CN115927164A - Culture method and application of vascularized tumor organoid - Google Patents

Culture method and application of vascularized tumor organoid Download PDF

Info

Publication number
CN115927164A
CN115927164A CN202310075071.7A CN202310075071A CN115927164A CN 115927164 A CN115927164 A CN 115927164A CN 202310075071 A CN202310075071 A CN 202310075071A CN 115927164 A CN115927164 A CN 115927164A
Authority
CN
China
Prior art keywords
tumor
culture
cells
endothelial cells
cell
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.)
Granted
Application number
CN202310075071.7A
Other languages
Chinese (zh)
Other versions
CN115927164B (en
Inventor
刘虹余
何春花
李胜
陆政昊
王嘉
杨云旭
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.)
Chengdu Nuoyeide Medical Laboratory Co ltd
Shenzhen Jingke Biotechnology Co ltd
Original Assignee
Guagnzhou Jingke Biotech Co ltd
Chengdu Nuoyeide Medical Laboratory Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Guagnzhou Jingke Biotech Co ltd, Chengdu Nuoyeide Medical Laboratory Co ltd filed Critical Guagnzhou Jingke Biotech Co ltd
Priority to CN202311143229.6A priority Critical patent/CN117025715A/en
Publication of CN115927164A publication Critical patent/CN115927164A/en
Application granted granted Critical
Publication of CN115927164B publication Critical patent/CN115927164B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • 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
    • C12N5/069Vascular Endothelial cells
    • C12N5/0691Vascular smooth muscle cells; 3D culture thereof, e.g. models of blood vessels
    • 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
    • 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
    • G01N33/5044Chemical 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 involving specific cell types
    • 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
    • G01N33/5044Chemical 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 involving specific cell types
    • G01N33/5064Endothelial 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • 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
    • C12N2513/003D culture
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

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

Abstract

The invention belongs to the technical field of cell culture, and particularly relates to a culture method and application of a vascularized tumor organoid. The method comprises the steps of taking biopsy tissues of a patient to prepare mixed cell masses, then respectively culturing tumor cells and endothelial cells by using the mixed cell masses, finally carrying out 3D co-culture on the tumor cells and the endothelial cells, and inducing the formation of a blood vessel network in the co-cultured tumor organoids in vitro.

