる。また、免疫賦活効果を有する補液との混合物の免疫がより好ましい。   The LR11 can be an immunogen for both monoclonal and polyclonal antibodies. The antibody can be prepared by a known method. For example, for the production of polyclonal antibodies, mice, rats, hamsters, rabbits, goats, sheep, chickens and the like are used as animals to be immunized. The antiserum containing an anti-LR11 polyclonal antibody can administer the LR11 once or multiple times subcutaneously, intradermally or abdominally in an animal. Further, immunization of a mixture with a replacement fluid having an immunostimulatory effect is more preferable.

また、モノクローナル抗体の作製には、公知のモノクローナル抗体作製方法、例えば、長宗香明、寺田弘共著、「単クローン抗体」廣川書店(1990年)や、Jame W.Golding,“Monoclonal Antibody”,3rd edition,Academic Press,(1996年)に従い作製することができる。
当該モノクローナル抗体は、常法に従い作製したハイブリドーマを培養し、培養上清から分離する方法、該ハイブリドーマをこれと適合性のある哺乳動物に投与し、腹水として回収する方法により製造できる。
For the production of monoclonal antibodies, known monoclonal antibody production methods such as Kamon Nagamune and Hiroko Terada, “Monoclonal Antibody” Yodogawa Shoten (1990), James W., et al. Golding, “Monoclonal Antibody”, 3rd edition, Academic Press, (1996).
The monoclonal antibody can be produced by culturing a hybridoma prepared according to a conventional method and separating it from the culture supernatant, or by administering the hybridoma to a compatible mammal and collecting it as ascites.

抗LR11抗体は、必要に応じて精製して使用することができる。抗体を精製・単離する手法としては、従来公知の方法、例えば、硫酸アンモニウム沈殿法などの塩析、セファデックスなどによるゲルろ過法、イオン交換クロマトグラフィー法、プロテインAカラムなどによるアフィニティ精製法などがある。   The anti-LR11 antibody can be purified and used as necessary. Methods for purifying and isolating antibodies include conventionally known methods such as salting out such as ammonium sulfate precipitation, gel filtration using Sephadex, ion exchange chromatography, affinity purification using protein A column, and the like. is there.

LR11を用いて抗LR11抗体の特異性を評価するには、免疫染色(ウエスタンブロット法)、酵素結合免疫吸着測定法(ELISA)、フローサイトメトリーなどが挙げられる。   To evaluate the specificity of the anti-LR11 antibody using LR11, immunostaining (Western blotting), enzyme-linked immunosorbent assay (ELISA), flow cytometry and the like can be mentioned.

また、LR11を標準品として用いるLR11の免疫学的測定法は特に限定されないが、抗LR11抗体を用いた免疫学的方法、RAP等の親和性を利用した方法、並びにそれらを組み合わせた方法により測定することが望ましい。免疫学的方法としては、例えば免疫染色(ウエスタンブロット法)、酵素結合免疫吸着測定法(ELISA)、免疫比濁法(TIAやLTIA)、エンザイムイムノアッセイ、化学発光イムノアッセイ、蛍光イムノアッセイなどに加えて、抗体とRAP等のLR11に親和性を有する物質を用いたサンドイッチELISAも利用できる。
本発明者らが構築した、界面活性剤により検体の前処理を行うELISA(特許文献3、非特許文献4)は、非常に簡便かつ精度よく測定できることから、特に好ましい。
Moreover, the immunological measurement method of LR11 using LR11 as a standard product is not particularly limited, but is measured by an immunological method using an anti-LR11 antibody, a method using affinity such as RAP, and a method combining them. It is desirable to do. Examples of immunological methods include immunostaining (Western blotting), enzyme-linked immunosorbent assay (ELISA), immunoturbidimetry (TIA and LTIA), enzyme immunoassay, chemiluminescence immunoassay, fluorescent immunoassay, etc. A sandwich ELISA using an antibody and a substance having affinity for LR11 such as RAP can also be used.
The ELISA (Patent Document 3 and Non-Patent Document 4) constructed by the present inventors for pretreatment of a specimen with a surfactant is particularly preferable because it can be measured very easily and accurately.

以下、実施例により、本発明を具体的に説明する。但し、本発明はこれらの実施例に限定されるものではない。   Hereinafter, the present invention will be described specifically by way of examples. However, the present invention is not limited to these examples.

