JPWO2017141789A1 - Storage method of blood-derived specimen and determination method of rare cells - Google Patents

Storage method of blood-derived specimen and determination method of rare cells Download PDF

Info

Publication number
JPWO2017141789A1
JPWO2017141789A1 JP2018500061A JP2018500061A JPWO2017141789A1 JP WO2017141789 A1 JPWO2017141789 A1 JP WO2017141789A1 JP 2018500061 A JP2018500061 A JP 2018500061A JP 2018500061 A JP2018500061 A JP 2018500061A JP WO2017141789 A1 JPWO2017141789 A1 JP WO2017141789A1
Authority
JP
Japan
Prior art keywords
cell
blood
cells
storage
sample
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
JP2018500061A
Other languages
Japanese (ja)
Inventor
茉奈美 増渕
茉奈美 増渕
淳吾 荒木
淳吾 荒木
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.)
Konica Minolta Inc
Original Assignee
Konica Minolta Inc
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 Konica Minolta Inc filed Critical Konica Minolta Inc
Publication of JPWO2017141789A1 publication Critical patent/JPWO2017141789A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/02Separating microorganisms from their culture media
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • 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
    • 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
    • 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
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/533Production of labelled immunochemicals with fluorescent label

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Dentistry (AREA)
  • Environmental Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

本発明は、採血後数日間経過しても適切な検査結果が得られる状態に血液由来検体を保管できる方法を提供することを課題とする。本発明の血液由来検体の保管方法は、血液由来検体から赤血球を除去して細胞検体を生成する分離工程と、前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.001〜0.4v/v%の範囲内となった状態で細胞検体を保管する保管工程とを有する。An object of the present invention is to provide a method capable of storing a blood-derived specimen in a state where an appropriate test result can be obtained even after several days have passed since blood collection. The blood-derived sample storage method of the present invention comprises a separation step of removing red blood cells from a blood-derived sample to produce a cell sample, a cell fixing solution added to the cell sample, and a formaldehyde concentration of 0.001 to 0.4 v Storage step of storing the cell specimen in a state of being in the range of / v%.

Description

本発明は、細胞検体を固定または保管する方法に関する。   The present invention relates to a method for fixing or storing a cell sample.

血液中には、通常、赤血球、白血球(例えば、好中球、好酸球、好塩基球、リンパ球、単球など)等の血液細胞が含まれているが、更に循環腫瘍細胞(CTC)、循環血管内皮細胞(CEC)、循環血管内皮前駆細胞(CEP)、又は各種幹細胞等の希少細胞が存在する場合もある。CTCとは、乳癌、肺癌、前立腺癌、膵臓癌等の患者の病態に応じて末梢血中に存在する細胞であり、血液中からCTCを検出することは、例えば癌に罹患しているか否か、又は患者に対する治療の効果を判断することができ、臨床的に有用である。しかしながら、血液中のCTCの存在数は極めて希少(例えば、全血10mLあたり1〜10個程度)である、その検出及び計数は容易ではない。   The blood usually contains blood cells such as red blood cells and white blood cells (for example, neutrophils, eosinophils, basophils, lymphocytes, monocytes, etc.), but also circulating tumor cells (CTC). There may be rare cells such as circulating vascular endothelial cells (CEC), circulating vascular endothelial progenitor cells (CEP), or various stem cells. CTC is a cell that exists in peripheral blood according to the pathology of patients such as breast cancer, lung cancer, prostate cancer, pancreatic cancer, etc., and detecting CTC in blood is, for example, whether or not it is affected by cancer. Or the effect of treatment on the patient can be determined and is clinically useful. However, the number of CTCs present in blood is extremely rare (for example, about 1 to 10 per 10 mL of whole blood), and its detection and counting are not easy.

希少細胞を検出するには、例えば、先行文献1及び2においては、対象細胞に特異的に磁性粒子を付加することにより対象細胞を血液由来検体から分離し、分離した当該対象細胞が特徴的に発現しているタンパク質に蛍光標識を付加して観察する検査方法が開示されている。タンパク質に蛍光標識を付加する方法として、蛍光標識された抗体を用いる免疫染色法が一般的に用いられている。免疫染色を適切に行うには、対象細胞の形態及び抗原性を保持することが重要であり、その達成手段として、多くの先行文献では、細胞固定液(非特許文献1では4%、非特許文献2では1%ホルムアルデヒド)を添加して細胞と反応させることで細胞固定を行うことが必須とされている。   In order to detect rare cells, for example, in the prior art documents 1 and 2, the target cells are separated from the blood-derived specimen by adding magnetic particles specifically to the target cells. An inspection method in which a fluorescent label is added to an expressed protein for observation is disclosed. As a method for adding a fluorescent label to a protein, an immunostaining method using a fluorescently labeled antibody is generally used. In order to perform immunostaining appropriately, it is important to maintain the morphology and antigenicity of the target cells. As a means for achieving this, many prior literatures have used cell fixatives (4% in Non-Patent Document 1 and non-patents). In Reference 2, it is essential to fix cells by adding 1% formaldehyde) and reacting with cells.

Wen Xu, Lu Cao, Lei Chen, Jing Li, Xiao-Feng Zhang, Hai-Hua Qian, Xiao-Yan Kang, Yu Zhang, Jian Liao, Le-Hua Shi, Ye-Fa Yang, Meng-Chao Wu and Zheng-Feng Yin. Isolation of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma Using a Novel Cell Separation Strategy. Clinical Cancer Research. 2011, vol. 17, p. 3783-3793.Wen Xu, Lu Cao, Lei Chen, Jing Li, Xiao-Feng Zhang, Hai-Hua Qian, Xiao-Yan Kang, Yu Zhang, Jian Liao, Le-Hua Shi, Ye-Fa Yang, Meng-Chao Wu and Zheng- Feng Yin. Isolation of Circulating Tumor Cells in Patients with Hepatocellular Carcinoma Using a Novel Cell Separation Strategy. Clinical Cancer Research. 2011, vol. 17, p. 3783-3793. Zhian Liu, Alberto Fusi, Eva Klopocki, Alexander Schmittel, Ingeborg Tinhofer, Anika Nonnenmacher and Ulrich Keilholz. Negative enrichment by immunomagnetic nanobeads for unbiased characterization of circulating tumor cells from peripheral blood of cancer patients. Journal of Translational Medicine. 2011, vol. 9.Zhian Liu, Alberto Fusi, Eva Klopocki, Alexander Schmittel, Ingeborg Tinhofer, Anika Nonnenmacher and Ulrich Keilholz.Negative enrichment by immunomagnetic nanobeads for unbiased characterization of cyclic tumor cells from peripheral blood of cancer patients. .

前述の通り、癌の診断又は治療効果を評価するには、血液中の希少細胞を観察、又は計数するような検査が臨床的に有用である。一般的に、当該検査において採血作業と検査作業は、その間には時間を置かずに連続して行われる。しかしながら、採血を行った病院等の医療機関の設備や技術が不十分な場合、医療機関から離れた検査施設に血液由来検体を輸送して、当該検査施設にて希少細胞を観察、又は計数するような検査を実施することもある。施設間の距離によって輸送時間が異なるが、一般的には数日間を要する。その場合、血液由来検体の輸送により細胞の形態や抗原性等が変化し、適切な検査結果を得ることができない。   As described above, a test that observes or counts rare cells in blood is clinically useful for evaluating the diagnosis or therapeutic effect of cancer. In general, in the examination, blood collection work and examination work are continuously performed without any time between them. However, if the equipment or technology of a medical institution such as a hospital that collected blood is insufficient, the blood-derived specimen is transported to a laboratory far from the medical institution, and rare cells are observed or counted at the laboratory. Such inspection may be performed. The transportation time varies depending on the distance between facilities, but generally it takes several days. In that case, the morphology and antigenicity of the cells change due to the transport of the blood-derived specimen, and appropriate test results cannot be obtained.

また、検査施設においては、検査待ちの検体が大量にある、或いは1検体についての検査項目が多いためその準備に時間を要する等、検査施設の都合により検査をすぐに実施できない場合がある。このような場合においても、採血から検査が実施されるまで数時間から数日間の待機期間が生じてしまい、施設間の輸送と同様に血液由来検体中の細胞の形態や抗原性等が変化し適切な検査結果を得ることができない。   In addition, in the inspection facility, there are cases in which the inspection cannot be performed immediately due to the convenience of the inspection facility, for example, because there are a large number of samples waiting for the inspection, or because there are many inspection items for one sample, preparation takes time. Even in such a case, a waiting period of several hours to several days occurs from the time of blood collection until the test is performed, and the form and antigenicity of cells in the blood-derived specimen change as in the case of transportation between facilities. Appropriate test results cannot be obtained.

上述の通り、いずれの場合においても、施設間の輸送中又は施設内の待機期間中に血液由来検体中の細胞には、アポトーシスや腐敗が起きる可能性がある。よって、これらの発生を防止若しくはなるべく遅延させるため、採血から長期間保管後に検査作業を行ったとしても適切な検査結果を得ることができるよう、血液由来検体を保管する方法が必要である。   As described above, in any case, apoptosis or decay may occur in cells in the blood-derived specimen during transportation between facilities or during a waiting period within the facility. Therefore, in order to prevent or delay these occurrences as much as possible, there is a need for a method for storing a blood-derived specimen so that an appropriate test result can be obtained even if a test operation is performed after long-term storage after blood collection.

上記課題を解決するために、本発明は一つの側面において、血液由来検体の保管方法であって、前記血液由来検体から赤血球を除去して細胞検体を生成する分離工程と、前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.001〜0.4v/v%の範囲内となった状態で細胞検体を保管する保管工程とを有する保管方法を提供する。   In order to solve the above problems, in one aspect, the present invention is a method for storing a blood-derived sample, the separation step of removing red blood cells from the blood-derived sample to generate a cell sample, There is provided a storage method including a storage step of storing a cell sample in a state where a fixative is added and a formaldehyde concentration is within a range of 0.001 to 0.4 v / v%.