Description

Culture method and application of vascularized tumor organoid
Chinese patent application No. CN2022112413917, entitled "a method of culturing and use of vascularized tumor organoids", filed on 10/11/2022, is hereby incorporated by reference in its entirety.
Technical Field
The invention belongs to the technical field of cell culture, and particularly relates to a culture method and application of vascularized tumor organoids.
Background
Research shows that during the process of occurrence and development of the tumor, a large number of new blood vessels are formed in the tumor, the new blood vessels provide nutrition and water for the growth of the tumor, and simultaneously, conditions are created for spreading tumor cells to a distance and forming new metastases at different parts in a body. Taking breast cancer as an example, the high proliferation and metastasis characteristics of breast cancer depend on continuously formed new blood vessels, so that the inhibition of angiogenesis is a key research content for overcoming the invasiveness of the microenvironment of breast cancer tumors. The existing research is cognizant on the concept of establishing a tumor-angiogenesis model, but most of the current research is to take umbilical vein endothelial cells (HUVEC), mesenchymal cells (MSC) and the like as the source of vascular endothelium, add breast cancer cell lines and transplant tumor models in vitro of animals so as to achieve the aim of generating blood vessels. The model established by the method depends on animals, namely the model is transplanted on the animals, so that the modeling period is long and the cost is high. Secondly, as HUVEC and MSC cell lines are cultured and passaged for a long time, the molecular and phenotypic characteristics of the HUVEC and MSC cell lines are changed, so that when the HUVEC and MSC cell lines are co-cultured with a breast cancer cell line, the HUVEC and MSC cell lines often face a microenvironment which cannot completely simulate the interaction between the tumor and the blood vessel in vivo due to the lack of the characteristics of the blood vessel in the tumor microenvironment, and the HUVEC and MSC cell lines also do not have tumor heterogeneity among different patients. Therefore, "tumor-angiogenesis" animal models constructed based on HUVECs and MSCs often do not translate fully into the clinic effectively.
A recently developed tumor-derived tumor organoid (PDO) model has characteristics of maintaining tumor heterogeneity and a high modeling success rate, and thus is widely used for preclinical tumor research and clinical drug screening. Current research suggests that tumor organoids have the potential to be used as an in vitro detection platform for predicting the response of personalized immunotherapy. However, due to the lack of important physiological processes in vivo and the lack of microenvironments such as immune cells and mesenchymal cells, organoids constructed in vitro cannot fully replicate all cell types and maturity of the real organ, and there is no distribution of blood vessels, lymphatic vessels and nervous system, which leads to difficulties in evaluating the true therapeutic response and pharmacological mechanism of immunomodulators in organoid models.
Although there are some prior art proposals for inducing organoid angiogenesis by co-culturing endothelial cells and organoids, these proposals are complicated and require long culture times, and do not solve the problem of maintaining heterogeneous inheritance of the original tumor tissue of different patients. For example, patent application No. CN2018800224776 entitled "stable three-dimensional vascularization and methods for forming same" discloses a technical solution for vascularizing organoids or decellularized organs by culturing organoids or decellularized organs with endothelial cells comprising an exogenous nucleic acid encoding an ETV2 transcription factor under conditions such that the exogenous ETV2 protein is expressed in the endothelial cells. This protocol first requires at least 3-4 weeks to construct an endothelial cell that encodes an exogenous nucleic acid for the ETV2 transcription factor (i.e., to construct a tailored endothelial cell), and the source of endothelial cells in the method includes the HUVEC cell line (lacking the vascular properties of the tumor microenvironment). After the construction of the endothelial cells is completed, the endothelial cells are further co-cultured with organoids, and under the culture condition, the endothelial cells need to express ETV2 protein to induce the formation of lumen-like and stable three-dimensional blood vessels. Therefore, the technical scheme for constructing the stable three-dimensional blood vessel formation provided by the patent method is complex in operation and long in time consumption. More importantly, the patented method is to surgically implant an in vitro constructed vascularized organoid into a patient, rather than constructing a platform in vitro for preclinical drug screening.
In view of the above, there is a need to develop a method and model that can approach the tumor angiogenesis characteristics of different patients and provide effective support for preclinical screening of anti-angiogenesis drugs in order to alleviate at least one of the problems in the prior art.
Disclosure of Invention
In view of the above, the present invention provides a culture method for inducing tumor organoid vascularization and the application thereof, and the specific technical scheme is as follows.
A culture method for inducing angiogenesis in a tumor organoid, comprising the steps of:
the method comprises the following steps: taking biopsy tissue of a patient, cleaning, cutting, then performing tissue digestion, and stopping the tissue digestion after 0.5-2h to obtain a mixed cell mass (the cell mass comprises tumor cells, a small amount of endothelial cells, immune cells, stromal cells and the like, so that the cell mass is called as a mixed cell mass);
step two: preparing the mixed cell mass obtained in the first step into a cell suspension, adding matrigel for heavy suspension and precipitation, inoculating the cell suspension into a pore plate, adding a tumor cell culture medium for amplification and culture of tumor cells, and carrying out passage after the tumor cells are fused at least 80% in density (observing under a common optical microscope, calculating the total number of tumor organoids and the total number of cells fused with each other under the same visual field to obtain a fusion ratio, and counting a plurality of visual fields to obtain an average value of the cell density fusion ratio);
step three: preparing the mixed cell mass obtained in the first step into cell suspension, adding an endothelial cell culture medium to resuspend and precipitate, inoculating the cell suspension into a culture plate coated with gelatin, culturing to obtain endothelial cells, and carrying out passage after the density of the endothelial cells is fused by at least 80% (calculating the cell density fusion percentage in the second step);
step four: further amplifying and culturing the endothelial cells obtained in the third step, and magnetically screening the endothelial cells by using immunomagnetic beads or purifying the endothelial cells by using a flow cytometer until the purity of the endothelial cells reaches more than 90 percent (the purity of the endothelial cells can influence the accuracy of an experimental result, because a mixed cell mass obtained by digesting the biopsy tissue of a patient contains other cells including tumor cells, immunocytes, stromal cells and the like, the magnetic bead sorting or the flow cytometer sorting is adopted to achieve the aim of purification so as to improve the accuracy of the experiment);