参考例1 ELISAによる可溶性LR11の測定
抗LR11モノクローナル抗体(M3:特許文献3)をPBSで10μg/mLに希釈し、マイクロプレート(NUNC社製)に1ウエルあたり100μL添加して室温で2時間静置した。0.05% Tween20を含むPBS(PBST)で洗浄後、1% BSAを含むPBST(BSA−PBST)を1ウエルあたり200μL添加し室温で1時間ブロッキングした。7% MEGA−9(同仁化学社製)とHBR(ScantibodiesLaboratory社製)を3:1に混合した検体処理液で、測定対象検体を希釈した。ウサギ血清から精製した可溶性LR11も前記検体処理液で段階希釈し、キャリブレーターとした(0〜4ng/mL)。これら希釈検体を、1ウエルあたり100μL添加し、室温で一晩反応させた後、ビオチン標識した抗LR11モノクローナル抗体(R14:特許文献3)をBSA−PBSTで希釈し0.4μg/mLとして、1ウエルあたり100μL添加し室温で4時間反応させた。PBSTで洗浄後、ペルオキシダーゼ標識ストレプトアビジン(PIERCE社製)をBSA−PBSTで希釈して0.2μg/mLとして1ウエルあたり100μL添加し、室温で1時間反応させた。PBSTで洗浄後、TMB基質液を1ウエルあたり100μL加え室温で30分間反応させ、1.5N硫酸を1ウエルあたり100μL加えて反応を停止させた。測定は、マイクロプレートリーダー(Abs.450nm)を用いて行った。キャリブレーターによる検量線より、検体中の可溶性LR11濃度を算出し、希釈倍率を掛けて、検体中の可溶性LR11濃度とした。
Reference Example 1 Measurement of soluble LR11 by ELISA Anti-LR11 monoclonal antibody (M3: Patent Document 3) was diluted to 10 μg / mL with PBS, added to a microplate (manufactured by NUNC) at 100 μL per well, and allowed to stand at room temperature for 2 hours. I put it. After washing with PBS containing 0.05% Tween 20 (PBST), 200 μL of PBST containing 1% BSA (BSA-PBST) was added per well and blocked at room temperature for 1 hour. The sample to be measured was diluted with a sample treatment solution in which 7% MEGA-9 (manufactured by Dojin Chemical Co., Ltd.) and HBR (manufactured by Scantibodies Laboratory) were mixed at a ratio of 3: 1. Soluble LR11 purified from rabbit serum was also serially diluted with the sample treatment solution to prepare a calibrator (0 to 4 ng / mL). These diluted specimens were added at 100 μL per well and allowed to react overnight at room temperature, and then biotin-labeled anti-LR11 monoclonal antibody (R14: Patent Document 3) was diluted with BSA-PBST to 0.4 μg / mL. 100 μL was added per well and reacted at room temperature for 4 hours. After washing with PBST, peroxidase-labeled streptavidin (manufactured by PIERCE) was diluted with BSA-PBST to add 0.2 μg / mL, 100 μL per well, and allowed to react at room temperature for 1 hour. After washing with PBST, 100 μL of TMB substrate solution was added per well and reacted at room temperature for 30 minutes, and 100 μL of 1.5 N sulfuric acid was added per well to stop the reaction. The measurement was performed using a microplate reader (Abs. 450 nm). The concentration of soluble LR11 in the sample was calculated from the calibration curve by the calibrator and multiplied by the dilution factor to obtain the soluble LR11 concentration in the sample.

実施例1 ヒト尿中の可溶性LR11の測定
健常者ボランティア(10名)の随時尿を数日間ランダムに収集し、参考例1に記載の検体処理液で4倍希釈した。参考例1に記載のELISA法と同様に、ヒト尿中の可溶性LR11濃度を測定した。濃度分布は0.1〜8.4ng/mLで、平均3.1ng/mLであった。ヒト尿中には、健常域の血中濃度(非特許文献4などによる)に匹敵する濃度で可溶性LR11が存在することが判明した。
Example 1 Measurement of Soluble LR11 in Human Urine Urine was collected at random from healthy volunteers (10 subjects) for several days and diluted 4-fold with the sample treatment solution described in Reference Example 1. In the same manner as the ELISA method described in Reference Example 1, the concentration of soluble LR11 in human urine was measured. The concentration distribution was 0.1 to 8.4 ng / mL, and the average was 3.1 ng / mL. It was found that soluble LR11 is present in human urine at a concentration comparable to the blood concentration in the normal range (according to Non-Patent Document 4 etc.).

