[go: up one dir, main page]

CN116463291A - Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof - Google Patents

Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof Download PDF

Info

Publication number
CN116463291A
CN116463291A CN202211462629.9A CN202211462629A CN116463291A CN 116463291 A CN116463291 A CN 116463291A CN 202211462629 A CN202211462629 A CN 202211462629A CN 116463291 A CN116463291 A CN 116463291A
Authority
CN
China
Prior art keywords
organoid
liver metastases
tumor
cell line
colorectal cancer
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
CN202211462629.9A
Other languages
Chinese (zh)
Inventor
程芳玲
许三鹏
陈孝平
张必翔
梁慧芳
董汉华
郭尔钢
江晶晶
张超
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.)
Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology
Original Assignee
Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology
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 Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology filed Critical Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology
Priority to CN202211462629.9A priority Critical patent/CN116463291A/en
Publication of CN116463291A publication Critical patent/CN116463291A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0271Chimeric vertebrates, e.g. comprising exogenous cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/067Hepatocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0679Cells of the gastro-intestinal tract
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/12Animals modified by administration of exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Environmental Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

本发明公开了一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系及其培养方法与应用,属于消化系统肿瘤细胞系技术领域。它包括从一例右半结肠癌合并同时性肝转移患者的结肠病灶和肝脏转移灶肿瘤组织中分离肿瘤细胞通过3D培养得到保藏于中国典型培养物保藏中心CCTCC,保藏号为CCTCC NO.C202218的人右半结肠癌原发灶类器官细胞系CWH22,和保藏号为CCTCC NO.C202219的配对人右半结肠癌肝转移灶类器官细胞系CLM22。这对类器官细胞系能够在体外稳定传代培养、裸鼠成瘤性良好,可用于结直肠癌及肝转移的发病机制、耐药机理、新药筛选等方面的研究。

The invention discloses a paired organoid cell line of human colorectal cancer primary focus and liver metastases, a culture method and application thereof, and belongs to the technical field of digestive system tumor cell lines. It includes separating tumor cells from the colon lesion and liver metastases tumor tissue of a patient with right colon cancer combined with simultaneous liver metastases and through 3D culture to obtain the human right colon cancer primary tumor organoid cell line CWH22 with the preservation number CCTCC NO.C202218 and the paired human right colon cancer liver metastases organoid cell line CLM22 with the preservation number CCTCC No. This pair of organoid cell lines can be stably subcultured in vitro, has good tumorigenicity in nude mice, and can be used for research on the pathogenesis, drug resistance mechanism, and new drug screening of colorectal cancer and liver metastasis.

Description

人结直肠癌原发灶与肝脏转移灶配对类器官细胞系及其培养 方法与应用Human colorectal cancer primary tumor and liver metastases paired organoid cell line and its culture Method and Application

技术领域technical field

本发明涉及一对右半结肠癌类器官和对应的肝脏转移灶类器官的培养建系过程,及构建的相关动物成瘤模型,属于消化系统肿瘤细胞系技术模型领域,具体涉及一种人结肠癌原发灶与肝脏转移灶配对类器官细胞系及其培养方法与应用。The present invention relates to a process of culturing and establishing a pair of right-sided colon cancer organoids and corresponding liver metastases organoids, and a related animal tumor formation model constructed thereof, belonging to the field of digestive system tumor cell line technology models, and specifically relates to a paired organoid cell line of a primary human colon cancer and a liver metastases, and a culture method and application thereof.

背景技术Background technique

结直肠癌是人类最常见的恶性肿瘤之一,目前在我国人群中的发病率一直呈上升趋势,在男性和女性中分别占所有癌种发病率的第五位和第四位,死亡率占所有癌种的第五位。结直肠癌的5年生存率不同分期差异很大:I期达90%,IV期仅为15%。肝脏是IV期结直肠癌最常见的转移部位,20-25%的结直肠癌患者在初诊时即伴有肝转移,50%的患者在原发灶切除后治疗过程中发生肝转移。虽然根治性手术切除是结直肠癌肝转移的标准治疗方法,但仅有不到20%的肝转移患者可以进行较好的转移灶切除,且都不可避免地继续肝脏复发。因此探讨结直肠癌肝转移的潜在机制、分子标志物和有效的治疗策略等,对提早阻断或延缓肝转移的发生、有效控制肿瘤的复发进展、延长患者的总生存等极为重要。Colorectal cancer is one of the most common malignant tumors in humans. At present, the incidence of colorectal cancer in the Chinese population has been on the rise. It accounts for the fifth and fourth incidence of all cancers in men and women, and the fifth death rate among all cancers. The 5-year survival rate of colorectal cancer varies greatly among different stages: 90% for stage I and only 15% for stage IV. The liver is the most common metastatic site of stage IV colorectal cancer. 20-25% of colorectal cancer patients have liver metastases when they are first diagnosed, and 50% of patients develop liver metastases during treatment after primary tumor resection. Although radical surgical resection is the standard treatment for liver metastases from colorectal cancer, less than 20% of patients with liver metastases can undergo good metastasectomy, and all of them inevitably continue to relapse in the liver. Therefore, exploring the potential mechanism, molecular markers and effective treatment strategies of colorectal cancer liver metastasis is extremely important for early blocking or delaying the occurrence of liver metastasis, effectively controlling tumor recurrence and progression, and prolonging the overall survival of patients.

在肿瘤的研究探索中,研究模型的可靠性是将基础研究有效地转化到临床应用的关键。肿瘤类器官被证明是用于研究疾病进展、受影响的信号通路、肿瘤微环境和相关药物治疗等的有用实验模型。结直肠癌类器官是从患者的结直肠原发灶或转移灶肿瘤组织中提取肿瘤细胞在基质胶中进行3D类器官培养,产生具有立体空间结构,且更接近体内生理水平的培养物,能最大程度地在体外模拟对应肿瘤的特性,实现更多的转化医学应用,如预测特定患者发生转移可能性的大小,对特定治疗方式敏感性的高低,筛选挽救性治疗的有效药物等,为有效预测患者的预后及探索个性化治疗提供帮助。In the research and exploration of tumors, the reliability of research models is the key to effectively transform basic research into clinical application. Tumor organoids have proven to be useful experimental models for studying disease progression, affected signaling pathways, tumor microenvironment, and relevant drug treatments, among others. Colorectal cancer organoids are extracted from patients’ colorectal primary or metastatic tumor tissues and cultured in 3D organoids in matrigel to produce cultures with a three-dimensional spatial structure that are closer to the physiological level in vivo, which can simulate the characteristics of corresponding tumors in vitro to the greatest extent, and realize more translational medical applications, such as predicting the possibility of metastasis in a specific patient, the sensitivity to a specific treatment method, and screening effective drugs for salvage therapy.

结直肠癌发生肝转移的机制尚不清楚,且无明确预测指标预判对标准治疗方案的敏感性,患者对标准多线治疗耐药后无其他可选药物进一步治疗是目前治疗的窘境。患者来源的类器官在结直肠癌转移机制及耐药机制的研究中的作用举足轻重,但目前尚无可公开使用的类器官细胞系。我们的建立的这对结肠癌原发灶及对应肝转移灶类器官细胞系将在提高治疗方案的针对性和有效性,进一步筛选有效治疗药物,降低复发率,延长患者生存期等研究中发挥重要作用。The mechanism of liver metastases in colorectal cancer is still unclear, and there is no clear predictor to predict the sensitivity to standard treatment regimens. Patients who are resistant to standard multi-line therapy and have no other options for further treatment are the dilemma of current treatment. Patient-derived organoids play an important role in the study of colorectal cancer metastasis and drug resistance mechanisms, but there are currently no publicly available organoid cell lines. The pair of colon cancer primary tumors and corresponding liver metastases organoid cell lines established by us will play an important role in improving the pertinence and effectiveness of the treatment plan, further screening effective therapeutic drugs, reducing the recurrence rate, and prolonging the survival of patients.

发明内容Contents of the invention

为实现上述技术目的,本发明公开了一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系及其培养方法与应用,该细胞系不仅能够在体外稳定传代培养,而且具备较强的增殖活性和裸鼠皮下、盲肠原位及肝脏转移成瘤能力。为进一步深入研究结直肠癌及肝转移的病因学、发病机制、耐药机理、新药筛选等提供新的体内外研究支撑。In order to achieve the above technical purpose, the present invention discloses a paired organoid cell line of human colorectal cancer primary focus and liver metastases and its culture method and application. The cell line can not only be stably subcultured in vitro, but also has strong proliferative activity and the ability to form nude mouse subcutaneous, cecal in situ and liver metastatic tumors. To provide new in vivo and in vitro research support for further in-depth research on the etiology, pathogenesis, drug resistance mechanism, and new drug screening of colorectal cancer and liver metastasis.