step five: mixing and re-suspending the tumor cells obtained in the second step and the endothelial cells obtained in the fourth step in matrigel, wherein the proportion of the re-suspended mixed cells in the matrigel is 4000-10000/25 mu L, and further inoculating the mixture into a co-culture medium for 3D co-culture, wherein the co-culture medium consists of FBS, penicillin/streptomycin, L-Glutamate, DMEM/F12 basal medium and angiogenesis promoting components, and the angiogenesis promoting components comprise VEGF and vitamin C (in the co-culture process, two processes are included, namely forming a tumor organoid and forming a vascular network between the interior of the tumor organoid and the organoid after the tumor organoid is induced by the endothelial cells);
step six: and (3) performing immunofluorescence staining identification on the blood vessels generated in the co-culture system.
It can be understood that the components of the tumor cell culture medium used in the method are different according to different tumor tissue sources, and the components of the culture medium are configured according to the existing literature data, so as to realize the purpose of culturing tumor cells into tumor organoids.
The "co-culture" in the method of the present invention is a 3D co-culture of tumor cells and endothelial cells. If the two cells are mixed and then subjected to conventional 2D co-culture, the tumor cells and the endothelial cells are likely to grow close to each other and adhere to each other, and thus the blood vessels cannot be effectively differentiated. However, after the 3D structure is formed, tumor cells form tumor organoids, endothelial cells induce angiogenesis of the tumor organoids, and a vascular network may be formed in the middle of the tumor organoid mass or between the mass and the mass, so that a partial microenvironment of the tumor organoids is simulated, a model closer to the actual behavior mode of the tumor is constructed on an in vitro platform, and higher accuracy is provided for subsequent screening of preclinical drugs.
Further, in the fifth step, the mixing ratio of the tumor cells to the endothelial cells is 1:1 to 1:2.
preferably, the mixing ratio of the tumor cells and the endothelial cells comprises 1.
Further, in the first step, the biopsy tissue of the patient is washed by a washing solution, and the washing solution comprises 1% of cyan/streptomycin and 99% of DPBS buffer solution in volume ratio.
Further, in the first step, tissue digestion is performed by using tissue digestion solution, wherein the tissue digestion solution comprises 2% of penicillin/streptomycin, 97% of DPBS buffer solution, 1% of BSA (bovine serum albumin) in mass fraction and 1mg/ml of collagenase type I.
Further, in the first step, the tissue digestion is stopped by using a stop solution, wherein the stop solution comprises 10% FBS and 90% DMEM/F12 medium.
Further, the co-culture medium in the fifth step is composed of 10% by volume of FBS, 1% by volume of penicillin/streptomycin, 0.2mM L-Glutamate, 89% by volume of DMEM/F12 basal medium, and 100ng/ml of VEGF and 60. Mu.g/. Mu.l of vitamin C.
Preferably, L-Glutamate is replaced by Glutamax, since it is unstable in the medium.
The co-culture medium provided by the method not only needs to provide nutrients and cytokines required for maintaining the growth of tumor cells and endothelial cells, but also provides components capable of inducing angiogenesis of tumor organoids.
Further, the co-culturing the mixed cells in the fifth step is carried out at 5% CO 2 、21%O 2 Normoxic conditions of (2) and 5% CO 2 、2% O 2 Under hypoxic conditions.
The vascularized tumor organoid model obtained by the culture method is characterized in that the vascularized tumor organoid model is a co-culture model of endothelial cells from the same patient for inducing the tumor organoids to generate blood vessels.
The vascularized tumor organoid model obtained by the method of the invention forms an interactive network of tumor and blood vessel related cells, rather than completely reducing the blood vessel tissues capable of transporting nutrient substances and oxygen in vivo.
Furthermore, the tumor cell sources of the tumor organoids comprise breast cancer, lung cancer, liver cancer, cholangiocarcinoma, gastric cancer, intestinal cancer, bladder cancer, kidney cancer, pancreatic cancer or esophageal cancer.
Furthermore, the vascularized tumor organoid model is applied to screening of anti-angiogenesis drugs before clinic.
Further, the vascularized tumor organoid model is applied to the preparation of an in vitro anti-tumor angiogenesis drug screening platform.
Preferably, the anti-angiogenic drug includes, but is not limited to, an Hsp90 inhibitor and/or a multiple kinase inhibitor of Raf-1 and B-Raf.
Advantageous technical effects
The existing research finds that the sensitivity of endothelial cells to drugs is really reflected, and the research can not be realized by only using a single cell line, so that the accuracy and precision of a plurality of existing clinical prodrug screening models are not enough. In order to simulate a microenvironment close to the interaction between a tumor and a blood vessel in a body and eliminate heterogeneity among individuals, and further provide a detection platform with higher in-vitro drug screening precision suitable for individualized precise medical treatment, the method provides a vascularized tumor organoid model of an individualized tumor organoid-endothelial cell. In the vascularized tumor organoid model provided by the present invention, the original tumor cells and endothelial cells are derived from the same patient. Since endothelial cells within tumors differ from those in normal tissues, tumor endothelial cells also exhibit different characteristics depending on the degree of malignancy of tumors, and thus have heterogeneity between individuals and within individuals. After long-term exploration, the tumor cells and endothelial cells from the same patient are pertinently used for 3D co-culture, and the constructed individualized tumor organoid-endothelial cell model can truly restore the blood vessel network and microenvironment in the tumor tissue in vitro in a short time (10 days) after induction, can better restore the heterogeneity between individuals and in the individuals, further remarkably improve the accuracy and precision of a clinical prodrug screening and detecting platform, and has great potential in promoting the development of individualized precise medical treatment.
In the research and development process of the invention, the research and development of the invention finds that co-culture of tumor cells and endothelial cells from the same patient has certain technical difficulty, including that the culture difficulty of primary tumor cells is high, and the sorting, purification and culture of endothelial cells from tumor sources have certain difficulty. For example, the purity of endothelial cells can affect the vascularization network that is ultimately formed; for another example, the characteristic of adherent growth of endothelial cells, which requires 3D culture of tumor cells into tumor organoids, requires 2D culture of tumor cells, and co-culturing of tumor cells and endothelial cells requires the simultaneous satisfaction of the different growth characteristics of both cells. After a large number of groping and experiments, the team of the invention realizes the culture of tumor organs with vascular network differentiation in a short time (10 days) by respectively performing 3D culture on the tumor organoids and 2D culture on endothelial cells, further sorting and purifying the endothelial cells, finally performing 3D co-culture on the two cells, optimally setting a specific tumor cell-endothelial cell ratio, a co-culture medium with specific components, specific culture conditions and the like, and can better reduce the heterogeneity among individuals and in the individuals.