実施例2 ヒト尿からの可溶性LR11の精製
ヒトRAP遺伝子を組み込んだpGEX2T(GEヘルスケア社製)ベクターを導入して形質転換した大腸菌DH5αを培養し、遠心分離により菌体を回収した。3Lの培養液から回収した菌体を、リゾチーム及びTritonX−100を含むPBSに懸濁して超音波処理により菌体を破砕した。この破砕液をGlutathione Sepharose 4 FF(GEヘルスケア社製)200mLに通して遠心上清中のRAP/GST融合タンパク質を吸着させた後、PBSで洗浄してRAP−セファロース樹脂を調製した。
1バッチ分として、このRAP−セファロース樹脂200mLに、ヒトボランティアプール尿を5〜8L通した。PBSで洗浄後、150mM NaClを含む、50mMクエン酸緩衝液(pH5.0)にてヒト尿由来可溶性LR11を溶出した。濃縮後、PBSで透析した。さらに、この液を、rProtein Sepharose A FF(GEヘルスケア社製)5mLに通し、得られた非吸着画分に、抗可溶性LR11モノクローナル抗体(M3)結合樹脂を加え、一晩攪拌した。樹脂をPBSで洗浄した後、150mMNaClを含む50mMクエン酸緩衝液(pH3.0)にてヒト尿由来可溶性LR11を溶出した。さらに、溶出液をゲルろ過クロマトグラフィー(Superdex200;GEヘルスケア社製)により、PBSで分離精製した。可溶性LR11の溶出フラクションを集め濃縮したものを、精製ヒト尿由来可溶性LR11とした。LR11含量は、参考例1に記載の方法に基づき算出された。最終的に12バッチ分の精製工程を行った結果、平均でヒト尿1Lあたり約0.8μgの精製ヒト尿由来可溶性LR11が得られた。参考例1においてキャリブレーターの一番高い濃度は4ng/mLであり、ヒト尿から免疫学的測定の標準品として十分な量の可溶性LR11を容易に確保できることが分かった。
Example 2 Purification of Soluble LR11 from Human Urine Escherichia coli DH5α transformed by introducing a pGEX2T (manufactured by GE Healthcare) vector incorporating a human RAP gene was cultured, and the cells were collected by centrifugation. The cells recovered from 3 L of the culture solution were suspended in PBS containing lysozyme and Triton X-100, and the cells were disrupted by ultrasonic treatment. This disrupted solution was passed through 200 mL of Glutathione Sepharose 4 FF (manufactured by GE Healthcare) to adsorb the RAP / GST fusion protein in the centrifugal supernatant, and then washed with PBS to prepare a RAP-Sepharose resin.
As a batch, 5-8 L of human volunteer pool urine was passed through 200 mL of this RAP-Sepharose resin. After washing with PBS, human urine-derived soluble LR11 was eluted with 50 mM citrate buffer (pH 5.0) containing 150 mM NaCl. After concentration, dialyzed against PBS. Further, this solution was passed through 5 mL of rProtein Sepharose A FF (manufactured by GE Healthcare), and an anti-soluble LR11 monoclonal antibody (M3) binding resin was added to the obtained non-adsorbed fraction, followed by stirring overnight. After washing the resin with PBS, human urine-derived soluble LR11 was eluted with 50 mM citrate buffer (pH 3.0) containing 150 mM NaCl. Further, the eluate was separated and purified with PBS by gel filtration chromatography (Superdex 200; manufactured by GE Healthcare). The soluble LR11-eluted fraction was collected and concentrated to obtain purified human urine-derived soluble LR11. The LR11 content was calculated based on the method described in Reference Example 1. As a result of the final purification process for 12 batches, about 0.8 μg of purified human urine-derived soluble LR11 was obtained per liter of human urine on average. In Reference Example 1, the highest concentration of the calibrator was 4 ng / mL, and it was found that a sufficient amount of soluble LR11 can be easily secured from human urine as a standard for immunological measurement.

実施例3 各種造血器腫瘍細胞株におけるLR11発現の確認
各種造血器腫瘍細胞株でのLR11の発現を、LR11の部分アミノ酸配列を有するペプチドを免疫して作製した抗LR11モノクローナル抗体(A2−2−3:特許文献3)を用いたウェスタンブロット法で確認した。細胞株は、急性骨髄性白血病由来3株(HL−60、ML−2、NB−4)、急性リンパ性白血病由来3株(NALL−1、MOLT−4、CCRF−SB)、リンパ腫由来2株(Daudi、U937)の合計8種を用いた。これらの細胞株は、HT培地(15% fetal bovine serum、HT supplement、penicillin/streptomycinを含むRPMI−1640)に懸濁し、5% 炭酸ガスインキュベーター内で37℃にて培養し、増殖させた。それぞれの細胞株の細胞数をカウントし、1×10個となるように分取し、1% プロテアーゼインヒビター(シグマアルドリッチ社製;P8340)を含むRIPA バッファー(25mM Tris−HCl pH7.6,150mM NaCl,1% NP−40,1% Sodium deoxycholate,0.1% SDS)を加えて、細胞を溶解させた。不溶物を遠心分離にて除去した後、細胞溶解液中のタンパク質濃度を測定して、1レーン当たり20μg分をSDS含有還元条件下で煮沸処理し、SDS−PAGE(2−15%)の各レーンにアプライして電気泳動した。泳動終了後、タンパク質をゲルからPVDF膜へ転写し、抗LR11モノクローナル抗体(A2−2−3)を反応させ、マウスIgG ABCキット(Vector社製)を用いてLR11を検出した。
一方、各細胞株の培養上清中に分泌された可溶性LR11の検出は、以下のように行った。上記と同様にHT培地で培養したそれぞれの細胞株を1×10個となるように分取し、RPMI−1640培地2mLにてさらに14時間培養した。培養上清中の可溶性LR11を、ウェスタンブロット法及び参考例1と同様のELISAで測定した。
各細胞株の細胞溶解液を用いたウェスタンブロットでは、分子量230kDa付近に若干異なる分子量を示す2本のバンドが検出された(図1)。さらに、NALL−1を除き培養上清中にも可溶性LR11が存在していることが確認され、図2に示すようにHL−60、ML−2、NB−4、CCRF−SB、Daudi、U937の6株においては、可溶性LR11の分泌量が培養上清2mLあたり1ngを越えていた。中でも、CCRF−SBでは分泌量が5ng/2mLを越えており、造血器腫瘍細胞を培養することによって、可溶性LR11を容易に得られることが分かった。
Example 3 Confirmation of LR11 Expression in Various Hematopoietic Tumor Cell Lines Anti-LR11 monoclonal antibody (A2-2) prepared by immunizing a peptide having a partial amino acid sequence of LR11 with LR11 expression in various hematopoietic tumor cell lines 3: Confirmed by Western blot using Patent Document 3). Cell lines are 3 strains derived from acute myeloid leukemia (HL-60, ML-2, NB-4), 3 strains derived from acute lymphocytic leukemia (NALL-1, MOLT-4, CCRF-SB), 2 strains derived from lymphoma A total of 8 types (Daudi, U937) were used. These cell lines were suspended in HT medium (RPMI-1640 containing 15% fetal bovine serum, HT supplement, penicillin / streptomycin), cultured at 37 ° C. in a 5% carbon dioxide incubator, and grown. The number of cells of each cell line was counted, sorted to 1 × 10 7 , and RIPA buffer (25 mM Tris-HCl pH 7.6, 150 mM) containing 1% protease inhibitor (Sigma Aldrich; P8340). NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS) was added to lyse the cells. After removing the insoluble matter by centrifugation, the protein concentration in the cell lysate was measured, and 20 μg per lane was boiled under reducing conditions containing SDS. Each of SDS-PAGE (2-15%) Applied to the lane and electrophoresed. After completion of the electrophoresis, the protein was transferred from the gel to a PVDF membrane, reacted with an anti-LR11 monoclonal antibody (A2-2-3), and LR11 was detected using a mouse IgG ABC kit (manufactured by Vector).
On the other hand, the detection of soluble LR11 secreted in the culture supernatant of each cell line was performed as follows. In the same manner as described above, each cell line cultured in HT medium was separated to 1 × 10 7 cells, and further cultured in 2 mL of RPMI-1640 medium for 14 hours. Soluble LR11 in the culture supernatant was measured by Western blotting and the same ELISA as in Reference Example 1.
In the Western blot using the cell lysate of each cell line, two bands having slightly different molecular weights were detected around the molecular weight of 230 kDa (FIG. 1). Further, it was confirmed that soluble LR11 was also present in the culture supernatant except for NALL-1, and as shown in FIG. 2, HL-60, ML-2, NB-4, CCRF-SB, Daudi, U937. In these 6 strains, the amount of soluble LR11 secreted exceeded 1 ng per 2 mL of culture supernatant. In particular, the secretion amount of CCRF-SB exceeded 5 ng / 2 mL, and it was found that soluble LR11 can be easily obtained by culturing hematopoietic tumor cells.