また、本発明はさらなる側面において、希少細胞の判定方法であって、血液由来検体から生成された細胞検体を保管する保管工程を含む前処理工程と、前記血液由来検体に含まれている対象細胞に特異的に結合する蛍光標識を付加する染色工程と、前記対象細胞に特異的に付加された前記蛍光標識の蛍光強度に基づいて前記対象細胞であるか否かを判定して前記対象細胞を特定する判定工程と、を有する判定方法を提供する。この判定方法において、前記保管工程には、前述した本発明の保管方法が用いられる。   In addition, the present invention provides a method for determining a rare cell, in a further aspect, a pretreatment step including a storage step of storing a cell sample generated from a blood-derived sample, and a target cell contained in the blood-derived sample. A staining step of adding a fluorescent label that specifically binds to the target cell, and determining whether or not the target cell is based on the fluorescence intensity of the fluorescent label specifically added to the target cell. And a determination method for specifying the determination method. In this determination method, the storage method of the present invention described above is used in the storage step.

本発明によれば、採血後数日間経過しても適切な検査結果が得られる状態に、血液由来検体を保管することが可能となる。   According to the present invention, it is possible to store a blood-derived specimen in a state where an appropriate test result can be obtained even after several days have passed since blood collection.

図1は、実施例2及び比較例2の保管方法により保管された細胞検体の保管0日から保管10日までの免疫染色蛍光強度の推移を示すグラフである。FIG. 1 is a graph showing the transition of immunostaining fluorescence intensity from the storage day 0 to the storage day 10 of the cell specimens stored by the storage method of Example 2 and Comparative Example 2.

<保管方法>
本発明の血液由来検体の保管方法は、少なくとも、分離工程および保管工程を含み、必要に応じてさらに、固定工程、その他の処理工程を含んでいてもよい。以下、本発明の血液由来検体の保管方法に含まれる各工程について、より詳細に説明する。
<Storage method>
The blood-derived specimen storage method of the present invention includes at least a separation step and a storage step, and may further include a fixing step and other processing steps as necessary. Hereinafter, each step included in the method for storing a blood-derived specimen of the present invention will be described in more detail.

(血液)
本明細書に記載の「血液」は、赤血球細胞、白血球細胞、血小板、内皮細胞、中皮細胞又は上皮細胞等を含んだ、全血の任意の成分を含むことを意図する。また、タンパク質、脂質、核酸、炭水化物等の血漿成分、及び、妊娠、臓器移植、感染、外傷又は疾患によって血液中に存在する可能性のある任意の他の細胞(例えば、CTC、CEC、CEP、その他の前駆細胞等の希少細胞等)をも含む。
(blood)
“Blood” as described herein is intended to include any component of whole blood, including red blood cells, white blood cells, platelets, endothelial cells, mesothelial cells or epithelial cells. Also, plasma components such as proteins, lipids, nucleic acids, carbohydrates, and any other cells that may be present in the blood due to pregnancy, organ transplantation, infection, trauma or disease (eg, CTC, CEC, CEP, Other rare cells such as progenitor cells).

(白血球)
本明細書に記載の「白血球」は、白血球、または網状赤血球若しくは血小板ではない造血系列の細胞である。白血球は、T細胞、B細胞及びナチュラルキラー(NK)細胞等のリンパ球を含む。白血球はまた、単球、マクロファージ、顆粒球等の貪食細胞及びマスト細胞を含む。
(White blood cells)
“Leukocytes” as used herein are leukocytes or cells of the hematopoietic lineage that are not reticulocytes or platelets. Leukocytes include lymphocytes such as T cells, B cells and natural killer (NK) cells. Leukocytes also include phagocytic cells and mast cells such as monocytes, macrophages, granulocytes.

(赤血球)
本明細書に記載の「赤血球」は、赤血球であり、特に非有核赤血球細胞を意味する。
(血液由来検体)
癌の診断または治療効果を評価するためには、血液中のCTC等の希少細胞の観察、又はその計数等の検査が有用である。本明細書に記載の「血液由来検体」は、血液中のCTC等の希少細胞の観察、又はその計数等の検査を行うために、本明細書に記載の「血液」から採取される検体である。
(Red blood cells)
As used herein, “erythrocytes” are erythrocytes, particularly non-nucleated red blood cells.
(Blood sample)
In order to evaluate the diagnosis or therapeutic effect of cancer, it is useful to observe rare cells such as CTC in blood, or to examine the count. The “blood-derived sample” described in the present specification is a sample collected from the “blood” described in the present specification in order to observe rare cells such as CTC in blood or to perform a test such as counting thereof. is there.

血液由来検体には、癌の診断上必要となる血液成分であり、検出の対象となるCTC等の希少細胞(対象細胞)、及び癌の診断上必要ではない血液成分であり、検出の対象とならない赤血球・白血球等の対象細胞以外の細胞(非対象細胞)が含まれている。   A blood-derived specimen is a blood component that is necessary for diagnosis of cancer, and is a rare cell (target cell) such as CTC to be detected, and a blood component that is not necessary for diagnosis of cancer. Cells other than target cells (non-target cells) such as red blood cells and white blood cells that are not included are included.

(抗凝固処理)
体外に取り出された血液(全血)は、そのまま空気に触れると時間の経過とともにゲル状になり、そこに含まれる細胞を回収して観察することができなくなってしまう。そのため、採取された血液は直ちに抗凝固処理することが好ましい。
(Anti-coagulation treatment)
The blood taken out of the body (whole blood), when exposed to air as it is, becomes a gel with the passage of time, and the cells contained therein cannot be collected and observed. Therefore, it is preferable that the collected blood is immediately subjected to anticoagulation treatment.

全血用の抗凝固剤として、エチレンジアミン四酢酸(EDTA)やクエン酸(ナトリウム塩等の塩を含む)に代表される、キレート作用によりカルシウムイオンと結合し反応系から除去することによって凝固を阻止するタイプの抗凝固剤と、ヘパリンに代表される、血漿中のアンチトロンビンIIIと複合体を形成しトロンビンの産生を抑制することにより凝固を阻止するタイプの抗凝固剤とが公知のものであり、一般的な濃度、処理時間等の条件に従って用いることができる。また、前述のような抗凝固剤が予め収容された採血管を利用しても良い。ただし、本発明において抗凝固剤はこれらに限定されるものではない。   Anticoagulant for whole blood, such as ethylenediaminetetraacetic acid (EDTA) and citric acid (including salts such as sodium salt), which inhibits coagulation by binding to calcium ions by chelation and removing it from the reaction system And anticoagulants of the type that block coagulation by forming a complex with antithrombin III in plasma and suppressing thrombin production, represented by heparin. It can be used according to conditions such as general concentration and processing time. Moreover, you may utilize the blood-collecting tube by which the above anticoagulants were previously accommodated. However, the anticoagulant is not limited to these in the present invention.

[分離工程]
分離工程は、血液由来検体から赤血球等を除去して、後述の細胞検体を生成する工程である。分離工程は、血液由来検体の中に含まれている対象細胞以外の細胞等の血液成分(赤血球等の非対象細胞)を除去して、観察の障害を軽減するために行われる処理であり、CTC等の対象細胞の検出性を高めることができる。
[Separation process]
The separation step is a step of removing a red blood cell or the like from a blood-derived sample to generate a cell sample described later. The separation step is a process performed to remove blood components (non-target cells such as red blood cells) such as cells other than the target cells contained in the blood-derived specimen, thereby reducing observation obstacles. Detectability of target cells such as CTC can be improved.

(赤血球除去)
観察の障害を軽減するために、血液由来検体の中に含まれている対象細胞以外の細胞(非対象細胞)等の血液成分をなるべく除きたい。本来であれば、対象細胞を血液由来検体から特異的に取り出すのが好ましいが、対象細胞を血液由来検体から取り出そうとすると、対象細胞の取りこぼしが生じる恐れがある。そのため、本発明においては、対象細胞を血液由来検体から取り出すのではなく、血液由来検体の中に含まれている観察の障害になり得る物質(例えば血中に大量に存在する赤血球、その他診断上に必要としない成分等)を除去する方法を採用する。
(Red blood cell removal)
In order to reduce the obstruction of observation, blood components such as cells (non-target cells) other than the target cells contained in the blood-derived specimen should be removed as much as possible. Originally, it is preferable to specifically extract the target cell from the blood-derived sample, but if the target cell is to be extracted from the blood-derived sample, the target cell may be lost. Therefore, in the present invention, the target cells are not taken out from the blood-derived sample, but are substances that can be an obstacle to observation contained in the blood-derived sample (for example, red blood cells present in large quantities in blood, other diagnostically A method of removing components that are not necessary for the above is adopted.

赤血球を除去する方法としては、塩化アンモニウム等を用いて赤血球を溶血させる方法と、遠心分離処理によって赤血球とそれ以外の細胞を分離させ、少なくとも対象細胞を含む細胞画分を精製する密度勾配遠心法とが挙げられるが、本発明においては、それらに限定されるものではない。なお、前述の方法により赤血球の除去を行ったとしても、必ずしも血液由来検体の中に含まれている赤血球が完全に除去されるわけではなく、微少量の赤血球が血液由来検体中に残る可能性はある。本発明は、このような状況下においても適用できるものである。   Methods for removing red blood cells include hemolyzing red blood cells using ammonium chloride and the like, and density gradient centrifugation that separates red blood cells and other cells by centrifugation and purifies the cell fraction containing at least the target cells. However, the present invention is not limited to these. Even if red blood cells are removed by the method described above, the red blood cells contained in the blood-derived sample are not necessarily completely removed, and a very small amount of red blood cells may remain in the blood-derived sample. There is. The present invention can also be applied under such circumstances.