Generally, the culture method provided by the invention is convenient to operate, short in modeling time and high in modeling success rate (the tumor organoid is differentiated into the vascular network as the standard for successful modeling), and the genetic engineering technology and the microfluidic platform are not required to be additionally used, so that the problems that other technical platforms need expensive instruments and complicated operation steps and the technical threshold is high are solved. Can be better and more widely applied to basic scientific research.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below. It is obvious that the drawings in the following description are some embodiments of the invention, and that for a person skilled in the art, other drawings can be derived from them without inventive exercise.
FIG. 1 is a schematic diagram of the general technical route of the cultivation method of the present invention;
FIG. 2 is a fluorescence staining graph of vascularization of different tumor organoids (from left to right, breast, bladder and liver cancer, in order; endothelial cells stained with CD31 antibody, shown as green fluorescence; tumor cells stained with Pan-CK antibody, shown as red fluorescence; DAPI staining of the nucleus, shown as blue fluorescence, indicating that all cells, non-cellular components are free of blue fluorescence);
FIG. 3 is a bright field diagram (scale bar: 100 μm) of cells from the same patient with a passaging density of tumor cells and endothelial cells > 80%;
FIG. 4 is a cell bright field diagram (from left to right, cell number < 4000, 4000-10000, 10000; scale bar: 100 μm);
FIG. 5 is a graph showing the effect of 3D co-culture using co-culture media without and with the addition of pro-angiogenic components, respectively (scale bar: 50 μm);
FIG. 6 is a graph of a tumor organoid and endothelial cells after 3D matrigel co-culture, counting plating, and 17-AAG treatment experiment;
FIG. 7 is a graph of experiments with counting plating and Sorafenib treatment after 3D matrigel co-culture of tumor organoids and endothelial cells;
FIG. 8 is a graph showing the difference in the endothelial network formation after the action of 17-AAG at high, medium and low drug concentrations (scale bar: 50 μm);
FIG. 9 is a graph showing the difference in the endothelial network formation after the action of high, medium and low drug concentrations of Sorafenib (scale bar: 50 μm).
Detailed Description
In order to make the objects, technical solutions and advantages of the embodiments of the present invention clearer, the technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention. It is to be understood that the embodiments described are only a few embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
It should be noted that, in this document, the terms "comprises," "comprising," or any other variation thereof, are intended to cover a non-exclusive inclusion, such that a process, method, article, or apparatus that comprises a list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, article, or apparatus. Without further limitation, an element defined by the phrases "comprising a component of' 8230; \8230;" does not exclude the presence of another like element in a process, method, article, or apparatus that comprises the element.
As used in this specification, the term "about" typically means +/-5% of the stated value, more typically +/-4% of the stated value, more typically +/-3% of the stated value, more typically +/-2% of the stated value, even more typically +/-1% of the stated value, and even more typically +/-0.5% of the stated value.
In this specification, certain embodiments may be disclosed in a range of formats. It should be understood that this description of "within a certain range" is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, the range
Figure SMS_1
The description should be read as having specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6, and the like, and ranges within this rangeIndividual numbers, such as 1,2,3,4,5 and 6. The above rules apply regardless of the breadth of the range.
The vascularized tumor organoid is to induce tumor organoid to generate blood vessel on the organoid platform constructed in vitro.
The "organoid" of the present invention refers to an in vitro model with tissue morphology formed by self-assembly of cells, and has a certain function because of inducing generation of partial vascular network.
The accuracy of the invention refers to that the vascularized tumor organoids cultured by the method of the invention have a certain vascular network differentiation and are closer to the real tumor internal environment, so that the vascularized tumor organoids can have higher accuracy in response to drug sensitivity and efficacy when being used for screening of preclinical drugs compared with a common organoid model.
The term "accuracy" as used herein means that the vascularized tumor organoids cultured by the method of the present invention have inter-and intra-individual heterogeneity, and therefore can be used in preclinical drug screening with accuracy that reflects drug sensitivity and efficacy in different patients, as compared to a universal drug screening model.
The Matrigel is Matrigel.
DMEM/F12, fetal Bovine Serum (FBS), trypLE, collagenase type I, DPBS, L-Glutamate, penicillin/streptomycin, ECGM-MV, VEGF, gelatin, matrigel Matrigel, human CD31 immunomagnetic beads, miltenyi Biotec and the cleaning solution, the tissue digestion solution and the stop solution in the culture method are products developed by Doudou medical science.
Example 1
1. Preparation of test materials
DMEM/F12, fetal Bovine Serum (FBS), trypLE, collagenase type I, DPBS, HPBS, BSA, L-Glutamate, penicillin/streptomycin, trypsin-EDTA from ThermoFisher.
Endothelial cell culture medium ECGM-MV was purchased from Promocell and comprised final concentrations of 0.05ml/ml FBS,0.004ml/ml ECGS,10ng/ml EGF, 90. Mu.g/ml heparin and 1. Mu.g/ml hydrocortisone.
Tumor cell culture media are described in various documents and are suitable for use with tumor cell culture media of a particular tumor type.
Matrigel was purchased from corning.
VEGF was purchased from PreproTech.
Gelatin was purchased from the next saint biotechnology.
Human CD31 immunomagnetic beads were purchased from Miltenyi Biotec.
2. Preparation of media and other reagents
1) Cleaning solution: comprises 1% by volume of penicillin/streptomycin and 99% by volume of DPBS buffer.
2) Tissue digestive juice: comprises 2% by volume of penicillin/streptomycin, 97% by volume of DPBS buffer, 1% by mass of BSA, and 1mg/ml of collagenase type I.
3) Stopping liquid: comprising 10% FBS and 90% DMEM/F12 medium.
4) Co-culture medium:
Figure SMS_2
Figure SMS_3
example 2
Example of a method for inducing angiogenesis in tumor organoids
1) Sample source: biopsy tissue sources were ethically required and patients informed and consented to fresh samples obtained on the day of patient surgery, and stored temporarily in storage solutions (4 ℃, <24 h). The tumor sample sources in this example include breast cancer tissue, bladder cancer tissue, and liver cancer tissue.
Tissue pretreatment: the tissue was washed 3 times with washing solution, the adipose tissue was removed and the tissue in the necrotic and hypoxic areas was avoided to be collected, and the tissue was cut into two parts with sterile surgical scissors, one part (tissue a) for isolated culture of tumor cells and the other part (group B) for isolated culture of endothelial cells.
2) Tissue digestion: shearing the tissues A and B, respectively using 8-10ml of tissue digestive juice, and incubating in two 50ml centrifuge tubes A and B at 37 deg.C water bath for 0.5-2h.
Collecting cell clusters: removing excessive tissue fragments from the tube A by using a 100-micron cell filter screen, cleaning the filter screen by using a cleaning solution, adding 2ml of stop solution to stop digestion, transferring the cell suspension to a 15ml centrifuge tube, centrifuging at 300g for 5min, and removing supernatant; tube B was not filtered, and 2ml of stop solution was added to stop digestion, transferred to a 15ml centrifuge tube, centrifuged at 300g for 5min and the supernatant discarded. If the erythrocyte is gathered at the bottom of the tube, 1ml of erythrocyte lysate is added to lyse the erythrocyte (the erythrocyte acts on ice for 2 min), then 4ml of cleaning solution is added to stop, 300g is added, and the supernatant is centrifuged and discarded after 5 min.