実施例4 各種造血器腫瘍細胞株のフローサイトメトリー
実施例3で用いた細胞株8種について、細胞表面にLR11が発現しているか否かを、フローサイトメトリーにて解析した。各細胞株を、5×10個になるよう50μL Staining Medium(1% FBS/PBS)に浮遊させた。1500rpmで5分間遠心分離した後、上清を除去し、ペレットに、1mg/mL FITC標識抗LR11モノクローナル抗体(M3)を3μL添加して、4℃で60分間反応させた。細胞をStaining Mediumにて2回洗浄後、使用説明書に従いJSAN(Bay bioscience社製)を用いて、フローサイトメトリー解析を実施した。FITC標識抗LR11モノクローナル抗体の陰性コントロールには、正常マウスIgG(BD pharmingen社製)を用いた。結果を図3に示す。実施例3で可溶性LR11の分泌が確認された7株の内、HL−60、ML−2、NB−4、MOLT−4、CCRF−SB、U937の6株については、強陽性から弱陽性の反応が見られ、これらの細胞の表面には、LR11が顕著に発現していることが判明した。
Example 4 Flow cytometry of various hematopoietic tumor cell lines For the eight cell lines used in Example 3, whether or not LR11 was expressed on the cell surface was analyzed by flow cytometry. Each cell line was suspended in 50 μL Staining Medium (1% FBS / PBS) so as to be 5 × 10 5 cells. After centrifuging at 1500 rpm for 5 minutes, the supernatant was removed, and 3 μL of 1 mg / mL FITC-labeled anti-LR11 monoclonal antibody (M3) was added to the pellet and reacted at 4 ° C. for 60 minutes. After the cells were washed twice with Staining Medium, flow cytometry analysis was performed using JSAN (manufactured by Bay bioscience) according to the instruction manual. As a negative control of FITC-labeled anti-LR11 monoclonal antibody, normal mouse IgG (BD pharmingen) was used. The results are shown in FIG. Among the seven strains in which secretion of soluble LR11 was confirmed in Example 3, six strains of HL-60, ML-2, NB-4, MOLT-4, CCRF-SB, and U937 were strongly positive to weakly positive. Reaction was observed, and it was found that LR11 was remarkably expressed on the surface of these cells.