为更好的实现本发明技术目的,本发明公开了一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系,包括保藏于中国典型培养物保藏中心CCTCC,保藏号为CCTCCNO.C202218的人右半结肠癌原发灶类器官细胞系CWH22与对应的保藏号为CCTCCNO.C202219的配对人右半结肠癌肝转移灶类器官细胞系CLM22。In order to better realize the technical purpose of the present invention, the present invention discloses a paired organoid cell line of the primary human colorectal cancer and liver metastases, including the organoid cell line CWH22 of the primary human right colon cancer with the preservation number CCTCCNO.

进一步地,所述细胞系为一例右半结肠癌合并同时性肝转移患者同时切除的原发灶和对应肝脏转移灶肿瘤组织进行体外3D类器官培养建立得到。Further, the cell line was established by in vitro 3D organoid culture of a patient with right colon cancer complicated with simultaneous liver metastases by simultaneously resecting the primary tumor and the tumor tissue of the corresponding liver metastases.

本发明的目的之二是公开一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系的培养方法,它包括如下步骤:The second object of the present invention is to disclose a method for cultivating a paired organoid cell line between the primary human colorectal cancer and liver metastases, which includes the following steps:

1)取材:术中分别留取该患者原发灶及对应肝脏转移灶肿瘤组织各0.5cm3于含Advanced DMEM/F12的无菌取样管中,置于4℃冰盒快速送到实验室;1) Material collection: 0.5 cm3 of tumor tissue from the patient’s primary tumor and corresponding liver metastases were collected during the operation, placed in a sterile sampling tube containing Advanced DMEM/F12, placed in a 4°C ice box, and quickly sent to the laboratory;

2)消化:在无菌超净台中用PBS清洗弃去步骤1)所得样本表面的黏液、坏死组织和残余血细胞,继而用活力碘(临床黏膜消毒用)冲洗一次及PBS冲洗三次后,在平皿中用剪刀将组织反复剪碎,加入2mL50%浓度消化酶TrypLE(2mLDMEM稀释)室温消化,每隔5min充分混匀含消化酶的组织悬液,约20min左右,加入等体积含Ca2+的HBSS缓冲液中和并再次混匀;2) Digestion: Wash and discard the mucus, necrotic tissue and residual blood cells on the surface of the sample obtained in step 1) in a sterile ultra-clean bench, then rinse once with iodine (for clinical mucous membrane disinfection) and three times with PBS, cut the tissue repeatedly with scissors in a plate, add 2 mL of 50% digestive enzyme TrypLE (diluted in 2 mL of DMEM) for digestion at room temperature, and fully mix the tissue suspension containing digestive enzyme every 5 minutes. About 20 minutes, add an equal volume of Ca 2 + HBSS buffer to neutralize and mix again;

3)过滤:用不小于200目网径的滤网过滤步骤2)所得悬液,并离心,沉淀用5mL培养基重悬,并反复洗涤离心弃去坏死细胞碎片,离心转速500~1500r/min;3) Filtration: filter the suspension obtained in step 2) with a filter of not less than 200 mesh diameter, and centrifuge, resuspend the precipitate with 5 mL of culture medium, wash and centrifuge repeatedly to discard necrotic cell fragments, and centrifuge at a speed of 500-1500 r/min;

4)种细胞:取上述洗涤所得细胞进行活细胞计数,用基质胶低温重悬细胞为8000个/30μL,种于已预热的24孔板中,30μL/孔,置于37℃培养箱30min待穹隆状基质胶凝固后,每孔添加培养基650μL;4) Seed cells: Take the above-mentioned washed cells for live cell counting, resuspend the cells with Matrigel at low temperature to 8000 cells/30 μL, seed in a preheated 24-well plate, 30 μL/well, place in a 37°C incubator for 30 minutes, and add 650 μL of medium to each well after the vaulted Matrigel solidifies;

5)常规传代培养:培养基每3~5天更换一次,在第10~14天时进行传代操作,用TrypLE消化酶250μL/孔进行消化,每隔5分钟吹打混匀一次,10~15min可消化为单个细胞悬液,以HBSS中和离心重悬后计数,以8000个细胞每孔继续传代,后续重复步骤4)和5)。5) Conventional subculture: the medium was changed every 3 to 5 days, and the subculture operation was performed on the 10th to 14th day. Digest with TrypLE digestive enzyme 250 μL/well, pipette and mix every 5 minutes, digest into a single cell suspension in 10 to 15 minutes, resuspend with HBSS neutralization and centrifugation, and count, continue to pass at 8000 cells per well, and then repeat steps 4) and 5).

本发明的目的之三是公开一种上述人结直肠癌原发灶与肝脏转移灶配对类器官细胞系用于构建盲肠原位成瘤动物模型和/或肝脏转移灶成瘤动物模型并用来研究直肠癌发生机理及耐药机理。The third objective of the present invention is to disclose a paired organoid cell line of the above-mentioned human colorectal cancer primary tumor and liver metastases, which is used to construct an animal model of cecal orthotopic tumor formation and/or an animal model of liver metastases tumor formation, and to study the pathogenesis and drug resistance mechanism of rectal cancer.

本发明的目的之四是公开一种肝脏转移灶成瘤动物模型的建立方法,它包括如下步骤:The fourth object of the present invention is to disclose a method for establishing a tumor-forming animal model of liver metastases, which includes the following steps:

1)类器官培养:将类器官细胞使用慢病毒感染稳定过表达荧光素酶后,常规培养类器官,于传代后第4、7、9、11天分别换液,取处于对数生长期的CWH22和CLM22类器官(每代次生长第10-12天)进行裸鼠脾脏注射转移成瘤操作;1) Organoid culture: After the organoid cells were stably overexpressed luciferase with lentivirus infection, the organoids were cultured routinely, and the medium was changed respectively on the 4th, 7th, 9th, and 11th day after passage, and the CWH22 and CLM22 organoids in the logarithmic growth phase (10-12 days of each passage) were taken for nude mouse spleen injection to transfer tumors;

2)类器官消化:消化收细胞过程同常规传代操作,尽量消化为单细胞悬液状态,并计数,以DMEM重悬为1.25*107/mL置4℃保温箱备用;2) Digestion of organoids: the process of digesting and harvesting cells is the same as the conventional subculture operation, try to digest into a single-cell suspension state, and count them, resuspend in DMEM to 1.25*10 7 /mL and put them in a 4°C incubator for later use;

3)脾脏注射:戊巴比妥麻醉裸鼠后,固定裸鼠并做腹部皮肤消毒,沿腹部左肋下缘腋前线前0.5cm处做纵向约1cm切口至腹腔,轻轻挑出脾脏后从脾脏远端注射混匀的细胞悬液约80μL/只裸鼠,10min后结扎脾脏血管剪除脾脏并缝合,密切观察确保顺利苏醒。3) Spleen injection: after anesthetizing the nude mice with pentobarbital, fix the nude mice and perform abdominal skin disinfection, make a longitudinal incision of about 1 cm along the left lower costal margin of the abdomen at the front of the axillary line 0.5 cm to the abdominal cavity, gently pick out the spleen and inject about 80 μL of mixed cell suspension from the distal end of the spleen. After 10 minutes, the splenic blood vessels were ligated, the spleen was cut off and sutured, and the spleen was closely observed to ensure smooth recovery.

4)成瘤监测:定期活体成像观察裸鼠肝转移成瘤情况并记录。4) Tumor formation monitoring: regular in vivo imaging was used to observe and record the tumor formation of liver metastases in nude mice.

本发明的目的之五是公开一种盲肠原位成瘤动物模型的构建方法,它包括如下步骤:The fifth object of the present invention is to disclose a method for constructing an animal model of cecum in situ tumor formation, which includes the following steps:

1)类器官培养:常规培养过表达荧光素酶的类器官CWH22和CLM22,于传代后第4、7、9、11天分别换液,取处于对数生长期的类器官进行裸鼠盲肠壁注射操作;其中,每代次生长第10~12天;1) Organoid culture: routinely culture luciferase-overexpressing organoids CWH22 and CLM22, and change the medium on the 4th, 7th, 9th, and 11th days after passage, and inject the organoids in the logarithmic growth phase into the cecum wall of nude mice; wherein, each passage grows on the 10th to 12th day;

2)类器官收集:于第10~12天收集类器官,以TrypLE消化酶常规消化5min后,中和收集类器官沉淀,约3*106细胞,预冷后重悬于60μL基质胶中置4℃保温箱备用;2) Organoid collection: Collect organoids on the 10th to 12th day, digest with TrypLE digestive enzyme for 5 minutes, neutralize and collect organoid precipitates, about 3*10 6 cells, resuspend in 60 μL Matrigel after pre-cooling, and place in a 4°C incubator for later use;

3)盲肠原位注射:戊巴比妥麻醉裸鼠后,固定裸鼠并做腹部皮肤消毒,沿下腹部正中线做纵向约1.5cm切口至腹腔并找到盲肠,将盲肠末端移至视野正中,以预冷胰岛素注射针抽取类器官悬液20μL做盲肠粘膜下注射。注射完毕观察无渗漏后,做腹部肌肉和皮肤缝合,并密切观察确保顺利苏醒;3) In situ injection of the cecum: after anesthetizing the nude mice with pentobarbital, fix the nude mice and perform abdominal skin disinfection, make a longitudinal incision about 1.5 cm along the midline of the lower abdomen to the abdominal cavity and find the cecum, move the end of the cecum to the center of the field of vision, and draw 20 μL of the organoid suspension with a pre-cooled insulin injection needle for submucosal injection of the cecum. After the injection is completed and no leakage is observed, the abdominal muscles and skin are sutured, and close observation is made to ensure a smooth recovery;

4)成瘤监测:定期活体成像观察裸鼠盲肠原位成瘤情况并记录。4) Tumor formation monitoring: Regular in vivo imaging was used to observe and record the in situ tumor formation in the cecum of nude mice.