3) Tumor cell culture and expansion (3D): the A tube was added with matrigel for resuspension, and the cell-containing gel drops were inoculated into 48-well plates (25. Mu.l/well), and cultured with tumor cell culture medium. Passage was performed by microscopic observation until the cell density fused to at least 80%. See fig. 3.
4) Endothelial cell culture (2D): endothelial cell culture medium was added to tube B to form a resuspension, and the cell suspension was inoculated into a gelatin-coated plate for 2D culture. Passages were performed by observing under a microscope that the cells fused to at least 80%. See fig. 3.
Endothelial cells have the characteristic of adherent growth, and are separated and cultured from biopsy tissues by utilizing the characteristic, and utilizing the conditions of an endothelial cell culture medium and gelatin coating for promoting adherent growth and the like, more endothelial cells are separated from tumor tissues, meanwhile, non-target cells cannot grow adherent and are suspended in the culture medium, so that better elution can be realized, and the purity of the endothelial cells cultured subsequently is ensured to be stored at a higher level.
5) Endothelial cell screening and expansion: cells were harvested after 0.25% trypsin/EDTA digestion (< 1 min) and digestion was stopped using stop solution, the supernatant was discarded by centrifugation at 300g,5min, 100 μ l 2% FBS in PBS was added to resuspend the pellet and purified using immunomagnetic bead screening or flow cytometry. Wherein the immunomagnetic beads can be CD31 marked immunomagnetic beads, and when the purity of endothelial cells reaches 90%, the endothelial cells are inoculated into a gelatin coated pore plate, and endothelial cell culture medium is added for maintenance culture.
6) 3D co-culture: digesting the tumor cells and endothelial cells obtained after the amplification culture by TrypLE and pancreatin respectively, adding 2 times of volume of cleaning solution and equal volume of stop solution to stop digestion, centrifuging, discarding supernatant, adding 2ml of FPBS (flash suspended solid) to resuspend and precipitate respectively, counting, mixing the tumor cells and the endothelial cells with determined cell number according to the required inoculation number to prepare suspension of two cells, centrifuging, resuspending by matrigel, inoculating into a 48-hole plate, adding a co-culture medium to perform 3D co-culture, and culturing at 37 ℃ and 5% by CO 2 ,21%O 2 After 24h, the cell culture plates were transferred to 37 ℃ C. And 5% CO 2 ,2%O 2 The hypoxic culture in the hypoxic incubator of (2) to induce uniform and rapid growth of blood vessels, 3 days later, the cell culture plate is returned to 37 ℃,5% 2 、21%O 2 The cultivation was continued for 10 days.
7) The vessels generated in the co-culture system were identified by immunofluorescence staining (vascularization evaluation).
The washing solution used in the above-mentioned culture method was a solution containing 1% by volume of penicillin/streptomycin and 99% by volume of DPBS buffer.
The tissue digest of the above culture method comprises 2% by volume of penicillin/streptomycin, 97% by volume of DPBS buffer, 1% by mass of BSA, and 1mg/ml of collagenase type I.
The stop solution of the above culture method is a medium comprising 10% FBS and 90% DMEM/F12.
The endothelial cell culture plate of the above culture method is a 6-well plate, and is coated with 1% or 1.5% gelatin for 1h, wherein the gelatin is prepared by 1 × PBS.
Endothelial cells were cultured and expanded in 2D as described above using a grown Medium MV Medium (Medium containing final concentrations of 0.05ml/ml FBS,0.004ml/ml ECGS,10ng/ml EGF, 90. Mu.g/ml heparin, 1. Mu.g/ml hydrocortisone).
The ratio of the number of endothelial cells to the number of tumor cells in the co-cultured mixed cells was 1, and the cells were resuspended in matrigel (about 4000-10000 cells/25. Mu.l) at a concentration of 8-10 mg/ml.
The above co-culture medium contained 10% by volume of FBS, 1% by volume of cyan/streptomycin, 0.2mM L-Glutamate,100ng/ml of VEGF, 60. Mu.g/. Mu.l of vitamin C and 89% by volume of DMEM/F12 basal medium, in which VEGF and vitamin C are angiogenesis promoting components.
The Matrigel used in the above culture method is Matrigel.
FIG. 1 shows a schematic technical route of the present invention, wherein the anti-vascularization assay means that the generated vascularized tumor organoid model is further used for screening and detecting anti-vascularization drugs in preclinical.
Example 3
An example of a method for inducing angiogenesis in a breast cancer tumor organoid is provided, which comprises two parts I and II.
Endothelial cells and breast cancer tumor cells derived from a biopsy of a patient's tissue are isolated.
Before endothelial cells and tumor cells are separated, a washing solution, a tissue digestion solution and a digestion stop solution (i.e., a stop solution) are prepared.
Preparation of gelatin-coated 6-well plates: according to the manufacturer's method of use, gelatin is formulated as a 2% gelatin solution in an Erlenmeyer flask and placed in an autoclave for autoclaving at 121 ℃ for 20min at a pressure of 15psi. 6-well plates were coated after dilution with 1 × PBS to 1.5% gelatin solution.
Tissue biopsy samples were obtained in accordance with ethical requirements and with the patient's knowledge and consent. The sample is preserved in a 15ml centrifuge tube filled with a preservation solution under the preservation condition of 4 ℃ and the time of less than 24h. Taking out the tissue from the preservation solution, uniformly mixing the preservation solution, taking 100 mu l of the mixed preservation solution, observing under a microscope to judge the cell shedding condition and the cell activity, transferring a tissue sample into a 50ml centrifuge tube, adding 10ml of cleaning solution to shake the centrifuge tube to clean the tissue (repeating for 3 times), dividing the tissue into two parts (A/B) by using sterile surgical scissors and forceps on an ultraclean workbench, using tissue A to separate tumor cells and tissue B to separate endothelial cells, placing a tissue block into two culture dishes, shearing the tissue block, adding 8-10ml of tissue digestive fluid, digesting in water bath at 37 ℃ for 1h, adding 2ml of stop solution to stop digestion, 300g, centrifuging at 5min, discarding supernatant, cleaning and precipitating for 2 times by using FPBS, finally embedding A cell precipitates in Matrigel (1 cell cluster with 4 or more than 4 cells, 1000 cell clusters/25 mu l), and inoculating the A cell precipitates in a 48-well plate. The B cell pellet was resuspended in endothelial cell culture medium and then plated in gelatin-coated 6-well plates.
And (3) purifying endothelial cells after 24-72h, wherein in the embodiment, CD31 magnetic beads are adopted for screening, and the specific steps comprise: the cells were collected in a 15ml centrifuge tube, 300g, centrifuged for 3min, and the cell pellet was resuspended in 1-2ml of medium and counted. 300g, centrifugating for 3min, discarding the supernatant, 1 × 10 7 cells were resuspended in 60. Mu.l of medium and screened according to the manufacturer's protocol. The selected endothelial cells were seeded in 6-well gelatin-coated plates and expanded to about 10 ℃ in culture with endothelial cell culture medium 5 The subsequent experiments can be carried out at any time.
Expanding the tumor cells to 2-3 passages and expanding the cell number to about 10 5 About one day, suspension co-culture of endothelial cells and tumor cells was performed.
Preferably, the preservation solution and the tissue washing solution are collected, 300g is centrifuged for 5min, the supernatant is discarded, and the pellet and the cell pellet obtained by digestion are inoculated into a 48-well plate together, so as to collect the tumor cells to the maximum extent.
Preferably, the digestion time is controlled in the interval of 0.5-2h, and the state and the amount of dissociated cells are observed by blowing with a 10ml pipette every 10 min.
Preferably, DPBS may be exchanged for dhands, which protect the cell viability index more strongly.
Preferably, endothelial cells are purified using a variety of screening methods, such as flow cytometric screening.
Co-culture of endothelial cells with tumor cells.
It is desirable to prepare in advance a co-culture medium consisting of FBS, penicillin/streptomycin, L-Glutamate, DMEM/F12 basal medium and pro-angiogenic components including VEGF and vitamin C.
Preferably, L-Glutamate is replaced by Glutamax, since it is unstable in the medium.
Tumor cells and endothelial cells were collected separately, resuspended and precipitated with co-culture medium, counted, mixed in the ratio of 1 (48 well plate, about 4000/well), 300g, centrifuged for 5min, supernatant discarded, resuspended by adding appropriate amount of Matrigel (48 well plate, 25 μ l/well), 300 μ l per well of pre-formulated co-culture medium was added after the gel drops had solidified, 5% co-co at 37 ℃,5% 2 ,21%O 2 Culturing in the incubator of (1), after 24h, transferring the cell culture plate to 37 ℃,5% 2 ,2%O 2 The hypoxic culture in the hypoxic incubator of (2) to induce uniform and rapid growth of blood vessels, 3 days later, the cell culture plate is returned to 37 ℃,5% 2 、21%O 2 The culture was continued for 10 days.
The method can be used for co-culturing the endothelial cells and the tumor cells in the normal oxygen state all the time, and finally, the endothelial network can be detected, but the endothelial network after the hypoxia treatment is more and the distribution in the aggregate is more uniform. Network formation of the endothelium will be detected on day 4 from the start of the gel drop seeding, and on day 7, aggregates of endothelial cells with tumor cells will be detected, at which time the endothelial network is largely expanded in tumor cell aggregates.
Endothelial network detection:
after co-culture to about day 10, organoid-containing gel drops were subjected to immunofluorescence staining using CD-31, pan-CK antibodies, DAPI staining solution, according to conventional staining procedures, and photographed under a fluorescence microscope, as shown in FIG. 2. It can be seen that endothelial cells and tumor cells are co-formed in the matrigel and cross each other, and that endothelial cells form vessel-like structures both inside and outside the organoid. This 3D co-culture format was shown to be successful and to some extent mimicking the interaction of human tumors with vascular networks.
Example 4
Screening experiment of vascularized tumor organoid model drug constructed by the invention
Anti-angiogenic drugs, such as 17-AAG and Sorafenib, were added to the constructed model and changes in the endothelial network were detected by immunofluorescence staining.
The 17-AAG is also called Tanespecycin and CP 127374, is an effective Hsp90 inhibitor, and can inhibit Akt activation and HER2 and ErbB2 expression, and shows obvious antitumor activity in vitro and in vivo.
Sorafenib (BAY 43-9006, NSC-724772) is a multi-kinase inhibitor of Raf-1 and B-Raf. Sorafenib also inhibits VEGFR-2, VEGFR-3, PDGFR-beta, flt-3 and c-KIT. Sorafenib induces autophagy, apoptosis and activates ferroptosis, and has anti-tumor activity in vivo.
In the embodiment, after 3D matrigel co-culture is carried out on the tumor organoid and the endothelial cells, counting and plating are carried out, 17-AAG and Sorafenib (Sorafenib) drug treatment are respectively carried out, 7 drug gradients are set, the sensitivity and IC50 data of the tumor organoid to the two drugs are obtained, and certain basic data support is provided for subsequent clinical medication and drug dosage.
The data for the drug addition tests in this example are shown in the following table:
17-AAG
concentration (nM) 300 150 75 37.5 18.75 9.375 4.6875
Survival rate 3.41 4.98 20.87 91.57 100.75 103.98 103.80
Sorafenib
Concentration (nM) 400 200 100 50 25 12.25 6.125
Survival rate 2.14 6.00 31.50 75.78 87.92 96.36 102.60
And (4) experimental conclusion: as shown in the above table, as the concentration of the added drug is decreased, the survival rate of tumor cells is increased, with a dose-dependent effect. The model can be used for screening relevant drugs in preclinical, and relevant results are shown in the experimental chart of fig. 6 and fig. 7.
Fig. 8 and 9 show the change of the endothelial network after adding 17-AAG or Sorafenib, respectively, and it can be seen that the green fluorescence representing the generation of the vascular network decreases after adding high concentration of the drug (left panel), indicating that the generation of the vascular network increases with decreasing drug concentration. Thus, the results were consistent with those shown in the drug laboratory tables.
Example 5
Verification of effect of co-culture medium components
The co-culture medium provided by the invention consists of 10% by volume of FBS, 1% by volume of cyan/streptomycin, 0.2mM L-Glutamate, 89% by volume of DMEM/F12 basal medium, 100ng/ml of VEGF and 60 mu g/mu L of vitamin C, wherein the VEGF and the vitamin C are angiogenesis promoting components. This example demonstrates the effect of adding and removing the pro-angiogenic component on co-culture results without changing the other formulation. See fig. 5.
The experimental results are as follows: in the figure, green represents the vessels produced, red represents the tumor cells, blue represents the nucleus, indicating all cells, and non-cellular components are free of blue fluorescence. It is clear that only tumor cells and a small number of endothelial cells are visible without the pro-angiogenic component (left panel) while the formation of a large number of vascular network structures is clearly visible after the pro-angiogenic component (right panel) is added during the same culture period.
Example 6
Verification of different paving proportions
In the co-culture method provided by the invention, the proportion of the mixed cells resuspended in the matrigel is 4000-10000/25 mul. This example demonstrates the construction of different decking ratios, see figure 4.
The experimental results are as follows: as can be seen, when the number of the co-cultured cells is less than 4000/25 μ l, the density of the tumor cells and the endothelial cells is low, and the co-cultured cells are difficult to interact to form a tumor-endothelial mixed organoid; when the number of the co-cultured cells is 4000-10000/25 mul, the tumor cells and the endothelial cells are fused and grow to effectively form tumor organoids with vascular networks; when the number of co-cultured cells is more than 10000 cells/25 mul, the cell density is too high, competitive inhibition is formed, the cells can not effectively proliferate, and organoids with mixed tumor and endothelium can not be formed. Therefore, the cell density of the planking has great influence on the growth of cells, the cell density is too low, the cells are difficult to survive and proliferate, the cell density is too high, the cells compete with each other for nutrients to generate contact inhibition, and the cells are difficult to effectively proliferate. 4000-10000/25 ul are the optimal cell numbers to ensure efficient proliferation of the co-cultured organoids, and more or less than this number are more difficult to achieve.
While the present invention has been described with reference to the embodiments shown in the drawings, the present invention is not limited to the embodiments, which are illustrative and not restrictive, and it will be apparent to those skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope of the invention as defined in the appended claims.