実施例5 各種上皮性悪性腫瘍細胞株のLR11タンパク質発現の確認
各種上皮性腫瘍細胞株でのLR11タンパク質発現を、実施例1で用いた抗LR11モノクローナル抗体(A2−2−3)によるウェスタンブロットで確認した。細胞株は、HLE(肝臓癌由来)、MKN1(胃癌由来)、T.Tn(食道癌由来)、COLO201(大腸癌由来)、AsPC−1(膵臓癌由来)、Caki−1(腎臓癌由来)、Oka−C−1(肺癌由来)、PC−3(前立腺癌由来)、KMBC−2(膀胱癌由来)、HeLa(子宮頸癌由来)、OVISE(卵巣癌由来)、YMB−1(乳癌由来)、KINGS−1(神経腫瘍由来)、G−361(黒色細胞腫由来)の合計14種を用いた。これら細胞株は、指定された培地(例えば、10%fetal bovine serum、penicillin/streptomycinを含むRPMI−1640)に懸濁し、5%炭酸ガスインキュベーター内で37℃にて適当な時間培養し、増殖させた。培養フラスコからの増殖した細胞の回収は、細胞をPBSで数回洗浄した後、トリプシン−EDTA溶液(インビトロジェン社製、25200−072)を用いて行った。それぞれの細胞株の細胞数をカウントし、1×10個となるように分取後、PBSを適当量加え懸濁させ、遠心分離で細胞を回収した。この操作を3回繰り返して細胞を洗浄した。その後、回収した細胞に、1%プロテアーゼインヒビター(シグマアルドリッチ社製、P8340)を含むPBS 100μLを加え、超音波振動器(ホモジナイザー)で細胞を破砕した。細胞の破砕断片を遠心分離にて除去した後、上清を超遠心分離(100,000g、10分間)にて膜画分を沈殿させた。上清を除去し、PBSにて沈殿を洗浄した後、1%プロテアーゼインヒビター(シグマアルドリッチ社製;P8340)及び1%MEGA−9を含むPBS 100μLを加え、超音波振動器(ホモジナイザー)にて膜画分を懸濁・溶解させ、再び超遠心分離(100,000g、10分間)にて不溶の膜画分を除去することで、可溶性膜画分溶液を得た。
各細胞株の可溶性膜画分溶液を、1レーン当たり2μL分をSDS含有還元条件下で煮沸処理し、その各処理液をSDS−PAGE(2−15%)の各レーンにアプライし分離した。泳動終了後、ゲルよりタンパク質をPVDF膜へ転写し、一次抗体として抗LR11モノクローナル抗体(A2−2−3)を反応させ、その後は、Biotinylated anti−mouse IgG ポリクローナル抗体(DAKO社製)及びHRP標識ストレプトアビジンを組み合わせて、最終的にジアミノベンチジンで発色させて、LR11タンパク質を検出した。その結果、14種すべての細胞株について、LR11タンパク質のバンドが検出された(図4)。
一方、各細胞株の培養上清中に分泌された可溶性LR11の検出は、以下のように行った。上記でLR11の発現が確認された細胞株の中から、HLE、COLO201、AsPC−1、KINGS−1、PC−3、KMBC−2を選択し、75cmの培養フラスコを用いて、それぞれ指定された培地40mLでコンフルエントになるまで培養した。また、COLO201は別途無血清のRPMI1640培地20mLに1×10個/mLになるように撒き、2日間培養した。培養上清中の可溶性LR11を、参考例1と同様にELISA法で測定した。結果、すべての細胞株の培養上清中に可溶性LR11が存在していることが確認された(図5)。検討した細胞株の中でも、COLO201細胞株は、無血清培地で培養することにより分泌量が1mL当たり5ngを越えていた。このように、培養細胞を用いる場合、培養条件を工夫することで産出量を増加させることもでき、また、血清成分などの夾雑物が存在しない条件でLR11を効率よく簡便に得ることが出来るので、抗体作製用の免疫原あるいは標準品の作製手段として有効である。
Example 5 Confirmation of LR11 protein expression in various epithelial malignant tumor cell lines The LR11 protein expression in various epithelial tumor cell lines was determined by Western blotting using the anti-LR11 monoclonal antibody (A2-2-3) used in Example 1. confirmed. Cell lines include HLE (derived from liver cancer), MKN1 (derived from stomach cancer), T. Tn (derived from esophageal cancer), COLO201 (derived from colon cancer), AsPC-1 (derived from pancreatic cancer), Caki-1 (derived from kidney cancer), Oka-C-1 (derived from lung cancer), PC-3 (derived from prostate cancer) , KMBC-2 (derived from bladder cancer), HeLa (derived from cervical cancer), OVISE (derived from ovarian cancer), YMB-1 (derived from breast cancer), KINGS-1 (derived from neuronal tumor), G-361 (derived from melanoma) 14) in total. These cell lines are suspended in a designated medium (for example, RPMI-1640 containing 10% fetal bovine serum, penicillin / streptomycin), cultured in a 5% carbon dioxide incubator at 37 ° C. for an appropriate time, and allowed to grow. It was. Recovery of the proliferated cells from the culture flask was performed using a trypsin-EDTA solution (Invitrogen, 25200-072) after the cells were washed several times with PBS. The number of cells in each cell line was counted, sorted to 1 × 10 7 cells, suspended in an appropriate amount of PBS, and the cells were collected by centrifugation. This operation was repeated 3 times to wash the cells. Thereafter, 100 μL of PBS containing 1% protease inhibitor (manufactured by Sigma Aldrich, P8340) was added to the collected cells, and the cells were disrupted with an ultrasonic vibrator (homogenizer). The cell fragments were removed by centrifugation, and then the supernatant was precipitated by ultracentrifugation (100,000 g, 10 minutes). After removing the supernatant and washing the precipitate with PBS, 100 μL of PBS containing 1% protease inhibitor (manufactured by Sigma-Aldrich; P8340) and 1% MEGA-9 was added, and the membrane was removed using an ultrasonic vibrator (homogenizer). The fraction was suspended and dissolved, and the insoluble membrane fraction was removed again by ultracentrifugation (100,000 g, 10 minutes) to obtain a soluble membrane fraction solution.
The soluble membrane fraction solution of each cell line was boiled under a reducing condition containing SDS at 2 μL per lane, and each treatment solution was applied to each lane of SDS-PAGE (2-15%) and separated. After completion of the electrophoresis, the protein is transferred from the gel to a PVDF membrane and reacted with an anti-LR11 monoclonal antibody (A2-2-3) as a primary antibody. After that, Biotinylated anti-mouse IgG polyclonal antibody (manufactured by DAKO) and HRP labeling Streptavidin was combined and finally developed with diaminobenzidine to detect LR11 protein. As a result, LR11 protein bands were detected in all 14 cell lines (FIG. 4).
On the other hand, the detection of soluble LR11 secreted in the culture supernatant of each cell line was performed as follows. HLE, COLO201, AsPC-1, KINGS-1, PC-3, and KMBC-2 are selected from the cell lines in which the expression of LR11 has been confirmed as described above, and each of them is designated using a 75 cm 2 culture flask. The culture medium was cultured until it became confluent at 40 mL. Moreover, COLO201 was separately seeded in 20 mL of serum-free RPMI1640 medium at 1 × 10 6 cells / mL and cultured for 2 days. Soluble LR11 in the culture supernatant was measured by ELISA as in Reference Example 1. As a result, it was confirmed that soluble LR11 was present in the culture supernatants of all cell lines (FIG. 5). Among the cell lines examined, the COLO201 cell line had a secretion amount exceeding 5 ng per mL when cultured in a serum-free medium. Thus, when cultured cells are used, the yield can be increased by devising the culture conditions, and LR11 can be obtained efficiently and simply under conditions where there are no contaminants such as serum components. It is effective as a means for preparing an immunogen or a standard for antibody production.