本发明目的之六是公开一种上述人结肠癌原发灶与肝脏转移灶配对类器官细胞系在制备用于筛选治疗结直肠癌和/或肝转移灶肿瘤药物中的应用。The sixth object of the present invention is to disclose the application of the above-mentioned organoid cell line paired with primary human colon cancer and liver metastases in the preparation of drugs for screening and treating colorectal cancer and/or liver metastases.

进一步地,将治疗药物施用于体外细胞模型中,施用后抑制细胞增殖或导致细胞凋亡的为治疗敏感性药物,或者将测试化合物施用于体外细胞模型中,施用后抑制细胞增殖或导致细胞凋亡的为候选化合物。Further, the therapeutic drug is administered to an in vitro cell model, and the drug that inhibits cell proliferation or causes cell apoptosis after administration is a therapeutically sensitive drug, or the test compound is administered to an in vitro cell model, and the candidate compound is a compound that inhibits cell proliferation or causes cell apoptosis after administration.

本发明目的之七是公开一种筛选用于治疗结直肠癌和/或肝转移灶肿瘤药物的方法,它包括取上述对数生长期的CWH22和CLM22类器官消化计数重悬于基质胶并种于96孔板内,2*105个cell/mL,10μL/孔,待基质胶凝固后,添加培养基100μL/孔,培养5~6天时,其中,类器官直径约100μm,替换成梯度药物培养基继续培养,3天换液一次,药物作用6天后通过3D细胞活性检测,计算对应药物IC50。The seventh object of the present invention is to disclose a method for screening tumor drugs for the treatment of colorectal cancer and/or liver metastases, which includes digesting and counting the above-mentioned CWH22 and CLM22 organoids in the logarithmic growth phase, resuspending them in Matrigel and planting them in a 96-well plate, 2*10 5 cells/mL, 10 μL/well, after the Matrigel is solidified, adding 100 μL/well of medium, and culturing for 5 to 6 days. The culture medium continued to be cultured, and the medium was changed once every 3 days. After 6 days of drug action, the 3D cell activity was detected to calculate the IC50 of the corresponding drug.

进一步地,所述梯度药物包含如下中的至少一种:5-氟尿嘧啶,奥沙利铂,伊立替康(SN-38),依托泊苷(VP-16),索托拉西布(sotorasib,AMG-510),瑞戈非尼(Regorafenib),达沙替尼(Dasatinib)或硼替佐米(Bortezomib,PS-341)。Further, the drug gradient comprises at least one of the following: 5-fluorouracil, oxaliplatin, irinotecan (SN-38), etoposide (VP-16), sotorasib (sotorasib, AMG-510), regorafenib, dasatinib or bortezomib (PS-341).

有益效果:Beneficial effect:

1、本发明培养得到的人右半结肠癌原发灶类器官CWH22和对应的肝脏转移灶类器官CLM22经STR鉴定为新的细胞株,且来自于同一患者,为人结直肠癌和肝转移的研究提供了新的与临床肿瘤生物学接近的实验材料。1. The human right colon cancer primary focus organoid CWH22 and the corresponding liver metastases organoid CLM22 cultured in the present invention are identified as new cell lines by STR, and come from the same patient, providing new experimental materials close to clinical tumor biology for the study of human colorectal cancer and liver metastasis.

2、本发明筛选得到的人结直肠癌原发灶类器官CWH22和对应的肝脏转移灶类器官CLM22具备较好3D类器官形态,且性状稳定,可持续稳定传代,可良好耐受病毒感染等特征,研究发现其具有非常强的盲肠原位成瘤和脾脏注射肝脏转移成瘤能力,可以成功制备结直肠癌肝转移动物模型,实验发现注射100万细胞于盲肠原位或者脾脏,25天左右,可形成明显的肿瘤,可用于在裸鼠体内进行结直肠癌转移机制及综合治疗的药物敏感性研究等。2. The human colorectal cancer primary tumor organoid CWH22 and the corresponding liver metastases organoid CLM22 screened by the present invention have good 3D organoid morphology, stable properties, sustainable and stable passage, and good tolerance to virus infection. Studies have found that they have very strong cecum in situ tumor formation and liver metastasis tumor formation ability by spleen injection, and can successfully prepare animal models of colorectal cancer liver metastasis. Experiments have found that injecting 1 million cells into the cecum in situ or in the spleen can form obvious tumors in about 25 days. It can be used to study the metastasis mechanism of colorectal cancer and the drug sensitivity of comprehensive treatment in nude mice.

3、本发明培养得到的人结直肠癌原发灶类器官细胞系CWH22和对应的转移灶类器官细胞系CLM22,全外显子(WES)测序提示均有KRAS外显子2错意突变(NM_033360.3:p.Gly12Cys/c.34G>T),APC基因移码突变(NM_000038.5:p.Tyr935fs/c.2805_2806delCA),SMAD4基因错义突变(:c.1610A>G),KMT2C错意突变(NM_170606.2:p.Gln3158His/c.9474G>T)及CDKN2B基因移码突变(NM_004064.4:p.Ser110fs/c.323_329dupATGTCAG)等。此外结直肠癌常见的相关驱动基因NRAS/HRAS/BRAF/PIK3CA,TP53等均为野生型。这组配对的类器官细胞系为典型的同时有KRAS、APC和SMAD4突变的结直肠癌类器官细胞系,故其可作为一有效的工具细胞,用于KRAS/APC/SMAD4三突变、TP53野生型亚群的结直肠癌发生发展及治疗耐药的相关机制研究。3. The human colorectal cancer primary focus organoid cell line CWH22 and the corresponding metastatic focus organoid cell line CLM22 cultivated in the present invention, all exome (WES) sequencing revealed that both had KRAS exon 2 missense mutation (NM_033360.3:p.Gly12Cys/c.34G>T), APC gene frameshift mutation (NM_000038.5:p.Tyr935fs/ c.2805_2806delCA), SMAD4 gene missense mutation (: c.1610A>G), KMT2C missense mutation (NM_170606.2:p.Gln3158His/c.9474G>T) and CDKN2B gene frameshift mutation (NM_004064.4:p.Ser110fs/c.323_329dupATGT CAG) etc. In addition, the common related driver genes NRAS/HRAS/BRAF/PIK3CA, TP53, etc. in colorectal cancer were all wild type. This group of paired organoid cell lines is a typical colorectal cancer organoid cell line with KRAS, APC and SMAD4 mutations at the same time, so it can be used as an effective tool cell for the study of the occurrence and development of colorectal cancer with KRAS/APC/SMAD4 triple mutations and TP53 wild-type subgroup and the mechanism of drug resistance.

附图说明Description of drawings

图1为本发明探究得到的CWH22和CLM22类器官细胞所来源的病人术前相关检查。其中,图1A为术前肠镜检查情况,图1B为病人术前肝脏CT及MR平扫情况。Figure 1 shows the preoperative examinations of the patients from which the CWH22 and CLM22 organoid cells obtained in the present invention are derived. Among them, Fig. 1A shows the preoperative colonoscopy examination, and Fig. 1B shows the preoperative liver CT and MR plain scan of the patient.

图2为图1的患者术中所切除原发灶及转移灶肿瘤组织对应的HE及免疫组化染色结果。CK20(+),CDX-2(+),Her-2(4B5)(-),Her-2(阳性对照)(+),P53(散在+),Ki-67(LI约60%),MLH1(+),PMS2(+),MSH2(+),MSH6(+),Villin(+)。根据形态学及免疫组化诊断为中分化结直肠腺癌。Figure 2 shows the results of HE and immunohistochemical staining corresponding to the resected primary tumor and metastatic tumor tissue of the patient in Figure 1. CK20(+), CDX-2(+), Her-2(4B5)(-), Her-2(positive control)(+), P53(scattered+), Ki-67(LI about 60%), MLH1(+), PMS2(+), MSH2(+), MSH6(+), Villin(+). Moderately differentiated colorectal adenocarcinoma was diagnosed based on morphology and immunohistochemistry.

图3为图2中所取原发灶和转移灶肿瘤组织培养所得类器官CWH22和CLM22显微镜下明场形态学及动态生长记录。其中比例尺分别为500μm和200μm。Figure 3 shows the bright field morphology and dynamic growth records of organoids CWH22 and CLM22 obtained from the primary tumor and metastatic tumor tissue cultured in Figure 2. The scale bars are 500 μm and 200 μm, respectively.