本来であれば、対象細胞には含まれない細胞である白血球も除去するのが好ましい。対象細胞がCTC等のがん細胞である場合、非有核細胞である赤血球は、その形態、密度及び表面性状ががん細胞と大きく異なるため除去することは可能である。一方、がん細胞と同様に有核細胞に属する白血球は、その形態、密度等が、がん細胞と類似していることから、溶血や密度勾配遠心分離法により除去しようとすると対象細胞も除去され取りこぼしが生じてしまい、不正確な計数により誤診等が発生する恐れがある。また、白血球特異的な抗体(例えば抗CD45抗体)による除去を試行した場合、確率的に非特異的な反応の発生により希少ながん細胞の取りこぼしが発生する可能性が高い。そのため、このような場合においては、血液由来検体の中に含まれている希少細胞をなるべく全て捕捉して取りこぼしを最低限に抑える観点から、白血球の除去を行わないことが好ましい。   Originally, it is preferable to remove leukocytes which are cells not included in the target cells. When the target cell is a cancer cell such as CTC, erythrocytes that are non-nucleated cells can be removed because their morphology, density, and surface properties are significantly different from those of cancer cells. On the other hand, leukocytes belonging to nucleated cells as well as cancer cells are similar in shape and density to cancer cells, so if you try to remove them by hemolysis or density gradient centrifugation, the target cells will also be removed May be missed and misdiagnosis may occur due to inaccurate counting. In addition, when removal with a leukocyte-specific antibody (for example, an anti-CD45 antibody) is attempted, there is a high possibility that a rare cancer cell may be missed due to the occurrence of a non-specific reaction. Therefore, in such a case, it is preferable not to remove leukocytes from the viewpoint of capturing all the rare cells contained in the blood-derived specimen as much as possible and minimizing the loss.

(細胞検体)
本明細書に記載の「細胞検体」は、前処理が施された対象細胞を含む細胞群のことを言う。例えば、培養細胞の場合は培養容器から剥がされ、洗浄された細胞群のことである。また、血液由来検体の場合は赤血球除去及び洗浄処理が施された主に白血球からなる細胞群のことであり、本細胞群には白血球だけでなく、CTC等のがん細胞が含まれる場合もある。なお、本明細書に記載の実施例において、培養細胞の前処理としては洗浄作業、血液由来検体の前処理としては赤血球除去及び洗浄作業を行ったが、前処理はこれに限定されるものではない。
(Cell sample)
The “cell specimen” described in the present specification refers to a group of cells including target cells that have been pretreated. For example, in the case of cultured cells, it is a group of cells that have been peeled off and washed from the culture vessel. In the case of a blood-derived sample, it is a group of cells mainly composed of white blood cells that have undergone red blood cell removal and washing treatment, and this cell group may contain not only white blood cells but also cancer cells such as CTC. is there. In the examples described in the present specification, washing work was performed as pretreatment of cultured cells, and red blood cell removal and washing work were performed as pretreatment of blood-derived specimens. However, the pretreatment is not limited thereto. Absent.

[保管工程]
保管工程は、細胞検体に適切な濃度のホルムアルデヒド溶液からなる細胞固定液を添加し、細胞固定液中で、細胞検体を保管する工程である。
[Storage process]
The storage step is a step of adding a cell fixing solution composed of a formaldehyde solution of an appropriate concentration to the cell sample and storing the cell sample in the cell fixing solution.

保管工程を行うことにより、採血から長期間経過した細胞検体の血液中のCTC等の希少細胞を観察、又は計数する検査を行っても、細胞の形態や抗原性の変化、アポトーシスや腐敗を防止して、適切な検査結果を得ることができる。   By performing the storage process, cell morphology and antigenicity changes, apoptosis, and corruption can be prevented even if a test is performed to observe or count rare cells such as CTC in the blood of a cell sample that has passed for a long time since blood collection. Thus, an appropriate inspection result can be obtained.

(細胞固定液)
本発明において「細胞固定液」とは、ホルムアルデヒド等の細胞固定剤をリン酸緩衝生理食塩水(PBS)等の適切な溶媒に溶解した(あるいはホルマリンのような元から水溶液で市販されているものを希釈した)水溶液をいう。
(Cell fixative)
In the present invention, the “cell fixing solution” refers to a cell fixing agent such as formaldehyde dissolved in a suitable solvent such as phosphate buffered saline (PBS) (or commercially available in the form of an aqueous solution such as formalin). Refers to an aqueous solution diluted.

保管工程において使用される細胞固定液としては、ホルムアルデヒド溶液が好ましい。
細胞検体を、従来の高濃度細胞固定液(例えば、ホルムアルデヒド濃度が1%又は4%)中で長期間保管した場合、細胞検体の免疫染色性が低下してしまう問題があった。そのために本発明では、保管工程において、細胞固定液中のホルムアルデヒドの濃度が希薄な低濃度細胞固定液を用いて保管を行う。つまり、本発明では、保管工程において、最終濃度が0.001v/v%〜0.4v/v%(0.001v/v%以上、かつ0.4v/v%以下)となる量のホルムアルデヒドを添加するようにする。保管工程中の細胞固定液中のホルムアルデヒドの最終濃度が0.001v/v%〜0.4v/v%の範囲内であることにより、その後に行われる染色工程における細胞検体の免疫染色性を低下させないようにすることができる。
As the cell fixing solution used in the storage step, a formaldehyde solution is preferable.
When a cell sample is stored for a long time in a conventional high-concentration cell fixing solution (for example, the formaldehyde concentration is 1% or 4%), there is a problem that the immunostaining property of the cell sample is lowered. Therefore, in the present invention, in the storage step, storage is performed using a low-concentration cell fixing solution in which the concentration of formaldehyde in the cell fixing solution is dilute. That is, in the present invention, in the storage step, the amount of formaldehyde is such that the final concentration is 0.001 v / v% to 0.4 v / v% (0.001 v / v% or more and 0.4 v / v% or less). Add. The final concentration of formaldehyde in the cell fixative during the storage process is within the range of 0.001 v / v% to 0.4 v / v%, thereby reducing the immunostaining of the cell specimen in the subsequent staining process You can avoid it.

本発明の一側面において、保管工程における細胞固定液中のホルムアルデヒドの最終濃度は、免疫染色時の蛍光強度の経時変化を少なくし、ほぼ一定に維持できるという観点から、0.1〜0.4v/v%とすることが好ましい。そのようにして蛍光強度をほぼ一定に維持できれば、保管日数が異なる細胞同士であっても、細胞画像処理条件の標準化や異なるデータ間の比較が容易になり、自動化計測において1種類の対象細胞の判定を、あらかじめ設定した1つの閾値に基づいて行うことが可能になる。   In one aspect of the present invention, the final concentration of formaldehyde in the cell fixing solution in the storage step is 0.1 to 0.4 v from the viewpoint that the change in fluorescence intensity with time during immunostaining can be reduced and maintained almost constant. / V% is preferable. If the fluorescence intensity can be maintained almost constant in this way, it becomes easy to standardize cell image processing conditions and compare different data even between cells having different storage days, and one type of target cell in automated measurement. The determination can be performed based on one preset threshold value.

本発明の別の側面において、保管工程における細胞固定液中のホルムアルデヒドの最終濃度は、免疫染色時の蛍光強度を高める観点から、0.001v/v%以上、0.1v/v%未満とすることが好ましい。ホルムアルデヒドの最終濃度がこのような範囲にある細胞固定液中で細胞を保管すると、保管日数が短いときよりも長いときの方が、免疫染色時の蛍光強度が高くなる傾向を示す。したがって、免疫染色時の蛍光強度を高める必要があるとき、または高めることが好ましいときは、その細胞固定液中で比較的長期間、細胞を保管すればよい。   In another aspect of the present invention, the final concentration of formaldehyde in the cell fixing solution in the storage step is 0.001 v / v% or more and less than 0.1 v / v% from the viewpoint of increasing the fluorescence intensity during immunostaining. It is preferable. When cells are stored in a cell fixative having a final formaldehyde concentration in such a range, the fluorescence intensity during immunostaining tends to be higher when the storage days are longer than when the storage days are shorter. Therefore, when it is necessary to increase the fluorescence intensity during immunostaining, or when it is preferable to increase the fluorescence intensity, the cells may be stored in the cell fixing solution for a relatively long period of time.

細胞固定液と細胞検体との接触時間(検体保管時間)も、検査等の目的に応じて適宜調節することができるが、例えば、室温で、1日〜14日程度である。
なお、本発明の保管工程では、細胞検体を低濃度細胞固定液中で保管するが、細胞のアポトーシスや腐敗を遅延させる面でも一定の効果を有するので、固定工程の機能も兼ねることができる。したがって本発明では、工程数削減のために、保管工程の前に、細胞検体に高濃度固定液を添加して細胞固定を行う固定工程を実施しなくともよい。ただし、より確実に細胞のアポトーシスや腐敗を遅延させる効果を得るためには、保管工程の前に固定工程を行ってもよい。
Although the contact time (sample storage time) between the cell fixing solution and the cell sample can be appropriately adjusted according to the purpose of examination or the like, for example, it is about 1 to 14 days at room temperature.
In the storage step of the present invention, the cell specimen is stored in a low-concentration cell fixing solution. However, since it has a certain effect in terms of delaying apoptosis and decay of cells, it can also function as a fixing step. Therefore, in the present invention, in order to reduce the number of steps, it is not necessary to perform a fixing step of fixing cells by adding a high concentration fixing solution to the cell specimen before the storage step. However, in order to obtain an effect of delaying cell apoptosis or decay more reliably, a fixing step may be performed before the storage step.

(免疫染色)
対象細胞を検出する方法として、色素が修飾された抗体(以下、標識抗体)をCTC等のがん細胞が有する標的抗原に結合させて染色する免疫染色が一般的に用いられている。当該免疫染色の結果によって遺伝子やタンパク質の発現異常等の診断が行なわれている。当該免疫染色においては、通常、以下に説明するように、固定工程、染色工程及び判定工程が行われている。
(Immunostaining)
As a method for detecting target cells, immunostaining is generally used in which a dye-modified antibody (hereinafter labeled antibody) is bound to a target antigen of a cancer cell such as CTC and stained. Diagnosis of abnormal expression of genes and proteins is performed based on the result of the immunostaining. In the immunostaining, a fixing process, a staining process, and a determination process are usually performed as described below.