実施例6 各種可溶性LR11の比較
実施例2で得られたヒト尿由来の可溶性LR11と、実施例3で得られた培養細胞(CCRF−SB)由来可溶性LR11、及び実施例5で得られた培養細胞(COLO201)由来可溶性LR11を、ウェスタンブロット法にて比較した。対照には、ヒト血清から抽出された可溶性LR11(非特許文献4)を用いた。その結果、図6に示すように、いずれの場合も分子量230kDa付近にバンドが認められた。
Example 6 Comparison of Various Soluble LR11 Soluble LR11 derived from human urine obtained in Example 2, soluble LR11 derived from cultured cells (CCRF-SB) obtained in Example 3, and culture obtained in Example 5 The soluble LR11 derived from cells (COLO201) was compared by Western blotting. As a control, soluble LR11 extracted from human serum (Non-Patent Document 4) was used. As a result, as shown in FIG. 6, a band was observed in the vicinity of a molecular weight of 230 kDa in all cases.

実施例7 細胞免疫
フローサイトメトリーで試験した実施例4でLR11を細胞表面に顕著に発現していた造血器腫瘍由来細胞の1つであるNB−4株をHT培地で拡大培養し、PBSにて2回洗浄後、1〜2×10個をPBSに懸濁して、BALB/cマウス(メス)の腹腔内に注入し、免疫した。隔週又は毎週免疫し、適切な時期に、次に示す方法によりマウス血清中の抗LR11抗体価を確認した。
マイクロプレート(NUNC社製)に、実施例2で得られたヒト尿由来精製可溶性LR11をPBSで50ng/mLに希釈して1ウエルあたり50μL添加し、室温で2時間静置した。0.05% Tween20を含むPBS(PBST)で洗浄後、1% BSAを含むPBST(BSA−PBST)を1ウエルあたり200μL添加し、室温で1時間ブロッキングした。マウスの尻尾より採血して得られた血清を、BSA−PBSTにより200、400、800、1600倍希釈して1ウエルあたり50μL添加し、室温で1時間反応させた。ウエルをPBSTで洗浄後、HRP標識抗マウスIgGポリクローナル抗体(Rockland社製)を10000倍希釈して1ウエルあたり50μL添加し、室温で1時間反応させた。PBSTで洗浄後、o−フェニレンジアミン基質液を1ウエルあたり50μL加えて室温で20分間発色させ、1.5N硫酸を1ウエルあたり50μL加えて反応を停止させた後、マイクロプレートリーダー(Abs.492nm)で測定した。
5回免疫した後のマウス血清中の抗LR11抗体価を測定した結果を図7に示す。免疫前のマウス血清では、固相化したヒト尿由来精製可溶性LR11への反応は認められないのに対し、NB−4細胞株を免疫したマウスの血清では、希釈濃度変化に応じた吸光度変化が認められたことから、抗LR11抗体が産生されたことが確認された。
Example 7 Cellular Immunity The NB-4 strain, one of the hematopoietic tumor-derived cells in which LR11 was significantly expressed on the cell surface in Example 4 tested by flow cytometry, was expanded and cultured in HT medium. After washing twice, 1-2 × 10 7 cells were suspended in PBS, injected into the abdominal cavity of BALB / c mice (female), and immunized. Immunization was performed every other week or weekly, and anti-LR11 antibody titer in mouse serum was confirmed at an appropriate time by the following method.
Purified soluble LR11 derived from human urine obtained in Example 2 was diluted to 50 ng / mL with PBS on a microplate (manufactured by NUNC), added 50 μL per well, and allowed to stand at room temperature for 2 hours. After washing with PBS containing 0.05% Tween 20 (PBST), 200 μL of PBST containing 1% BSA (BSA-PBST) was added per well and blocked at room temperature for 1 hour. Serum obtained by collecting blood from the tail of the mouse was diluted 200, 400, 800, 1600 times with BSA-PBST, added at 50 μL per well, and allowed to react at room temperature for 1 hour. After washing the wells with PBST, HRP-labeled anti-mouse IgG polyclonal antibody (manufactured by Rockland) was diluted 10,000 times, added at 50 μL per well, and allowed to react at room temperature for 1 hour. After washing with PBST, 50 μL of o-phenylenediamine substrate solution was added per well, color was allowed to develop for 20 minutes at room temperature, and the reaction was stopped by adding 50 μL of 1.5 N sulfuric acid per well, followed by microplate reader (Abs. 492 nm). ).
The result of measuring the anti-LR11 antibody titer in mouse serum after 5 immunizations is shown in FIG. In the mouse sera before immunization, no reaction was observed with the solid-phased human urine-derived purified soluble LR11, whereas in the sera of mice immunized with the NB-4 cell line, the change in absorbance corresponding to the change in dilution concentration was observed. As a result, it was confirmed that anti-LR11 antibody was produced.