图4为图1中所取肿瘤组织及图3中培养所得类器官的相关染色情况对比。图4A为图1中所取原发灶和转移灶肿瘤组织肉眼所见形态及组织切片HE染色,CK20和CDX2表达情况。图4B为培养的类器官明场形态学和类器官切片对应的HE染色,CK20和CDX2表达情况。Figure 4 is a comparison of the relevant staining conditions of the tumor tissue obtained in Figure 1 and the cultured organoids in Figure 3. Figure 4A shows the macroscopic morphology of the primary tumor and the metastatic tumor tissue taken in Figure 1, HE staining of the tissue section, and the expression of CK20 and CDX2. Figure 4B shows the bright-field morphology of cultured organoids and corresponding HE staining of organoid slices, and the expression of CK20 and CDX2.

图5为CWH22和CLM22全外显子测序,与对应肿瘤组织突变一致性及突变特征分析。Figure 5 shows the whole exon sequencing of CWH22 and CLM22, and the mutation consistency and mutation characteristics analysis of corresponding tumor tissues.

图6为CWH22和CLM22细胞裸鼠体内肝转移成瘤相关研究。图6A为经脾脏分别注射100万CWH22和CLM22细胞至两组裸鼠(5只/组),第45天活体成像信号及取肝脏观察转移成瘤情况;图6B为对应HE染色,及对应的CK20和CDX2表达情况。Fig. 6 is a study on liver metastasis and tumor formation of CWH22 and CLM22 cells in nude mice. Figure 6A shows the intrasplenic injection of 1 million CWH22 and CLM22 cells into two groups of nude mice (5 mice/group), live imaging signals on day 45 and observation of metastatic tumor formation in the liver; Figure 6B shows the corresponding HE staining and the corresponding expression of CK20 and CDX2.

图7为CWH22类器官细胞盲肠注射原位成瘤后肉眼所见及HE染色情况。Figure 7 shows the macroscopic findings and HE staining of CWH22 organoid cells after cecum injection to form tumors in situ.

图8为图3中细胞CWH22和CLM22在对应浓度梯度下,5-氟尿嘧啶,奥沙利铂,伊立替康(SN-38),依托泊苷(VP-16),索托拉西布(sotorasib,AMG-510),瑞戈非尼(Regorafenib),达沙替尼(Dasatinib)和硼替佐米(Bortezomib,PS-341)的剂量效应反应曲线及对应的IC50检测结果。Figure 8 is the dose-response curves of 5-fluorouracil, oxaliplatin, irinotecan (SN-38), etoposide (VP-16), sotorasib (AMG-510), regorafenib, dasatinib and bortezomib (PS-341) under the corresponding concentration gradient of cells CWH22 and CLM22 in Figure 3 And the corresponding IC50 test results.

图9为临床患者术前及术后8个疗程mFOLFOX+Bevacizumab治疗过程中影像学复查情况对比。Figure 9 is a comparison of imaging re-examinations of clinical patients before and after 8 courses of mFOLFOX+Bevacizumab treatment.

具体实施方式Detailed ways

下面结合具体实例,进一步阐述本发明。这些实施例仅用于说明本发明而不用于限制本发明的范围。Below in conjunction with specific example, further set forth the present invention. These examples are only for illustrating the present invention and are not intended to limit the scope of the present invention.

下列实施例中未注明具体条件的实验方法,可采用本领域中的常规方法,例如参考《分子克隆实验指南》(第三版,纽约,冷泉港实验室出版社,New York:ColdSpringHarbor Laboratory Press,1989)或按照供应商所建议的条件。For the experimental methods that do not indicate specific conditions in the following examples, conventional methods in the art can be used, for example, refer to "Molecular Cloning Experiment Guide" (Third Edition, New York, Cold Spring Harbor Laboratory Press, New York: Cold Spring Harbor Laboratory Press, 1989) or according to the conditions suggested by the supplier.

本发明涉及生物材料保藏,其中,保藏单位为中国典型培养物保藏中心,地址为中国武汉,武汉大学,邮编:430072,保藏时间为2022年10月21日。The present invention relates to the preservation of biological materials, wherein the preservation unit is the China Typical Culture Collection Center, the address is Wuhan University, Wuhan, China, postcode: 430072, and the preservation time is October 21, 2022.

下列实施例中未特别说明的各种仪器、原料和试剂均为本领域熟知的市售产品,可通过商业途径获得。在下列实施例中列出的所使用的具体材料及其来源,仅仅是示例性的,并不意图限制本发明,与如下组织、细胞、试剂和仪器的类型、型号、品质、性质或功能相同或相似的材料均可以用于实施本发明。Various instruments, raw materials and reagents not specifically described in the following examples are commercially available products well known in the art and can be obtained through commercial channels. The specific materials used and their sources listed in the following examples are exemplary only and are not intended to limit the present invention. Materials identical or similar to the type, model, quality, property or function of the following tissues, cells, reagents and instruments can be used to implement the present invention.

实施例1Example 1

公开了一例人右半结肠癌原发灶类器官细胞系CWH22和对应的肝转移类器官细胞系CLM22的建立方法,它包括如下步骤:A method for establishing a primary human right colon cancer organoid cell line CWH22 and a corresponding liver metastasis organoid cell line CLM22 is disclosed, which includes the following steps:

1.1标本的来源:1.1 The source of the specimen:

该肿瘤组织标本来源于一例65岁右半结肠占位合并肝脏转移的患者,2021年1月因“体检发现肝脏占位半月余”入院,CT及MR平扫提示“肝左叶-尾叶巨大分块状肿块影,考虑肿瘤性病变”,肠镜检查示“升结肠新生物,Ca?”。后于我院行同期“右半结肠切除术+左半肝切除术”。术后病检示:右半结肠中分化腺癌,侵及肠壁浆膜下;(1)吻合环近端切缘:未见癌;(2)吻合环远端切缘:未见癌;(3)放射状切缘/系膜血管断端:未见癌,局灶紧邻切缘;(4)脉管侵犯:可见;(5)神经侵犯:未见;(6)肠系膜淋巴结:13枚未见癌;(7)淋巴结外肿瘤种植(ENTD):可见,N=1。(8)肝IV段、V段、VIII段、肝总动脉旁均可见癌;(9)胆囊、阑尾、肝十二指肠韧带淋巴结1枚均未见癌。免疫组化:MLH1(+),PMS2(+),MSH2(+),MSH6(+),CDX-2(+),Her-2(4B5)(-),Her-2(阳性对照)(+),P53(散在+),Ki-67(LI约60%)。基因检测提示:KRAS基因外显子2突变;MSS,BRAF扩增;TMB 4.61Muts/Mb,排位高于80.98%的患者。The tumor tissue sample came from a 65-year-old patient with a right-semi-colon mass and liver metastases. He was admitted to the hospital in January 2021 because of "a physical examination found that the liver mass occupied more than half a month." Plain CT and MR scans showed "a huge blocky mass in the left lobe-caudal lobe of the liver, considering neoplastic lesions." Colonoscopy showed "new growths in the ascending colon, Ca?". Later, "right hemicolectomy + left hemihepatectomy" was performed in our hospital at the same time. Postoperative pathological examination showed: moderately differentiated adenocarcinoma of the right colon, invading the subserosa of the intestinal wall; (1) Proximal resection margin of the anastomotic ring: no cancer; (2) Distal resection margin of the anastomotic ring: no cancer; (3) Radial resection margin/mesangial vessel stump: no cancer, focally adjacent to the resection margin; (4) vessel invasion: visible; (5) nerve invasion: no; (6) mesenteric lymph nodes: 13 no cancer; (7) extralymphatic tumor implantation ( ENTD): Visible, N=1. (8) Carcinoma was seen in segment IV, segment V, segment VIII of the liver, and beside the common hepatic artery; (9) Cancer was not found in one lymph node of the gallbladder, appendix, and hepatoduodenal ligament. Immunohistochemistry: MLH1(+), PMS2(+), MSH2(+), MSH6(+), CDX-2(+), Her-2(4B5)(-), Her-2(positive control)(+), P53(scattered+), Ki-67(LI about 60%). Genetic testing revealed: KRAS gene exon 2 mutation; MSS, BRAF amplification; TMB 4.61Muts/Mb, ranking higher than 80.98% of patients.

病人术前肠镜、CT及MR影像学表现见图1。病人肿瘤组织切片染色情况见图2。The patient's preoperative colonoscopy, CT and MR imaging findings are shown in Figure 1. The staining of the patient's tumor tissue sections is shown in Figure 2.