Claims (10)

1. A culture method for inducing angiogenesis of tumor organoids in vitro, comprising the steps of:
the method comprises the following steps: taking biopsy tissues of a patient, cleaning, shearing, then performing tissue digestion, and stopping the tissue digestion after 0.5-2h to obtain a mixed cell mass;
step two: preparing the mixed cell mass obtained in the first step into cell suspension, adding matrigel to resuspend and precipitate, inoculating the cell suspension into a pore plate, adding a tumor cell culture medium to amplify and culture tumor cells, and carrying out passage after the tumor cells are fused at least 80% in density;
step three: preparing the mixed cell mass obtained in the first step into a cell suspension, adding an endothelial cell culture medium to resuspend and precipitate, inoculating the cell suspension into a culture plate coated with gelatin, culturing to obtain endothelial cells, and carrying out passage after the endothelial cells are fused at least 80% in density;
step four: further amplifying and culturing the endothelial cells obtained in the third step, and performing magnetic screening by using immunomagnetic beads or purifying the endothelial cells by using a flow cytometer until the purity of the endothelial cells reaches over 90 percent;
step five: mixing and re-suspending the tumor cells obtained in the second step and the endothelial cells obtained in the fourth step in matrigel, wherein the proportion of the re-suspended mixed cells in the matrigel is 4000-10000/25 mu L, and further inoculating the mixture into a co-culture medium for 3D co-culture, wherein the co-culture medium consists of FBS, penicillin/streptomycin, L-Glutamate, DMEM/F12 basal medium and angiogenesis promoting components, and the angiogenesis promoting components comprise VEGF and vitamin C;
step six: and (3) performing immunofluorescence staining identification on the blood vessels generated in the co-culture system.
2. The culture method according to claim 1, wherein the mixing ratio of the tumor cells and the endothelial cells in the step five is 1:1 to 1:2.
3. the method of claim 1, wherein in step one, the patient biopsy is washed with a wash solution comprising 1% by volume cyan/streptomycin and 99% dpbs buffer.
4. The culture method according to claim 1, wherein in the first step, the tissue digestion is performed using a tissue digestion solution comprising 2% by volume of penicillin/streptomycin, 97% by volume of a DPBS buffer, 1% by mass of BSA, and 1mg/ml of collagenase type I.
5. The culture method of claim 1, wherein in step one, the tissue digestion is terminated with a stop solution comprising 10% fbs and 90% dmem/F12 medium.
6. The culture method according to claim 1, wherein the co-culture medium in the fifth step consists of 10% by volume of FBS, 1% by volume of cyan/streptomycin, 0.2mM L-Glutamate, 89% by volume of DMEM/F12 basal medium, and 100ng/ml of VEGF and 60 μ g/μ L of vitamin C.
7. The culture method according to claim 1, wherein the co-cultured mixed cells in the step five are at 5% CO 2 、21%O 2 Normoxic conditions of (2) and 5% CO 2 、2%O 2 Under hypoxic conditions.
8. A vascularized tumor organoid model obtained by the culture method according to any one of claims 1 to 7, wherein said vascularized tumor organoid model is a co-culture model of endothelial cells derived from the same patient inducing vascularization of the tumor organoid.
9. The vascularized tumor organoid model of claim 8 wherein the tumor cell source of said tumor organoid comprises breast cancer, lung cancer, liver cancer, bile duct cancer, stomach cancer, intestinal cancer, bladder cancer, kidney cancer, pancreatic cancer or esophageal cancer.
10. The use of the vascularized tumor organoid model of claim 8 for preclinical screening of anti-tumor angiogenesis drugs.
CN202310075071.7A 2022-10-11 2023-01-12 Culture method and application of vascularized tumor organoids Active CN115927164B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311143229.6A CN117025715A (en) 2022-10-11 2023-01-12 Method for screening medicines based on vascularized organoids

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211241391 2022-10-11
CN2022112413917 2022-10-11

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202311143229.6A Division CN117025715A (en) 2022-10-11 2023-01-12 Method for screening medicines based on vascularized organoids

Publications (2)

Publication Number Publication Date
CN115927164A true CN115927164A (en) 2023-04-07
CN115927164B CN115927164B (en) 2023-09-26

Family

ID=86550944

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202311143229.6A Pending CN117025715A (en) 2022-10-11 2023-01-12 Method for screening medicines based on vascularized organoids
CN202310075071.7A Active CN115927164B (en) 2022-10-11 2023-01-12 Culture method and application of vascularized tumor organoids

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202311143229.6A Pending CN117025715A (en) 2022-10-11 2023-01-12 Method for screening medicines based on vascularized organoids

Country Status (1)

Country Link
CN (2) CN117025715A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075286A2 (en) * 1999-06-03 2000-12-14 National Research Council Of Canada 3-dimensional in vitro models of mammaliam tissues
US20210079358A1 (en) * 2019-09-16 2021-03-18 POSTECH Research and Business Development Foundation Assembloid - 3d mimetic tissue structure based on patient- derived multiple cell types and method of manufacturing the same
CN114746543A (en) * 2019-11-13 2022-07-12 延世大学校产学协力团 Method for isolating pure cultured vascular endothelial cells, medium for maintaining characteristics of vascular endothelial cells, and culture method comprising same
US20220284832A1 (en) * 2021-03-05 2022-09-08 Advanced Solutions Life Sciences, Llc Models and methods to establish perfused vascularized tissues in three-dimensional in vitro culture

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075286A2 (en) * 1999-06-03 2000-12-14 National Research Council Of Canada 3-dimensional in vitro models of mammaliam tissues
US20210079358A1 (en) * 2019-09-16 2021-03-18 POSTECH Research and Business Development Foundation Assembloid - 3d mimetic tissue structure based on patient- derived multiple cell types and method of manufacturing the same
CN114746543A (en) * 2019-11-13 2022-07-12 延世大学校产学协力团 Method for isolating pure cultured vascular endothelial cells, medium for maintaining characteristics of vascular endothelial cells, and culture method comprising same
US20220284832A1 (en) * 2021-03-05 2022-09-08 Advanced Solutions Life Sciences, Llc Models and methods to establish perfused vascularized tissues in three-dimensional in vitro culture

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MANGALA HEGDE等: "Human breast tumor derived endothelial cells exhibit distinct biological properties", BIOL CELL., vol. 114, no. 2, pages 74 *

Also Published As

Publication number Publication date
CN115927164B (en) 2023-09-26
CN117025715A (en) 2023-11-10

Similar Documents

Publication Publication Date Title
dos Santos et al. Bioreactor design for clinical‐grade expansion of stem cells
EP3158056B1 (en) Single cell-derived organoids
US20150284689A1 (en) Strategy for engineering various 3d tissues, organoids and vasculature
US8278101B2 (en) Stem cell bioprocessing and cell expansion
CN111876386A (en) Method for culturing breast cancer organoid and co-culturing tumor-associated fibroblast
CN106414722B (en) In vitro expansion of erythroid cells
CN108504625B (en) Mouse fibroblast and application thereof
Bertoncello et al. Isolation and clonal assay of adult lung epithelial stem/progenitor cells
Song et al. Influence of gene modification in biological behaviors and responses of mouse lung telocytes to inflammation
US7179643B2 (en) Device and a process for expansion of haemopoeitic stem cells for therapeutic use
CN117004572A (en) Construction method and application of patient-derived transplanted tumor organoid model PDXO
CN108486039B (en) Method for inducing human adipose-derived stem cells to differentiate into testicular interstitial cells by using small molecules
CN112852709B (en) Method for culturing mouse lung organoid
CN113583947B (en) Mesenchymal stem cell and hematopoietic stem cell in vitro culture method and system
CN112553289A (en) Method for evaluating effectiveness of CAR-T cells
JP2008531007A (en) Method for obtaining human hematopoietic stem cell population
CN115927164B (en) Culture method and application of vascularized tumor organoids
CN115125192B (en) Bone marrow supernatant and application thereof in cell culture
CN114292804B (en) Vascularized fat organoid culture method
WO2021221179A1 (en) Establishment of mouse model using human pancreatic cancer organoid
TW200810775A (en) Matrix and method for isolation of hepatic progenitor cells
WO2012026687A2 (en) Human blood-derived blood cell mass assay using cell coagulation, blood adult stem cells using same, method for expanding progenitor cells, and stem cells produced using the method
Chitteti et al. In vitro construction of 2D and 3D simulations of the murine hematopoietic niche
CN113897331A (en) Differentiation method for inducing human artery endothelial cells
CN109182268B (en) Application of umbilical artery endothelial cells in constructing microenvironment of blood stem cells

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
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: 610000 No. 701, 702, 704, Floor 7, Unit 2, Building 1, No. 818, Xingshi Road, Wuhou District, Chengdu, Sichuan

Patentee after: Chengdu nuoyeide medical laboratory Co.,Ltd.

Country or region after: China

Patentee after: Shenzhen Jingke Biotechnology Co.,Ltd.

Address before: 610043 No. 701, 702, 704, 7th Floor, Unit 2, Building 1, No. 818, Xingshi Road, Wuhou District, Chengdu, Sichuan Province

Patentee before: Chengdu nuoyeide medical laboratory Co.,Ltd.

Country or region before: China

Patentee before: GUAGNZHOU JINGKE BIOTECH CO.,LTD.