Claims (14)

悪性腫瘍細胞由来又は尿由来LR11を用いることを特徴とする抗LR11抗体の製造法。   A method for producing an anti-LR11 antibody, comprising using malignant tumor cell-derived or urine-derived LR11. 悪性腫瘍細胞由来又は尿由来LR11が、免疫原として及び/又は抗LR11抗体の特異性評価用として使用される請求項1記載の製造法。   The method according to claim 1, wherein malignant cell-derived or urine-derived LR11 is used as an immunogen and / or for evaluating the specificity of an anti-LR11 antibody. 悪性腫瘍細胞由来LR11が、細胞表面に発現しているものである請求項1又は2記載の製造法。   The method according to claim 1 or 2, wherein the malignant tumor cell-derived LR11 is expressed on the cell surface. 悪性腫瘍細胞由来LR11が、培養上清中に分泌されるものである請求項1〜3のいずれか1項記載の製造法。   The method according to any one of claims 1 to 3, wherein the malignant cell-derived LR11 is secreted into the culture supernatant. 悪性腫瘍細胞が、造血器腫瘍細胞又は上皮性悪性腫瘍細胞である請求項1〜4のいずれか1項記載の製造法。   The method according to any one of claims 1 to 4, wherein the malignant tumor cells are hematopoietic tumor cells or epithelial malignant tumor cells. 造血器腫瘍細胞が、急性白血病由来又は悪性リンパ腫由来である請求項5記載の製造法。   6. The method according to claim 5, wherein the hematopoietic tumor cell is derived from acute leukemia or malignant lymphoma. 上皮性悪性腫瘍細胞が、胃癌、肝臓癌、膵臓癌、肺癌、前立腺癌、膀胱癌、食道癌、乳癌、子宮頸癌、卵巣癌、結腸癌、大腸癌、腎臓癌、胆嚢癌、神経腫瘍、黒色細胞腫からなる群より選択される、請求項5記載の製造法。   Epithelial malignant tumor cells are stomach cancer, liver cancer, pancreatic cancer, lung cancer, prostate cancer, bladder cancer, esophageal cancer, breast cancer, cervical cancer, ovarian cancer, colon cancer, colon cancer, kidney cancer, gallbladder cancer, nerve tumor, The production method according to claim 5, which is selected from the group consisting of melanoma. 尿由来LR11が、ヒト尿由来可溶性LR11である請求項1又は2記載の製造法。   The method according to claim 1 or 2, wherein the urine-derived LR11 is human urine-derived soluble LR11. 悪性腫瘍細胞由来又は尿由来LR11を含有するLR11の免疫学的測定用標準品。   A standard product for immunological measurement of LR11 containing LR11 derived from malignant tumor cells or urine. 悪性腫瘍細胞由来LR11が、培養上清中に分泌されるものである請求項9記載の標準品。   The standard product according to claim 9, wherein the LR11 derived from malignant tumor cells is secreted into the culture supernatant. 悪性腫瘍細胞が、造血器腫瘍細胞又は上皮性悪性腫瘍細胞である請求項9又は10記載の標準品。   The standard product according to claim 9 or 10, wherein the malignant tumor cells are hematopoietic tumor cells or epithelial malignant tumor cells. 造血器腫瘍細胞が、ヒト急性白血病由来又はヒト悪性リンパ腫由来である請求項11記載の標準品。   The standard product according to claim 11, wherein the hematopoietic tumor cell is derived from human acute leukemia or human malignant lymphoma. 上皮性悪性腫瘍細胞が、胃癌、肝臓癌、膵臓癌、肺癌、前立腺癌、膀胱癌、食道癌、乳癌、子宮頸癌、卵巣癌、結腸癌、大腸癌、腎臓癌、胆嚢癌、神経腫瘍、黒色細胞腫からなる群より選択される、請求項11記載の標準品。   Epithelial malignant tumor cells are stomach cancer, liver cancer, pancreatic cancer, lung cancer, prostate cancer, bladder cancer, esophageal cancer, breast cancer, cervical cancer, ovarian cancer, colon cancer, colon cancer, kidney cancer, gallbladder cancer, nerve tumor, The standard product according to claim 11, which is selected from the group consisting of melanoma. 尿由来LR11が、ヒト尿由来可溶性LR11である請求項請求項8又は9記載の標準品。   The standard product according to claim 8 or 9, wherein the urine-derived LR11 is human urine-derived soluble LR11.
JP2012524618A 2010-07-15 2011-07-15 Production method of anti-LR11 antibody and standard for immunological measurement Pending JPWO2012008595A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2010161038 2010-07-15
JP2010161038 2010-07-15
PCT/JP2011/066277 WO2012008595A1 (en) 2010-07-15 2011-07-15 Method for producing anti-lr11 antibody and standard for immunoassay