1.2标本处理:1.2 Specimen processing:

1)取材:术中分别留取该患者原发灶及转移灶肿瘤组织各0.5cm3于含AdvancedDMEM/F12的无菌取样管中,置于4℃冰盒快速送到实验室;1) Material collection: 0.5 cm 3 of the patient's primary tumor and metastatic tumor tissue were collected during the operation, placed in a sterile sampling tube containing AdvancedDMEM/F12, placed in a 4°C ice box, and quickly sent to the laboratory;

2)消化:在无菌超净台中用PBS清洗弃去步骤1)所得样本表面的黏液、坏死组织和残余血细胞,继而用活力碘(临床黏膜消毒用)冲洗一次及PBS冲洗三次后,在平皿中用剪刀将组织反复剪碎,加入2mL50%浓度消化酶TrypLE(2mLDMEM稀释)室温消化,每隔5min充分混匀含消化酶的组织悬液,约20min左右,加入等体积含Ca2+的HBSS缓冲液中和并再次混匀;2) Digestion: Wash and discard the mucus, necrotic tissue and residual blood cells on the surface of the sample obtained in step 1) in a sterile ultra-clean bench, then rinse once with iodine (for clinical mucous membrane disinfection) and three times with PBS, cut the tissue repeatedly with scissors in a plate, add 2 mL of 50% digestive enzyme TrypLE (diluted in 2 mL of DMEM) to digest at room temperature, fully mix the tissue suspension containing digestive enzyme every 5 minutes, about 20 minutes, add an equal volume of Ca 2 + HBSS buffer to neutralize and mix again;

3)过滤:用不小于200目网径的滤网过滤步骤2)所得悬液,并离心,沉淀用5mL培养基重悬,并反复洗涤离心弃去坏死细胞碎片,离心转速500~1500r/min;3) Filtration: filter the suspension obtained in step 2) with a filter of not less than 200 mesh diameter, and centrifuge, resuspend the precipitate with 5 mL of culture medium, wash and centrifuge repeatedly to discard necrotic cell fragments, and centrifuge at a speed of 500-1500 r/min;

4)种细胞:取上述洗涤所得细胞进行活细胞计数,用基质胶低温重悬细胞为8000个/30μL,种于已预热的24孔板中,30μL/孔,置于37℃培养箱30min待穹隆状基质胶凝固后,每孔添加培养基650μL;4) Seed cells: Take the above-mentioned washed cells for live cell counting, resuspend the cells with Matrigel at low temperature to 8000 cells/30 μL, seed in a preheated 24-well plate, 30 μL/well, place in a 37°C incubator for 30 minutes, and add 650 μL of medium to each well after the vaulted Matrigel solidifies;

5)常规传代培养:培养基每3~5天更换一次,在第10~14天时进行传代操作,用TrypLE消化酶250μL/孔进行消化,每隔5分钟吹打混匀一次,10~15min可消化为单个细胞悬液,以HBSS中和离心重悬后计数,以8000个细胞每孔继续传代,后续重复步骤4)和5)。即培养得到具备较好3D类器官形态,且性状稳定,可持续传代,并良好耐受病毒感染等的类器官细胞系。5) Conventional subculture: the medium was changed every 3 to 5 days, and the subculture operation was performed on the 10th to 14th day. Digest with TrypLE digestive enzyme 250 μL/well, pipette and mix every 5 minutes, digest into a single cell suspension in 10 to 15 minutes, resuspend with HBSS neutralization and centrifugation, and count, continue to pass at 8000 cells per well, and then repeat steps 4) and 5). That is, organoid cell lines with good 3D organoid morphology, stable properties, sustainable passage, and good tolerance to virus infection can be obtained.

实施例2Example 2

公开了一例人右半结肠癌原发灶类器官细胞系CWH22和对应肝转移类器官细胞系CLM22的生物学特性。The biological characteristics of the organoid cell line CWH22 of the primary focus of human right colon cancer and the corresponding organoid cell line CLM22 of liver metastases are disclosed.

2.1CWH22和CLM22类器官细胞系的形态学观察2.1 Morphological observation of CWH22 and CLM22 organoid cell lines

取传代培养的CWH22和CLM22类器官,在日本Olympus IMT-2倒置显微镜下观察记录类器官生长形态学变化情况,结果见图3。该类器官以单细胞形式传代,在第4天时可见少数较小腔状腺上皮上分化结构,第7天后明显进入对数生长期。The subcultured CWH22 and CLM22 organoids were taken, and the morphological changes of organoid growth were observed and recorded under the Japanese Olympus IMT-2 inverted microscope. The results are shown in Figure 3. The organoids were passaged in the form of single cells, and a few small luminal epithelial epithelial differentiated structures could be seen on the 4th day, and obviously entered the logarithmic growth phase after the 7th day.

2.2CWH22和CLM22类器官结构与原发肿瘤的相似性2.2 Similarity of CWH22 and CLM22 organoid structures to primary tumors

将临床取回的结直肠癌原发灶及肝转移灶组织进行固定包埋制片,后行HE,CK20和CDX2染色;类器官正常培养14天后进行固定并用Thermo的组织包埋胶包埋后制备蜡块切片行HE,CK20和CDX2染色,结果见图4。类器官与对应的肿瘤组织具有一致的上皮样排列分化表现及CK20和CDX2的阳性表达。The retrieved primary colorectal cancer and liver metastases tissues were fixed and embedded and stained with HE, CK20, and CDX2. After 14 days of normal culture, the organoids were fixed and embedded with Thermo tissue embedding gel, and then prepared into wax block sections for HE, CK20, and CDX2 staining. The results are shown in Figure 4. Organoids and corresponding tumor tissues have consistent epithelial-like arrangement differentiation and positive expression of CK20 and CDX2.

2.5CWH22和CLM22类器官细胞系的基因检测2.5 Gene detection of CWH22 and CLM22 organoid cell lines

取正常培养第四代的CWH22和CLM22类器官沉淀,术中切除并速冻的病人肿瘤组织及正常粘膜组织分别提取DNA,质检合格后行全外显子测序,图5A为人类肿瘤驱动基因研究数据库中(http://driverdb.tms.cmu.edu.tw/cancer)结直肠癌高频突变基因(APC,KRAS,SMAD4,CDKN1B,KMT2C等)在该患者肿瘤组织及对应的类器官CWH22、CLM22中的阳性突变情况汇总,图5B为培养的类器官与原发肿瘤突变一致性分析结果,提示类器官较好保留了原发肿瘤的突变特性,且已建立的该患者来源的这对类器官为APC/KRAS/SMAD4三突变,TP53野生型的结肠癌类器官细胞系及对应肝转移灶来源的类器官细胞系。The fourth generation of normal cultured CWH22 and CLM22 organoids were collected, and DNA was extracted from the patient’s tumor tissue and normal mucosal tissue that were resected and quick-frozen during the operation. Whole-exome sequencing was performed after passing the quality inspection. Figure 5A shows the high-frequency mutation genes (APC, KRAS, SMAD4, CDKN1B, KMT2C, etc.) And the positive mutations in the corresponding organoids CWH22 and CLM22 are summarized. Figure 5B shows the results of the mutation consistency analysis between the cultured organoids and the primary tumor, suggesting that the organoids better retain the mutation characteristics of the primary tumor, and the established pair of organoids derived from this patient are APC/KRAS/SMAD4 triple mutations, TP53 wild-type colon cancer organoid cell line and the organoid cell line corresponding to the source of liver metastases.

2.6CWH22和CLM22类器官细胞系的STR检测2.6 STR Detection of CWH22 and CLM22 Organoid Cell Lines

短串联重复序列((short tandem repeat,STR))又称为微卫星DNA,是指染色体上,由数个碱基对作为核心单位,一般2~6个碱基对,串联重复形成的一类DNA序列,重复次数为10~60多次,基因片段在400碱基对以下;每个核心单位重复的次数会出现个体差异,从而形成片段长度不同的等位基因。因此,一组STR序列的重复次数在不同个体中几乎是唯一的,是个体的基因身份特征,也是细胞生物学对细胞身份和来源进行鉴定的主要方法。Short tandem repeat ((short tandem repeat, STR)), also known as microsatellite DNA, refers to a type of DNA sequence formed by tandem repetition of several base pairs as the core unit on the chromosome, generally 2 to 6 base pairs. The number of repetitions is more than 10 to 60, and the gene fragment is less than 400 base pairs. There will be individual differences in the number of repetitions of each core unit, thus forming alleles with different fragment lengths. Therefore, the number of repetitions of a group of STR sequences is almost unique among different individuals, and it is an individual's genetic identity feature, and it is also the main method of cell biology to identify cell identity and origin.

将收集的第4代CWH22和CLM22类器官细胞及术中切除并速冻的病人正常粘膜组织,寄送至中国典型培养物保藏中心CCTCC,进行STR检测。STR位点包括:D19S433、D5S818、D21S11、D18S51、D6S1043、AMEL、D3S1358、D13S317、D7S820、D16S539、CSF1PO、Penta D、D2S441、vWA、D8S1179、TPOX、Penta E、TH01、D12S391、D2S1338、FGA。The collected 4th passage CWH22 and CLM22 organoid cells and normal mucosal tissues of patients excised and frozen during operation were sent to CCTCC, China Center for Type Culture Collection, for STR detection. STR loci include: D19S433, D5S818, D21S11, D18S51, D6S1043, AMEL, D3S1358, D13S317, D7S820, D16S539, CSF1PO, Penta D, D2S441, vWA, D8S1179, TPOX, Penta E, TH01, D12S 391, D2S1338, FGA.