[固定工程]
固定工程は、細胞のアポトーシスや腐敗を遅延させるために、細胞染色の前に、後述する細胞固定剤を用いて調製した細胞固定液を使用した細胞固定が行われる工程である。また、細胞のアポトーシスや腐敗を遅延させるという観点から、本発明の保管方法において、前記保管工程を行う前に、固定工程を行うことが好ましい。特に、生細胞と同様な形態及び抗原性の保持の観点から、細胞を採取した時点で細胞固定を行うことが好ましい。
[Fixing process]
The fixing step is a step in which cell fixation using a cell fixing solution prepared using a cell fixing agent described later is performed before cell staining in order to delay cell apoptosis and decay. In addition, from the viewpoint of delaying apoptosis and decay of cells, in the storage method of the present invention, it is preferable to perform a fixing step before performing the storage step. In particular, from the viewpoint of maintaining the same form and antigenicity as a living cell, it is preferable to perform cell fixation at the time of collecting the cell.

細胞固定剤として、ホルムアルデヒド、グルタルアルデヒド、グリオキサール等のアルデヒド類、アセトン、メチルエチルケトン等のケトン類、エタノール、メタノール等のアルコール類が一般的に用いられている。このうち、ホルムアルデヒド等のアルデヒド類及びアセトン等のケトン類は、アルデヒド基及びケトン基が特定のアミノ酸残基と反応して共有結合を形成し、架橋剤として作用する固定剤であり、タンパク質構造を安定化するとともに細胞の原形質をゲル化して酵素活性を抑えることができるため、本発明における好ましい固定化剤である。ただし、本発明において細胞固定剤はこれらに限定されるものではない。   As cell fixing agents, aldehydes such as formaldehyde, glutaraldehyde and glyoxal, ketones such as acetone and methyl ethyl ketone, and alcohols such as ethanol and methanol are generally used. Among these, aldehydes such as formaldehyde and ketones such as acetone are fixatives that act as a crosslinking agent by reacting an aldehyde group and a ketone group with a specific amino acid residue, and have a protein structure. It is a preferred immobilizing agent in the present invention because it can stabilize and gel the cell protoplasm to suppress enzyme activity. However, in the present invention, the cell fixing agent is not limited to these.

また、それ自体が固定剤として直接作用するものではないが、加水分解等を受けることにより固定剤を遊離する供与体(例えばホルムアルデヒド供与体)を固定剤の一形態として用いることもできる。   In addition, although it does not act directly as a fixing agent, a donor that releases the fixing agent upon hydrolysis or the like (for example, formaldehyde donor) can be used as one form of the fixing agent.

細胞固定は、適切な濃度の固定化剤を適切な時間、細胞検体に接触させることで行うことができる。細胞固定液中の固定化剤の濃度は適宜調節することができるが、例えば0.1〜10v/v%の範囲内である。細胞固定液と細胞検体との接触時間(細胞固定時間)も適宜調節することができるが、例えば、室温で、6時間以下で行うのが好ましい。   Cell fixation can be performed by contacting a cell sample with an appropriate concentration of an immobilizing agent for an appropriate time. Although the density | concentration of the fixing agent in a cell fixing solution can be adjusted suitably, it exists in the range of 0.1-10 v / v%, for example. The contact time between the cell fixing solution and the cell specimen (cell fixing time) can also be adjusted as appropriate. For example, the contact time is preferably 6 hours or less at room temperature.

[染色工程]
染色工程は、血液由来検体から赤血球を除去して生成された細胞検体に対して、色素で修飾された抗体(標識抗体)を対象細胞又は非対象細胞に特異的に結合させてそれらの細胞を特異的に染色する工程である。
[Dyeing process]
In the staining process, an antibody modified with a dye (labeled antibody) is specifically bound to a target cell or a non-target cell to a cell sample generated by removing red blood cells from a blood-derived sample, and these cells are bound to each other. This is a step of specifically staining.

当該標識抗体としては、公知の蛍光色素等によって標識された抗体を用いることができる。蛍光色素を用いた場合には、対象細胞を染色する標識抗体の蛍光色素と、非目的細胞を染色する標識抗体の蛍光色素とを分離して測定できるように、それぞれ異なる励起波長、蛍光波長を有する蛍光色素を用いることが好ましい。   As the labeled antibody, an antibody labeled with a known fluorescent dye or the like can be used. When fluorescent dyes are used, different excitation wavelengths and fluorescent wavelengths are used so that the fluorescent dyes of labeled antibodies that stain target cells and the fluorescent dyes of labeled antibodies that stain non-target cells can be measured separately. It is preferable to use a fluorescent dye.

対象細胞を特異的に染色する方法としては、対象細胞が有する標的抗原に特異的に結合する標識抗体を含む溶液と細胞検体とを接触させることで対象細胞に特異的に蛍光標識を付加することができる。対象細胞が有する抗原と特異的な反応を示す抗体として、抗サイトケラチン抗体、抗EpCAM抗体、抗ビメンチン抗体等を用いることが可能であるが、本発明においては特に限定されるものではない。   As a method for specifically staining the target cell, a fluorescent label is specifically added to the target cell by contacting a cell sample with a solution containing a labeled antibody that specifically binds to the target antigen of the target cell. Can do. An anti-cytokeratin antibody, an anti-EpCAM antibody, an anti-vimentin antibody, or the like can be used as an antibody that shows a specific reaction with the antigen of the target cell, but is not particularly limited in the present invention.

非対象細胞を特異的に染色する方法としては、非対象細胞が有する標的抗原に特異的に結合する標識抗体を含む溶液と細胞検体とを接触させることで行うことができる。非対象細胞が有する抗原と特異的な反応を示す抗体としては、抗CD45抗体、抗CD2抗体、抗CD13抗体、抗CD14抗体、抗CD15抗体、抗CD16抗体、抗CD19抗体、抗CD203c抗体等を用いることが可能であるが、本発明においては特に限定されるものではない。   The method of specifically staining non-target cells can be performed by contacting a cell sample with a solution containing a labeled antibody that specifically binds to a target antigen possessed by the non-target cells. Examples of antibodies showing a specific reaction with the antigen of non-target cells include anti-CD45 antibody, anti-CD2 antibody, anti-CD13 antibody, anti-CD14 antibody, anti-CD15 antibody, anti-CD16 antibody, anti-CD19 antibody, anti-CD203c antibody and the like. Although it can be used, it is not particularly limited in the present invention.

また、全細胞(対象細胞、非対象細胞ともに)に対して染色を施す方法としては、細胞核を染色することで行うことができる。全細胞の細胞核を染色する色素の例として、DAPI(4',6-diamidino-2-phenylindole)、Hoechst系色素(Hoechst 33342、Hoechst 33258等)、Acridine Orange、DAMO、Ethidium Bromide(臭化エチジウム)、Ethidium Homodimer、Propidium Iodide等が挙げられる。   In addition, as a method for staining all cells (both target cells and non-target cells), staining can be performed by staining cell nuclei. Examples of dyes that stain the nuclei of whole cells include DAPI (4 ', 6-diamidino-2-phenylindole), Hoechst dyes (Hoechst 33342, Hoechst 33258, etc.), Acridine Orange, DAMO, Ethidium Bromide (Ethidium bromide) , Ethidium Homodimer, Propidium Iodide and the like.

[判定工程]
判定工程は、染色工程において付加した、血液由来検体に含まれている対象細胞に特異的に結合する蛍光標識の蛍光強度に基づいて、血液由来検体に含まれる細胞が前記対象細胞であるか否かを判定して、前記対象細胞を特定(検出)する工程である。
[Judgment process]
In the determination step, based on the fluorescence intensity of the fluorescent label that is added in the staining step and specifically binds to the target cell contained in the blood-derived sample, whether or not the cell contained in the blood-derived sample is the target cell. And determining (detecting) the target cell.

(細胞検出)
対象細胞を検出する方法として、染色後の細胞の細胞撮影を行って、その後画像解析により対象細胞を検出する方法がある。
(Cell detection)
As a method for detecting a target cell, there is a method of performing cell photography of the stained cell and then detecting the target cell by image analysis.

例えば、蛍光色素が標識された抗体を用いて染色を行った場合には、まず、蛍光標識が付加された細胞検体に対して、対象細胞を染色する標識抗体の蛍光色素と、非対象細胞を染色する標識抗体の蛍光色素、全細胞を染色する蛍光色素のそれぞれを励起する励起光を順次照射し、それぞれの蛍光色素から発光する蛍光に基づく細胞画像を撮影する。その後、撮影して得られた対象細胞を特異的に染色した細胞画像、非対象細胞を特異的に染色した細胞画像、全細胞を染色した細胞画像等の画像解析を行い、予め設定された蛍光強度の閾値に基づいて対象細胞を検出する。   For example, when staining is performed using an antibody labeled with a fluorescent dye, first, the fluorescent dye of the labeled antibody that stains the target cell and the non-target cell are stained with respect to the cell sample to which the fluorescent label is added. Excitation light that excites each of the fluorescent dye of the labeled antibody to be stained and the fluorescent dye that stains all cells is sequentially irradiated, and a cell image based on the fluorescence emitted from each fluorescent dye is taken. After that, image analysis such as cell images specifically stained for target cells obtained by photographing, cell images specifically stained for non-target cells, cell images stained for all cells, etc. are performed, and preset fluorescence is set. Target cells are detected based on the intensity threshold.

<判定方法>
本発明の希少細胞の判定方法は、前記保管工程を含む前処理工程、前記染色工程および前記判定工程を含み、必要に応じてさらに、前記固定工程、その他の処理工程を含んでいてもよい。
<Judgment method>
The rare cell determination method of the present invention includes a pretreatment step including the storage step, the staining step, and the determination step, and may further include the fixing step and other processing steps as necessary.