Publications (1)

Publication Number Publication Date
JPWO2012008595A1 true JPWO2012008595A1 (en) 2013-09-09

Family

ID=45469588

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2012524618A Pending JPWO2012008595A1 (en) 2010-07-15 2011-07-15 Production method of anti-LR11 antibody and standard for immunological measurement

Country Status (2)

Country Link
JP (1) JPWO2012008595A1 (en)
WO (1) WO2012008595A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101894417B1 (en) 2011-05-09 2018-09-03 세키스이 메디칼 가부시키가이샤 Method for immunologically measuring soluble lr11
CA3018859A1 (en) * 2017-12-01 2019-06-01 Turun Yliopisto Treatment of her2-dependent cancers by sorl1 inhibition

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008155891A1 (en) * 2007-06-18 2008-12-24 Sekisui Medical Co., Ltd. Novel marker for arteriosclerotic disease
WO2009116268A1 (en) * 2008-03-21 2009-09-24 積水メディカル株式会社 Method for quantification of soluble lr11

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2012007009A (en) * 2009-12-16 2012-07-03 Sekisui Medical Company Ltd Method for diagnosing malignant tumor.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008155891A1 (en) * 2007-06-18 2008-12-24 Sekisui Medical Co., Ltd. Novel marker for arteriosclerotic disease
WO2009116268A1 (en) * 2008-03-21 2009-09-24 積水メディカル株式会社 Method for quantification of soluble lr11

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JPN6011041494; Biochem. Biophys. Res. Commun. Vol.275, No.2, 20000828, pp.365-373 *
JPN6011053313; Histochem. Cell Biol. Vol.118, No.3, 200209, p.183-191 *
JPN6011053314; J. Biol. Chem. Vol.282, No.3, 20070119, p.1873-1881 *
JPN7011002846; J. Cell Sci. Vol.113, No.24, 200012, p.4475-4485 *

Also Published As

Publication number Publication date
WO2012008595A1 (en) 2012-01-19

Similar Documents

Publication Publication Date Title
AU2015254257B2 (en) Anti-MUC1 antibody or antigen-binding fragment of same, and use thereof
KR101832199B1 (en) Method for diagnosing malignant tumor
WO1994004563A1 (en) PEPTIDES CONTAINING RESPECTIVE AMINO ACID SEQUENCES SELECTED FROM AMONG THOSE OF LIPOPROTEIN(a) AND APOLIPOPROTEIN(a), ANTIBODIES RESPECTIVELY RECOGNIZING THESE AMINO ACID SEQUENCES, AND METHOD OF ASSAYING WITH THESE ANTIBODIES
AU2015376851B2 (en) Glypican epitopes and uses thereof
US8865424B2 (en) Anti-FDP monoclonal antibody, FDP measurement reagent and reagent kit using same, and FDP measurement method
CN115724967A (en) Monoclonal antibody for identifying Tau protein N-terminal region and application thereof
US9952230B2 (en) Method of inhibiting nonspecific reaction in PIVKA-II assay reagent
TW201403068A (en) Kit for diagnosis of melanoma
CN116535510B (en) Anti-human PLA2R antibody, kit and application thereof
WO2012008595A1 (en) Method for producing anti-lr11 antibody and standard for immunoassay
JP5864918B2 (en) Autotaxin isoform-specific antibody and detection method
EP2187216B1 (en) Novel liver cancer marker
JP6553491B2 (en) Method for predicting morbidity or risk of diseases associated with final glycation products (AGEs)
JP6630116B2 (en) Monoclonal anti-AGEs antibody and method for producing the same
JP6913631B2 (en) Immunoassay antibody and its preparation method
CN112062848B (en) anti-CD47 monoclonal antibody and application thereof
CN113046325B (en) Vitamin K 3 Monoclonal antibody hybridoma cell strain and application thereof
CN109022415B (en) PDGFR beta monoclonal heavy chain antibody hybridoma cell and preparation method and application thereof
CN117362424A (en) Helicobacter pylori resistant monoclonal antibody and application thereof
JP2000125867A (en) Monoclonal antibody against carp vitellogenin
CN114213542A (en) CPS-I antibodies and uses thereof

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20140714

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20150818

A521 Written amendment

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20151014

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20160126