分析结果见表1,可知本发明培养得到的CWH22和CLM22类器官细胞系是真实存在的,与亲本组织具有一致的谱型,为同一病人来源。在德国微生物菌种保藏中心DSMZ数据库(Deutsche Sammlung vonMikroorganismen und Zellkulturen)、美国典型培养物保藏中心(ATCC)数据库,进行STR序列检索,未发现相同STR检测结果。即,未找到与该细胞STR位点完全匹配的细胞系,表明此为新细胞株;未发现多等位点,表明此细胞株为单一细胞,无其他细胞污染。The analysis results are shown in Table 1. It can be seen that the CWH22 and CLM22 organoid cell lines cultivated in the present invention are real, have the same spectrum type as the parental tissue, and are derived from the same patient. In the German Microorganism Culture Collection DSMZ database (Deutsche Sammlung vonMikroorganismen und Zellkulturen) and the American Type Culture Collection (ATCC) database, the STR sequence search was performed, but no identical STR detection results were found. That is, no cell line that completely matches the STR locus of the cell was found, indicating that this is a new cell line; no multi-allelic sites were found, indicating that the cell line was a single cell without other cell contamination.

表1CWH22,CLM22及患者正常粘膜组织STR检测结果列表Table 1 CWH22, CLM22 and the list of STR detection results of normal mucosal tissues of patients

2.7CWH22和CLM22类器官细胞脾脏注射肝脏转移成瘤性检测2.7 Detection of tumorigenicity of liver metastasis by CWH22 and CLM22 organoid cells injected into spleen

类器官表达荧光素酶(过表达荧光素酶病毒(PLVX-Luc-IRES-Neo)购买于上海典君生物科技有限公司):在96孔板中预铺1:2稀释的基质胶*10孔,30μL/孔铺满底部,置于37℃培养箱30min待其成胶;同步消化细胞为单细胞悬液,并用培养基重悬为3*104/mL,100μL/孔种于上述10孔中;48h后,取其中9孔(另一孔为空白对照)换上述含病毒培养液100μL/孔(加polybrene 5μg/mL);作用12小时后,换回常规类器官培养基,继续培养24小时后,进行G418(500μg/mL)筛选5-7天左右,待对照组全部死亡后,继续加G418至传代撤除;取1*105细胞加入20μL活体成像底物中,荧光素酶报告基因发光值>50000即为阳性表达。Organoids expressing luciferase (overexpressed luciferase virus (PLVX-Luc-IRES-Neo) was purchased from Shanghai Dianjun Biotechnology Co., Ltd.): pre-spread 1:2 diluted Matrigel*10 wells in a 96-well plate, cover the bottom with 30 μL/well, and place it in a 37°C incubator for 30 minutes until it gels; synchronously digested cells into a single-cell suspension, and resuspended in medium to 3*104/mL, 100 μL/well was planted in the above 10 wells; after 48 hours, 9 of the wells were taken (the other well was the blank control) and replaced with 100 μL/well of the above-mentioned virus-containing culture medium (with polybrene 5 μg/mL); after 12 hours of action, the conventional organoid medium was replaced, and after 24 hours of continuous culture, G418 (500 μg/mL) was screened for about 5-7 days. 1*105Cells were added to 20 μL in vivo imaging substrate, and the luciferase reporter gene luminescence value > 50,000 was positive expression.

将过表达荧光素酶的CWH22和CLM22类器官常规传代培养,取对数生长期的类器官细胞消化为单细胞悬液后计数,用DMEM重悬为1.25*107/mL,经脾脏注射80μL/只裸鼠,10min后结扎脾脏血管剪除脾脏并缝合。定期活体成像观察裸鼠肝转移成瘤情况并记录。图6A分别为原发灶和转移灶类器官经脾脏注射45天后肝脏转移成瘤情况,图6B为两组细胞肝脏成瘤HE染色及CK20和CDX2的表达情况。The luciferase-overexpressing CWH22 and CLM22 organoids were conventionally subcultured, and the organoid cells in the logarithmic growth phase were digested into a single cell suspension and counted, resuspended in DMEM to 1.25*10 7 /mL, injected 80 μL/nude mouse through the spleen, and 10 minutes later, the splenic blood vessels were ligated, the spleen was cut off, and sutured. Regular in vivo imaging was used to observe and record the liver metastases in nude mice. Figure 6A shows the liver metastases of the primary tumor and metastatic organoids 45 days after spleen injection, and Figure 6B shows the HE staining and the expression of CK20 and CDX2 in the liver tumors of the two groups of cells.

2.8CWH22类器官细胞盲肠注射原位成瘤2.8 In situ tumor formation of CWH22 organoid cells injected into cecum

将CWH22和CLM22类器官常规传代培养,取对数生长期的类器官用TrypLE酶消化5分钟释放类器官结构后,约3*106细胞离心洗涤并预冷后重悬于60μL基质胶中置4℃;以预冷胰岛素注射针抽取类器官悬液20μL做盲肠粘膜下注射。第45天将老鼠安乐死后,取盲肠观察成瘤情况,并行HE染色鉴定。图7为第45天,盲肠原位成瘤肉眼所见及HE染色情况,提示在粘膜下层可见中分化样腺瘤细胞排列。CWH22 and CLM22 organoids were conventionally subcultured, and the organoids in the logarithmic growth phase were digested with TrypLE enzyme for 5 minutes to release the organoid structure. About 3*10 6 cells were centrifuged and washed, pre-cooled, resuspended in 60 μL Matrigel and placed at 4°C; 20 μL of the organoid suspension was drawn with a pre-cooled insulin injection needle for submucosal injection in the cecum. After the mice were euthanized on the 45th day, the cecum was removed to observe the tumor formation, and HE staining was performed for identification. Figure 7 shows the macroscopic findings and HE staining of in situ tumor formation in the cecum on day 45, suggesting that the arrangement of moderately differentiated adenoma cells can be seen in the submucosa.

实施例3Example 3

公开了具备上述生物特征的一对人右半结肠癌原发灶类器官细胞系CWH22和对应的肝转移灶类器官细胞系CLM22在用于判断治疗结直肠癌药物敏感性的应用。Disclosed is the application of a pair of organoid cell lines CWH22 for the primary focus of human right colon cancer and the corresponding organoid cell line CLM22 for liver metastases having the above biological characteristics in judging drug sensitivity for the treatment of colorectal cancer.

3.1相关化疗药物IC50检测3.1 Detection of IC50 of related chemotherapeutic drugs

取对数生长期的CWH22和CLM22类器官消化计数重悬于基质胶并种于96孔板内,2*105个cell/mL,10μL/孔,待基质胶凝固后,添加培养基100μL/孔,培养5-6天时(类器官直径约100μm)替换成梯度药物培养基继续培养,3天换液一次,药物作用6天后通过3D细胞活性检测,计算对应药物IC50。见图8。The CWH22 and CLM22 organoids in the logarithmic growth phase were digested and counted, resuspended in Matrigel and planted in a 96-well plate, 2* 105 cells/mL, 10 μL/well. After the Matrigel solidified, 100 μL/well of medium was added. After 5-6 days of culture (the diameter of the organoid was about 100 μm), the gradient drug medium was replaced to continue the culture. The medium was changed once every 3 days. See Figure 8.

CWH22和CLM22的IC50如下:5-氟尿嘧啶分别为3.3μm和3.78μm;奥沙利铂分别为15.47μm和18.38μm;SN-38分别为7.54nm和10.67nm;VP-16分别为5.84μm和6.98μm;瑞戈非尼分别为3.32μm和3.68μm;AMG-510分别为15.02μm和10.95μm;PS-341分别为8.06nm和7.71nm。The IC50 of CWH22 and CLM22 are as follows: 5-fluorouracil is 3.3 μm and 3.78 μm; oxaliplatin is 15.47 μm and 18.38 μm; SN-38 is 7.54 nm and 10.67 nm; VP-16 is 5.84 μm and 6.98 μm; regorafenib is 3.32 μm and 3.68 μm; 510 are 15.02μm and 10.95μm respectively; PS-341 are 8.06nm and 7.71nm respectively.

与GDSC(https://www.cancerrxgene.org/)中其他结直肠癌细胞系如Lovo、SW620、T84和SW480等的IC50相比,明确了CWH22和CLM22对5-氟尿嘧啶和奥沙利铂的药物敏感性。如图9所示,临床患者术后复查提示肝脏散在病灶,接受8个疗程5-氟尿嘧啶+奥沙利铂+贝伐单抗(mFOLFOX+Bevacizumab)治疗,病灶稳定并明显消退,提示该肿瘤初期对5-氟尿嘧啶和奥沙利铂治疗敏感。Compared with the IC50 of other colorectal cancer cell lines such as Lovo, SW620, T84 and SW480 in GDSC (https://www.cancerrxgene.org/), the drug sensitivity of CWH22 and CLM22 to 5-fluorouracil and oxaliplatin was clarified. As shown in Figure 9, the postoperative re-examination of the clinical patient showed scattered lesions in the liver. After receiving 8 courses of 5-fluorouracil + oxaliplatin + bevacizumab (mFOLFOX + Bevacizumab) treatment, the lesions were stable and regressed significantly, suggesting that the tumor was initially sensitive to 5-fluorouracil and oxaliplatin treatment.