前処理工程は、少なくとも、前記保管工程を含む。検体として培養細胞を使用する場合は洗浄処理、血液由来検体を使用する場合は赤血球除去及び洗浄処理等が前処理工程で実施される処理工程に含まれるが、必要に応じてさらに、その他の処理工程を含んでいてもよい。   The pretreatment process includes at least the storage process. When using cultured cells as specimens, washing treatment is included. When using blood-derived specimens, erythrocyte removal and washing treatment are included in the pretreatment process, but other treatments may be performed as necessary. A process may be included.

以下、本発明について実施例を挙げて更に詳細に説明する。
[実施例1]
実施例1の保管方法は、血液由来検体から赤血球を除去して生成した細胞検体に高濃度固定液を用いた細胞固定を施した後に、細胞固定が施された当該細胞検体を低濃度固定液の中で数日保管することを特徴とする。
Hereinafter, the present invention will be described in more detail with reference to examples.
[Example 1]
The storage method of Example 1 is such that a cell sample generated by removing red blood cells from a blood-derived sample is subjected to cell fixation using a high-concentration fixing solution, and then the cell sample subjected to cell fixation is treated with a low-concentration fixing solution. It is characterized by being stored for several days.

(細胞)
実施例1においては、ヒト乳がん培養細胞株(MDA−MB−231)と、抗凝固剤が予め充填された市販の採血管(テルモ社)で採血した健常者ボランティア血液由来の細胞との2種類の細胞を用いた。MDA−MB−231は、トリプシン処理及びリン酸緩衝生理食塩水(PBS)による後述する洗浄処理を2回施した培養細胞を用いた。血液由来の細胞は、赤血球を除去する操作が施された主に白血球からなる細胞検体を用いた。血液由来検体から赤血球を除去するために、以下に説明するように赤血球除去処理を行った。
(cell)
In Example 1, two types of cells derived from human breast cancer cell line (MDA-MB-231) and cells derived from blood of healthy volunteers collected with a commercially available blood collection tube (Terumo) pre-filled with an anticoagulant. Cells were used. MDA-MB-231 used cultured cells that were subjected to trypsin treatment and washing treatment described later twice with phosphate buffered saline (PBS). As the blood-derived cells, a cell specimen mainly composed of white blood cells subjected to an operation for removing red blood cells was used. In order to remove red blood cells from the blood-derived specimen, red blood cell removal treatment was performed as described below.

[分離工程]
(赤血球除去)
免疫染色において、後述する細胞固定液を使用して細胞固定を行うが、血液由来検体の中に対象細胞以外の細胞(例えば赤血球など)が大量に存在すると、それと比べるとCTCの個数は極めて少ないため、細胞固定の効果が十分に現れない場合がある。更に、細胞固定ができている場合においても、細胞固定が施された血液由来検体は、細胞膜の性状、形態及び比重が変化することから、赤血球等の成分を密度勾配遠心法により分離することは困難であり、事前に赤血球を除去することが望ましい。そのため、本実施形態においては赤血球を除去してから細胞固定を行うようにしている。
[Separation process]
(Red blood cell removal)
In immunostaining, cell fixation is performed using a cell fixing solution, which will be described later. When a large amount of cells (for example, erythrocytes) other than the target cells are present in the blood-derived specimen, the number of CTCs is extremely small compared to that. Therefore, the effect of cell fixation may not be sufficiently exhibited. Furthermore, even when cells have been fixed, blood-derived specimens to which cells have been fixed change the properties, morphology, and specific gravity of the cell membrane, so that components such as red blood cells cannot be separated by density gradient centrifugation. It is difficult and it is desirable to remove red blood cells in advance. Therefore, in this embodiment, cell fixation is performed after removing red blood cells.

赤血球を除去するために、まずは、採血管で採取した血液のうち2mLを、密度勾配遠心用分離液(Ficoll、比重1.077)を3mL予め充填した遠沈管(ベクトンディッキンソン社)の該分離液の上から重層し、室温、400×gで40分間遠心分離を行った。遠心後、赤血球が多く含まれる層以外の層を全て採取し、0.1%ウシ血清アルブミン(BSA)を含有するPBSを用いて、室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除き、PBSを1mL加えて洗浄する洗浄処理を2回実施した。洗浄後、PBSで細胞検体の懸濁液を作製した。   In order to remove erythrocytes, first, 2 mL of blood collected by a blood collection tube and 3 mL of a density gradient centrifugal separation solution (Ficoll, specific gravity 1.077) pre-filled with a centrifuge tube (Becton Dickinson) The mixture was layered from above and centrifuged at 400 × g for 40 minutes at room temperature. After centrifugation, all the layers other than the layer containing a large amount of red blood cells are collected, and centrifuged at 300 × g for 1 minute at room temperature using PBS containing 0.1% bovine serum albumin (BSA) to obtain a cell pellet. The supernatant was removed, and washing was performed twice by adding 1 mL of PBS and washing. After washing, a cell specimen suspension was prepared with PBS.

(細胞固定液)
高濃度固定液として、20%ホルマリン液(8%ホルムアルデヒドを含有するもの)(和光純薬工業)をリン酸緩衝生理食塩水(PBS)で、ホルムアルデヒド濃度が4v/v%になるように希釈して調製したものを用いた。低濃度固定液としては、後述するように、同材料で遠心管(プロテオセーブ)の中でのホルムアルデヒド濃度が約0.008v/v%になるように調製した。
(Cell fixative)
As a high concentration fixative, 20% formalin solution (containing 8% formaldehyde) (Wako Pure Chemical Industries) is diluted with phosphate buffered saline (PBS) so that the formaldehyde concentration is 4v / v%. Was used. As described later, the low concentration fixing solution was prepared so that the formaldehyde concentration in the centrifuge tube (Proteo Save) was about 0.008 v / v% with the same material.

[固定工程]
(細胞固定方法)
プロテオセーブ(住友ベークライト)に細胞検体の懸濁液をそれぞれ1mL入れて、室温、300×gで1分間遠心分離を行った。遠心後、細胞ペレットの上清液を取り除き、高濃度固定液を1mL加え、ローテーターで回転させながら室温で20分間反応させた。高濃度固定液は、理想的にはプロテオセーブに加えてからも再度、所定の濃度になるように微調整を行った方が好ましい。ただし、高濃度固定液はホルムアルデヒド濃度が高いため、プロテオセーブに添加後、濃度の微調整を行わなくとも、上清液を取り除いた細胞ペレットの中に含まれるPBSの量によるホルムアルデヒド濃度への影響は僅かである。
[Fixing process]
(Cell fixation method)
1 mL of each cell sample suspension was placed in Proteo Save (Sumitomo Bakelite) and centrifuged at 300 × g for 1 minute at room temperature. After centrifugation, the supernatant of the cell pellet was removed, 1 mL of a high concentration fixative was added, and the mixture was reacted at room temperature for 20 minutes while rotating with a rotator. Ideally, the high-concentration fixing solution should be finely adjusted to a predetermined concentration again after being added to the proteo save. However, since the high-concentration fixative has a high formaldehyde concentration, the amount of PBS contained in the cell pellet from which the supernatant was removed has no effect on the formaldehyde concentration without fine-tuning the concentration after addition to Proteo Save. Is slight.

[保管工程]
(検体保管方法)
細胞固定反応終了後、室温、300×gで1分間遠心分離を行った。遠心後、高濃度固定液が20μL程度残るように細胞ペレットの上清液を取り除き、最終的に低濃度固定液のホルムアルデヒド濃度が約0.008v/v%となるようにPBSを加えた。一方、高濃度固定液とは異なり、低濃度固定液はホルムアルデヒド濃度が低いため、上清液を取り除いた細胞ペレットの中に含まれるPBSの量によってホルムアルデヒド濃度が大きく左右される場合がある。そのため、本実施形態においては、微調整をしながらPBSを加えるようにしている。目安として、高濃度固定液を20μL残した細胞ペレットに対して約1000μLのPBSを加えた。
[Storage process]
(Specimen storage method)
After completion of the cell fixation reaction, centrifugation was performed at room temperature at 300 × g for 1 minute. After centrifugation, the cell pellet supernatant was removed so that about 20 μL of the high concentration fixative remained, and PBS was finally added so that the formaldehyde concentration of the low concentration fixative was about 0.008 v / v%. On the other hand, unlike the high-concentration fixing solution, the low-concentration fixing solution has a low formaldehyde concentration, and thus the formaldehyde concentration may be greatly influenced by the amount of PBS contained in the cell pellet from which the supernatant is removed. Therefore, in the present embodiment, PBS is added while performing fine adjustment. As a guide, about 1000 μL of PBS was added to the cell pellet in which 20 μL of the high concentration fixative was left.

ホルムアルデヒド濃度の微調整後、低濃度固定液の中で、固定日を0日目として細胞検体を0、3、又は7日間保管した。細胞検体を、前述のような従来の高濃度固定液(ホルムアルデヒド濃度が1%又は4%のもの)中で長期間にわたって保管した場合、その後細胞の免疫染色性(蛍光輝度)が低下してしまうため、本実施形態においては、ホルムアルデヒド濃度が極めて希薄な低濃度固定液を敢えて用いて検体保管を行った。このことにより、固定液の中で長期間にわたって検体保管を行った後でも、細胞検体の免疫染色性が低下することはない。   After fine adjustment of the formaldehyde concentration, the cell specimen was stored for 0, 3, or 7 days in the low concentration fixative, with the fixed date as day 0. When cell specimens are stored for a long time in the conventional high-concentration fixing solution (formaldehyde concentration of 1% or 4%) as described above, the immunostaining properties (fluorescence luminance) of the cells are subsequently lowered. Therefore, in the present embodiment, the specimen is stored by using a low concentration fixative with extremely dilute formaldehyde concentration. As a result, the immunostaining property of the cell sample does not deteriorate even after the sample is stored in the fixing solution for a long period of time.

なお、前述した固定工程及び保管工程において、対象細胞の取りこぼしを最低限に抑えるために、細胞検体を遠心管から移動させず当該遠心管に高濃度固定液又は低濃度固定液を添加して細胞固定及び検体保管を行ったが、細胞固定及び検体保管の進め方はこれらに限定されない。例えば、固定液が収容されている容器に細胞検体を移動させても良く、固定液と細胞検体とを接触させて反応を進めることができれば良い。   In the above-described fixing step and storage step, in order to minimize the target cells from being missed, the cell sample is not moved from the centrifuge tube, and a high concentration fixing solution or a low concentration fixing solution is added to the centrifuge tube. Although fixation and specimen storage were performed, the method of cell fixation and specimen storage is not limited to these. For example, the cell sample may be moved to a container in which the fixing solution is accommodated, and it is sufficient if the reaction can be advanced by bringing the fixing solution and the cell sample into contact with each other.

(免疫染色)
保管工程後の細胞の免疫染色性を評価するために、以下に説明するように免疫染色を行った。
(Immunostaining)
In order to evaluate the immunostaining properties of the cells after the storage step, immunostaining was performed as described below.

先ずは、室温、300×gで1分間遠心分離を行い、遠心後、細胞ペレットの上清液を取り除き、PBSを添加後、再度室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除いた。次いで、1次抗体反応液を添加し1次抗体反応を行った。1次抗体反応液として、MDA−MB−231細胞検体には抗サイトケラチン(CK)抗体(ベクトンディッキンソン社)、健常者の血液由来検体から生成された細胞検体には抗CD45抗体(BioLegend社)を添加して室温で30分間反応させた。   First, centrifuge at 300 × g for 1 minute at room temperature, remove the cell pellet supernatant after centrifugation, add PBS, and centrifuge again at room temperature at 300 × g for 1 minute. The supernatant was removed. Subsequently, the primary antibody reaction solution was added to perform the primary antibody reaction. As a primary antibody reaction solution, an anti-cytokeratin (CK) antibody (Becton Dickinson) is used for MDA-MB-231 cell samples, and an anti-CD45 antibody (BioLegend) is used for cell samples generated from blood samples of healthy individuals. Was added and allowed to react at room temperature for 30 minutes.

1次抗体反応後、室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除き、PBSを1mL加えて洗浄する洗浄処理を3回実施した。
洗浄後、2次抗体反応を行った。2次抗体反応を行うには、MDA−MB−231細胞検体には1次抗体反応にて使用した抗CK抗体に特異的に結合するAlexaFluor647標識抗マウスIgG2a抗体(インビトロジェン社)、血液由来検体から生成された細胞検体には1次抗体反応にて使用した抗CD45抗体に特異的に結合するAlexaFluor488標識抗マウスIgG1抗体(インビトロジェン社)を添加した。そして、細胞核を染色するために、MDA−MB−231細胞検体及び血液由来検体から生成された細胞検体に、細胞核中のDNAに結合し染色するHoechst33342(インビトロジェン社)を添加して室温で30分間反応させた。
After the primary antibody reaction, centrifugation was performed at 300 × g for 1 minute at room temperature, the cell pellet supernatant was removed, and washing was performed 3 times by adding 1 mL of PBS and washing.
After washing, a secondary antibody reaction was performed. To perform the secondary antibody reaction, the MDA-MB-231 cell specimen was analyzed using AlexaFluor 647-labeled anti-mouse IgG2a antibody (Invitrogen) that specifically binds to the anti-CK antibody used in the primary antibody reaction. AlexaFluor488-labeled anti-mouse IgG1 antibody (Invitrogen) that specifically binds to the anti-CD45 antibody used in the primary antibody reaction was added to the generated cell specimen. In order to stain the cell nucleus, Hoechst33342 (Invitrogen) that binds to and stains DNA in the cell nucleus is added to the cell sample generated from the MDA-MB-231 cell sample and the blood-derived sample, and then for 30 minutes at room temperature. Reacted.

2次抗体反応後、室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除き、PBSを1mL加えて洗浄する洗浄処理を2回実施し、最後にPBSを加えて105〜106cells/mLの細胞懸濁液を作製した。After the secondary antibody reaction, the mixture is centrifuged at 300 × g for 1 minute at room temperature, the supernatant of the cell pellet is removed, 1 mL of PBS is added and washing is performed twice, and finally PBS is added to add 10 A cell suspension of 5 to 10 6 cells / mL was prepared.

(免疫染色性評価)
免疫染色後、以下に説明するように蛍光顕微鏡にて免疫染色性の評価を行った。
先ずは、免疫染色後の細胞検体懸濁液から20μLを血球計算盤に充填して蛍光顕微鏡にて細胞画像の撮影を行った。
(Immunostaining evaluation)
After the immunostaining, the immunostaining property was evaluated with a fluorescence microscope as described below.
First, 20 μL from the cell specimen suspension after immunostaining was filled in a hemocytometer, and cell images were taken with a fluorescence microscope.

次に、撮影して得られた細胞画像の画像解析を行い、個々の細胞について、MDA−MB−231細胞についてはAlexaFluor647の蛍光強度、血液由来検体の細胞についてはAlexaFluor488の蛍光強度を、下記の式1により算出した。そして、それぞれの細胞検体において、30細胞の蛍光強度の平均値を、該当細胞検体の免疫染色性の指標として算出した。保管日数0日目の値を1として規格化したものを表1に示す。   Next, image analysis of the cell images obtained by imaging was performed. For each cell, the fluorescence intensity of AlexaFluor 647 for MDA-MB-231 cells, and the fluorescence intensity of AlexaFluor 488 for blood-derived specimen cells Calculated according to Equation 1. In each cell sample, the average value of the fluorescence intensity of 30 cells was calculated as an index of immunostaining of the cell sample. Table 1 shows the values normalized with the value of the storage day 0 as 1.

(式1)
[(1細胞についての蛍光強度の最大値)−(バックグラウンドの蛍光強度の平均値)]/(バックグラウンドの蛍光強度の標準偏差)
[比較例1]
実施例1において、検体保管方法を実施しなかった以外は実施例1と同様にして各細胞検体の免疫染色性を評価した。すなわち、実施例1において、細胞固定後には遠心分離を行わず、高濃度固定液のままで細胞検体を0、3、又は7日間保管した後に免疫染色や蛍光強度の算出等を行った。その結果を表1に示す。
(Formula 1)
[(Maximum fluorescence intensity per cell) − (average background fluorescence intensity)] / (standard deviation of background fluorescence intensity)
[Comparative Example 1]
In Example 1, the immunostaining property of each cell sample was evaluated in the same manner as in Example 1 except that the sample storage method was not performed. That is, in Example 1, after cell fixation, centrifugation was not performed, and cell samples were stored for 0, 3, or 7 days in a high concentration fixative solution, and then immunostaining and calculation of fluorescence intensity were performed. The results are shown in Table 1.

Figure 2017141789
Figure 2017141789

表1からも分かるように、実施例1の方法によって固定及び保管された検体は、比較例1と比較して、保管日数7日後でも蛍光強度の低下はほとんどなく、特に細胞質に存在する抗原に対する抗体(MDA−MB−231細胞検体に使用した抗CK抗体)で顕著であった。この結果から、血液由来検体から赤血球が除去された状態で保管する、及び、低濃度固定液中で保管するといった本発明の構成により、免疫染色性が低下することなく、細胞検体を保管できることが分かる。   As can be seen from Table 1, the specimen fixed and stored by the method of Example 1 has almost no decrease in fluorescence intensity even after 7 days of storage compared to Comparative Example 1, and particularly against the antigen present in the cytoplasm. It was remarkable with the antibody (anti-CK antibody used for the MDA-MB-231 cell specimen). From this result, it is possible to store a cell sample without reducing immunostaining by the configuration of the present invention such as storing in a state in which red blood cells are removed from a blood-derived sample and storing in a low concentration fixative. I understand.

[実施例2]
実施例2の保管方法は、血液由来検体から赤血球を除去して生成した細胞検体に高濃度固定液を用いた細胞固定を施さず、当該細胞検体を低濃度固定液の中に保存することで、細胞固定の目的も兼ねて細胞検体をそのまま放置して保管日数まで保管することを特徴とする。
[Example 2]
In the storage method of Example 2, cell samples generated by removing red blood cells from a blood-derived sample are not subjected to cell fixation using a high concentration fixative, and the cell sample is stored in a low concentration fixative. The cell specimen is also left as it is for the purpose of cell fixation, and is stored until the storage days.

(細胞)
実施例1と同様の状態のヒト乳がん培養細胞株(MDA−MB−231)を用いた。
(細胞固定液)
実施例2においては、20%ホルマリン液(8%ホルムアルデヒドを含有するもの)(和光純薬工業)をリン酸緩衝生理食塩水(PBS)で、ホルムアルデヒド濃度が(A)0.4v/v%、(B)0.04v/v%、(C)0.01v/v%、(D)0.001v/v%になるように希釈して調製した4種類の低濃度固定液(A)〜(D)を用いた。
(cell)
A human breast cancer cell line (MDA-MB-231) in the same state as in Example 1 was used.
(Cell fixative)
In Example 2, 20% formalin solution (containing 8% formaldehyde) (Wako Pure Chemical Industries) was phosphate buffered saline (PBS), and the formaldehyde concentration was (A) 0.4 v / v%, (B) Four types of low-concentration fixing solutions (A) to (D) prepared by diluting to 0.04 v / v%, (C) 0.01 v / v%, and (D) 0.001 v / v%. D) was used.

[保管工程]
(細胞固定を兼ねた検体保管方法)
プロテオセーブ(住友ベークライト)に細胞検体を1.5mLずつ入れて、室温、300×gで1分間遠心分離を行った。遠心後、細胞ペレットの上清液を取り除き、前記低濃度固定液(A)〜(D)をそれぞれ1mL加えてローテーターで回転させながら室温で20分間反応させた。前述したように、低濃度固定液はホルムアルデヒド濃度が低いため、上清液を取り除いた細胞ペレットの中に含まれるPBSの量によってホルムアルデヒド濃度が大きく左右される場合がある。そのため、実施例2においても、プロテオセーブに添加後のホルムアルデヒド濃度の微調整を行うようにしている。
[Storage process]
(Specimen storage method also used for cell fixation)
Cell samples were put in 1.5 mL each in Proteo Save (Sumitomo Bakelite), and centrifuged at 300 × g for 1 minute at room temperature. After centrifugation, the supernatant of the cell pellet was removed, 1 mL each of the low concentration fixatives (A) to (D) was added, and the mixture was reacted at room temperature for 20 minutes while rotating with a rotator. As described above, since the low concentration fixative has a low formaldehyde concentration, the formaldehyde concentration may be greatly affected by the amount of PBS contained in the cell pellet from which the supernatant has been removed. Therefore, also in Example 2, fine adjustment of the formaldehyde concentration after addition to Proteo Save is performed.

細胞固定を兼ねた検体保管において低濃度固定液(A)、(B)を添加後、細胞検体をそのまま放置し、固定液を添加した日を0日目として0、1、3、又は7日間保管した。また、低濃度固定液(C)、(D)を添加後、細胞検体をそのまま放置し、固定液を添加した日を0日目として0又は10日間保管した。   In sample storage that also serves as cell fixation, after adding low concentration fixatives (A) and (B), the cell sample is left as it is, and 0, 1, 3, or 7 days, starting on day 0 when the fixative is added Stored. In addition, after adding the low-concentration fixing solutions (C) and (D), the cell specimen was left as it was, and stored for 0 or 10 days on the 0th day when the fixing solution was added.

(免疫染色)
保管日数経過後、細胞検体の免疫染色性を評価するために、以下に説明するように免疫染色を行った。
(Immunostaining)
After elapse of storage days, immunostaining was performed as described below in order to evaluate the immunostaining property of the cell specimen.

先ずは、1次抗体反応として、抗サイトケラチン抗体(CK)又はフィコエリスリン(PE)標識抗ビメンチン(Vmt)抗体(CSTジャパン株式会社)を添加して室温で30分間反応させた。なお、低濃度固定液(C)、(D)を用いて固定及び保管された細胞検体には、抗CK抗体による標識のみを行った。   First, as a primary antibody reaction, an anti-cytokeratin antibody (CK) or a phycoerythrin (PE) labeled anti-vimentin (Vmt) antibody (CST Japan Co., Ltd.) was added and reacted at room temperature for 30 minutes. In addition, the cell sample fixed and stored using the low concentration fixing solutions (C) and (D) was only labeled with an anti-CK antibody.

1次抗体反応後、室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除き、PBSを1mL加えて洗浄する洗浄処理を3回実施した。
洗浄後、2次抗体反応として、抗CK抗体結合MDA−MB−231細胞検体には抗CK抗体に特異的に結合するAlexaFluor647標識抗マウスIgG2a抗体(インビトロジェン社)、及び、細胞核中のDNAに結合し染色するHoechst33342(インビトロジェン社)を添加して室温で30分間反応させた。抗Vmt抗体結合MDA−MB−231細胞検体にはHoechst33342(インビトロジェン社)のみを添加して室温で30分間反応させた。
After the primary antibody reaction, centrifugation was performed at 300 × g for 1 minute at room temperature, the cell pellet supernatant was removed, and washing was performed 3 times by adding 1 mL of PBS and washing.
After washing, as a secondary antibody reaction, the anti-CK antibody-conjugated MDA-MB-231 cell specimen binds to AlexaFluor647-labeled anti-mouse IgG2a antibody (Invitrogen) that specifically binds to the anti-CK antibody, and to DNA in the cell nucleus Hoechst 33342 (Invitrogen) was added and allowed to react at room temperature for 30 minutes. Only Hoechst 33342 (Invitrogen) was added to the anti-Vmt antibody-bound MDA-MB-231 cell specimen and allowed to react at room temperature for 30 minutes.

2次抗体反応後、室温、300×gで1分間遠心分離を行い、細胞ペレットの上清液を取り除き、PBSを1mL加えて洗浄する洗浄処理を2回実施し、最後にPBSを加えて105〜106cells/mLの細胞懸濁液を作製した。After the secondary antibody reaction, the mixture is centrifuged at 300 × g for 1 minute at room temperature, the supernatant of the cell pellet is removed, 1 mL of PBS is added and washing is performed twice, and finally PBS is added to add 10 A cell suspension of 5 to 10 6 cells / mL was prepared.

(免疫染色性評価)
免疫染色後、以下に説明するように蛍光顕微鏡にて免疫染色性の評価を行った。
先ずは、免疫染色後の細胞検体懸濁液から20μLを血球計算盤に充填して蛍光顕微鏡にて撮影を行った。
(Immunostaining evaluation)
After the immunostaining, the immunostaining property was evaluated with a fluorescence microscope as described below.
First, 20 μL of the cell specimen suspension after immunostaining was filled in a hemocytometer and photographed with a fluorescence microscope.

次に、前述したように、式1を用いて、個々の細胞についてのAlexaFluor647又はPEの蛍光強度を算出し、30細胞の蛍光強度の平均値を該当細胞検体の免疫染色性の指標として算出した。保管日数0日目の値を1として規格化したものを表2に示す。   Next, as described above, the fluorescence intensity of AlexaFluor 647 or PE for each cell was calculated using Equation 1, and the average value of the fluorescence intensity of 30 cells was calculated as an index of immunostaining of the corresponding cell specimen. . Table 2 shows the values normalized with the value on the 0th storage day set to 1.

[比較例2]
実施例2において、低濃度固定液の代わりに高濃度固定液(ホルムアルデヒド濃度が4v/v%)を使用した以外は実施例2と同様にして各細胞検体の免疫染色性及び免疫染色割合を評価した。すなわち、比較例2において、各細胞検体は高濃度固定液で固定され、そのまま保管日数7日間まで放置された。その後に免疫染色を行い、免疫染色性の評価を行った。その結果を表2に示す。また、実施例2及び比較例2のデータをまとめて図1に示す。
[Comparative Example 2]
In Example 2, the immunostaining and the immunostaining ratio of each cell specimen were evaluated in the same manner as in Example 2 except that a high concentration fixing solution (formaldehyde concentration was 4 v / v%) was used instead of the low concentration fixing solution. did. That is, in Comparative Example 2, each cell sample was fixed with a high concentration fixative and left as it was for 7 days. Thereafter, immunostaining was performed, and the immunostaining property was evaluated. The results are shown in Table 2. The data of Example 2 and Comparative Example 2 are collectively shown in FIG.

Figure 2017141789
Figure 2017141789

表2からも分かるように、実施例2の方法によって固定を兼ねた検体保管処理を行った細胞検体は、低濃度固定液(A)〜(D)のいずれについても、比較例2と比較して、保管日数7日後でも蛍光強度が維持され、若しくは向上した。特に、低濃度固定液(A)で固定及び保管された細胞検体は、保管日数0日目からの蛍光強度変化が少なく、その値がほぼ一定に維持された。また、低濃度固定液(C)及び(D)で固定及び保管された細胞検体は、保管日数10日で保管日数0日の約12倍と、蛍光強度が顕著に向上した。   As can be seen from Table 2, the cell samples subjected to the sample storage process that also fixed by the method of Example 2 were compared with Comparative Example 2 for any of the low concentration fixing solutions (A) to (D). Thus, the fluorescence intensity was maintained or improved even after 7 days of storage. In particular, the cell specimen fixed and stored with the low-concentration fixing solution (A) showed little change in fluorescence intensity from the 0th storage day, and the value was maintained almost constant. In addition, the fluorescence intensity of the cell specimen fixed and stored with the low concentration fixing solutions (C) and (D) was remarkably improved, with the storage days being 10 days and the storage days being about 12 times.

この結果から、予め従来の高濃度固定液を用いた細胞固定を行わなくても、血液由来検体から赤血球が除去された状態で、且つ、低濃度固定液中で細胞検体を保管すれば、細胞固定が有効に進行し、免疫染色性が低下することなく検体を保管できることが明らかである。特に、低濃度固定液(A)で固定及び保管された細胞検体は、蛍光強度がほぼ一定値に維持されるため、細胞画像処理条件の標準化や異なるデータ間の比較が可能となる。   From this result, it is possible to store cells in a low concentration fixative in a state where erythrocytes have been removed from the blood-derived sample without performing cell fixation using a conventional high concentration fixative in advance. It is clear that the fixation proceeds effectively and the specimen can be stored without a decrease in immunostaining. In particular, since the fluorescence intensity of the cell specimen fixed and stored in the low concentration fixing solution (A) is maintained at a substantially constant value, it is possible to standardize the cell image processing conditions and compare different data.

例えば、免疫染色においては対象細胞に特異的に結合する抗体を使用しており、理想的には非対象細胞は染色されないのが好ましいが、非特異的吸着等により非対象細胞が染色されることもある。このような状況においては、染色された細胞の蛍光強度に基づいて対象細胞であるか否かを判定して対象細胞を特定する必要がある。蛍光強度の閾値を予め設定し、当該蛍光強度の閾値に基づいて前述した対象細胞の特定の自動化を図る場合、検査ごとに蛍光強度が変わると、当該閾値に基づいた対象細胞であるか否かの判定を適切に行うことができなくなってしまう。すなわち、予め設定した1つの閾値に基づいて対象細胞であるか否かの判定を行う場合に、異なる蛍光強度を示す細胞を同じ細胞として判断することは困難である。一方、保管日数等によって変化する蛍光強度に対応して対象細胞の自動検出を行おうとするのであれば、検査ごとにキャリブレーションを行い異なる閾値を設定する必要がある。   For example, in immunostaining, antibodies that specifically bind to target cells are used, and ideally, non-target cells are preferably not stained, but non-target cells are stained by non-specific adsorption or the like. There is also. In such a situation, it is necessary to determine whether the cell is a target cell based on the fluorescence intensity of the stained cell to identify the target cell. When a threshold value of fluorescence intensity is set in advance and the above-described target cell is automated based on the threshold value of fluorescence intensity, whether the target cell is based on the threshold value when the fluorescence intensity changes for each examination. Cannot be properly determined. That is, when determining whether or not a target cell is based on one preset threshold value, it is difficult to determine cells having different fluorescence intensities as the same cell. On the other hand, if the target cell is to be automatically detected in response to the fluorescence intensity that changes depending on the number of storage days, etc., it is necessary to perform calibration for each examination and set different threshold values.

しかしながら、本発明によれば、保管日数0日目からの蛍光強度変化が少ないため、自動化計測において1種類の対象細胞の判定を、予め設定した1つの閾値に基づいて行うことが可能となる。   However, according to the present invention, since the change in fluorescence intensity from the 0th storage day is small, it is possible to determine one type of target cell based on one preset threshold value in automated measurement.

また、低濃度固定液中で保管することにより、輸送の途中でも細胞固定が進行するため、工数削減の効果も得られる。   Further, by storing in a low concentration fixing solution, cell fixation proceeds even during transportation, so that the effect of reducing the number of steps can be obtained.

Claims (7)

血液由来検体の保管方法であって、
前記血液由来検体から赤血球を除去して細胞検体を生成する分離工程と、
前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.001〜0.4v/v%の範囲内となった状態で細胞検体を保管する保管工程と、
を有する保管方法。
A method for storing a blood-derived specimen,
A separation step of removing red blood cells from the blood-derived sample to produce a cell sample;
A storage step of adding a cell fixing solution to the cell sample and storing the cell sample in a state where the formaldehyde concentration is within a range of 0.001 to 0.4 v / v%;
Storage method.
前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.1〜10v/v%の範囲内となった状態で細胞検体を固定する固定工程を更に有し、
前記保管工程を行う前に前記固定工程を行うことを特徴とする請求項1に記載の保管方法。
A cell fixing solution is further added to the cell sample, and the cell sample is further fixed in a state where the formaldehyde concentration is in the range of 0.1 to 10 v / v%,
The storage method according to claim 1, wherein the fixing step is performed before the storage step.
前記保管工程は、前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.1〜0.4v/v%の範囲内となった状態で細胞検体を保管する保管工程であることを特徴とする請求項1に記載の保管方法。   The storage step is a storage step of adding a cell fixing solution to the cell sample and storing the cell sample in a state where the formaldehyde concentration is within a range of 0.1 to 0.4 v / v%. The storage method according to claim 1. 前記固定工程は、細胞検体を6時間以下固定することを特徴とする請求項2に記載の保管方法。   The storage method according to claim 2, wherein in the fixing step, the cell specimen is fixed for 6 hours or less. 前記保管工程は、前記細胞検体に細胞固定液を添加し、ホルムアルデヒド濃度が0.001v/v%以上、0.1v/v%未満の範囲内となった状態で保管する保管工程であることを特徴とする請求項1、2又は4のいずれか一項に記載の保管方法。   The storage step is a storage step in which a cell fixing solution is added to the cell sample and stored in a state where the formaldehyde concentration is within a range of 0.001 v / v% or more and less than 0.1 v / v%. The storage method according to claim 1, wherein the storage method is characterized by the following. 血液由来検体から生成された細胞検体を保管する保管工程を含む前処理工程と、
前記血液由来検体に含まれている対象細胞に特異的に結合する蛍光標識を付加する染色工程と、
前記対象細胞に特異的に付加された前記蛍光標識の蛍光強度に基づいて前記対象細胞であるか否かを判定して前記対象細胞を特定する判定工程と、
を有する希少細胞の判定方法であって、
前記保管工程は、請求項1から5のいずれか一項に記載の保管方法が用いられていることを特徴とする判定方法。
A pretreatment step including a storage step for storing a cell sample generated from a blood-derived sample;
A staining step of adding a fluorescent label that specifically binds to a target cell contained in the blood-derived specimen;
A determination step of determining whether or not the target cell is based on the fluorescence intensity of the fluorescent label specifically added to the target cell and identifying the target cell;
A method for determining a rare cell having
The determination method according to claim 1, wherein the storage method uses the storage method according to claim 1.
前記判定工程は、前記蛍光強度と、前記対象細胞の種類に応じて予め設定された特定の閾値とを比較して前記対象細胞であるか否かを判定することを特徴とする請求項6に記載の判定方法。   The said determination step compares the said fluorescence intensity with the specific threshold value preset according to the kind of said object cell, and determines whether it is the said object cell, It is characterized by the above-mentioned. The determination method described.
JP2018500061A 2016-02-19 2017-02-08 Storage method of blood-derived specimen and determination method of rare cells Pending JPWO2017141789A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2016030055 2016-02-19
JP2016030055 2016-02-19
PCT/JP2017/004595 WO2017141789A1 (en) 2016-02-19 2017-02-08 Method for storing blood-derived specimens and method for determining rare cells

Publications (1)

Publication Number Publication Date
JPWO2017141789A1 true JPWO2017141789A1 (en) 2018-12-13

Family

ID=59626051

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2018500061A Pending JPWO2017141789A1 (en) 2016-02-19 2017-02-08 Storage method of blood-derived specimen and determination method of rare cells

Country Status (2)

Country Link
JP (1) JPWO2017141789A1 (en)
WO (1) WO2017141789A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486477A (en) * 1991-06-13 1996-01-23 Carver, Jr.; Edward L. Reagent system for improved multiple species blood analysis
JP2005536550A (en) * 2002-08-23 2005-12-02 コールター インターナショナル コーポレイション Formaldehyde-ammonium salt complex for stabilization of blood cells
JP2008511829A (en) * 2004-08-27 2008-04-17 ベックマン コールター,インコーポレーテッド Whole blood products for cytometric analysis of cell signaling pathways
WO2014185151A1 (en) * 2013-05-13 2014-11-20 コニカミノルタ株式会社 Cell-staining method and specimen collection tube for use in method
CN104178424A (en) * 2013-05-24 2014-12-03 益善生物技术股份有限公司 Red blood cell lysis solution and lysis method thereof
CN104178454A (en) * 2013-05-24 2014-12-03 益善生物技术股份有限公司 Enrichment and analysis method for circulating tumor cells
JP2015500999A (en) * 2011-12-21 2015-01-08 ベックマン コールター, インコーポレイテッド Method for labeling intracellular and extracellular targets of leukocytes

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2813891B1 (en) * 2000-09-14 2005-01-14 Immunotech Sa MULTIFUNCTIONAL REAGENT FOR ERYTHROCYTES INVOLVING CARBAMATES AND APPLICATIONS
US6653137B2 (en) * 2001-12-03 2003-11-25 Streck Laboratories Inc. Hematology reference control
WO2013168767A1 (en) * 2012-05-10 2013-11-14 コニカミノルタ株式会社 Process for removing red blood cells and centrifugal tube for blood collection
WO2014007190A1 (en) * 2012-07-03 2014-01-09 コニカミノルタ株式会社 Cell-spreading device and method for detecting rare cell
WO2014061675A1 (en) * 2012-10-17 2014-04-24 コニカミノルタ株式会社 Method for recovering rare cells and method for detecting rare cells

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486477A (en) * 1991-06-13 1996-01-23 Carver, Jr.; Edward L. Reagent system for improved multiple species blood analysis
JP2005536550A (en) * 2002-08-23 2005-12-02 コールター インターナショナル コーポレイション Formaldehyde-ammonium salt complex for stabilization of blood cells
JP2008511829A (en) * 2004-08-27 2008-04-17 ベックマン コールター,インコーポレーテッド Whole blood products for cytometric analysis of cell signaling pathways
JP2015500999A (en) * 2011-12-21 2015-01-08 ベックマン コールター, インコーポレイテッド Method for labeling intracellular and extracellular targets of leukocytes
WO2014185151A1 (en) * 2013-05-13 2014-11-20 コニカミノルタ株式会社 Cell-staining method and specimen collection tube for use in method
CN104178424A (en) * 2013-05-24 2014-12-03 益善生物技术股份有限公司 Red blood cell lysis solution and lysis method thereof
CN104178454A (en) * 2013-05-24 2014-12-03 益善生物技术股份有限公司 Enrichment and analysis method for circulating tumor cells

Also Published As

Publication number Publication date
WO2017141789A1 (en) 2017-08-24

Similar Documents

Publication Publication Date Title
JP6179508B2 (en) Method for detecting rare cells
Grisendi et al. Detection of microparticles from human red blood cells by multiparametric flow cytometry
JP2005501236A (en) Stabilization of cells and biological specimens for analysis
CN107449713B (en) Method for mixed antibody dependent sorting and enrichment of circulating tumor cells
JPH0737978B2 (en) Viable cell labeling
CN106872338A (en) A kind of blood cell stabilizer and its application
Tong et al. Isolation and characterization of extracellular vesicles from ex vivo cultured human placental explants
WO2018232589A1 (en) Method and device for identifying platelet aggregation, and cell analyzer
JP6617516B2 (en) Method for detecting target cells contained in blood sample
KR20180083853A (en) Blood products and profiling
EP2450457B1 (en) Method of analyzing genetically abnormals cells
CA2926304C (en) Use of a reagent for the lysis of erythrocytes as well as methods and kits relating thereto
Miller et al. Detection of C4d deposition in cardiac allografts: a comparative study of immunofluorescence and immunoperoxidase methods
CN104833805A (en) Circulating tumor cell detection and identification kit and application thereof
JP7143678B2 (en) Methods for detecting target cells
WO2016104308A1 (en) Cell image analysis method and cell image analysis device
JP6617495B2 (en) Method for detecting tumor cells
JP6582486B2 (en) Method for detecting rare cells in blood
CN112305232A (en) Mixed quality control substance of blood and specific protein and quality control method thereof
Siwaponanan et al. Enumeration and phenotyping of circulating microvesicles by flow cytometry and nanoparticle tracking analysis: Plasma versus serum
RU2437100C1 (en) Diagnostic technique for abnormal chromatin packaging in male infertility
WO2017141789A1 (en) Method for storing blood-derived specimens and method for determining rare cells
CN112504793B (en) Reagent for permeating and fixing blood cells and analysis method
Santise et al. Identification of mesothelial cells in intraoperative blood salvage
JP7279542B2 (en) Method for quantifying target cells contained in a sample

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20190327

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20200407

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20200603

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20201110

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20210511