综上所述,本发明筛选得到的人结直肠癌原发灶类器官CWH22和对应的肝脏转移灶类器官CLM22具有较好3D类器官形态,且性状稳定,可持续稳定传代,可良好耐受病毒感染等操作。研究发现其具有非常强的盲肠原位成瘤和脾脏注射肝脏转移成瘤能力,可以成功制备结直肠癌肝转移动物模型。为进一步深入研究结直肠癌及肝转移的病因学、发病机制、耐药机理、新药筛选等提供新的体内外研究支撑。To sum up, the organoid CWH22 of primary human colorectal cancer and the corresponding organoid CLM22 of liver metastases screened by the present invention have better 3D organoid morphology, stable properties, sustainable and stable passage, and good tolerance to virus infection and other operations. Studies have found that it has a very strong ability to form tumors in cecum in situ and liver metastasis by spleen injection, and can successfully create an animal model of colorectal cancer liver metastasis. To provide new in vivo and in vitro research support for further in-depth research on the etiology, pathogenesis, drug resistance mechanism, and new drug screening of colorectal cancer and liver metastasis.

以上所述仅为本发明较佳的具体实施方式,但本发明的保护范围并不局限于此,任何熟悉本技术领域的技术人员在本发明揭露的技术范围内,可轻易想到的变化或替换,都应涵盖在本发明的保护范围之内。The above description is only a preferred embodiment of the present invention, but the scope of protection of the present invention is not limited thereto. Any changes or substitutions that can be easily conceived by those skilled in the art within the technical scope disclosed in the present invention shall be covered within the scope of protection of the present invention.

Claims (10)

1.一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系,其特征在于,包括保藏于中国典型培养物保藏中心CCTCC,保藏号为CCTCC NO.C202218的人右半结肠癌原发灶类器官细胞系CWH22,与对应的保藏号为CCTCC NO.C202219的配对人右半结肠癌肝转移灶类器官细胞系CLM22。1. A paired organoid cell line of primary human colorectal cancer and liver metastases, characterized in that it includes the organoid cell line CWH22 of the primary human right colon cancer with the preservation number CCTCC No. 2.根据权利要求1所述人结直肠癌原发灶与肝脏转移灶配对类器官细胞系,其特征在于,所述细胞系为原发灶和对应肝脏转移灶肿瘤组织进行体外3D类器官培养建立得到。2. The organoid cell line paired with the primary human colorectal cancer and liver metastases according to claim 1, wherein the cell line is obtained by in vitro 3D organoid culture of the primary tumor and the corresponding liver metastases. 3.一种人结直肠癌原发灶与肝脏转移灶配对类器官细胞系的培养方法,其特征在于,它包括如下步骤:3. A method for cultivating a paired organoid cell line of a human colorectal cancer primary focus and liver metastases, characterized in that it comprises the following steps: 1)取材:术中分别留取原发灶及对应肝脏转移灶肿瘤组织各0.5cm3于含AdvancedDMEM/F12的无菌取样管中,置于4℃冰盒快速送到实验室;1) Material collection: 0.5 cm3 of the tumor tissue from the primary tumor and the corresponding liver metastases were collected during the operation, placed in a sterile sampling tube containing AdvancedDMEM/F12, placed in a 4°C ice box, and sent to the laboratory quickly; 2)消化:在无菌超净台中用PBS清洗弃去步骤1)所得样本表面的黏液、坏死组织和残余血细胞,继而用活力碘冲洗一次及PBS冲洗三次后,在平皿中用剪刀将组织反复剪碎,加入2mL 50%浓度消化酶TrypLE室温消化,每隔5min充分混匀含消化酶的组织悬液,约20min左右,加入等体积含Ca2+的HBSS缓冲液中和并再次混匀;2) Digestion: Wash and discard the mucus, necrotic tissue and residual blood cells on the surface of the sample obtained in step 1) in a sterile ultra-clean bench, then rinse once with iodine and PBS three times, cut the tissue repeatedly with scissors in a plate, add 2 mL of 50% digestive enzyme TrypLE to digest at room temperature, and mix the tissue suspension containing digestive enzyme every 5 minutes. About 20 minutes, add an equal volume of HBSS buffer containing Ca 2+ to neutralize and mix again ; 3)过滤:用不小于200目网径的滤网过滤步骤2)所得悬液,并离心,沉淀用5mL培养基重悬,并反复洗涤离心弃去坏死细胞碎片,离心转速500~1500r/min;3) Filtration: filter the suspension obtained in step 2) with a filter of not less than 200 mesh diameter, and centrifuge, resuspend the precipitate with 5 mL of culture medium, wash and centrifuge repeatedly to discard necrotic cell fragments, and centrifuge at a speed of 500-1500 r/min; 4)种细胞:取上述洗涤所得细胞进行活细胞计数,用基质胶低温重悬细胞为8000个/30μL,种于已预热的24孔板中,30μL/孔,置于37℃培养箱30min待穹隆状基质胶凝固后,每孔添加培养基650μL;4) Seed cells: Take the above-mentioned washed cells for live cell counting, resuspend the cells with Matrigel at low temperature to 8000 cells/30 μL, seed in a preheated 24-well plate, 30 μL/well, place in a 37°C incubator for 30 minutes, and add 650 μL of medium to each well after the vaulted Matrigel solidifies; 5)常规传代培养:培养基每3~5天更换一次,在第10~14天时进行传代操作,用TrypLE消化酶250μL/孔进行消化,每隔5分钟吹打混匀一次,10~15min可消化为单个细胞悬液,以HBSS中和离心重悬后计数,以8000个细胞每孔继续传代,后续重复步骤4)和5)。5) Conventional subculture: the medium was changed every 3 to 5 days, and the subculture operation was performed on the 10th to 14th day. Digest with TrypLE digestive enzyme 250 μL/well, pipette and mix every 5 minutes, digest into a single cell suspension in 10 to 15 minutes, resuspend with HBSS neutralization and centrifugation, and count, continue to pass at 8000 cells per well, and then repeat steps 4) and 5). 4.一种权利要求1或2所述人结直肠癌原发灶与肝脏转移灶配对类器官细胞系或权利要求3所述方法培养得到的所述细胞系用于构建盲肠原位成瘤动物模型和/或肝脏转移灶成瘤动物模型并用来研究结直肠癌发生机理及耐药机理。4. A paired organoid cell line of human colorectal cancer primary focus and liver metastases according to claim 1 or 2 or the cell line obtained by cultivating the method according to claim 3 is used to construct an animal model of cecum in situ tumor formation and/or an animal model of liver metastases tumor formation and to study the mechanism of colorectal cancer occurrence and drug resistance. 5.一种肝脏转移灶成瘤动物模型的建立方法,其特征在于,它包括如下步骤:5. A method for establishing a tumor-forming animal model of liver metastases, characterized in that it comprises the steps of: 1)类器官培养:将类器官细胞使用慢病毒感染稳定过表达荧光素酶后,常规培养类器官,于传代后第4、7、9、11天分别换液,取处于对数生长期的CWH22和CLM22类器官进行裸鼠脾脏注射转移成瘤操作;1) Organoid culture: After the organoid cells were stably overexpressed luciferase with lentivirus infection, the organoids were cultured routinely, and the medium was changed on the 4th, 7th, 9th, and 11th day after subculture, and the CWH22 and CLM22 organoids in the logarithmic growth phase were taken for nude mouse spleen injection and tumor transfer operation; 2)类器官消化:消化细胞过程与常规传代操作保持相同,消化为单细胞悬液状态,并计数,以DMEM重悬为1.25*107/mL置4℃保温箱备用;2) Digestion of organoids: the process of digesting cells is the same as that of conventional subculture operations, digested into a single cell suspension state, and counted, resuspended in DMEM to 1.25*10 7 /mL and placed in a 4°C incubator for standby; 3)脾脏注射:戊巴比妥麻醉裸鼠后,固定裸鼠并做腹部皮肤消毒,沿腹部左肋下缘腋前线前0.5cm处做纵向约1cm切口至腹腔,挑出脾脏后从脾脏远端注射混匀的细胞悬液约80μL/只裸鼠,10min后结扎脾脏血管剪除脾脏并缝合,密切观察确保顺利苏醒。3) Spleen injection: After anesthetizing the nude mice with pentobarbital, fix the nude mice and perform abdominal skin disinfection, make a longitudinal incision about 1 cm along the left lower costal margin of the abdomen at the front of the axillary line 0.5 cm to the abdominal cavity, pick out the spleen and inject about 80 μL/nude mouse with a mixed cell suspension from the distal end of the spleen, ligate the splenic blood vessels 10 minutes later, cut off the spleen and suture it, and observe closely to ensure smooth recovery. 4)成瘤监测:定期活体成像观察裸鼠肝转移成瘤情况并记录。4) Tumor formation monitoring: regular in vivo imaging was used to observe and record the tumor formation of liver metastases in nude mice. 6.一种盲肠原位成瘤动物模型的构建方法,其特征在于,它包括如下步骤:6. A method for constructing an animal model of cecum in situ tumor formation, characterized in that it comprises the steps of: 1)类器官培养:常规培养过表达荧光素酶的类器官CWH22和CLM22,于传代后第4、7、9、11天分别换液,取处于对数生长期的类器官进行裸鼠盲肠壁注射操作;1) Organoid culture: routinely culture luciferase-overexpressing organoids CWH22 and CLM22, and change the medium on the 4th, 7th, 9th, and 11th days after subculture, and inject the organoids in the logarithmic growth phase into the cecum wall of nude mice; 2)类器官收集:于第10~12天收集类器官,以TrypLE消化酶常规消化5min后,中和收集类器官沉淀,约3*106细胞,预冷后重悬于60μL基质胶中置4℃保温箱备用;2) Organoid collection: collect organoids on the 10th to 12th day, digest with TrypLE digestive enzyme for 5 minutes, neutralize and collect organoid precipitates, about 3*10 6 cells, resuspend in 60 μL Matrigel after pre-cooling, and place in a 4°C incubator for later use; 3)盲肠原位注射:戊巴比妥麻醉裸鼠后,固定裸鼠并做腹部皮肤消毒,沿下腹部正中线做纵向约1.5cm切口至腹腔并找到盲肠,将盲肠末端移至视野正中,以预冷胰岛素注射针抽取类器官悬液20μL做盲肠粘膜下注射,注射完毕观察无渗漏后,做腹部肌肉和皮肤缝合,并密切观察确保顺利苏醒;3) Caecum in situ injection: after pentobarbital anesthetized the nude mice, fix the nude mice and perform abdominal skin disinfection, make a longitudinal incision about 1.5 cm along the midline of the lower abdomen to the abdominal cavity and find the cecum, move the end of the cecum to the center of the field of vision, draw 20 μL of the organoid suspension with a pre-cooled insulin injection needle for submucosal injection of the cecum, and observe that there is no leakage after the injection, suture the abdominal muscles and skin, and closely observe to ensure smooth recovery; 4)成瘤监测:定期活体成像观察裸鼠盲肠原位成瘤情况并记录。4) Tumor formation monitoring: Regular in vivo imaging was used to observe and record the in situ tumor formation in the cecum of nude mice. 7.一种权利要求1或2所述人结直肠癌原发灶与肝脏转移灶配对类器官细胞系或权利要求3所述方法培养得到的所述细胞系在制备用于筛选治疗结直肠癌和/或肝转移灶肿瘤药物中的应用。7. The use of a paired organoid cell line of human colorectal cancer primary focus and liver metastases according to claim 1 or 2 or the cell line cultured by the method according to claim 3 in the preparation of tumor drugs for screening colorectal cancer and/or liver metastases. 8.根据权利要求7所述的应用,其特征在于,将治疗药物施用于体外细胞模型中,施用后抑制细胞增殖或导致细胞凋亡的为治疗敏感性药物,或者将测试化合物施用于体外细胞模型中,施用后抑制细胞增殖或导致细胞凋亡的为候选化合物。8. The application according to claim 7, characterized in that, the therapeutic drug is applied to an in vitro cell model, and the drug that inhibits cell proliferation or causes cell apoptosis is a therapeutic sensitive drug after application, or the test compound is applied to an in vitro cell model, and the candidate compound is a candidate compound that inhibits cell proliferation or causes cell apoptosis after application. 9.一种筛选用于治疗结直肠癌和/或肝转移灶肿瘤药物的方法,其特征在于,它包括取权利要求3培养的对数生长期的CWH22和CLM22类器官消化计数重悬于基质胶并种于96孔板内,2*105个cell/mL,10μL/孔,待基质胶凝固后,添加培养基100μL/孔,培养5~6天时替换成梯度药物培养基继续培养,3天换液一次,药物作用6天后通过3D细胞活性检测,计算对应药物IC50。9. A method for screening tumor drugs for the treatment of colorectal cancer and/or liver metastases, characterized in that it comprises taking the CWH22 and CLM22 organoids in the logarithmic growth phase cultivated in claim 3, digesting and counting them, resuspending them in matrigel and planting them in a 96-well plate, 2* 105 cells/mL, 10 μL/well, after the matrigel is solidified, adding 100 μL/well of the medium, and replacing it with a gradient drug medium to continue culturing for 5 to 6 days, and changing the medium once every 3 days , after 6 days of drug action, through 3D cell activity detection, calculate the corresponding drug IC50. 10.根据权利要求9所述的方法,其特征在于,所述梯度药物培养基包含以下药物中的至少一种:5-氟尿嘧啶,奥沙利铂,伊立替康,依托泊苷,索托拉西布,瑞戈非尼,达沙替尼或硼替佐米。10. The method according to claim 9, wherein the drug gradient medium comprises at least one of the following drugs: 5-fluorouracil, oxaliplatin, irinotecan, etoposide, sotoracib, regorafenib, dasatinib or bortezomib.
CN202211462629.9A 2022-11-21 2022-11-21 Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof Pending CN116463291A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211462629.9A CN116463291A (en) 2022-11-21 2022-11-21 Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211462629.9A CN116463291A (en) 2022-11-21 2022-11-21 Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof

Publications (1)

Publication Number Publication Date
CN116463291A true CN116463291A (en) 2023-07-21

Family

ID=87179462

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211462629.9A Pending CN116463291A (en) 2022-11-21 2022-11-21 Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof

Country Status (1)

Country Link
CN (1) CN116463291A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116751682A (en) * 2023-08-17 2023-09-15 内蒙古医科大学附属医院(内蒙古自治区心血管研究所) Colorectal cancer tissue culture device, colorectal cancer tissue culture method, colorectal cancer tissue culture monitoring method and colorectal cancer tissue culture model
CN118571500A (en) * 2024-07-31 2024-08-30 南方医科大学南方医院 Colorectal cancer chemotherapy response prediction system and storage medium

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116751682A (en) * 2023-08-17 2023-09-15 内蒙古医科大学附属医院(内蒙古自治区心血管研究所) Colorectal cancer tissue culture device, colorectal cancer tissue culture method, colorectal cancer tissue culture monitoring method and colorectal cancer tissue culture model
CN116751682B (en) * 2023-08-17 2023-11-14 内蒙古医科大学附属医院(内蒙古自治区心血管研究所) Colorectal cancer tissue culture device, colorectal cancer tissue culture method, colorectal cancer tissue culture monitoring method and colorectal cancer tissue culture model
CN118571500A (en) * 2024-07-31 2024-08-30 南方医科大学南方医院 Colorectal cancer chemotherapy response prediction system and storage medium
CN118571500B (en) * 2024-07-31 2024-09-27 南方医科大学南方医院 Colorectal cancer chemotherapy response prediction system and storage medium

Similar Documents

Publication Publication Date Title
CN116463291A (en) Human colorectal cancer primary focus and liver transfer focus pairing organoid cell line and culture method and application thereof
CN113234679B (en) Crizotinib-resistant human lung adenocarcinoma cell strain and preparation and application thereof
JP2011225613A (en) Method of inhibiting migration of tumor cell and medicament used therefor
US20180142214A1 (en) In vitro method for screening testing compound to evaluate its potential as liver drug
CN105296430B (en) A kind of human colon cancer cells system DXH-1 and its application
CN110106150B (en) A kind of preparation method and application of synovial sarcoma cell line hSS-005R
CN105255832B (en) Human bile duct cancer cell line and application thereof
CN114214281B (en) Human glioblastoma cell line GWH04 and culture method and application thereof
US20220125877A1 (en) Method for treating colorectal cancer
CN107541494B (en) Human bile duct cancer cell line and application thereof
WO2021004372A1 (en) Application of metformin in treatment of kras mutant colorectal cancer
WO2021004373A1 (en) Use of mate1 gene in treatment of colorectal cancer
CN110499290B (en) A human Ewing sarcoma cell line
CN107541496A (en) A kind of human esophageal carcinoma cell line and its application
CN115747139A (en) Multi-mutation human primary lung cancer cell strain and application thereof
CN105969734A (en) Esophageal cancer cell line and application thereof
CN107460171A (en) Human thyroid undifferentiated cancer cell system and its application
CN106047815A (en) Esophageal cancer cell line and application thereof
US20180143181A1 (en) In vitro method for screening testing compound to evaluate its potential as liver drug
CN106011067A (en) Esophageal cancer cell line and application thereof
CN112029866B (en) Application of WWP1 in pancreatic cancer
CN106011069A (en) A kind of esophageal cancer cell line and application thereof
CN118879636B (en) Cell strain AlmoR1 and application thereof
CN115322992A (en) Group of mutant genes related to gallbladder cancer and application thereof
CN112266960A (en) Application of a kind of Msi1 in the preparation of preparations for the treatment of gastric cancer and the prediction of chemotherapeutic drug tolerance

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination