WO2024007388A1 - Method and kit for detecting cancer cell-specific t cell - Google Patents

Method and kit for detecting cancer cell-specific t cell Download PDF

Info

Publication number
WO2024007388A1
WO2024007388A1 PCT/CN2022/108956 CN2022108956W WO2024007388A1 WO 2024007388 A1 WO2024007388 A1 WO 2024007388A1 CN 2022108956 W CN2022108956 W CN 2022108956W WO 2024007388 A1 WO2024007388 A1 WO 2024007388A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
nanoparticles
antigen
cell
loaded
Prior art date
Application number
PCT/CN2022/108956
Other languages
French (fr)
Chinese (zh)
Inventor
刘密
Original Assignee
苏州尔生生物医药有限公司
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 苏州尔生生物医药有限公司 filed Critical 苏州尔生生物医药有限公司
Publication of WO2024007388A1 publication Critical patent/WO2024007388A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Optical investigation techniques, e.g. flow cytometry
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N2015/1006Investigating individual particles for cytology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Optical investigation techniques, e.g. flow cytometry
    • G01N2015/1486Counting the particles

Definitions

  • the present invention relates to the field of detection technology, and in particular, to a detection method and kit for cancer cell-specific T cells.
  • T cells are the main cells in the body that specifically recognize and kill cancer cells.
  • Each clone of cancer cell-specific T cells can specifically recognize an antigenic epitope.
  • Cancer patients especially those who have undergone immunotherapy or radiotherapy, contain a certain number of cancer cell-specific T cells. Studies have shown that the number of cancer cell-specific T cells in cancer patients treated with immunotherapy and other methods is positively correlated with the prognosis of cancer patients. Therefore, it is particularly important to detect the number of cancer cell-specific T cells in cancer patients.
  • the inventor has previously proposed using nanoparticles or microparticles loaded with whole cell components of cancer cells to assist in the detection of cancer cell-specific T cells (Application No.
  • the present invention provides a method for detecting and killing nanoparticles and/or microparticles prepared by using antigen-presenting cells activated by nanoparticles (NP) or microparticles (MP) loaded with cancer cell whole cell antigens.
  • the method of effective (effector) cancer cell-specific T cells (T eff ) uses nanoparticles or microparticles prepared by activated antigen-presenting cells to first activate cancer cell-specific T cells, and then uses the activated killing cells Markers specifically expressed by cancer cell-specific T cells (T eff ) analyze the content and proportion of cancer cell-specific T cells in the cells to be tested.
  • the first object of the present invention is to provide a method for detecting cancer cell-specific T cells using particles prepared from activated antigen-presenting cells, which includes the following steps:
  • step S3 Incubate the nanovesicles and/or second particles loaded with cell membrane components in step S2 with the cells to be tested, activate broad-spectrum cancer cell-specific T cells that can recognize antigens, and then use appropriate detection technology to analyze the cells to be tested. Markers inside or on the surface of activated cancer cell-specific T cells are analyzed to obtain the number and proportion of the cancer cell-specific T cells by analyzing the number and proportion of T cells containing the markers.
  • markers inside cells or on cell surfaces include proteins or nucleic acids.
  • the specific surface markers are proteins, they include but are not limited to interferon- ⁇ , interleukins, granzymes, perforin, CD69, CD25, OX40 (CD134), CD39, CD103, CD56, CD279, CD278, CD244, CD27, CD154, TCF-1, CD137, CD44, CD28, etc.
  • Technologies that use surface marker analysis to detect the number and proportion of cancer cell-specific T cells include but are not limited to flow cytometry, magnetic bead sorting, enzyme-linked immunospot assay (ELISPOT), enzyme-linked immunosorbent assay (ELISA), etc. .
  • the cells to be tested may be T cells or a cell mixture containing T cells, such as T cells or a cell mixture containing T cells derived from peripheral blood, peripheral immune organs or tumor-infiltrating lymphocytes.
  • the cells to be tested are co-incubated with the products of S2, they can be sorted to sort out the T cells.
  • flow cytometry or magnetic bead sorting is used to select cells from peripheral blood and peripheral immune tissues. , sorting CD3 + cells from tumor infiltrating lymphocytes, sorting CD45 + CD3 + cells, sorting CD3 + CD8 + cells, sorting CD45 + CD3 + CD8 + cells, sorting CD3 + CD4 + cells or sort CD45 + CD3 + CD4 + cells.
  • the co-incubation system of the antigen-presenting cells and the first particles in step S1 may contain cytokines or antibodies.
  • the co-incubation system contains granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-2, IL-7 and IL-12.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • IL-2 granulocyte-macrophage colony-stimulating factor
  • IL-7 granulocyte-macrophage colony-stimulating factor
  • cytokines include, but are not limited to, interleukin 2 (IL-2), interleukin 7 (IL-7), interleukin 14 (IL-14), interleukin 4 (IL-4), interleukin 15 (IL-15), interleukin 21 (IL-21), interleukin 17 (IL-17), interleukin 12 (IL-12), interleukin 6 (IL-6), interleukin 33 (IL-33), gamma interferon (IFN- ⁇ ), TNF- ⁇ .
  • IL-2 interleukin 2
  • IL-7 interleukin 7
  • IL-14 interleukin 14
  • IL-4 interleukin 4
  • interleukin 15 interleukin 15
  • IL-21 interleukin 21
  • IL-17 interleukin 17
  • IL-12 interleukin 12
  • IL-6 interleukin 6
  • IL-33 interleukin 33
  • IFN- ⁇ gamma interferon
  • antibodies include, but are not limited to, ⁇ CD-3 antibody, ⁇ CD-4 antibody, ⁇ CD-8 antibody, ⁇ CD-28 antibody, ⁇ CD-40 antibody, ⁇ OX-40 antibody, and ⁇ OX-40L antibody.
  • the first particle or the second particle can also be loaded with a bacterial component or a bacterial outer vesicle component, and the bacterial component or bacterial outer vesicle component is lysed by a lysis solution containing a lysis agent.
  • the lysing agent is urea, guanidine hydrochloride, deoxycholate, dodecyl sulfate (such as SDS), glycerol, protein degrading enzyme, albumin, lecithin, Triton, Tween, amino acids , glycosides, choline, bacteria including but not limited to Bacillus Calmette-Guérin, Escherichia coli, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium lactis, Lactobacillus acidophilus, Lactobacillus formati, Lactobacillus reuteri, Lactobacillus rhamnosus Bacilli etc.
  • the activated antigen-presenting cell membrane component can also be mixed with the cancer cell cell membrane component or the cancer cell extracellular vesicle membrane component to prepare a mixed membrane component and then be prepared into nanovesicles or loaded on second particles. surface.
  • the first particle or the second particle is also loaded with an immune-enhancing adjuvant.
  • the immune-enhancing adjuvant includes but is not limited to pattern recognition receptor agonist, BCG, BCG cell wall skeleton, BCG methanol extraction residue, BCG muramyl Dipeptides, Mycobacterium phlei, polyantigen, mineral oil, virus-like particles, immune-enhancing reconstituted influenza virions, cholera enterotoxin, saponins and their derivatives, Resiquimod, thymosin, neonatal bovine liver active peptide, Imiquimod, polysaccharide, curcumin, immune adjuvant CpG, immune adjuvant poly(I:C), immune adjuvant poly ICLC, Corynebacterium parvum vaccine, hemolytic streptococcus preparation, coenzyme Q10, levamisole, polycysteine Glycolic acid, manganese adjuvant, aluminum adjuvant, calcium adjuvant, cytokine, interleukin
  • the immune-enhancing adjuvant includes (1) Poly(I:C) or Poly(ICLC); (2) CpG-ODN, wherein CpG-ODN is type A CpG-ODN, type B CpG-ODN and type C At least two types of CpG-ODN, and at least one of them is type B CpG-ODN or type C CpG-ODN.
  • type A CpG-ODN is selected from CpG-ODN 2216, CpG-ODN 1585 or CpG-ODN 2336
  • type B CpG-ODN is selected from CpG-ODN 1018, CpG-ODN 2006, CpG-ODN 1826, CpG-ODN 1668 , CpG-ODN 2007, CpG-ODN BW006 or CpG-ODN SL01
  • Class C CpG-ODN is selected from CpG-ODN 2395, CpG-ODN SL03 or CpG-ODN M362.
  • the first particle or the second particle is also loaded with positively charged polypeptides (such as KALA polypeptide, RALA polypeptide, melittin, etc.), arginine, polyarginine, lysine, and polylysine. , histidine, polyhistidine, NH 4 HCO 3 , protamine or histone, etc.
  • positively charged polypeptides such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • arginine such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • arginine such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • arginine such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • arginine such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • arginine such as KALA polypeptide, RALA polypeptide, melittin, etc.
  • the first particle or the second particle is also loaded with a target that actively targets antigen-presenting cells.
  • the target may be mannose, mannan, CD19 antibody, CD20 antibody, BCMA antibody, CD32 antibody, or CD11c antibody. , CD103 antibody, CD44 antibody, etc.
  • first particle or the second particle can be prepared from the following materials: organic synthetic polymer materials including but not limited to PLGA, PLA, PGA, PEG, PCL, Poloxamer, PVA, PVP, PEI, PTMC, polyanhydride, PDON, PPDO, PMMA, polyamino acids, synthetic peptides, etc.; natural polymer materials include but are not limited to lecithin, cholesterol, alginate, albumin, collagen, gelatin, cell membrane components, starch, sugars, peptides, etc.; inorganic materials include But it is not limited to ferric oxide, ferric oxide, carbonate, phosphate, etc.
  • the particle size of the first particle or the second particle is nanometer or micron, which can ensure that the particles are engulfed by the antigen-presenting cells.
  • the particle size should be within an appropriate range.
  • the particle size of nanoparticles (NP) is 1nm-1000nm, more preferably, the particle size is 30nm-1000nm, most preferably, the particle size is 100nm-600nm; the particle size of microparticles (MP)
  • the size is 1 ⁇ m-1000 ⁇ m, more preferably, the particle size is 1 ⁇ m-100 ⁇ m, more preferably, the particle size is 1 ⁇ m-10 ⁇ m, and most preferably, the particle size is 1 ⁇ m-5 ⁇ m.
  • step S2 the activated antigen-presenting cells are subjected to mechanical destruction, membrane filtration or gradient centrifugation to prepare nanovesicles, or the activated antigen-presenting cells are subjected to mechanical destruction, membrane filtration or gradient centrifugation,
  • the product is combined with the second particle to obtain the second particle loaded with cell membrane components.
  • the mechanical destruction method is selected from one or more of ultrasonic, homogenization, homogenization, high-speed stirring, high-pressure destruction, high-shear force destruction, swelling, chemical substances, and shrinkage.
  • the co-action method is selected from one or more of co-incubation, co-extrusion, ultrasound, stirring, homogenization and homogenization.
  • the antigen-presenting cell components cover the original nanoparticles. New nanoparticles or microparticles are formed on the surface of particles or microparticles.
  • the cancer cells or tumor tissues are frozen at -20°C to -273°C, and water or a solution without a dissolving agent is added, followed by repeated freeze-thaw lysis.
  • the resulting supernatant is a water-soluble component, which is dissolved during the precipitation.
  • the agent is dissolved, the soluble part becomes the non-water-soluble component.
  • the water-soluble component and the non-water-soluble component are combined to obtain the whole cell component of the cancer cells.
  • the dissolving agent is selected from the group consisting of urea, guanidine hydrochloride, deoxycholate, dodecyl sulfate (such as SDS), glycerin, protein degrading enzyme, albumin, lecithin, inorganic salts (0.1-2000mg/mL), Triton, and methane. At least one of temperature, amino acid, glycoside and choline.
  • the antigen-presenting cells include at least one of B cells, dendritic cells (DC) and macrophages, preferably two or more including DC, and more preferably a combination of three types of cells.
  • DC dendritic cells
  • macrophages preferably two or more including DC, and more preferably a combination of three types of cells.
  • the obtained cancer cell-specific T cells include CD4 + T cells and/or CD8 + T cells, and preferably include both CD4 + T cells and CD8 + T cells.
  • nanoparticles and/or microparticles loaded with cancer cell whole cell antigens are used to specifically activate antigen-presenting cells, and then the antigen-presenting cells are prepared into nanoparticles or microparticles.
  • the prepared nanoparticles or micron particles are The particles are loaded with whole-cell antigen epitopes of cancer cells, and then nanoparticles or microparticles prepared from antigen-presenting cells are used to activate cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor-infiltrating lymphocytes, and then used Characteristics of activated cancer cell-specific T cells that highly express certain molecules within the cell or on the cell surface.
  • the antigen-presenting cells can be derived from the same type as the cancer cell-specific T cells, allogeneic, cell lines, or transformed from stem cells.
  • At least one of the cancer cells or tumor tissues used to prepare the antigen is the same as the disease type corresponding to the detected cancer cell-specific T cells.
  • the second object of the present invention is to provide a kit for detecting cancer cell-specific T cells, which kit includes at least one of the following (1)-(2):
  • Nanovesicles prepared from activated antigen-presenting cells prepared from activated antigen-presenting cells
  • the nanovesicles are prepared by co-incubating the antigen-presenting cells with the first particles to obtain activated antigen-presenting cells, and then extracting the cell membrane components of the activated antigen-presenting cells;
  • the particles loaded with cell membrane components of activated antigen-presenting cells are obtained by cooperating the cell membrane components of activated antigen-presenting cells with second particles, so that the cell membrane components are loaded on the second particles;
  • the first particles or second particles are independently selected from nanoparticles or microparticles loaded with tumor tissue and/or whole cell components of cancer cells.
  • the present invention breaks through the limitations of existing detection methods, allowing particles to be loaded with all antigens and activated antigen-presenting cell membranes, capable of detecting a wider spectrum and variety of cancer cell-specific T cells, and is highly specific and effective in immune detection. Better, thus providing a potential biomarker detection method for immunotherapy.
  • the present invention at least has the following advantages:
  • the present invention provides a technology for in vitro detection of cancer cell-specific T cells in immune cells using a nanoscale or micron-scale particle delivery system.
  • the analyzed and detected cancer cell-specific T cells are broad-spectrum and highly specific, including all clones that can Specific recognition and killing of cancer cell effector (killer) cancer cell-specific T cells (T eff ); and, compared with particles prepared without loaded activated antigen-presenting cells, particles loaded with activated antigen-presenting cells
  • T eff cancer cell effector cancer cell-specific T cells
  • the particles of the present invention to detect cancer cell-specific T cells to avoid the situation where some cancer cell-specific T cells cannot be detected because the specific markers expressed after activation are weak. Therefore, the detection method of the present invention The detection accuracy of the method is higher. On this basis, the present invention also optimizes the activation process of antigen-presenting cells, the incubation process with T cells, and the loading materials of the first particle and the second particle, so that the method can detect more comprehensive cancer cell-specific T cells, and The signal is stronger and more accurate during detection.
  • Figure 1 is a schematic diagram of the preparation process and application of the cell system of the present invention
  • a is a schematic diagram of collecting and preparing nanoparticles or microparticles from water-soluble components and non-water-soluble components respectively
  • b is dissolving using a dissolving solution containing a dissolving agent
  • c is after activating antigen-presenting cells using nanoparticles and/or microparticles prepared in a or b, and preparing the activated antigen-presenting cells into particles.
  • Figures 2-14 respectively show the experimental results of using nanoparticles or microparticles to detect cancer cell-specific T cells in Examples 1-13; * represents p ⁇ 0.05, indicating a significant difference.
  • the present invention is used to detect cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor infiltrating lymphocytes.
  • the cancer cell-specific T cells are first prepared with activated antigen-presenting cells during detection. Nanoparticles and/or microparticles are co-incubated, and then flow cytometry, enzyme-linked immunospot assay (ELISPOT), or enzyme-linked immunosorbent assay are used to analyze molecules that are highly expressed after cancer cell-specific T cells are specifically activated by antigens. A broad spectrum of cancer cell-specific T cell information can be obtained.
  • the antigen-presenting cells used to prepare nanoparticles or microparticles are first activated by nanoparticles and/or microparticles loaded with tumor tissue and/or cancer cell whole cell antigens or mixtures thereof.
  • the process and application fields of detecting cancer cell-specific T cells are shown in Figure 1.
  • the cells or tissues can be lysed and the water-soluble components and water-insoluble antigens can be collected separately to prepare nanoparticle or microparticle systems respectively; alternatively, a solution containing a dissolving agent can be directly used.
  • the lysis solution directly lyses cells or tissues and dissolves whole cell antigens of cancer cells to prepare nano or micro particle systems.
  • the cancer cell whole cell antigen of the present invention can be subjected to a process including but not limited to inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, nuclease treatment, protease treatment before or (and) after lysis.
  • Nanoparticles or microparticles can be prepared after endolysis or degradation; or without any inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, nucleic acid treatment before or after cell lysis. Nanoparticles or microparticles are directly prepared by enzymatic treatment, proteolytic digestion or degradation.
  • tumor tissue cells are inactivated or/and denatured before lysis. In actual use, they can also be inactivated or/and denatured after cell lysis, or the cells can also be lysed.
  • Inactivation or (and) denaturation treatment is performed before and after lysis; in some embodiments of the present invention, the inactivation or (and) denaturation treatment method before or (and) after cell lysis is ultraviolet irradiation and high-temperature heating.
  • Treatment methods including but not limited to radiation irradiation, high pressure, solidification, biomineralization, ionization, chemical modification, nuclease treatment, collagenase treatment, protease endolysis or degradation, freeze-drying and other treatment methods can also be used in the process.
  • Treatment methods including but not limited to radiation irradiation, high pressure, solidification, biomineralization, ionization, chemical modification, nuclease treatment, collagenase treatment, protease endolysis or degradation, freeze-drying and other treatment methods can also be used in the process.
  • Those skilled in the art can understand that during actual application, the skilled person can make appropriate adjustments according to specific circumstances.
  • the antigen-presenting cells When preparing activated antigen-presenting cells into nanoparticles or micron particles, the antigen-presenting cells are first mechanically destroyed, and then centrifuged and/or filtered with a filter membrane of a certain pore size. Optionally, the antigen-presenting cells are combined with the nanoparticles or micron particles. Particles work together.
  • Activated antigen-presenting cells contain a certain cell membrane structure after mechanical destruction.
  • the antigen-presenting cells prepared into nanoparticles or microparticles and the antigen-presenting cells used to incubate with T cells can be derived from autologous or allogeneic sources, or from cell lines or stem cells.
  • Antigen-presenting cells can be DC cells, B cells, macrophages, or any mixture of the above three, or other cells with antigen-presenting functions.
  • the system can contain cytokines and/or antibodies to improve activation efficiency.
  • nanoparticles or microparticles loaded with cancer cell whole cell antigens are used to first activate antigen-presenting cells, and then the antigen-presenting cells are prepared into nanoparticles or microparticles, and the nanoparticles or microparticles prepared by the antigen-presenting cells are used.
  • the specific method of using micron particles to detect cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor-infiltrating lymphocytes is as follows:
  • Step 1 Add a first predetermined volume of an aqueous phase solution containing a first predetermined concentration to a second predetermined volume of an organic phase containing a second predetermined concentration of the raw material for preparing particles.
  • the aqueous solution may contain each component of the cancer cell/tumor tissue lysate and an immune-enhancing adjuvant; each component of the lysate is a water-soluble component during preparation or is dissolved in a solution containing The original non-water-soluble components in the solution of dissolving agents such as urea or guanidine hydrochloride.
  • the concentration of water-soluble components or the original concentration of non-water-soluble components contained in the aqueous phase solution, that is, the first predetermined concentration requires the concentration of protein polypeptide to be greater than 1ng/mL, and can load enough cancer cell whole cell antigens to activate related cells. .
  • the concentration of the immune-enhancing adjuvant in the initial aqueous phase is greater than 0.01ng/mL.
  • the second predetermined concentration of raw materials for preparing particles ranges from 0.5 mg/mL to 5000 mg/mL, preferably 100 mg/mL.
  • the second predetermined volume of the organic phase is set according to its ratio to the first predetermined volume of the aqueous phase.
  • the range of the ratio of the first predetermined volume of the aqueous phase to the second predetermined volume of the organic phase is It is 1:1.1-1:5000, preferably 1:10.
  • the first predetermined volume, the second predetermined volume and the ratio of the first predetermined volume to the second predetermined volume can be adjusted as needed to adjust the size of the prepared nanoparticles or microparticles.
  • the solvent is DMSO, acetonitrile, ethanol, chloroform, methanol, DMF, isopropyl alcohol, dichloromethane, propanol, ethyl acetate, etc., preferably dichloromethane;
  • the concentration of the organic phase is 0.5 mg/mL ⁇ 5000mg/mL, preferably 100mg/mL.
  • Step 2 subject the mixed solution obtained in Step 1 to ultrasonic treatment for more than 2 seconds or stirring or homogenization treatment or microfluidic treatment for more than 1 minute.
  • the stirring is mechanical stirring or magnetic stirring
  • the stirring speed is greater than 50 rpm
  • the stirring time is greater than 1 minute.
  • the stirring speed is 50 rpm ⁇ 1500 rpm
  • the stirring time is 0.1 hour ⁇ 24 hours
  • the ultrasonic power is greater than 5W
  • the time Greater than 0.1 seconds such as 2 to 200 seconds
  • Step 3 Add the mixture obtained after step 2 to a third predetermined volume of aqueous solution containing a third predetermined concentration of emulsifier and perform ultrasonic treatment for more than 2 seconds or stirring for more than 1 minute or perform homogenization or microfluidic treatment. deal with.
  • the mixture obtained in step 2 is added to the aqueous emulsifier solution and continued to be ultrasonically or stirred to form nanometers or micrometers.
  • the ultrasonic time is greater than 0.1 seconds, such as 2 to 200 seconds
  • the stirring speed is greater than 50 rpm, such as 50 rpm to 500 rpm
  • the stirring time is greater than 1 minute, such as 60 to 6000 seconds.
  • the stirring speed is greater than 50rpm, and the stirring time is greater than 1 minute.
  • the stirring speed is 50rpm to 1500rpm, and the stirring time is 0.5 to 5 hours; during ultrasonic treatment, the ultrasonic power is 50W to 500W.
  • the time is greater than 0.1 seconds, such as 2 to 200 seconds; when homogenizing, use a high-pressure/ultra-high-pressure homogenizer or high-shear homogenizer.
  • the pressure is greater than 20 psi, such as 20 psi to 100 psi.
  • the rotation speed is greater than 1000rpm, such as 1000rpm ⁇ 5000rpm; when using microfluidic processing, the flow rate is greater than 0.01mL/min, such as 0.1mL/min-100mL/min.
  • Ultrasonic or stirring or homogenization treatment or microfluidic treatment can be used to nanonize or micronize the particles.
  • the length of ultrasonic time or stirring speed or homogenization process pressure and time can control the size of the prepared nano or micron particles. Too large or too small will cause Changes in particle size.
  • the emulsifier aqueous solution is a polyvinyl alcohol (PVA) aqueous solution
  • the third predetermined volume is 5 mL
  • the third predetermined concentration is 20 mg/mL.
  • the third predetermined volume is adjusted according to its ratio to the second predetermined volume.
  • the range between the second predetermined volume and the third predetermined volume is set to 1:1.1-1:1000, preferably 2:5.
  • the ratio of the second predetermined volume and the third predetermined volume can be adjusted.
  • the ultrasonic time or stirring time, the volume and concentration of the emulsifier aqueous solution in this step are all based on obtaining nanoparticles or microparticles of suitable size.
  • Step 4 Add the liquid obtained after the treatment in Step 3 to a fourth predetermined volume of the emulsifier aqueous solution with a fourth predetermined concentration, and stir until the predetermined stirring conditions are met.
  • the emulsifier aqueous solution is PVA solution or other solutions.
  • the predetermined stirring condition of this step is until the volatilization of the organic solvent is completed, that is, the volatilization of methylene chloride in step 1 is completed.
  • Step 5 After centrifuging the mixed liquid that meets the predetermined stirring conditions in Step 4 at a rotation speed of greater than 100 RPM for more than 1 minute, remove the supernatant, and resuspend the remaining sediment in a fifth predetermined volume of Five predetermined concentrations of an aqueous solution containing a lyoprotectant or a sixth predetermined volume of PBS (or physiological saline).
  • Step 6 After freeze-drying the suspension containing the lyoprotectant obtained in Step 5, the freeze-dried material is used for later use.
  • Step 7 Resuspend a sixth predetermined volume of the nanoparticle-containing suspension obtained in Step 5 in PBS (or physiological saline) or use a sixth predetermined volume of PBS (or physiological saline) to resuspend the nanoparticle-containing suspension obtained in Step 6
  • PBS or physiological saline
  • the freeze-dried substance containing nanoparticles or microparticles and a lyoprotectant is used directly; or the above sample is mixed with a seventh predetermined volume of water-soluble components or dissolved original non-water-soluble components and used.
  • the volume ratio of the sixth predetermined volume to the seventh predetermined volume is 1:10000 to 10000:1, the preferred volume ratio is 1:100 to 100:1, and the optimal volume ratio is 1:30 to 30:1 .
  • Step 8 Incubate the antigen-presenting cells with the nanoparticles and/or microparticles prepared above for a certain period of time.
  • the tumor tissue and/or cancer cells and antigen-presenting cells used to prepare nanoparticles and/or microparticles can be from autologous or allogeneic sources.
  • Step 9 Collect the co-incubated cells and perform mechanical damage such as sonication, homogenization, and mechanical stirring.
  • Step 10 Centrifuge the ultrasonicated sample and/or filter it with a filter membrane of a certain pore size, and/or interact with nanoparticles and/or microparticles loaded with whole cell components of cancer cells to prepare a sample based on antigen-presenting cells. Nanoparticles or microparticles.
  • Step 11 Obtain peripheral blood, peripheral immune tissue or tumor tissue, and collect T cells or immune cells containing T cells in the above tissues.
  • the above peripheral blood, peripheral immune tissue or tumor tissue can be from autologous or allogeneic.
  • Step 12 Mix the nanoparticles and/or microparticles prepared in step 10 with the T cells or immune cells containing T cells obtained in step 11 and incubate them together for a certain period of time.
  • Step 13 Use flow cytometry, enzyme-linked immunospot method, enzyme-linked immunosorbent assay, or magnetic bead sorting method to analyze the content of cancer cell-specific T cells activated by the antigen.
  • Example 1 Nanoparticles are used to detect cancer cell-specific T cells in peripheral immune organs
  • This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticles prepared by nanoparticle-activated antigen-presenting cells to detect cancer cell-specific T cells in mouse splenocytes.
  • B16F10 melanoma tumor tissue was lysed to prepare water-soluble components and non-water-soluble components of the tumor tissue.
  • the organic polymer material PLGA was used as the nanoparticle skeleton material, and Polyinosinic-polycytidylic acid (poly(I :C))
  • a solvent evaporation method is used to prepare a nanoparticle system loaded with water-soluble components and non-water-soluble components of tumor tissue, and then the nanoparticles are used to activate antigen-presenting cells, and the antigen-presenting cells are mechanically Nanoparticles are prepared by centrifugation after destruction, and the nanoparticles are used to assist in the detection of cancer cell-specific T cells in peripheral immune organs.
  • the nanoparticle 1 is prepared by the double emulsion method in the solvent evaporation method.
  • nanoparticles loaded with water-soluble components in whole cell antigens of cancer cells and nanoparticles loaded with non-water-soluble components in whole cell antigens of cancer cells are prepared separately, and then used together.
  • the molecular weight of PLGA, the material used to prepare the nanoparticles, is 24KDa-38KDa.
  • the immune adjuvant used is poly(I:C) and poly(I:C) is only distributed inside the nanoparticles.
  • the preparation method is as described above. During the preparation process, the double emulsion method is first used to load cell components and adjuvants inside the nanoparticles.
  • nanoparticles After loading the cell lysis components inside, 100 mg of nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of nanoparticles containing Resuspend in 4% trehalose ultrapure water and freeze-dry for 48 h.
  • the average particle size of the nanoparticles 1 is about 280nm, and each 1 mg of PLGA nanoparticles is loaded with approximately 100 ⁇ g of protein or peptide components.
  • the poly(I:C) immune adjuvant used per 1 mg of PLGA nanoparticles is 0.02 mg.
  • This example uses the preparation of dendritic cells from mouse bone marrow cells as an example to illustrate how to prepare BMDC.
  • a 6-8 week old C57 mouse was sacrificed by cervical dislocation.
  • the tibia and femur of the hind legs were surgically removed and placed in PBS.
  • the muscle tissue around the bones was removed with scissors and tweezers.
  • the needles are inserted into the bone marrow cavity from both ends of the bone, and the bone marrow is repeatedly flushed into the culture dish.
  • RPMI 1640 (10% FBS) medium to stop lysis, centrifuge at 400 g for 3 min, and discard the supernatant.
  • the cells were placed in a 10 mm culture dish and cultured in RPMI 1640 (10% FBS) medium with recombinant mouse GM-CSF (20 ng/mL) added at 37 degrees Celsius and 5% CO 2 for 7 days. On the third day, shake the culture bottle gently and add the same volume of RPMI 1640 (10% FBS) medium containing GM-CSF (20ng/mL).
  • Nanoparticles loaded with whole cell components of cancer cells derived from tumor tissue 250 ⁇ g nanoparticles loaded with water-soluble components + 250 ⁇ g nanoparticles loaded with non-water-soluble components were completely cultured with BMDC (10 million) in 15 mL RPMI1640.
  • the base was incubated for a total of 48 hours (37°C, 5% CO 2 ); the incubation system contained cytokine combination 1: granulocyte-macrophage colony-stimulating factor (GM-CSF, 500U/mL), IL-2 (500U/mL) ), IL-7 (500U/mL), IL-12 (500U/mL) or containing cytokine component 2: GM-CSF (500U/mL), IL-4 (500U/mL), tumor necrosis factor ⁇ ( TNF- ⁇ , 500U/mL), IL10 (500U/mL).
  • cytokine combination 1 granulocyte-macrophage colony-stimulating factor (GM-CSF, 500U/mL), IL-2 (500U/mL) ), IL-7 (500U/mL), IL-12 (500U/mL) or containing cytokine component 2: GM-CSF (500U/mL), IL-4 (
  • step (3) collect the DCs (10 million) prepared in step (3) without any nanoparticle or microparticle activation, then wash the cells twice with physiological saline, resuspend the cells in physiological saline and incubate at 4°C and 7.5W. Sonicate for 20 minutes to disrupt cells and prepare samples containing cell membrane components. The sample was then centrifuged at 2000g for 20 minutes and the supernatant was collected. The supernatant was centrifuged at 7000g for 20 minutes and the supernatant was collected. The supernatant was mixed with 40 mg of nanoparticles loaded with cancer cell whole cell components prepared in step (2).
  • Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 ⁇ m filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticle 3 with a particle size of 300 nm.
  • Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 ⁇ m filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticles. Among them, the nanoparticle 4 obtained by incubating the nanoparticle 1 with the membrane component has a particle size of 300 nm.
  • Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 ⁇ m filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticle 5 with a particle size of 300 nm.
  • mice 0.5 ⁇ 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and splenocytes were harvested. Mouse splenocytes were prepared into a single cell suspension, and then CD3 + T cells were sorted from the mouse splenocyte single cell suspension using flow cytometry.
  • Nanoparticle 1 50 ⁇ g nanoparticles loaded with water-soluble components + 50 ⁇ g nanoparticles loaded with non-water-soluble components
  • nanoparticles 2 100 ⁇ g
  • nanoparticles 3 100 ⁇ g
  • nanoparticles 4 100 ⁇ g
  • nanoparticles 5 100 ⁇ g
  • splenocyte-derived T cells 1 million
  • RPMI 1649 10 mL
  • the cells were then collected and centrifuged at 400g for 5 minutes. The cells were resuspended in PBS and the T cells were first treated with Fc block to avoid non-specific loading.
  • CD3 antibody, CD4 antibody, and CD8 antibody were used for extracellular staining of mouse splenocytes, and then the cells were fixed and membrane-broken, and FN- ⁇ antibody was used for intracellular staining of T cells.
  • the sample T cells were then detected using flow cytometry. Analyze the proportion of CD4 + T cells that can secrete IFN- ⁇ after activation among all CD4 + T cells and the proportion of CD8 + T cells that can secrete IFN- ⁇ after activation among all CD8 + T cells. Proportion of T cells.
  • the above CD4 + IFN- ⁇ + T cells and CD8 + IFN- ⁇ + T cells are cancer cell-specific T cells.
  • splenocyte-derived T cells (1 million) were incubated in 10 mL of RPMI 1649 complete medium for 72 hours (37°C, 5% CO 2 ). The cells were then collected and centrifuged at 400g for 5 minutes. The cells were resuspended in PBS and the T cells were first treated with Fc block to avoid non-specific loading. Then use anti-mouse CD3 antibody, anti-mouse CD4 antibody, and anti-mouse CD8 antibody connected with specific fluorescent probes to perform extracellular staining of mouse splenocytes.
  • the cells were fixed and membrane-broken, and the cells were fixed and membrane-broken using the anti-mouse CD4 antibody and anti-mouse CD8 antibody connected with specific fluorescent probes.
  • Flow cytometry is then used to detect T cells in the sample that contain fluorescent signals linked to IFN- ⁇ antibodies. Analyze the proportion of CD4 + T cells that can secrete IFN- ⁇ after activation among all CD4 + T cells and the proportion of CD8 + T cells that can secrete IFN- ⁇ after activation among all CD8 + T cells. Proportion of T cells.
  • the above CD4 + IFN- ⁇ + T cells and CD8 + IFN- ⁇ + T cells are cancer cell-specific T cells.
  • antigen-presenting cells After nanoparticles/microparticles loaded with tumor tissue and/or cancer cell whole cell components are engulfed by antigen-presenting cells, the antigen will be degraded into polypeptide epitopes and combined with major histocompatibility complex (MHC) molecules. Presented to the surface of antigen-presenting cell membranes. Since whole-cell antigens loaded with nanoparticles and microparticles can be cross-presented and released, cancer cell antigen epitopes can be presented to the surface of antigen-presenting cell membranes through two pathways, MHC I and MCH II.
  • MHC major histocompatibility complex
  • the MHC molecules and antigen peptide complexes loaded therein can directly bind to T cell surface receptors on the surface of T cells that can specifically recognize cancer cell antigens. Moreover, if the T cells are specific T cells that can kill cancer cells, the cancer cells will begin to secrete killer substances such as IFN- ⁇ and granzymes. By analyzing the T cells secreting killer substances, we can obtain the characteristics of cancer cells. Proportion of effector cancer cell-specific T cells with recognition and killing capabilities.
  • Nanoparticle 2 As shown in Figure 2, the cancer cell-specific T cells in the T cell alone control group and the nanoparticle 1 detection group were very low.
  • Nanoparticle 2, nanoparticle 3, nanoparticle 4 and nanoparticle 5 can all assist in detecting a certain amount of cancer cell-specific T cells.
  • nanoparticle 5 has the best effect, and nanoparticle 5 has a better effect than nanoparticle 2, nanoparticle 3 and nanoparticle 4.
  • Nanoparticle 5 is better than nanoparticle 4, indicating that the effect of adding cytokine combination 1 during the activation of antigen-presenting cells is better than cytokine component 2; nanoparticle 5 is better than nanoparticle 2, indicating that the surface of nanoparticles that assist in the detection of T cells is loaded with antigens.
  • the effect of solid nanoparticles loaded with cell components inside is better than that of nanovesicle structures that only load membrane components on the surface; Nanoparticles 4 and 5 are better than Nanoparticles 3, indicating that whole cells of cancer cells are loaded.
  • the nanoparticles prepared by the antigen-presenting cells activated by the nanoparticles of the component are more effective than the nanoparticles prepared by the unactivated antigen-presenting cells.
  • the particle system of the present invention can be used to detect cancer cell-specific T cells.
  • the antigen-presenting cells activated by the nanoparticles loaded with cancer cell whole cell components will degrade and present the broad-spectrum whole-cell antigens in the cancer cell whole cell components loaded with the phagocytosed nanoparticles, which are presented to Cancer cell antigen epitopes on the cell membrane surface have been bound to major histocompatibility complex (MHC) molecules.
  • MHC major histocompatibility complex
  • the cell membrane components in the above antigen-presenting cells will form nanoparticles or microparticles, and be loaded with MHC molecules and degraded. Therefore, it can directly activate cancer cell-specific T cells for detecting this type of cells without the assistance of antigen-presenting cells.
  • Example 2 Antigen-presenting cell-based particles are used for the detection of cancer cell-specific T cells
  • This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticle-activated antigen-presenting cells to prepare nanoparticles to assist in the detection of cancer cell-specific T cells.
  • B16F10 melanoma tumor tissue was lysed to prepare water-soluble components and non-water-soluble components of the tumor tissue.
  • PLGA was used as the nanoparticle framework material
  • poly(I:C) and CpG1018 were used as immune adjuvants.
  • the agent uses a solvent evaporation method to prepare a nanoparticle system loaded with water-soluble components and non-water-soluble components of tumor tissue, then uses nanoparticles to activate antigen-presenting cells, and prepares the activated antigen-presenting cells into nanoparticle detection Cancer cell-specific T cells among peripheral blood immune cells.
  • nanoparticles were prepared by solvent evaporation method.
  • the immune adjuvants used are poly(I:C) and CpG1018, and the adjuvants are contained inside the nanoparticles.
  • the preparation method is as described above. During the preparation process, the double emulsion method is first used to load the antigen and adjuvant inside the nanoparticles.
  • nanoparticles After loading the antigen (cleavage component) inside, 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL containing Resuspend in 4% trehalose ultrapure water and freeze-dry for 48 h.
  • the average particle size of the nanoparticles 1 is about 280nm; each 1 mg of PLGA nanoparticles is loaded with approximately 100 ⁇ g of protein and peptide components, and each 1 mg of PLGA nanoparticles uses 0.02 mg of poly(I:C) and CpG1018 immune adjuvant; nanoparticles 1 Also called nanovaccine1.
  • polypeptide neoantigens B16-M20 (Tubb3, FRRKAFLHWYTGEAMDEMEFTEAESNM), B16-M24 (Dag1, TAVITPPTTTTKKARVSTPKPATPSTD), B16-M46 (Actn4, NHSGLVTFQAFIDVMSRETTDTDTADQ) and TRP2:180-188 (SVYDFFVWL) were loaded with equal mass.
  • Peptide nanoparticle 2 is used as a control nanoparticle. Its preparation materials and preparation methods are the same as nanoparticle 1.
  • the particle size of control nanoparticle 2 is about 280 nm, and it is loaded with 100 ⁇ g of peptide component and an equal amount of adjuvant.
  • the preparation materials and methods of blank nanoparticle 3 are the same as those of nanoparticle 1.
  • the particle size is about 280 nm. It only carries the same amount of immune adjuvant but does not load any antigen component.
  • Bone marrow-derived dendritic cells (BMDC) and B cells were used as antigen-presenting cells.
  • the preparation of BMDC is the same as in Example 1.
  • the B cell extraction process is as follows: kill the mouse and remove the mouse spleen, then prepare a mouse splenocyte single cell suspension, and then use magnetic bead sorting to sort CD19 + B cells from the splenocyte single cell suspension. Mix BMDC and B cells at a ratio of 1:1 and use them as mixed antigen-presenting cells.
  • nanoparticle 1 500 ⁇ g or peptide nanoparticle 2 (500 ⁇ g) or blank nanoparticle 3 (500 ⁇ g) + free lysate with 20 million mixed antigen-presenting cells (10 million BMDC + 10 million B cells) in 15 mL Incubate in RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contains a combination of cytokines: IL-15 (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL) mL), IL-12 (1000U/mL).
  • IL-15 500U/mL
  • IL-2 500U/mL
  • IL-7 500U/mL
  • IL-12 1000U/mL
  • nanoparticle 1 and 20 million BMDCs were incubated in 15mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contained a combination of cytokines: IL-15 (500U/mL), IL-2 ( 500U/mL), IL-7 (500U/mL), IL-12 (1000U/mL).
  • IL-15 500U/mL
  • IL-2 500U/mL
  • IL-7 500U/mL
  • IL-12 1000U/mL
  • Particle 1 50 mg or peptide nanoparticle 2 (50 mg) or blank nanoparticle 3 (50 mg) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 ⁇ m filter. The extruded liquid was centrifuged at 15000g for 60 minutes and discarded. After removing the supernatant, resuspend in physiological saline and the resulting precipitate is nanoparticles.
  • the nanoparticles prepared by using the mixed antigen-presenting cell membrane components activated by nanoparticle 1 and nanoparticle 1 are nanoparticles 4, with a particle size of 300 nm; the mixed antigen-presenting cell membrane activated by peptide nanoparticles 2
  • the nanoparticles prepared by the interaction between the components and the polypeptide nanoparticles 2 are nanoparticles 5, with a particle size of 300 nm; they are prepared by the interaction between the mixed antigen-presenting cell membrane components activated by the blank nanoparticles 3 and the blank nanoparticles 3.
  • the obtained nanoparticles were nanoparticle 6, with a particle size of 300 nm.
  • step (2) Collect the filtrate and incubate it with the nanoparticle 1 (50 mg) prepared in step (2) for 10 minutes, and then Repeated co-extrusion using a 0.45 ⁇ m filter membrane, centrifuge the extrudate at 15,000 g for 60 minutes, discard the supernatant and resuspend in physiological saline, and the resulting precipitate is Nanoparticle 7, with a particle size of 300 nm.
  • mice On day 0, 1.5 ⁇ 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse.
  • the mice On days 10 and 14, the mice were subcutaneously injected with 100 ⁇ L of 1 mg PLGA nanovaccine loaded with whole cell components of cancer cells1. Or 100 ⁇ L PBS; monitor the tumor growth rate of mice in the PBS group and vaccine group during the process. The mice were sacrificed on day 18, and their peripheral blood was collected.
  • Peripheral blood mononuclear cells (PBMC) were isolated from the mouse peripheral blood using density gradient centrifugation, and then CD3 + T cells were isolated from the PBMC using flow cytometry. .
  • the CD3 + T cells in the group treated with Nano Vaccine 1 are the vaccine treatment group
  • the T cells in the group treated with PBS are the PBS control group.
  • nanoparticle 1, or nanoparticle 2, or nanoparticle 3 prepared in step (2) with B cells (5 million) and 400,000 T cells from tumor-infiltrating lymphocytes (vaccine group or PBS group) were incubated in 5 mL RPMI1640 complete medium for a total of 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to sort the incubated CD3 + IFN- ⁇ + T cells.
  • MFI mean fluorescence intensity
  • step (5) antigen-presenting cell-based nanoparticles prepared in step (5) (nanoparticle 4, or nanoparticle 5, or nanoparticle 6, or nanoparticle 7) with 400,000 T cells derived from tumor-infiltrating lymphocytes ( Vaccine group or PBS group) were incubated in 5 mL RPMI1640 complete medium for a total of 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to sort the incubated CD3 + IFN- ⁇ + T cells.
  • MFI mean fluorescence intensity
  • nanoparticle 1 prepared in step (2) with 400,000 T cells (vaccine group) derived from tumor infiltrating lymphocytes in 5 mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), and then use flow cytometry to sort the incubated CD3 + IFN- ⁇ + T cells.
  • T cells vaccine group
  • the tumor growth rate of mice in the PBS group was significantly faster than that of the vaccine group. This shows that there are significantly more cancer cell-specific T cells in the mice in the vaccine group than in the PBS group, and thus the mouse tumors can be delayed and controlled. Volume growth.
  • the PBS group has not been induced by vaccines, so it contains fewer cancer cell-specific T cells, while the vaccine group will have more cancer cell-specific T cells after vaccine induction.
  • the PBS control group and vaccine group There are differences in the results after testing different nanoparticles.
  • Nanoparticle 4 has the best effect and can detect the most comprehensive cancer cell-specific T cells. Nanoparticle 1 cannot detect cancer cell-specific T cells without the assistance of antigen-presenting cells.
  • Nanoparticle 4, nanoparticle 5 and nanoparticle 7 can also detect cancer cell-specific T cells without the assistance of antigen-presenting cells, and the effect of nanoparticle 4 is significantly better than the other two nanoparticles. Moreover, the effect of nanoparticle 4 on detecting cancer cell-specific T cells without the assistance of antigen-presenting cells was better than that of nanoparticle 7 on detecting cancer cell-specific T cells with the assistance of antigen-presenting cells. Moreover, there is little difference in the cancer cell-specific T cells that can be detected by Nanoparticle 4, Nanoparticle 5 and Nanoparticle 7 with or without the assistance of antigen-presenting cells.
  • Nanoparticles prepared by using antigen-presenting cells activated by nanoparticles loaded with cancer cell whole-cell antigens are conducive to better detection of cancer cell-specific T cells; and the effect of mixed antigen-presenting cells of DC and B cells is better than Single DC.
  • the method of detecting broad-spectrum cancer cell-specific T cells described in the present invention may not rely on antigen-presenting cells.
  • Nanoparticles or microparticles prepared by activated antigen-presenting cells have an advantage in detecting cancer cell-specific T cells.
  • Nanoparticles loaded with cancer cell whole cell antigens and activated by antigen-presenting cells are more effective in detecting cancer cell-specific T cells than nanoparticles loaded with four antigen peptides. Nanoparticles prepared by activated antigen-presenting cells are better at detecting cancer cells. Cell-specific T cells. This shows that the types of cancer cell-specific T cell clones that can be detected by nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with four neoantigen peptides are limited. Nanoparticles prepared by activated antigen-presenting cells loaded with nanoparticles loaded with cancer cell whole cell antigens can detect a wider spectrum of cancer cell-specific T cells, so the number of T cell clones that can be obtained after expansion is also broader. .
  • nanoparticles loaded with activated antigen-presenting cell membrane components and whole cell components of cancer cells 4 were used.
  • the fluorescence signal (mean fluorescence intensity, MFI) of the fluorescent probe connected to the IFN- ⁇ antibody that can be detected by activated cancer cell-specific T cells is stronger than that of nanoparticles simply loaded with whole cell components of cancer cells. 1.
  • the activated cancer cell-specific T cells can detect the fluorescent signal of the fluorescent probe connected to the IFN- ⁇ antibody.
  • the fluorescence signal detected by the cancer cell-specific T cells activated by nanoparticle 4 is stronger than that of the cancer cell-specific T cells activated by nanoparticle 1. , which shows that the cancer cell-specific T cells activated by nanoparticle 4 express more specific markers and are therefore easier to detect during detection.
  • the nanoparticles prepared by the antigen-presenting cells activated by the nanoparticles loaded with whole cell components according to the present invention can better detect cancer cell-specific T cells with the ability to recognize and kill cancer cells.
  • the cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen presenting cells and presented to the surface of the antigen presenting cells.
  • Specific T cells that can recognize the cancer cell whole cell antigens can recognize them.
  • Cancer cells are activated after whole-cell antigen epitopes and express specific surface markers. By analyzing the proportion of T cells that highly express specific surface markers through flow cytometry, we can know that the activated T cells can recognize and have killing efficacy. Number and proportion of cancer cell-specific T cells.
  • This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticles prepared by nanoparticle-activated antigen-presenting cells to detect cancer cell-specific T cells.
  • B16F10 melanoma tumor tissue and cancer cells are first lysed to prepare a mixture of water-soluble components (mass ratio 1:1) and a mixture of non-water-soluble components (mass ratio 1:1) of tumor tissue and cancer cells, And mix the water-soluble component mixture and the water-insoluble component mixture at a mass ratio of 1:1.
  • PLGA is used as the nanoparticle skeleton material
  • Poly(I:C) and CpG2006 are used as adjuvants to prepare nanoparticles loaded with lysate components, and then the nanoparticles are incubated with antigen-presenting cells for a period of time to activate antigen presentation. cells, and prepare antigen-presenting cells into nanoparticles to detect cancer cell-specific T cells.
  • B16F10 cells When collecting tumor tissue, 1.5 ⁇ 10 5 B16F10 cells were first subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. The tumor tissue was cut into sections. Grind, add an appropriate amount of pure water through a cell strainer and freeze and thaw repeatedly 5 times, and can be accompanied by ultrasound to destroy the lysed sample; when collecting the cultured B16F10 cancer cell line, first centrifuge to remove the medium, then wash twice with PBS and centrifuge Cancer cells were collected, resuspended in ultrapure water, frozen and thawed three times, and destroyed and lysed by ultrasound.
  • the lysate After the tumor tissue or cancer cells are lysed, centrifuge the lysate at 5000g for 5 minutes and take the supernatant to obtain the water-soluble component that is soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate.
  • the non-water-soluble components that are insoluble in pure water are converted into soluble in 8M urea aqueous solution.
  • the water-soluble components of the tumor tissue and the water-soluble components of the cancer cells are mixed at a mass ratio of 1:1; the water-insoluble components of the tumor tissue and the non-water-soluble components of the cancer cells are mixed at a mass ratio of 1:1. Mixing the water-soluble component mixture and the water-insoluble component mixture at a mass ratio of 1:1 is the source of the antigen raw material for preparing nanoparticles.
  • centrifuge Bifidobacterium longum at 5000g for 30 minutes, then discard the precipitate and collect the supernatant. Filter the supernatant with a 1 ⁇ m filter, ultrasonicate at 4°C for 5 minutes at 20W, and then centrifuge at 16000g for 90 minutes. , resuspend the pellet in PBS to form the collected bacterial outer vesicle membrane components, and then use Tween 80 aqueous solution to lyse and dissolve the bacterial membrane components.
  • nanoparticle 1 is prepared by the double emulsion method.
  • the molecular weight of the preparation material PLGA is 7KDa-17KDa.
  • the immune adjuvants used are poly(I:C) and CpG2006 and the adjuvants are encapsulated in the nanoparticles.
  • the preparation method is as mentioned above.
  • the double emulsion method is first used to load the lysis solution components and adjuvants inside the nanoparticles. Then 100 mg of nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of ultrapure solution containing 4% trehalose is used. Resuspend in water and freeze-dry for 48 hours before use.
  • the average particle size of nanoparticles 1 is about 250nm.
  • Each 1 mg of PLGA nanoparticles 1 is loaded with approximately 130 ⁇ g of protein or peptide components.
  • Each 1 mg of PLGA nanoparticles 1 carries 0.02 mg of poly(I:C) and CpG2006 immune adjuvant.
  • Nano Particle 1 is also called nanovaccine 1 when injected.
  • the preparation materials and preparation method of nanoparticle 2 are the same as nanoparticle 1.
  • the nanoparticle 2 is internally loaded with the antigen component prepared in step (1) and the 8M urea-dissolved bacterial outer vesicle membrane component prepared in step (2) at the same time, and the mass ratio of the two is 1:1.
  • the immune adjuvants used were poly(I:C) and CpG2006, and the adjuvants were encapsulated in nanoparticles.
  • the double emulsion method was first used to load the tumor tissue inside the nanoparticles with lysate components, bacterial external vesicle components and adjuvants.
  • nanoparticles were centrifuged at 10,000g for 20 minutes, and 10 mL containing 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 hours before use.
  • the average particle size of nanoparticles 2 is about 250nm.
  • Each 1 mg of PLGA nanoparticles 2 is loaded with approximately 130 ⁇ g of protein or peptide components.
  • Each 1 mg of PLGA nanoparticles 2 carries 0.02 mg of poly(I:C) and CpG2006 immune adjuvants each.
  • the preparation materials and preparation methods of nanoparticle 3 are the same as nanoparticle 1.
  • the nanoparticle 3 is simultaneously loaded with the antigen component prepared in step (1) and the bacterial outer vesicle membrane component dissolved in Tween 80 prepared in step (2), and the mass ratio of the two is 1:1.
  • the immune adjuvants used were poly(I:C) and CpG2006, and the adjuvants were encapsulated in nanoparticles.
  • the double emulsion method was first used to load the tumor tissue inside the nanoparticles with lysate components, bacterial external vesicle components and adjuvants.
  • nanoparticles 3 100 mg were centrifuged at 10,000g for 20 minutes, and 10 mL containing 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 hours before use.
  • the average particle size of nanoparticles 3 is about 250nm.
  • Each 1 mg of PLGA nanoparticles 3 is loaded with approximately 130 ⁇ g of protein or peptide components.
  • Each 1 mg of PLGA nanoparticles 3 is loaded with 0.02 mg of poly(I:C) and CpG2006 immune adjuvant.
  • blank nanoparticle 4 The preparation materials and methods of blank nanoparticle 4 are the same as nanoparticle 1, but blank nanoparticle 4 only carries the same amount of adjuvant and does not load any tumor tissue lysate components.
  • the particle size of the nanoparticles 4 is approximately 250 nm.
  • mice After the C57BL/6 mice were sacrificed, the spleens of the mice were removed, a single cell suspension of mouse splenocytes was prepared, and CD19 + B cells in the splenocytes were isolated using magnetic bead sorting.
  • 500 ⁇ g of nanoparticle 1, or 500 ⁇ g of nanoparticle 2, or 500 ⁇ g of nanoparticle 3, or 500 ⁇ g of nanoparticle 4 were incubated with B cells (10 million cells) in 15 mL of RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), albumin (50ng /mL), and CD80 antibody (10ng/mL).
  • the incubated B cells were collected by centrifugation at 400g for 5 minutes, then washed with PBS three times, resuspended in PBS water and sonicated at low power (10W) for 15 minutes. Then centrifuge the sample at 500g for 5 minutes and collect the supernatant. The supernatant is filtered through membranes with pore diameters of 30 ⁇ m, 10 ⁇ m, 5 ⁇ m, 0.45 ⁇ m, and 0.22 ⁇ m. The resulting filtrate sample is centrifuged at 18000g for 60 minutes and discarded. The supernatant and the pellet were resuspended in PBS to obtain nanoparticles.
  • the nanoparticles prepared by using antigen-presenting cells activated by nanoparticle 1 are nanoparticles 5, with a particle size of 110 nanometers;
  • the nanoparticles prepared by using antigen-presenting cells activated by nanoparticles 2 are nanoparticles 6, with a particle size of 110 nanometers.
  • the nanoparticles prepared using antigen-presenting cells activated by nanoparticle 3 are nanoparticles 7, with a particle size of 110 nanometers;
  • the nanoparticles prepared using antigen-presenting cells activated with nanoparticles 4 are nanoparticles 8, with a particle size of 110 nanometers nanometer.
  • mice Inoculate 1.5 ⁇ 10 5 B16F10 cells subcutaneously on the back of each C57BL/6 mouse on day 0, and subcutaneously inject 0.7 mg of PLGA prepared in step (3) into the mice on day 7, day 12, and day 17 respectively.
  • Nanoparticle 1 Nanovaccine 1
  • 100 ⁇ L PBS 100 ⁇ L PBS. The mice were sacrificed on the 21st day, the mouse spleens were removed and mouse spleen single cell suspension was prepared, and the CD3 + T cells in the spleen cells were sorted using magnetic bead sorting method.
  • the isolated T cells (4 million) and 100 ⁇ g of nanoparticles prepared in step (6) (nanoparticle 5, or nanoparticle 6, or nanoparticle 7, or nanoparticle 8) in 40 mL of high-glucose DMEM complete medium
  • the CCP was incubated for 48 hours (37°C, 5% CO 2 ), and then centrifuged at 400 g for 5 minutes to collect the cells.
  • flow cytometry was used to analyze the CD3 + IFN ⁇ + T cells in the incubated T cells, which were the The number and proportion of cancer cell-specific T cells specifically activated by cancer cell whole-cell antigens.
  • the cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen-presenting cells and presented to the surface of the antigen-presenting cell membrane.
  • the nanoparticles prepared by the antigen-presenting cells are loaded with the above-mentioned degraded and presented products.
  • the antigenic epitope can be recognized by cancer cell-specific T cells and activate cancer cell-specific T cells. After activation, they secrete killer cytokines.
  • IFN- ⁇ is the most important cytokine secreted by antigen-specific T cells that are activated after recognizing antigens.
  • CD3 + IFN- ⁇ + T cells analyzed using flow cytometry are cancer cell-specific T cells that can recognize and kill cancer cells.
  • the tumor growth rate of mice in the PBS control group was very fast, while the tumor growth rate of the nanoparticle-treated group was slower, which shows that the nanoparticles induce cancer cell-specific T cells and can control the growth of tumors.
  • Nanoparticle 8 could hardly activate cancer cell-specific T cells; while nanoparticle 5, nanoparticle 6, and nanoparticle 7 could detect more cancer cell-specific T cells.
  • the effect of Nanoparticle 6 is better than that of Nanoparticle 5 and Nanoparticle 7, which shows that the activated antigen-presenting cells after loading the bacterial outer vesicle components lysed and dissolved using appropriate methods into the nanoparticles are beneficial to the specific detection of cancer cells.
  • Sexual T cells are beneficial to the specific detection of cancer cells.
  • Example 4 Nanoparticles or microparticles detect cancer cell-specific T cells
  • 6M guanidine hydrochloride was first used to cleave the whole cell antigen of B16F10 melanoma cancer cells. Then, a micron particle system loaded with cancer cell whole cell antigens was prepared using PLGA as the micron particle skeleton material, CpG BW006 (B type), CPG2216 (A type) and Poly ICLC as immune adjuvants. After using micron particles to activate antigen-presenting cells, the antigen-presenting cells are prepared into nanoparticles or micron particles to detect cancer cell-specific T cells.
  • the cultured B16F10 melanoma cancer cell line was collected and centrifuged at 350g for 5 minutes, then the supernatant was discarded and washed twice with PBS, and then the cancer cells were resuspended and lysed with 6M guanidine hydrochloride.
  • the whole cell antigen of the cancer cells was lysed and dissolved in 6M Guanidine hydrochloride is the source of antigen raw materials for preparing micron particle systems.
  • the micron particles are prepared by the double emulsion method.
  • the molecular weight of PLGA, the material used to prepare micron particles 1, is 38KDa-54KDa, and the immune adjuvants used are CpG BW006, CPG2216 and Poly ICLC.
  • Poly ICLC is a toll-like receptor 3 agonist, while various CpGs are Toll-like receptor 9 agonists, and both Toll-like receptor 3 and Toll-like receptor 9 are located in the endosome membrane structure within the cell.
  • the lysate components and immune adjuvant were loaded into the micron particles, and then centrifuged at 10,000g for 15 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and then freeze-dried for 48 hours; before use, the particles were Resuspend in 7 mL of PBS and then add 3 mL of cancer cell lysate component (protein concentration 50 mg/mL) and incubate at room temperature for 10 min to obtain micron particles 1 loaded with lysate both inside and outside.
  • cancer cell lysate component protein concentration 50 mg/mL
  • the average particle size of the microparticles is about 2.50 ⁇ m, and the surface potential is about -2mV; each 1 mg of PLGA microparticles is loaded with approximately 140 ⁇ g of protein or peptide components, and the loaded CpG BW006 (Class B), CPG2216 (Class A) and Poly ICLC 0.02mg each.
  • control microparticle 2 The preparation materials and preparation methods of control microparticle 2 are the same, and the loaded immune adjuvants are CpG2336 (Class A), CPG2216 (Class A) and Poly ICLC.
  • the diameter of the control micron particles 2 is about 2.50 ⁇ m, and the surface potential is about -2mV.
  • Each 1 mg PLGA micron particle is loaded with approximately 140 ⁇ g of protein or peptide components.
  • Each 1 mg PLGA micron particle is loaded with CpG2336 (Class A) and CPG2216 (Class A). and Poly ICLC immune adjuvant are 0.02mg each.
  • control microparticle 3 The preparation materials and preparation methods of control microparticle 3 are the same, and the loaded immune adjuvants are CpG BW006 (category B) and CPG2216 (category A).
  • the adjuvant used in control microparticle 3 is 0.02 mg per 1 mg PLGA microparticle, the particle size is about 2.50 ⁇ m, and the surface potential is about -2mV.
  • Each 1 mg PLGA microparticle is loaded with approximately 140 ⁇ g of protein or peptide components.
  • Each 1 mg PLGA microparticle contains The loaded CpG BW006 (Category B) and CPG2216 (Category A) are 0.03mg each.
  • mice After the mice were killed, mouse lymph nodes and spleens were collected. The mouse lymph nodes or spleens were minced and ground, and filtered through a cell mesh to prepare a single cell suspension. After mixing the lymph node single cell suspension and the spleen single cell suspension, flow cytometry was used. CD19 + B cells and CD11c + DC were sorted by cytometry.
  • Micron particles (500 ⁇ g) loaded with whole cell components of cancer cells were incubated with prepared DCs (10 million) and B cells (10 million) in 20 mL high-glucose DMEM complete medium for 72 hours (37°C, 5% CO2 ), the incubation system contains granulocyte-macrophage colony-stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL ) and CD86 antibody (10ng/mL).
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • IL-2 500U/mL
  • IL-7 200U/mL
  • IL-12 200U/mL
  • CD86 antibody 10ng/mL
  • the incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors.
  • PBS 4°C phosphate buffer solution
  • the cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 22.5W) Ultrasonic for 1 minute. Then centrifuge the sample at 3000g for 15 minutes and collect the supernatant. Centrifuge the supernatant at 8000g for 15 minutes and collect the supernatant. Then collect the supernatant after centrifugation at 16000g for 90 minutes. Discard the supernatant and collect the pellet. Place the pellet in PBS. After resuspension, nanoparticles are obtained.
  • the nanoparticles prepared by the mixed antigen-presenting cells activated by micron particles 1 are nanoparticles 1, and the particle size is 110 nanometers; among them, the nanoparticles prepared by the mixed antigen-presenting cells activated by micron particles 2 are nanoparticles 2, and the particle size is 110 nanometers. Nano; the nanoparticles prepared by mixed antigen-presenting cells activated by micron particles 3 are nanoparticles 3, with a particle size of 110 nanometers.
  • Micron particles 4 the particle size is about 2.55 ⁇ m.
  • B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse.
  • days 10, 15, and 20 the mice were subjected to radiotherapy or injected with 100 ⁇ L PBS at the tumor site. The mice were sacrificed on the 24th day, and the peripheral blood of mice in each group was collected. Peripheral blood mononuclear cells (PBMC) were then prepared, and CD3 + T cells were sorted using magnetic bead sorting.
  • PBMC Peripheral blood mononuclear cells
  • the sorted T cells (5 million), nanoparticles 1-3 (100 ⁇ g) or microparticles 4 (100 ⁇ g) prepared in step (5) were incubated in 2 mL RPMI1640 complete medium for 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to detect CD3 + IFN- ⁇ + T cells in T cells, which are cancer cell-specific T cells activated by cancer cell whole cell antigens.
  • MFI mean fluorescence intensity
  • nanoparticles prepared from antigen-presenting cells activated by microparticles loaded with CpG adjuvant and Poly ICLC mixed adjuvant were better at detecting cancer cell-specific T cells than microparticles loaded with two CpG mixed adjuvants.
  • Nanoparticles prepared from activated antigen-presenting cells were better than those prepared using micron particles loaded with two type A CpG and PolyICLC mixed adjuvants.
  • Nanoparticles prepared from particle-activated antigen-presenting cells are more effective at loading whole cell components of cancer cells internally.
  • the analysis results of the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN- ⁇ + in CD3 + IFN- ⁇ + T cells were consistent with the above trend ( Figure 5b). This shows that the nanoparticles activated by antigen-presenting cells loaded with mixed adjuvants of two different toll-like receptors can produce better nanoparticles, and that nanoparticles containing class B CpG and Toll-like receptor 3 agonists are used as mixed adjuvants. Nanoparticles prepared by activating antigen-presenting cells with micron particles are more effective.
  • 8M urea was first used to lyse B16F10 melanoma tumor tissue and dissolve the tumor tissue lysate components. Then, PLGA was used as the nanoparticle skeleton material, and Poly(I:C), CpG2006 (B type) and CpGSL01 (B type) were used as immune adjuvants to prepare nanoparticles loaded with cancer cell whole cell antigens, and the nanoparticles were used to activate the antigens. After presenting the cells, the nanoparticles are prepared and then detected for cancer cell-specific T cells in the tumor tissue.
  • nanoparticles were prepared by solvent evaporation method.
  • the molecular weight of PLGA, the material used to prepare nanoparticle 1, is 7KDa-17KDa.
  • the immune adjuvants used are Poly(I:C), CpG2006 and CpGSL01, and the lysate components and adjuvants are encapsulated inside the nanoparticles.
  • the preparation method is as described above.
  • each 1 mg of PLGA nanoparticles is loaded with approximately 80 ⁇ g of protein or peptide components, and each 1 mg of PLGA nanoparticles uses Poly(I:C), CpG2006 and CpGSL01 is 0.02 mg each.
  • the preparation materials and preparation method of control nanoparticle 2 are the same as above.
  • the particle size is about 270nm. It is loaded with an equal amount of lysate components.
  • the loaded immune adjuvant is Poly(I:C). Each 1mg of PLGA is loaded with 0.06mg of Poly(I:C). .
  • the preparation materials and preparation method of control nanoparticle 3 are the same as above.
  • the particle size is about 270nm. It is loaded with equal amounts of lysate components.
  • the loaded immune adjuvants are Poly(I:C), CpG1585 (Class A) and CpG2216 (Class A). Each 1mg of PLGA loads 0.02mg each of Poly (I:C), CpG1585 (Class A) and CpG2216 (Class A).
  • mouse lymph nodes were removed to prepare mouse lymph node single cell suspension, and then flow cytometry was used to sort CD11c + DC and CD19 + B cells from the lymph node cell single cell suspension.
  • Nanoparticles 1 (500 ⁇ g), nanoparticles 2 (500 ⁇ g), or nanoparticles 3 (500 ⁇ g) loaded with cancer cell whole cell components were mixed with DCs (5 million cells) and B cells (5 million cells) in 20 mL of high-glucose DMEM.
  • the culture medium was incubated for a total of 72 hours (37°C, 5% CO 2 ); the incubation system contained granulocyte-macrophage colony-stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL- 7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL).
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • IL-2 500U/mL
  • IL- 7 200U/mL
  • IL-12 200U/mL
  • CD86 antibody 10ng/mL
  • nanoparticles 4 were prepared using antigen-presenting cells activated by nanoparticles 1, with a particle size of 150 nanometers; nanoparticles 5 were prepared using antigen-presenting cells activated with nanoparticles 2, and the particle size was 150 nanometers; The antigen-presenting cells activated by particle 3 were prepared as nanoparticles 6, with a particle size of 150 nanometers.
  • mice On day 0, 1.5 ⁇ 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. On days 8, 10, 12, 14, and 16, mice were injected subcutaneously with 100 ⁇ L. ⁇ PD-1 antibody (10 mg/kg) or 100 ⁇ L PBS. The mice were sacrificed on the 20th day, and the tumor tissues of the mice were collected respectively. The tumor tissues were cut into small pieces and filtered through a cell mesh to prepare a single cell suspension of tumor tissue, and then CD3 was sorted from it using a magnetic bead sorting method. + T cells.
  • the sorted T cells (500,000) were co-incubated with allogeneic B cells (2.5 million), the nanoparticles prepared in step (5) (100 ⁇ g), or in 20 mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ). Then centrifuge at 400g for 5 minutes, collect the supernatant, and analyze the concentration of IFN- ⁇ in the supernatant using ELISA.
  • cancer cell-specific T cells secrete killer substances such as IFN- ⁇ after being activated.
  • the concentration of this type of killer substance represents the amount of activated cancer-specific T cells and the strength of the killing ability of cancer cells.
  • Nanoparticles prepared by antigen-presenting cells activated by nanoparticles loaded with cancer cell whole cell antigens are better than that of two types of type A CpG with Poly(I:C) as a mixed adjuvant.
  • Nanoparticles prepared by mixing adjuvant nanoparticles or nanoparticles loading only Poly(I:C) as an adjuvant to activate antigen-presenting cells are better than that of two types of type A CpG with Poly(I:C) as a mixed adjuvant.
  • This example uses MC38 mouse colon cancer as a cancer model to illustrate how to use nanoparticles prepared from nanoparticle-activated antigen-presenting cells to detect a broad spectrum of cancer cell-specific T cells.
  • Colon cancer tumor tissue and lung cancer cancer cells are first lysed to prepare water-soluble components and water-insoluble components, and the antigen is first degraded into polypeptides using protease in vitro. In practical applications, other enzymes or other methods can also be used to first degrade the proteins in the whole cell fraction into peptides. Then prepare a mixture of water-soluble components (mass ratio 1:1) and non-water-soluble components (mass ratio 1:1), and mix the water-soluble component mixture and the non-water-soluble component mixture in a mass ratio of 1:1 mix.
  • PLA was used as the nanoparticle skeleton material
  • CpGM362, CPG1018 and Poly ICLC were used as immune adjuvants to prepare nanoparticles, and the nanoparticles were used to detect cancer cell-specific T cells in vitro.
  • the water-insoluble components of pure water are converted into soluble in 8M urea aqueous solution.
  • trypsin 0.5 mg/mL
  • chymotrypsin Chymotrypsin, 0.5 mg/mL
  • the water-soluble components from colon cancer tumor tissue and lung cancer cancer cells were mixed at a mass ratio of 1:1; the water-insoluble components dissolved in 8M urea were also mixed at a mass ratio of 1:1. Then, the water-soluble component mixture and the water-insoluble component mixture are mixed at a mass ratio of 1:1, and this mixture is the source of raw materials for preparing nanoparticles.
  • Collect BCG use 8M urea aqueous solution to cleave BCG, and then dissolve the cleavage components for later use.
  • nanoparticle 1 is prepared by solvent evaporation method.
  • the molecular weight of PLA, the material for preparing nanoparticle 1, is 20KDa.
  • the nanoparticles are loaded with tumor tissue and cancer cell lysate, bacterial lysate and immune adjuvant inside, and the tumor tissue and cancer cell lysate components are loaded on the surface.
  • the immune adjuvants used were CpGM362, CPG1018 and poly ICLC, and the adjuvants were loaded inside the nanoparticles.
  • the mass ratio of tumor tissue and cancer cell lysate to bacterial lysate used in preparing the nanoparticles was 1:1. The preparation method is as mentioned above.
  • the lysate mixture, bacterial lysate components and adjuvants are first loaded inside the nanoparticles using the double emulsion method. After loading the lysates and adjuvants inside, 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose and freeze-dried for 48 h. Before use, resuspend 20 mg of nanoparticles in 0.9 mL of PBS and mix and incubate at room temperature for 5 minutes with 0.1 mL of samples containing equal amounts of cancer cell and tumor tissue lysate mixture and bacterial lysate components (80 mg/mL). use.
  • the average particle size of nanoparticles 1 is about 290nm.
  • Each 1 mg of PLGA nanoparticles 1 is loaded with approximately 140 ⁇ g of protein or peptide components.
  • Each 1 mg of PLGA nanoparticles contains 0.04 mg each of CpGM362, CPG1018 and Poly ICLC immune adjuvant.
  • PBMC Peripheral blood mononuclear cells
  • CD11c + DC and CD19 + B cells were sorted from PBMC using flow cytometry.
  • BMDC and BMDM were used simultaneously as antigen-presenting cells.
  • the preparation method of BMDC is the same as in Example 1.
  • the preparation method of BMDM is as follows:
  • mice were anesthetized and sacrificed by dislocation.
  • the mice were disinfected with 75% ethanol.
  • a small opening was made on the back of the mouse with scissors.
  • the skin was directly torn open to the calf joint of the mouse by hand, and the foot joint and foot joint of the mouse were removed. skin.
  • Macrophage colony-stimulating factor stimulates bone marrow cells to differentiate into mononuclear macrophages at a concentration of 40ng/mL. After culturing for 8 days, the morphological changes of macrophages were observed under a light microscope. Digest and collect the cells after 8 days, use anti-mouse F4/80 antibody and anti-mouse CD11b antibody, incubate at 4°C for 30 minutes in the dark, and use flow cytometry to identify the proportion of successfully induced macrophages.
  • M-CSF Macrophage colony-stimulating factor
  • Nanoparticle 1 (1000 ⁇ g) was incubated with peripheral blood-derived DC (20 million) and BMDC (20 million) in RPMI1640 complete medium for 72 hours (37°C, 5% CO 2 ); the incubation system contained GM- CSF (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL), IL-12 (500U/mL) and CD80 antibody (10ng/mL).
  • GM- CSF 500U/mL
  • IL-2 500U/mL
  • IL-7 500U/mL
  • IL-12 500U/mL
  • CD80 antibody 10ng/mL
  • incubate nanoparticle 1 1000 ⁇ g
  • peripheral blood-derived DCs 10 million
  • BMDC 10 million
  • BMDM 10 million
  • B cells 10 million
  • the incubation system contains GM-CSF (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL), and IL-12 (500U/mL) and CD80 antibody (10ng/mL).
  • peripheral blood-derived DCs (20 million) and BMDCs (20 million) were collected by centrifugation at 400g for 5 minutes, and then the cells were washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors.
  • PBS 4°C phosphate buffer solution
  • the cells were resuspended in PBS water and treated in a high-pressure homogenizer (5000 bar) for 5 minutes.
  • the sample was then centrifuged at 2000g for 15 minutes and the supernatant was collected.
  • the supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected.
  • the supernatant was mixed with the nanoparticle 1 (50mg) prepared in step (3) at 4°C.
  • each C57BL/6 mouse was subcutaneously inoculated with 1.5 ⁇ 10 5 MC38 cells on the back.
  • the mice were subcutaneously injected with 100 ⁇ L of 1 mg PLGA nanoparticles 1 to activate the mice. Cancer cell-specific T cells in the body. The mice were sacrificed on day 24, and the draining lymph nodes and spleens of the mice were collected. Single-cell suspensions of draining lymph nodes and spleen cells were prepared and T cells were isolated from them using the magnetic bead method.
  • T cells (4 million), 200 ⁇ g of nanoparticles (nanoparticle 1, or nanoparticle 2, or nanoparticle 3), IL-2 (500 U/mL), IL-7 (500 U/mL) and IL- 15 (5000U/mL) was incubated in 5mL DMEM complete medium for 24 hours, and then flow cytometry was used to sort the incubated CD3 + IFN- ⁇ + T cells, which are cancer cell-specific cells activated by cancer cell whole cell antigens. T cells.
  • MFI mean fluorescence intensity
  • the obtained T cells (4 million), peripheral blood-derived DCs (1 million), BMDC (1 million), B cells (1 million), BMDM (1 million), and 200 ⁇ g of nanoparticles (nano Particle 1, or nanoparticle 3), IL-2 (500U/mL), IL-7 (500U/mL) and IL-15 (5000U/mL) were incubated in 5mL DMEM complete medium for 24 hours, and then flow cytometry was used CD3 + IFN- ⁇ + T cells after surgical sorting and incubation are cancer cell-specific T cells activated by cancer cell whole cell antigens. At the same time, the mean fluorescence intensity (MFI) of the IFN- ⁇ + -linked fluorescent probe in CD3 + IFN- ⁇ + T cells was analyzed.
  • MFI mean fluorescence intensity
  • Nanoparticle 3 outperforms Nanoparticle 1 and Nanoparticle 2.
  • nanoparticle 1 cannot be detected, but both nanoparticle 2 and nanoparticle 3 can be detected.
  • nanoparticle 1, nanoparticle 2 and nanoparticle 3 can all detect T cells.
  • nanoparticle 2 and nanoparticle 3 are better than nanoparticle 1, and nanoparticle 3 is better than nanoparticle 3.
  • Particle 2. even when detecting T cells without antigen-presenting cells, the effect of nanoparticle 2 is better than that of nanoparticles 1 and 2 when detecting T cells when they contain antigen-presenting cells.
  • the analysis results of average fluorescence intensity are consistent with the above trend. This shows that loading activated antigen-presenting cell membrane components on the particle surface and using mixed antigen-presenting cell membrane components can improve the effectiveness of nanoparticles or microparticles in detecting cancer cell-specific T cells.
  • This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how micron particles loaded with cancer cell whole cell antigens activate antigen-presenting cells and then are prepared into micron particles to detect cancer cell-specific T cells from peripheral immune organs.
  • the cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water, and then repeatedly frozen and thawed 5 times with ultrasound to lyse cancer cells.
  • the double emulsion method is used to prepare micron particles.
  • the molecular weight of the micron particle 1 framework material PLGA is 38KDa-54KDa, and the immune adjuvants used are CpG2395 (Category C), CpGM362 (Category C) and Poly (I:C).
  • the double emulsion method is used to prepare micron particles loaded with lysate components and adjuvants internally, and then 100 mg of micron particles are centrifuged at 9000g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and dried for 48 hours before use.
  • the average particle size of this microparticle system is about 2.5 ⁇ m, and the surface potential is about -6mV; each 1 mg of PLGA micron particles is loaded with approximately 110 ⁇ g of protein or polypeptide components, and each of CpG2395, CpGM362 and Poly(I:C) is loaded with 0.02 mg.
  • the preparation materials and preparation method of control micron particles 2 are the same as above, the particle size is about 2.5 ⁇ m, and the surface potential is about -6mV; every 1 mg of PLGA micron particles is loaded with approximately 110 ⁇ g of protein or peptide components, and every 1 mg of PLGA is loaded with CpG1585 (Class A), CpG2336 ( Class A) and Poly(I:C) 0.02mg each.
  • mice peripheral splenocytes were used. After the mice were sacrificed, the spleens were removed, and then a single cell suspension of mouse spleen cells was prepared, and the CD19 + B cells in the single cell suspension were sorted using magnetic bead sorting.
  • micron particles (800 ⁇ g) loaded with cancer cell whole cell antigen components were incubated with the B cells (10 million) prepared in step (3) in 15 mL high-sugar DMEM complete medium for 48 hours (37°C, 5% CO 2 ) , the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL).
  • GM-CSF 2000U/mL
  • IL-2 500U/mL
  • IL-7 200U/mL
  • IL-12 200U/mL
  • CD86 antibody 10ng/mL
  • the incubated B cells (10 million) were collected by centrifugation at 400g for 5 minutes, then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, and resuspended in PBS water at 4°C. Ultrasonicate at low power (20W) for 1 minute and then use a homogenizer at 1000rpm for 3 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected. The supernatant was mixed with the microparticles (60mg) prepared in step (2) and DSPE-PEG.
  • PBS 4°C phosphate buffer solution
  • micron particles prepared by using the antigen-presenting cell membrane component activated by micron particle 1 and micron particle 1 are micron particles 3, with a particle size of 2.6 ⁇ m; using the antigen-presenting cell membrane component activated by micron particle 2 and micron particle The micron particles prepared by the combined action of 2 are micron particles 4, with a particle size of 2.6 ⁇ m.
  • each BALB/c mouse was subcutaneously inoculated with 1 ⁇ 10 6 4T1 cells on the back.
  • the mice were subcutaneously injected with 100 ⁇ L of 1 mg PLGA micron particles 1 (micron vaccine 1). ) or 100 ⁇ L of PBS.
  • the mice were sacrificed on day 22, their spleens were collected, and a single cell suspension of splenocytes was prepared.
  • Spleen cell single cell suspension (6 million cells), DC2.4 (2 million cells) and micron particles (50 ⁇ g) were incubated in 2 mL DMEM complete medium for 72 hours (37°C, 5% CO 2 ), and then used flow CD3 + IFN- ⁇ + T cells, which are cancer cell-specific T cells activated by whole-cell antigens of cancer cells, are sorted out by cytometry.
  • micron particles prepared by micron particle-activated antigen-presenting cells can effectively detect more cancer cell-specific T cells in mice in the treatment group.
  • the effect of microparticles 3 prepared by using two types of CpG and Poly(I:C) as mixed adjuvants was better than that of two types of A CpG and Poly(I:C).
  • the micron particles 4 were prepared by mixing the micron particles 2 with the adjuvant to activate the antigen-presenting cells.
  • Mannose is used as an active target in the micron vaccine of this embodiment.
  • any target with the ability to target target cells such as CD32 antibodies, mannans, CD205 antibodies, and CD19 antibodies, can also be used.
  • This example uses mannose as the target to illustrate how to detect cancer cell-specific T cells in peripheral blood using nanoparticles prepared by activating antigen-presenting cells using active targeting nanoparticles.
  • the cultured Pan02 pancreatic cancer cells were collected and 10% octylglucoside was used to lyse the cancer cells and dissolve the whole cell antigens derived from the cancer cells.
  • the nanoparticle system was prepared using the double emulsion method.
  • the nanoparticle preparation materials are PLGA and mannose-modified PLGA, both of which have molecular weights of 7KDa-17KDa. When the two are used together to prepare nanoparticles with a target, the mass ratio is 4:1.
  • the immune adjuvants used were Poly(I:C) and CpG SL03. The preparation method is as described above. The lysate components and adjuvants are loaded into the nanoparticles using the double emulsion method. Then 100 mg of the nanoparticles are centrifuged at 10,000 g for 20 minutes and resuspended in 10 mL of ultrapure water containing 4% trehalose.
  • Nanoparticles 1 with the target is about 270nm.
  • Each 1 mg of PLGA nanoparticles is loaded with approximately 80 ⁇ g of protein and peptide components, including 0.04 mg each of Poly(I:C) and CpGSL03.
  • Nanoparticles 2 that are not loaded with adjuvants but have a mannose target are also about 270nm in size. They are prepared using the same amount of cell lysis components but do not contain any immune adjuvants.
  • Each 1 mg of PLGA nanoparticles is loaded with approximately 80 ⁇ g of protein and peptides. components.
  • This example uses BMDC and BMDM as antigen-presenting cells.
  • the preparation methods of BMDC and BMDM are the same as above.
  • Nanoparticle 1 (1000 ⁇ g) or nanoparticle 2 (1000 ⁇ g) were incubated with BMDC (10 million pcs), BMDM (10 million pcs) and IL-7 (500 U/mL) respectively in 15 mL high-glucose DMEM complete medium for 48 hours. (37°C, 5% CO 2 ).
  • BMDC 10 million pcs
  • BMDM 10 million pcs
  • IL-7 500U/mL
  • Both of the above incubation systems contain IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN- ⁇ (500U/mL) and CD80 antibody (10ng/mL ).
  • the one prepared using mixed antigen-presenting cells that have not been activated by nanoparticles is nanoparticle 3, with a particle size of 110 nanometers;
  • the one prepared using mixed antigen-presenting cells activated with nanoparticles 2 is nanoparticle 4, with a particle size of 110 nanometers.
  • Nanoparticle 5 was prepared using mixed antigen-presenting cells activated by nanoparticle 1, with a particle size of 110 nanometers.
  • mice On day 0, each C57BL/6 mouse was subcutaneously inoculated with 1 ⁇ 10 6 Pan02 pancreatic cancer cells on the back. On day 10, day 15, day 20, and day 27, mice were injected subcutaneously with 100 ⁇ L of 1 mg PLGA. Nanoparticles. The mice were sacrificed on day 24 and peripheral blood was collected. Peripheral blood mononuclear cells (PBMC) were prepared from the peripheral blood, and then CD3 + T cells were isolated from the PBMC using flow cytometry.
  • PBMC Peripheral blood mononuclear cells
  • T cells (5 million) were incubated with 100 ⁇ g of nanoparticles (nanoparticle 1, or nanoparticle 3, or nanoparticle 4, or nanoparticle 5) in DMEM high-glucose medium for 72 hours (37°C, 5% CO 2 ), the incubation system contains IL-2 (500U/mL) and IL-7 (500U/mL). Then flow cytometry is used to sort out CD3 + IFN- ⁇ + T cells from the incubated cells, which are cancer cell-specific T cells.
  • nanoparticles 4 and 5 are better than nanoparticles 1 and 3.
  • Nanoparticle 5 is better than Nanoparticle 4.
  • the prepared nanoparticles can effectively detect cancer cell-specific T cells, but the antigens activated by nanoparticles with adjuvants Nanoparticles prepared by presenting cells are more effective.
  • Example 9 Nanoparticles detect lung cancer cancer cell-specific T cells
  • This example illustrates the detection of cancer cell-specific T cells in mouse splenocytes using calcified nanoparticles.
  • other biomineralization technologies cross-linking, gelation and other modified particles can also be used.
  • mouse lung cancer tumor tissue was lysed with 8M urea (containing 200mM sodium chloride), dissolved and loaded into a nanoparticle system. After using the particles to activate antigen-presenting cells, the antigen-presenting cells were prepared into nanoparticles. Detection of cancer cell-specific T cells.
  • mice Inoculate 1 ⁇ 10 LLC mouse lung cancer cells on the back of female C57BL/6 mice aged 6 to 8 weeks. When the tumor volume reaches 1000 mm3 , the mice will be sacrificed. The mouse tumor tissue will be removed, cut into pieces and ground. Then filter it through a cell mesh to prepare a single cell suspension, use ultraviolet irradiation for 5 minutes and then heat at 80°C for 10 minutes, and then use 8M urea (containing 200mM sodium chloride) to lyse and dissolve the tumor tissue single cell suspension to obtain Cancer cell whole cell antigen.
  • 8M urea containing 200mM sodium chloride
  • the nanoparticles are loaded with cancer cell whole cell antigens inside and on the surface, and then the nanoparticles are biocalcified.
  • the nanoparticles are prepared by a solvent evaporation method.
  • the molecular weight of the nanoparticle preparation material PLGA is 7KDa-17KDa.
  • the immune adjuvants CpG2006 and Poly(I:C) are loaded inside the nanoparticles.
  • the preparation method is as follows. During the preparation process, the double emulsion method is first used to load the antigen inside the nanoparticles. After loading the lysis components inside, 100mg PLGA nanoparticles are centrifuged at 13000g for 20 minutes and resuspended in 18mL PBS.
  • This example uses BMDC and B as antigen-presenting cells.
  • the preparation method of BMDC is the same as in Example 1.
  • B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
  • Nanoparticles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ;
  • the incubation system contains cytokine component 1: IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN- ⁇ (500U/mL).
  • nanoparticles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); does not contain any cytokines or antibodies in the incubation system.
  • Nanoparticles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ;
  • the incubation system contains cytokine combination 2: IL-4 (500U/mL), IL-10 (200U/mL), IL-37 (200U/mL), and TGF- ⁇ (500U/mL).
  • the incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors.
  • PBS 4°C phosphate buffer solution
  • the cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 20W) Ultrasonic for 2 minutes.
  • the sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected.
  • the supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected.
  • the supernatant was filtered through a 0.45 ⁇ m membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, filter and concentrate.
  • the filtered and concentrated sample with the nanoparticles 1 prepared in step (2) and treat it with a high-pressure homogenizer (10000bar) for 3 minutes. Then centrifuge at 13000g for 30 minutes and discard the supernatant for collection. Precipitate and resuspend the precipitate in PBS to obtain nanoparticles.
  • the antigen-presenting cells containing cytokine combination 1 in the system are prepared as nanoparticles 2, with a particle size of 300 nanometers; when particles are used to activate antigen-presenting cells, the system does not contain cytokines.
  • the nanoparticles 3 prepared by the antigen-presenting cells have a particle size of 300 nanometers; the nanoparticles 4 prepared by the antigen-presenting cells containing the cytokine combination 2 in the system when activating the antigen-presenting cells using particles are nanoparticles 4 with a particle size of 300 nanometers. 300 nm.
  • each C57BL/6 mouse was subcutaneously inoculated with 0.5 ⁇ 10 6 LLC lung cancer cells on the back.
  • the mice were subcutaneously injected with 100 ⁇ L of 1 mg PLGA nanoparticles.
  • the mice were sacrificed and spleen cells were harvested and prepared into single cell suspension, and then flow cytometry was used to isolate CD45 + CD3 + T cells.
  • T cells (5 million) were incubated with nanoparticles 2 (100 ⁇ g) or nanoparticles 3 (100 ⁇ g) or nanoparticles 4 (100 ⁇ g) prepared by antigen-presenting cells in DMEM high-glucose medium for 12 hours (37°C, 5 %CO 2 ), the system contains IL-2 (500U/mL) and IL-7 (500U/mL) during the incubation process;
  • T cells (5 million) and nanoparticles 2 (100 ⁇ g) prepared from antigen-presenting cells were incubated in DMEM high-glucose medium for 12 hours (37°C, 5% CO2), and the incubation system did not contain any cytokines or Antibody;
  • incubate T cells (5 million), 10 million mixed antigen-presenting cells prepared in step (3) and nanoparticle 1 (100 ⁇ g) in DMEM high-glucose medium for 12 hours (37°C, 5% CO 2 )
  • the system contained IL-2 (500U/mL) and IL-7 (500U/mL) during the incubation process.
  • nanoparticles prepared from antigen-presenting cells activated by calcified nanoparticles could detect more cancer cell-specific T cells.
  • the system containing cytokine combination 1 is better than the system without cytokines and/or antibodies; when nanoparticles activate antigen-presenting cells, the system contains cytokines.
  • combination 1 or the system without cytokines is better than that of the system with cytokine combination 2; moreover, when the nanoparticles prepared by antigen-presenting cells are co-incubated with T cells, the effect of the system with cytokine combination 2 is better than that of the system without cytokine combination 2.
  • cytokines Contains cytokines.
  • Example 10 Nanoparticles detect cancer cell-specific T cells in melanoma
  • the supernatant part is the water-soluble component; the precipitate part uses 10% sodium deoxycholate aqueous solution to dissolve the non-water-soluble component.
  • the water-soluble component and the non-water-soluble component after dissolving sodium deoxycholate are miscible at a mass ratio of 1:1, which is the source of the antigen raw material for preparing the nanoparticle system.
  • the nanoparticles are prepared by the double emulsion method and have the ability to target dendritic cells.
  • the nanoparticle preparation materials used are PLGA and mannan-modified PLGA, both of which have molecular weights of 24KDa-38KDa. When used, the mass ratio of unmodified PLGA to mannan-modified PLGA is 9:1.
  • the immune adjuvants used are poly(I:C), CpG1018 and CpG2216.
  • the substance that increases lysosomal immune escape is KALA polypeptide (WEAKLAKALAKALAKHLAKALAKALKACEA), and the adjuvants and KALA polypeptide are encapsulated in nanoparticles.
  • the preparation method is as mentioned above.
  • the double emulsion method is first used to load the lysis solution components, adjuvants, and KALA polypeptide inside the nanoparticles. After loading the above components inside, 100 mg of the nanoparticles are centrifuged at 12,000g for 25 minutes. And resuspended in 10 mL of ultrapure water containing 4% trehalose and freeze-dried for 48 h.
  • the average particle size of the nanoparticles is about 250nm, and the surface potential is about -5mV; each 1 mg of PLGA nanoparticles is loaded with approximately 100 ⁇ g of protein or peptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I:C), CpG1018 and CpG2216 immune
  • the adjuvants are 0.02mg each, and the loaded KALA polypeptide is 0.05mg.
  • the preparation materials and methods of Nanoparticle 2 are the same. Its particle size is about 250nm, its surface potential is about -5mV, it does not load KALA polypeptide, and it loads equal amounts of adjuvants and cell lysis components.
  • the preparation materials and preparation methods of nanoparticle 3 are the same, about 250nm, and the surface potential is about -5mV; each 1 mg of PLGA nanoparticles is loaded with approximately 100 ⁇ g of protein and peptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I:C) 0.02mg, the loaded CpG1018 is 0.04mg, and the loaded KALA polypeptide is 0.05mg.
  • This example uses BMDC and BMDM as antigen-presenting cells.
  • the preparation methods of BMDC and BMDM are the same as above.
  • Nanoparticles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (10 million) and BMDM (10 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 );
  • the incubation system contains GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN - ⁇ (500U/mL) and CD80 antibody (10ng/mL).
  • the antigen-presenting cell membrane component activated by nanoparticle 1 and nanoparticle 1 were used to prepare nanoparticle 4, with a particle size of 260 nanometers; the antigen-presenting cell membrane component activated by nanoparticle 2 was used together with nanoparticle 2.
  • the nanoparticle 5 prepared by the interaction has a particle size of 260 nanometers; the nanoparticle 6 is prepared by using the antigen-presenting cell membrane component activated by the nanoparticle 3 and the nanoparticle 3 to interact with the nanoparticle 3, and the particle size is 260 nanometers.
  • mice Female C57BL/6 mice aged 6-8 weeks were selected and injected subcutaneously with 0.5 mg of PLGA nanoparticles (lysate-loaded group) on days 0, 7, 14, 21 and 28. points, Poly(I:C) and two CpG adjuvants and KALA polypeptide). The mice were sacrificed on day 32, and their peripheral blood and lymph nodes were collected. Gradient centrifugation was used to isolate PBMCs from the peripheral blood of mice. The lymph nodes were cut into small pieces and ground through a cell mesh to prepare a single cell suspension. The PBMCs were then mixed with the single cell suspension of lymph node cells. CD45 + CD3 + T cells were then sorted using magnetic bead sorting.
  • the sorted CD3 + T cells (5 million cells) were incubated with nanoparticles (40 ⁇ g) and IL-7 (10 ng/mL) in 2 mL RPMI1640 complete medium for 96 hours. Then flow cytometry is used to sort the CD3 + IFN- ⁇ + T cells in the incubated T cells, which are cancer cell-specific T cells that can recognize cancer cell whole cell antigens.
  • nanoparticles prepared by activated antigen-presenting cells loaded with nanoparticles loaded with whole cell components can detect more cancer cell-specific T cells.
  • the effect of nanoparticles 4 prepared by adding antigen-presenting cells activated by nanoparticles that increase lysosome escape substances is better than that of nanoparticles prepared by adding nanoparticles activated by antigen-presenting cells without adding lysosome escape substances 5; using two Nanoparticles prepared with CpG and Poly(I:C) as mixed adjuvants activated antigen-presenting cells.
  • Nanoparticles 4 are more effective than nanoparticles activated with only one CpG and Poly(I:C) mixed adjuvant. Presentation of Cell-Prepared Nanoparticles 6.
  • This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how to use microparticles loaded with cancer cell whole cell antigens to activate the antigen-presenting cell membrane component and microparticles prepared from cancer cell membrane components to detect cancer cell-specific T cell.
  • breast cancer cells are first inactivated and denatured, and then the cells are lysed, and octylglucoside is used to dissolve and lyse the non-water-soluble components in the cancer cells.
  • PLGA was used as the micron particle skeleton material
  • CpG2007, CpG1018, and Poly ICLC were used as immune adjuvants
  • polyarginine and polylysine were used as substances that enhance lysosomal escape to prepare whole cell antigens loaded with cancer cells.
  • Micron particles are used to activate antigen-presenting cells to prepare mixed micron particles based on antigen-presenting cell membrane components and cancer cell membrane components, and then the micron particles are used to detect cancer cell-specific T cells.
  • the cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water.
  • the obtained cancer cells were inactivated and denatured using ultraviolet light and high-temperature heating at 60°C for 5 minutes, and then ultrapure water was added and repeatedly frozen and thawed 5 times, supplemented by ultrasound to lyse the cancer cells.
  • the cell lysate was centrifuged at 5000g for 10 minutes, and the supernatant The liquid is the water-soluble component. Dissolve the precipitate with 10% octyl glucoside to become the original dissolved non-water-soluble component. Mix the water-soluble component and the non-water-soluble component at a mass ratio of 2:1 , which is the lysate component required to prepare micron particles.
  • the micron particles 1 are prepared using the double emulsion method.
  • the molecular weight of the micron particle skeleton material PLGA is 38KDa-54KDa.
  • the immune adjuvants used are CpG2007, CpG1018 and Poly ICLC.
  • the lysosomal escape increasing substance used is poly. Arginine and polylysine.
  • During preparation first use the double emulsion method to prepare microparticles internally loaded with lysate components, adjuvants and KALA polypeptides. After loading lysate and adjuvants internally, centrifuge 100mg of microparticles at 9000g for 20 minutes, and use 10mL containing 4% trehalose.
  • the average particle size of the micron particle 1 system is about 3.1 ⁇ m, and the surface potential of the micron particle system is about -7mV; every 1 mg of PLGA micron particles is loaded with approximately 110 ⁇ g of protein or peptide components, including 0.01 mg each of CpG2007, CpG1018 and Poly ICLC. 0.02mg each of arginine and polylysine.
  • This example uses BMDC and DC2.4 as antigen-presenting cells.
  • the preparation method of BMDC is the same as in Example 1.
  • Micron particles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (5 million cells) and DC2.4 cells (5 million cells) in 15 mL of high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contains GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL ), IFN- ⁇ (500U/mL) and CD80 antibody (10ng/mL).
  • GM-CSF 2000U/mL
  • M-CSF 2000U/mL
  • IL-2 500U/mL
  • IL-7 200U/mL
  • IL-12 200U/mL
  • IFN- ⁇ 500U/mL
  • CD80 antibody 10ng/mL
  • micron particles 1 were ultrasonicated at 10W for 10 seconds and incubated for 10 minutes, and then repeatedly co-extruded using a 5 ⁇ m membrane.
  • the extruded liquid was collected after centrifugation at 9000g for 120 minutes. The supernatant was discarded to collect the precipitate, and the precipitate was placed in PBS. After medium resuspension, micron particles 2 were obtained, with a particle size of 3.15 ⁇ m.
  • step (2) collect 20 million DCs after incubation by centrifugation at 400 g for 5 minutes, wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water and incubate at 4°C with low power (20W ) Ultrasound for 2 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was collected after centrifugation at 5000g for 10 minutes. The supernatant was repeatedly co-extruded through a 0.22 ⁇ m membrane, and then 30mg of step (2) was added.
  • PBS phosphate buffer solution
  • micron particles 1 were ultrasonicated at 10W for 10 seconds and incubated for 10 minutes, and then repeatedly co-extruded using a 5 ⁇ m membrane.
  • the extruded liquid was collected after centrifugation at 9000g for 120 minutes. The supernatant was discarded to collect the precipitate, and the precipitate was placed in PBS. After medium resuspension, micron particles 3 were obtained, with a particle size of 3.15 ⁇ m.
  • mice Select 6-8 week old female BALB/c mice and inoculate 2 ⁇ 10 6 4T1 breast cancer cells subcutaneously on the back of the mice on day 0; subcutaneously inject 0.3mg on days 7, 14 and 21 respectively.
  • PLGA micron particles1 On the 25th day, the mice were sacrificed, peripheral blood was collected, PBMC were isolated from the peripheral blood, and CD3 + T cells were sorted from the PBMC.
  • CD3 + T cells (1 million), 100 ⁇ g of microparticles 2 or microparticles 3 were incubated in 10 mL of RPMI1640 complete medium for 24 hours (37°C, 5% CO 2 ).
  • the cell membrane was stained with anti-mouse CD4 antibodies and anti-mouse CD8 antibodies modified with different fluorescent probes.
  • intracellular staining with anti-mouse granzyme B (Granzyme B) antibodies modified with fluorescent probes was performed.
  • CD8 + Granzyme B + T cells and CD4 + Granzyme B + T cells were cancer cell-specific T cells that can recognize cancer cell whole cell antigens.
  • Cancer cell-specific T cells will begin to synthesize and express killer substances after being activated by antigens.
  • Granzyme B is one of these killer substances and has the strongest activity in inducing cell apoptosis.
  • MFI mean fluorescence intensity
  • micron particles 2 prepared by mixing the antigen-presenting cell membrane components activated by micron particles and the cancer cell membrane components can better detect cancer cell-specific T cells than micron particles 3, and the cancer cells activated by them are better than those of micron particles 3.
  • Cell-specific T cells can synthesize more killer substances.
  • cancer cell-specific T cells activated by micron particles 2 can synthesize more killer substances, because they are easier to detect and the detection will be more accurate.
  • the cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water.
  • the obtained cancer cells were inactivated and denatured using ultraviolet light and high-temperature heating respectively, and then an 8M urea aqueous solution (containing 500mM sodium chloride) was used to lyse the cancer cells and dissolve the lysate components, which are the antigen components for preparing the micron particle system.
  • Centrifuge Lactobacillus acidophilus at 5000g for 30 minutes, then discard the precipitate and collect the supernatant. Filter the supernatant with a 1 ⁇ m filter membrane, and then centrifuge at 16000g for 90 minutes. The precipitate is treated with 8M urea aqueous solution (containing 500mM chloride). Sodium) cleaves and solubilizes bacterial outer vesicle components.
  • the double emulsion method is used to prepare micron particles.
  • the skeleton material of micron particle 1 is unmodified PLA and mannose-modified PLA, both with a molecular weight of 40KDa.
  • the ratio of unmodified PLA to mannose-modified PLA is 4:1.
  • the immune adjuvants used were CpG2006, CpG2216 and Poly ICLC, and the lysosomal escape-increasing substances used were arginine and histidine.
  • the mass ratio of cancer cell lysate components and bacterial external vesicle components used in the preparation of micron particles is 1:1.
  • the double emulsion method is first used to prepare micron particles that are internally loaded with cancer cell lysate components, bacterial external vesicle components, adjuvants, arginine and histidine. Then, 100mg of micron particles are centrifuged at 9000g for 20 minutes. Use 10 mL of ultrapure water containing 4% trehalose to resuspend and dry for 48 hours to obtain micron particles 1. The average particle size is about 1.5 ⁇ m. Each 1 mg of PLGA micron particles 1 is loaded with approximately 100 ⁇ g of protein or peptide components, including CpG2006 and CpG2216. and Poly ICLC 0.02mg each, containing 0.05mg each of arginine and histidine.
  • control micron particle 2 The preparation materials and preparation methods of control micron particle 2 are the same as those of micron particle 1.
  • the particle size is about 1.5 ⁇ m, and it is loaded with equal amounts of arginine, histidine and equal amounts of cancer cell lysate components and bacterial outer vesicle components. However, Does not contain any adjuvants.
  • This example uses BMDC, B cells and BMDM as antigen-presenting cells.
  • the preparation methods of BMDC and BMDM are the same as above.
  • B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
  • microparticles 1 or 2 were incubated with 40 million mixed antigen-presenting cells (containing 20 million BMDCs, 10 million B cells and 10 million BMDMs) in 15 mL of high-glucose DMEM complete medium for 48 hours. (37°C, 5% CO 2 ); the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN- ⁇ (500U/mL), CD80 antibody (10ng/mL) and CD40 antibody (20mg/mL).
  • mixed antigen-presenting cells containing 20 million BMDCs, 10 million B cells and 10 million BMDMs
  • Nanoparticles Use a high-pressure homogenizer (10,000 bar) to process the filtered and concentrated sample for 3 minutes. Then, centrifuge at 13,000g for 30 minutes. Discard the supernatant to collect the precipitate. Resuspend the precipitate in PBS. Nanoparticles. Among them, the mixed antigen-presenting cells activated using micron particles 1 were prepared as nanoparticles 1, with a particle size of 250 nanometers; the mixed antigen-presenting cells activated using micron particles 2 were prepared as nanoparticles 2, with a particle size of 250 nanometers.
  • step (2) Use a high-pressure homogenizer (10,000 bar) to treat the filtered and concentrated sample for 3 minutes, and then react it with 60 mg of micron particles 1 or 2 prepared in step (2) for 10 minutes before use.
  • the 2 ⁇ m filter membrane was repeatedly co-extruded, and then the extruded liquid was centrifuged at 10,000 g for 20 minutes, the supernatant was discarded, and the precipitate was collected. The precipitate was resuspended in PBS to obtain micron particles.
  • the mixed antigen-presenting cell membrane component activated by micron particle 1 and the micron particle 1 are combined to prepare micron particle 3, with a particle size of 1.6 ⁇ m; the mixed antigen-presenting cell membrane component activated by micron particle 2 and micron particle 2 are prepared by the combined action of micron particles 4, with a particle size of 1.6 ⁇ m.
  • mice that are 6-8 weeks old and subcutaneously inject 100 ⁇ L of micron particles containing 0.4 mg of PLGA prepared in step (2) on days 0, 7, 14, 21, and 28. 1.
  • the mice were sacrificed on day 32, peripheral blood and spleen were collected, and then a single cell suspension of PBMC and spleen cells was prepared and mixed, and then CD3 + T cells were sorted out using magnetic bead sorting method.
  • the sorted CD3 + T cells (2 million), 100 ⁇ g nanoparticles or microparticles (nanoparticle 1, or nanoparticle 2, or microparticle 3, or microparticle 4), DC2.4 cells (1 million)
  • 10mL RPMI1640 complete medium for a total of 48 hours (37°C, 5% CO 2 ).
  • the incubation system contains IL-2 (200U/mL), IL-7 (200U/mL), and IL-15 (200U/mL). and CD80 antibody (10ng/mL).
  • flow cytometry was used to analyze the CD3 + IFN- ⁇ + T cells in the incubated CD3 + T cells, which are cancer cell-specific T cells that can recognize cancer cell whole cell antigens.
  • MFI mean fluorescence intensity
  • the cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen-presenting cells and presented to the surface of the antigen-presenting cell membrane.
  • the nanoparticles prepared by the antigen-presenting cells are loaded with the above-mentioned degraded and presented products.
  • the antigenic epitope can be recognized by cancer cell-specific T cells and activate cancer cell-specific T cells. After activation, they secrete killer cytokines.
  • IFN- ⁇ is the most important cytokine secreted by antigen-specific T cells that are activated after recognizing antigens.
  • CD3 + IFN- ⁇ + T cells analyzed using flow cytometry are cancer cell-specific T cells that can recognize and kill cancer cells. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN- ⁇ in CD3+IFN- ⁇ +T cells was analyzed.
  • MFI mean fluorescence intensity
  • the proportion of cancer cell-specific T cells is related to the efficacy in the figure, which shows that the T cells detected using the particles of the present invention are cancer cell-specific T cells that can specifically recognize and kill cancer cells.
  • nanoparticle 1 is better than nanoparticle 2; microparticle 3 is better than microparticle 4.
  • Adjuvant micron particles are prepared by activating antigen-presenting cells.
  • micron particles 3 are better than nanoparticles 1
  • microparticles 4 are better than nanoparticles 2, which shows that solid particles loaded with cancer cell lysis components inside and activated antigen-presenting cell components on the surface detect cancer cell-specific T
  • the cellular effect is better than vesicle particles simply loaded with activated antigen-presenting cell components.
  • MFI mean fluorescence intensity
  • This example uses mouse colon cancer as a cancer model to illustrate how to detect cancer cell-specific T cells using nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with colon cancer whole-cell antigens.
  • 8M urea aqueous solution is first used to lyse colon cancer tumor tissue and dissolve the lysed components.
  • PLGA is used as the skeleton material
  • Poly(I:C) CpG2336 and CpG2006 are used as adjuvants
  • NH 4 HCO 3 is used as the adjuvant.
  • Add lysosomal escape substances to prepare nanoparticles use the nanoparticles to activate antigen-presenting cells, prepare the antigen-presenting cells into nanoparticles, and then use the nanoparticles to detect cancer cell-specific T cells.
  • the nanoparticles were prepared using the double emulsion method.
  • the preparation material of nanoparticles is PLGA with a molecular weight of 7KDa-17KDa, Poly(I:C) and CpG as adjuvants, NH 4 HCO 3 as a substance that increases lysosomal escape, and the adjuvants and NH 4 HCO 3 are loaded on the nanoparticles Within; the preparation method is as described above.
  • the lysis solution components and adjuvants are first loaded inside the nanoparticles, and then 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of ultrapure water containing 4% trehalose is used to rehydrate the nanoparticles.
  • each 1 mg of PLGA nanoparticles is loaded with approximately 90 ⁇ g of protein and polypeptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I :C), CpG2336 and CpG2006 immune adjuvant 0.02mg each, loaded with NH 4 HCO 3 0.01mg.
  • nanoparticle 2 The preparation materials and methods of nanoparticle 2 are the same as nanoparticle 1.
  • the particle size is about 260nm and the surface potential is about -7mV.
  • Each 1 mg PLGA nanoparticle is loaded with approximately 90 ⁇ g of protein and peptide components.
  • Each 1 mg PLGA nanoparticle is loaded with NH 4 HCO. 3 0.01mg, loaded with 0.03mg each of CpG2336 and CpG2006.
  • This example uses BMDC and B as antigen-presenting cells.
  • the preparation method of BMDC is the same as in Example 1.
  • B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
  • Nanoparticles (1000 ⁇ g) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ;
  • the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN- ⁇ (500U/mL) and CD80 antibody (10ng/mL) and CD40 antibody (20mg/mL).
  • the incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors.
  • PBS 4°C phosphate buffer solution
  • the cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 20W) Ultrasonic for 2 minutes.
  • the sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected.
  • the supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected.
  • the supernatant was filtered through a 0.45 ⁇ m membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, filter and concentrate.
  • the one prepared by using the antigen-presenting cells activated by the nanoparticle 1 and the nanoparticle 1 is nanoparticle 3, with a particle size of 300 nanometers; the one prepared by using the antigen-presenting cells activated by the nanoparticle 2 and the nanoparticle 2. is nanoparticle 4, with a particle size of 300 nanometers.
  • mice Female C57BL/6 mice aged 6-8 weeks were selected and subcutaneously injected with 100 ⁇ L of nanoparticles containing 0.5 mg PLGA (loaded lysate component, mixed adjuvant) on days 0, 7, 14, and 28. and substances that increase lysosomal escape) or 100 ⁇ L PBS. Mice were sacrificed on day 32, peripheral blood was collected and peripheral blood mononuclear cells (PBM C) were isolated, and then flow cytometry was used to sort CD3 + T cells from PBMC.
  • PLGA loaded lysate component, mixed adjuvant
  • PBM C peripheral blood mononuclear cells
  • This example uses enzyme-linked immunospot method (ELISPOT) to detect cancer cell-specific T cells.
  • ELISPOT enzyme-linked immunospot method
  • the mice injected with particles loaded with whole cell components contained more cancer cell-specific T cells, and the antigen-presenting cells activated by the nanoparticles of the present invention
  • the prepared nanoparticles can effectively detect cancer cell-specific T cells.
  • nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with mixed adjuvants, lysate components and lysosomal escape substances are more effective in assisting in the isolation of cancer cell-specific T cells than those loaded with lysate components and both.
  • Nanoparticles of CpG adjuvants and lysosomal escape substances are more effective in assisting in the isolation of cancer cell-specific T cells than those loaded with lysate components and both.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Dispersion Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

A method and kit for detecting a cancer cell-specific T cell. The method comprises: co-incubating T cells in peripheral blood, peripheral immune organs or tumor-infiltrating lymphocytes with a particle prepared from an antigen-presenting cell activated by the particle loaded with whole cell components, so as to activate the cancer cell-specific T cell that can recognize and kill cancer cells, and then detecting and analyzing the number and proportion of the activated cancer cell-specific T cell by means of using techniques, such as ELISPOT, flow cytometry and ELISA, and thereby evaluating the strength of the cancer cell-specific immunity in patient. According to the present invention, the problem that the broad-spectrum polyclonal specific T cell in peripheral blood, peripheral immune organs or tumor-infiltrating lymphocytes cannot be effectively screened clinically at present is overcome, and the broad-spectrum effector cell-specific T cell having a specific killing function can be detected in cells. The particle has the characteristics of easiness to separate and obtain and a high specificity, and can be used as an effective biomarker.

Description

一种癌细胞特异性T细胞的检测方法及试剂盒A detection method and kit for cancer cell-specific T cells 技术领域Technical field
本发明涉及检测技术领域,尤其涉及一种癌细胞特异性T细胞的检测方法及试剂盒。The present invention relates to the field of detection technology, and in particular, to a detection method and kit for cancer cell-specific T cells.
背景技术Background technique
T细胞尤其是癌细胞特异性T细胞发挥着抗癌主力军的作用。T细胞是机体特异性识别和杀灭癌细胞的主要细胞,每一种癌细胞特异性T细胞的克隆可以特异性识别一种抗原表位。癌症患者体内尤其是经过免疫治疗或者放疗的患者体内都含有一定数量的癌细胞特异性T细胞。研究表明,经过免疫治疗等方法处理的癌症患者体内的癌细胞特异性T细胞的多少与癌症患者的预后正相关,因而检测癌症患者体内癌细胞特异性T细胞数量就显得尤为重要。发明人之前提出过使用负载癌细胞全细胞组分的纳米粒子或微米粒子辅助检测癌细胞特异性T细胞(申请号202011027741.0,肿瘤特异性T细胞的检测方法),但是在上述检测体系中需同时含有抗原提呈细胞(Antigen-presenting cells,APC),这就使得检测体系中同时含有多种细胞,而且抗原激活T细胞辅助检测的过程为间接过程而不是直接过程。为了解决上述问题,申请人提出本发明。T cells, especially cancer cell-specific T cells, play the main role in the fight against cancer. T cells are the main cells in the body that specifically recognize and kill cancer cells. Each clone of cancer cell-specific T cells can specifically recognize an antigenic epitope. Cancer patients, especially those who have undergone immunotherapy or radiotherapy, contain a certain number of cancer cell-specific T cells. Studies have shown that the number of cancer cell-specific T cells in cancer patients treated with immunotherapy and other methods is positively correlated with the prognosis of cancer patients. Therefore, it is particularly important to detect the number of cancer cell-specific T cells in cancer patients. The inventor has previously proposed using nanoparticles or microparticles loaded with whole cell components of cancer cells to assist in the detection of cancer cell-specific T cells (Application No. 202011027741.0, detection method of tumor-specific T cells), but in the above detection system it is necessary to simultaneously It contains Antigen-presenting cells (APC), which makes the detection system contain a variety of cells at the same time, and the process of antigen-activated T cells to assist detection is an indirect process rather than a direct process. In order to solve the above problems, the applicant proposes the present invention.
发明内容Contents of the invention
为解决上述技术问题,本发明提供了一种使用被负载癌细胞全细胞抗原的纳米粒子(NP)或微米粒子(MP)激活过的抗原提呈细胞制备的纳米粒子和/或微米粒子检测杀伤性(效应性)癌细胞特异性T细胞(T eff)的方法,该方法使用被激活的抗原提呈细胞制备的纳米粒子或微米粒子先激活癌细胞特异性T细胞,然后利用被激活的杀伤性癌细胞特异性T细胞(T eff)特异性表达的标志物分析待测细胞中癌细胞特异性T细胞的含量和比例。有效地解决了如何检测外周血、外周免疫器官或肿瘤浸润淋巴细胞中具有识别和杀伤癌细胞能力的广谱和多克隆的癌细胞特异性T细胞的问题。而且,由于用于检测癌细胞特异性T细胞的纳米粒子或微米粒子负载抗原提呈细胞膜组分,所以在使用本发明所述纳米粒子或微米粒子与T细胞共孵育时体系中可以没有抗原提呈细胞的辅助,是更直接的检测方法。 In order to solve the above technical problems, the present invention provides a method for detecting and killing nanoparticles and/or microparticles prepared by using antigen-presenting cells activated by nanoparticles (NP) or microparticles (MP) loaded with cancer cell whole cell antigens. The method of effective (effector) cancer cell-specific T cells (T eff ) uses nanoparticles or microparticles prepared by activated antigen-presenting cells to first activate cancer cell-specific T cells, and then uses the activated killing cells Markers specifically expressed by cancer cell-specific T cells (T eff ) analyze the content and proportion of cancer cell-specific T cells in the cells to be tested. It effectively solves the problem of how to detect broad-spectrum and polyclonal cancer cell-specific T cells with the ability to recognize and kill cancer cells in peripheral blood, peripheral immune organs or tumor-infiltrating lymphocytes. Moreover, since the nanoparticles or microparticles used to detect cancer cell-specific T cells are loaded with antigen-presenting cell membrane components, there may be no antigen-presenting cell membrane components in the system when the nanoparticles or microparticles of the present invention are used to co-incubate with T cells. With the help of cells, it is a more direct detection method.
本发明的第一个目的是提供一种由激活的抗原提呈细胞制备的粒子检测癌细胞特异性T细胞的方法,包括以下步骤:The first object of the present invention is to provide a method for detecting cancer cell-specific T cells using particles prepared from activated antigen-presenting cells, which includes the following steps:
S1、将抗原提呈细胞与第一粒子共孵育,得到激活后的抗原提呈细胞;其中,第一粒子为负载肿瘤组织和/或癌细胞全细胞组分的纳米粒子或微米粒子;S1. Co-incubate the antigen-presenting cells with the first particles to obtain activated antigen-presenting cells; wherein the first particles are nanoparticles or microparticles loaded with tumor tissue and/or cancer cell whole cell components;
S2、将激活后的抗原提呈细胞的细胞膜组分制备成纳米囊泡;或将激活后的抗原提呈细胞的细胞膜组分与第二粒子共作用,使细胞膜组分负载于第二粒子上,得到负载细胞膜组分的第二粒子;其中,第二粒子为负载肿瘤组织和/或癌细胞全细胞组分的纳米粒子或微 米粒子;S2. Preparing the activated cell membrane components of the antigen-presenting cells into nanovesicles; or cooperating the activated cell membrane components of the antigen-presenting cells with the second particles to load the cell membrane components on the second particles. , obtaining second particles loaded with cell membrane components; wherein the second particles are nanoparticles or microparticles loaded with tumor tissue and/or cancer cell whole cell components;
S3、将步骤S2的纳米囊泡和/或负载细胞膜组分的第二粒子与待测细胞共孵育,将其中可以识别抗原的广谱癌细胞特异性T细胞激活,然后使用适当检测技术分析被激活的癌细胞特异性T细胞内部或者表面的标志物,通过分析含有标志物的T细胞的数量和比例得到所述癌细胞特异性T细胞的数量和比例。S3. Incubate the nanovesicles and/or second particles loaded with cell membrane components in step S2 with the cells to be tested, activate broad-spectrum cancer cell-specific T cells that can recognize antigens, and then use appropriate detection technology to analyze the cells to be tested. Markers inside or on the surface of activated cancer cell-specific T cells are analyzed to obtain the number and proportion of the cancer cell-specific T cells by analyzing the number and proportion of T cells containing the markers.
进一步地,上述细胞内部或者细胞表面的标志物包括蛋白质或核酸。Furthermore, the above-mentioned markers inside cells or on cell surfaces include proteins or nucleic acids.
进一步地,特异性表面标志物为蛋白质时,包括但不限于干扰素-γ、白介素、颗粒酶、穿孔素、CD69、CD25、OX40(CD134)、CD39、CD103、CD56、CD279、CD278、CD244、CD27、CD154、TCF-1、CD137、CD44、CD28等。利用表面标志物分析检测癌细胞特异性T细胞数量和比例的技术包括但不限于流式细胞术、磁珠分选法、酶联免疫斑点法(ELISPOT)、酶联免疫吸附法(ELISA)等。Further, when the specific surface markers are proteins, they include but are not limited to interferon-γ, interleukins, granzymes, perforin, CD69, CD25, OX40 (CD134), CD39, CD103, CD56, CD279, CD278, CD244, CD27, CD154, TCF-1, CD137, CD44, CD28, etc. Technologies that use surface marker analysis to detect the number and proportion of cancer cell-specific T cells include but are not limited to flow cytometry, magnetic bead sorting, enzyme-linked immunospot assay (ELISPOT), enzyme-linked immunosorbent assay (ELISA), etc. .
进一步地,待测细胞可为T细胞或者含有T细胞的细胞混合物,如来源于外周血、外周免疫器官或肿瘤浸润淋巴细胞的T细胞或者含有T细胞的细胞混合物。Further, the cells to be tested may be T cells or a cell mixture containing T cells, such as T cells or a cell mixture containing T cells derived from peripheral blood, peripheral immune organs or tumor-infiltrating lymphocytes.
进一步地,待测细胞与S2的产物共孵育之前,可对其进行分选,分选出其中的T细胞,具体地,使用流式细胞术或者磁珠分选法从外周血、外周免疫组织、肿瘤浸润淋巴细胞中分选CD3 +的细胞、分选出CD45 +CD3 +的细胞、分选出CD3 +CD8 +的细胞、分选出CD45 +CD3 +CD8 +的细胞、分选出CD3 +CD4 +的细胞或分选出CD45 +CD3 +CD4 +的细胞。 Furthermore, before the cells to be tested are co-incubated with the products of S2, they can be sorted to sort out the T cells. Specifically, flow cytometry or magnetic bead sorting is used to select cells from peripheral blood and peripheral immune tissues. , sorting CD3 + cells from tumor infiltrating lymphocytes, sorting CD45 + CD3 + cells, sorting CD3 + CD8 + cells, sorting CD45 + CD3 + CD8 + cells, sorting CD3 + CD4 + cells or sort CD45 + CD3 + CD4 + cells.
进一步地,步骤S1的抗原提呈细胞与第一粒子的共孵育体系中可含有细胞因子或抗体。Furthermore, the co-incubation system of the antigen-presenting cells and the first particles in step S1 may contain cytokines or antibodies.
进一步地,在步骤S3中,S2的产物与待测细胞共孵育体系中可含有细胞因子或抗体。Further, in step S3, the co-incubation system of the product of S2 and the cells to be tested may contain cytokines or antibodies.
优选地,共孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF)、IL-2、IL-7和IL-12。Preferably, the co-incubation system contains granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-2, IL-7 and IL-12.
进一步地,细胞因子包括但不限于白介素2(IL-2)、白介素7(IL-7)、白介素14(IL-14)、白介素4(IL-4)、白介素15(IL-15)、白介素21(IL-21)、白介素17(IL-17)、白介素12(IL-12)、白介素6(IL-6)、白介素33(IL-33)、γ干扰素(IFN-γ)、TNF-α。Further, cytokines include, but are not limited to, interleukin 2 (IL-2), interleukin 7 (IL-7), interleukin 14 (IL-14), interleukin 4 (IL-4), interleukin 15 (IL-15), interleukin 21 (IL-21), interleukin 17 (IL-17), interleukin 12 (IL-12), interleukin 6 (IL-6), interleukin 33 (IL-33), gamma interferon (IFN-γ), TNF- α.
进一步地,抗体包括但不限于αCD-3抗体、αCD-4抗体、αCD-8抗体、αCD-28抗体、αCD-40抗体、αOX-40抗体、αOX-40L抗体。Further, antibodies include, but are not limited to, αCD-3 antibody, αCD-4 antibody, αCD-8 antibody, αCD-28 antibody, αCD-40 antibody, αOX-40 antibody, and αOX-40L antibody.
进一步地,在上述制备方法中,第一粒子或第二粒子上还可负载细菌组分或细菌外囊泡组分,该细菌组分或细菌外囊泡组分经含有裂解剂的裂解液裂解细菌或细菌外囊泡得到,裂解剂为尿素、盐酸胍、脱氧胆酸盐、十二烷基硫酸盐(如SDS)、甘油、蛋白质降解酶、白蛋白、卵磷脂、Triton、吐温、氨基酸、糖苷、胆碱,细菌包括但不限于卡介苗、大肠杆菌、长双歧杆菌、短双歧杆菌、乳双歧杆菌、嗜酸乳杆菌、格式乳杆菌、罗伊氏乳杆菌、鼠李糖乳杆菌等。Further, in the above preparation method, the first particle or the second particle can also be loaded with a bacterial component or a bacterial outer vesicle component, and the bacterial component or bacterial outer vesicle component is lysed by a lysis solution containing a lysis agent. Obtained from bacteria or bacterial external vesicles, the lysing agent is urea, guanidine hydrochloride, deoxycholate, dodecyl sulfate (such as SDS), glycerol, protein degrading enzyme, albumin, lecithin, Triton, Tween, amino acids , glycosides, choline, bacteria including but not limited to Bacillus Calmette-Guérin, Escherichia coli, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium lactis, Lactobacillus acidophilus, Lactobacillus formati, Lactobacillus reuteri, Lactobacillus rhamnosus Bacilli etc.
进一步地,被激活的抗原提呈细胞膜组分还可以与癌细胞细胞膜组分或癌细胞细胞外囊泡膜组分混合后制备成混合膜组分后制备成纳米囊泡或者负载于第二粒子表面。Furthermore, the activated antigen-presenting cell membrane component can also be mixed with the cancer cell cell membrane component or the cancer cell extracellular vesicle membrane component to prepare a mixed membrane component and then be prepared into nanovesicles or loaded on second particles. surface.
进一步地,第一粒子或第二粒子上还负载有免疫增强佐剂,免疫增强佐剂包括但不限于模式识别受体激动剂、卡介苗、卡介苗细胞壁骨架、卡介苗甲醇提取残余物、卡介苗胞壁酰二肽、草分枝杆菌、多抗甲素、矿物油、病毒样颗粒、免疫增强的再造流感病毒小体、霍乱肠毒素、皂苷及其衍生物、Resiquimod、胸腺素、新生牛肝活性肽、米喹莫特、多糖、姜黄素、免疫佐剂CpG、免疫佐剂poly(I:C)、免疫佐剂poly ICLC、短小棒状杆菌苗、溶血性链球菌制剂、辅酶Q10、左旋咪唑、聚胞苷酸、锰佐剂、铝佐剂、钙佐剂、细胞因子、白细胞介素、干扰素、聚肌苷酸、聚腺苷酸、明矾、磷酸铝、羊毛脂、角鲨烯、植物油、内毒素、脂质体佐剂、MF59、双链RNA、双链DNA、CAF01、人参、黄芪的有效成分等。Further, the first particle or the second particle is also loaded with an immune-enhancing adjuvant. The immune-enhancing adjuvant includes but is not limited to pattern recognition receptor agonist, BCG, BCG cell wall skeleton, BCG methanol extraction residue, BCG muramyl Dipeptides, Mycobacterium phlei, polyantigen, mineral oil, virus-like particles, immune-enhancing reconstituted influenza virions, cholera enterotoxin, saponins and their derivatives, Resiquimod, thymosin, neonatal bovine liver active peptide, Imiquimod, polysaccharide, curcumin, immune adjuvant CpG, immune adjuvant poly(I:C), immune adjuvant poly ICLC, Corynebacterium parvum vaccine, hemolytic streptococcus preparation, coenzyme Q10, levamisole, polycysteine Glycolic acid, manganese adjuvant, aluminum adjuvant, calcium adjuvant, cytokine, interleukin, interferon, polyinosinic acid, polyadenylic acid, alum, aluminum phosphate, lanolin, squalene, vegetable oil, internal Toxins, liposome adjuvants, MF59, double-stranded RNA, double-stranded DNA, CAF01, ginseng, active ingredients of astragalus, etc.
优选地,免疫增强佐剂包括(1)Poly(I:C)或Poly(ICLC);(2)CpG-ODN,其中,CpG-ODN为A类CpG-ODN、B类CpG-ODN和C类CpG-ODN中的至少两种,且至少其中一种为B类CpG-ODN或C类CpG-ODN。其中,A类CpG-ODN选自CpG-ODN 2216、CpG-ODN 1585或CpG-ODN 2336,B类CpG-ODN选自CpG-ODN 1018、CpG-ODN 2006、CpG-ODN 1826、CpG-ODN 1668、CpG-ODN 2007、CpG-ODN BW006或CpG-ODN SL01,C类CpG-ODN选自CpG-ODN 2395、CpG-ODN SL03或CpG-ODN M362。Preferably, the immune-enhancing adjuvant includes (1) Poly(I:C) or Poly(ICLC); (2) CpG-ODN, wherein CpG-ODN is type A CpG-ODN, type B CpG-ODN and type C At least two types of CpG-ODN, and at least one of them is type B CpG-ODN or type C CpG-ODN. Among them, type A CpG-ODN is selected from CpG-ODN 2216, CpG-ODN 1585 or CpG-ODN 2336, and type B CpG-ODN is selected from CpG-ODN 1018, CpG-ODN 2006, CpG-ODN 1826, CpG-ODN 1668 , CpG-ODN 2007, CpG-ODN BW006 or CpG-ODN SL01, Class C CpG-ODN is selected from CpG-ODN 2395, CpG-ODN SL03 or CpG-ODN M362.
进一步地,第一粒子或第二粒子上还负载有带正电荷的多肽(如KALA多肽、RALA多肽、蜂毒肽等)、精氨酸、聚精氨酸、赖氨酸、聚赖氨酸、组氨酸、聚组氨酸、NH 4HCO 3、鱼精蛋白或组蛋白等。 Furthermore, the first particle or the second particle is also loaded with positively charged polypeptides (such as KALA polypeptide, RALA polypeptide, melittin, etc.), arginine, polyarginine, lysine, and polylysine. , histidine, polyhistidine, NH 4 HCO 3 , protamine or histone, etc.
进一步地,第一粒子或第二粒子上还负载有主动靶向抗原提呈细胞的靶头,靶头可为甘露糖、甘露聚糖、CD19抗体、CD20抗体、BCMA抗体、CD32抗体、CD11c抗体、CD103抗体、CD44抗体等。Furthermore, the first particle or the second particle is also loaded with a target that actively targets antigen-presenting cells. The target may be mannose, mannan, CD19 antibody, CD20 antibody, BCMA antibody, CD32 antibody, or CD11c antibody. , CD103 antibody, CD44 antibody, etc.
进一步地,第一粒子或第二粒子可由以下材料制备得到:有机合成高分子材料包括但不限于PLGA、PLA、PGA、PEG、PCL、Poloxamer、PVA、PVP、PEI、PTMC、聚酸酐、PDON、PPDO、PMMA、聚氨基酸、合成多肽等;天然高分子材料包括但不限于卵磷脂、胆固醇、海藻酸盐、白蛋白、胶原蛋白、明胶、细胞膜成分、淀粉、糖类、多肽等;无机材料包括但不限于三氧化二铁、四氧化三铁、碳酸盐、磷酸盐等。Further, the first particle or the second particle can be prepared from the following materials: organic synthetic polymer materials including but not limited to PLGA, PLA, PGA, PEG, PCL, Poloxamer, PVA, PVP, PEI, PTMC, polyanhydride, PDON, PPDO, PMMA, polyamino acids, synthetic peptides, etc.; natural polymer materials include but are not limited to lecithin, cholesterol, alginate, albumin, collagen, gelatin, cell membrane components, starch, sugars, peptides, etc.; inorganic materials include But it is not limited to ferric oxide, ferric oxide, carbonate, phosphate, etc.
进一步地,第一粒子或第二粒子的粒径大小为纳米级或微米级,这样能保证粒子被抗原提呈细胞吞噬,而为了提高吞噬效率,粒径大小要在适宜的范围内。纳米粒子(Nanoparticle,NP)的粒径大小为1nm-1000nm,更优选地,粒径大小为30nm-1000nm,最优选地,粒径大小为100nm-600nm;微米粒子(Microparticle,MP)的粒径大小为1μm-1000μm,更优选地,粒径大小为1μm-100μm,更优选地,粒径大小为1μm-10μm,最优选地,粒径大小为1μm-5μm。Furthermore, the particle size of the first particle or the second particle is nanometer or micron, which can ensure that the particles are engulfed by the antigen-presenting cells. In order to improve the phagocytosis efficiency, the particle size should be within an appropriate range. The particle size of nanoparticles (NP) is 1nm-1000nm, more preferably, the particle size is 30nm-1000nm, most preferably, the particle size is 100nm-600nm; the particle size of microparticles (MP) The size is 1 μm-1000 μm, more preferably, the particle size is 1 μm-100 μm, more preferably, the particle size is 1 μm-10 μm, and most preferably, the particle size is 1 μm-5 μm.
进一步地,在步骤S2中,将被激活的抗原提呈细胞经过机械破坏、膜过滤或梯度离心制备得到纳米囊泡,或将被激活的抗原提呈细胞经过机械破坏、膜过滤或梯度离心,将产物与第二粒子共作用,得到负载细胞膜组分的第二粒子。Further, in step S2, the activated antigen-presenting cells are subjected to mechanical destruction, membrane filtration or gradient centrifugation to prepare nanovesicles, or the activated antigen-presenting cells are subjected to mechanical destruction, membrane filtration or gradient centrifugation, The product is combined with the second particle to obtain the second particle loaded with cell membrane components.
进一步地,机械破坏方式选自超声、均质化、匀浆、高速搅拌、高压破坏、高剪切力 破坏、溶胀、化学物质、皱缩中的一种或多种。共作用方式选自共孵育、共挤出、超声、搅拌、均质化和匀浆中的一种或多种,与纳米粒子或微米粒子共作用后抗原提呈细胞组分覆盖于原有纳米粒子或微米粒子表层形成新的纳米粒子或微米粒子。Further, the mechanical destruction method is selected from one or more of ultrasonic, homogenization, homogenization, high-speed stirring, high-pressure destruction, high-shear force destruction, swelling, chemical substances, and shrinkage. The co-action method is selected from one or more of co-incubation, co-extrusion, ultrasound, stirring, homogenization and homogenization. After co-action with nanoparticles or microparticles, the antigen-presenting cell components cover the original nanoparticles. New nanoparticles or microparticles are formed on the surface of particles or microparticles.
进一步地,将癌细胞或肿瘤组织在-20℃~-273℃下冷冻,加水或不含溶解剂的溶液后进行反复冻融裂解,得到的上清液为水溶性组分,沉淀中经溶解剂溶解后转为可溶的部分为非水溶性组分,水溶性组分和非水溶性组分合并后得到癌细胞全细胞组分。溶解剂选自尿素、盐酸胍、脱氧胆酸盐、十二烷基硫酸盐(如SDS)、甘油、蛋白质降解酶、白蛋白、卵磷脂、无机盐(0.1-2000mg/mL)、Triton、吐温、氨基酸、糖苷、胆碱中的至少一种。Further, the cancer cells or tumor tissues are frozen at -20°C to -273°C, and water or a solution without a dissolving agent is added, followed by repeated freeze-thaw lysis. The resulting supernatant is a water-soluble component, which is dissolved during the precipitation. After the agent is dissolved, the soluble part becomes the non-water-soluble component. The water-soluble component and the non-water-soluble component are combined to obtain the whole cell component of the cancer cells. The dissolving agent is selected from the group consisting of urea, guanidine hydrochloride, deoxycholate, dodecyl sulfate (such as SDS), glycerin, protein degrading enzyme, albumin, lecithin, inorganic salts (0.1-2000mg/mL), Triton, and methane. At least one of temperature, amino acid, glycoside and choline.
进一步地,抗原提呈细胞包括B细胞、树突状细胞(DC)和巨噬细胞中的至少一种,优选为包括DC在内的两种及以上,更优选为三种细胞的组合。Further, the antigen-presenting cells include at least one of B cells, dendritic cells (DC) and macrophages, preferably two or more including DC, and more preferably a combination of three types of cells.
进一步地,得到的癌细胞特异性T细胞包括CD4 +T细胞和/或CD8 +T细胞,优选为同时包括CD4 +T细胞和CD8 +T细胞。 Further, the obtained cancer cell-specific T cells include CD4 + T cells and/or CD8 + T cells, and preferably include both CD4 + T cells and CD8 + T cells.
本发明中,使用负载癌细胞全细胞抗原的纳米粒子和/或微米粒子先特异性激活抗原提呈细胞,再将抗原提呈细胞制备成纳米粒子或微米粒子,所制备成的纳米粒子或微米粒子就负载癌细胞全细胞抗原表位,然后使用由抗原提呈细胞制备而成的纳米粒子或微米粒子激活外周血、外周免疫组织或者肿瘤浸润淋巴细胞中的癌细胞特异性T细胞,再利用被激活的癌细胞特异性T细胞细胞内或者细胞表面高表达某些分子的特性,利用流式细胞术等手段分析最多样和广谱的具有识别和杀伤癌细胞功能的癌症特异性T细胞的数量和比例。In the present invention, nanoparticles and/or microparticles loaded with cancer cell whole cell antigens are used to specifically activate antigen-presenting cells, and then the antigen-presenting cells are prepared into nanoparticles or microparticles. The prepared nanoparticles or micron particles are The particles are loaded with whole-cell antigen epitopes of cancer cells, and then nanoparticles or microparticles prepared from antigen-presenting cells are used to activate cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor-infiltrating lymphocytes, and then used Characteristics of activated cancer cell-specific T cells that highly express certain molecules within the cell or on the cell surface. Use flow cytometry and other methods to analyze the most diverse and broad-spectrum cancer-specific T cells that have the function of recognizing and killing cancer cells. Quantity and proportion.
进一步地,抗原提呈细胞可以与癌细胞特异性T细胞来源于同体、同种异体,细胞系或由干细胞转化而来。Furthermore, the antigen-presenting cells can be derived from the same type as the cancer cell-specific T cells, allogeneic, cell lines, or transformed from stem cells.
进一步地,第一粒子或第二粒子中,用于制备抗原的癌细胞或肿瘤组织中至少有一种与所检测的癌细胞特异性T细胞对应的疾病类型相同。Further, in the first particle or the second particle, at least one of the cancer cells or tumor tissues used to prepare the antigen is the same as the disease type corresponding to the detected cancer cell-specific T cells.
本发明的第二个目的是提供一种用于检测癌细胞特异性T细胞的试剂盒,该试剂盒中包括以下(1)-(2)中的至少一种:The second object of the present invention is to provide a kit for detecting cancer cell-specific T cells, which kit includes at least one of the following (1)-(2):
(1)由激活的抗原提呈细胞制备的纳米囊泡;(1) Nanovesicles prepared from activated antigen-presenting cells;
(2)负载激活的抗原提呈细胞细胞膜组分的粒子;(2) Particles loaded with activated antigen-presenting cell membrane components;
其中,in,
所述纳米囊泡通过将抗原提呈细胞与第一粒子共孵育,得到激活后的抗原提呈细胞,再提取激活后的抗原提呈细胞的细胞膜组分制备得到;The nanovesicles are prepared by co-incubating the antigen-presenting cells with the first particles to obtain activated antigen-presenting cells, and then extracting the cell membrane components of the activated antigen-presenting cells;
所述负载激活的抗原提呈细胞细胞膜组分的粒子通过将激活后的抗原提呈细胞的细胞膜组分与第二粒子共作用,使所述细胞膜组分负载于第二粒子上得到;The particles loaded with cell membrane components of activated antigen-presenting cells are obtained by cooperating the cell membrane components of activated antigen-presenting cells with second particles, so that the cell membrane components are loaded on the second particles;
所述第一粒子或第二粒子分别独立地选自负载肿瘤组织和/或癌细胞全细胞组分的纳米粒子或微米粒子。The first particles or second particles are independently selected from nanoparticles or microparticles loaded with tumor tissue and/or whole cell components of cancer cells.
本发明突破现有检测方式的限制,使粒子上负载全部抗原和经激活的抗原提呈细胞细 胞膜,能够检测更广谱和多样的癌细胞特异性T细胞,且高度特异,在免疫检测中效果更佳,从而为免疫治疗提供了一种潜在的生物标志物检测方法。The present invention breaks through the limitations of existing detection methods, allowing particles to be loaded with all antigens and activated antigen-presenting cell membranes, capable of detecting a wider spectrum and variety of cancer cell-specific T cells, and is highly specific and effective in immune detection. Better, thus providing a potential biomarker detection method for immunotherapy.
借由上述方案,本发明至少具有以下优点:Through the above solutions, the present invention at least has the following advantages:
本发明提供了一种使用纳米级或微米级粒子递送系统体外检测免疫细胞中癌细胞特异性T细胞的技术,所分析检测的癌细胞特异性T细胞广谱而且高度特异,包含所有克隆的可以特异性识别和杀伤癌细胞效应性(杀伤性)癌细胞特异性T细胞(T eff);而且,与不负载激活的抗原提呈细胞制备的粒子相比,负载激活的抗原提呈细胞制备的粒子所激活的T细胞分泌的特异性标志物含量更高,这也就使得其更容易被检测出,避免了信号较差而无法检测出的情况。上述优势使得本发明所述粒子检测癌细胞特异性T细胞时避免了部分癌细胞特异性T细胞因为被激活后表达的特异性标志物较弱无法检测出的情况,因而本发明所述的检测方法的检测准确性更高。本发明还在此基础上对抗原提呈细胞激活过程、与T细胞孵育过程、第一粒子和第二粒子负载物质进行优化,使该方法能够检测出更全面的癌细胞特异性T细胞,且检测时信号更强更准确。 The present invention provides a technology for in vitro detection of cancer cell-specific T cells in immune cells using a nanoscale or micron-scale particle delivery system. The analyzed and detected cancer cell-specific T cells are broad-spectrum and highly specific, including all clones that can Specific recognition and killing of cancer cell effector (killer) cancer cell-specific T cells (T eff ); and, compared with particles prepared without loaded activated antigen-presenting cells, particles loaded with activated antigen-presenting cells The T cells activated by the particles secrete a higher content of specific markers, which makes them easier to detect and avoids the situation where the signal is poor and cannot be detected. The above advantages allow the particles of the present invention to detect cancer cell-specific T cells to avoid the situation where some cancer cell-specific T cells cannot be detected because the specific markers expressed after activation are weak. Therefore, the detection method of the present invention The detection accuracy of the method is higher. On this basis, the present invention also optimizes the activation process of antigen-presenting cells, the incubation process with T cells, and the loading materials of the first particle and the second particle, so that the method can detect more comprehensive cancer cell-specific T cells, and The signal is stronger and more accurate during detection.
上述说明仅是本发明技术方案的概述,为了能够更清楚了解本发明的技术手段,并可依照说明书的内容予以实施,以下以本发明的较佳实施例并配合详细附图说明如后。The above description is only an overview of the technical solutions of the present invention. In order to have a clearer understanding of the technical means of the present invention and implement them according to the contents of the description, the preferred embodiments of the present invention are described below with detailed drawings.
附图说明Description of the drawings
为了使本发明的内容更容易被清楚的理解,下面根据本发明的具体实施例并结合附图,对本发明作进一步详细的说明。In order to make the content of the present invention easier to understand clearly, the present invention will be described in further detail below based on specific embodiments of the present invention and in conjunction with the accompanying drawings.
图1为本发明细胞系统的制备过程及应用示意图;其中,a为水溶性组分和非水溶性组分分别收集和制备纳米粒子或微米粒子的示意图;b为采用含有溶解剂的溶解液溶解癌细胞全细胞抗原和制备纳米粒子或微米粒子的示意图;c为使用a或b中制备的纳米粒子和/或微米粒子激活抗原提呈细胞,并将被激活的抗原提呈细胞制备成粒子后用于检测癌细胞特异性T细胞的示意图;Figure 1 is a schematic diagram of the preparation process and application of the cell system of the present invention; a is a schematic diagram of collecting and preparing nanoparticles or microparticles from water-soluble components and non-water-soluble components respectively; b is dissolving using a dissolving solution containing a dissolving agent Schematic diagram of cancer cell whole cell antigen and preparation of nanoparticles or microparticles; c is after activating antigen-presenting cells using nanoparticles and/or microparticles prepared in a or b, and preparing the activated antigen-presenting cells into particles. Schematic diagram for detection of cancer cell-specific T cells;
图2-14分别为实施例1-13中用纳米粒子或微米粒子检测癌细胞特异性T细胞实验结果;*代表p≤0.05,有显著性差异。Figures 2-14 respectively show the experimental results of using nanoparticles or microparticles to detect cancer cell-specific T cells in Examples 1-13; * represents p≤0.05, indicating a significant difference.
具体实施方式Detailed ways
下面结合附图和具体实施例对本发明作进一步说明,以使本领域的技术人员可以更好地理解本发明并能予以实施,但所举实施例不作为对本发明的限定。The present invention will be further described below in conjunction with the accompanying drawings and specific examples, so that those skilled in the art can better understand and implement the present invention, but the examples are not intended to limit the present invention.
本发明所述的用于检测外周血、外周免疫组织或者肿瘤浸润淋巴细胞中的癌细胞特异性T细胞,该类癌细胞特异性T细胞在检测时先与激活过的抗原提呈细胞所制备的纳米粒子和/或微米粒子共孵育,然后使用流式细胞术或者酶联免疫斑点法(ELISPOT)或者酶联免疫吸附法等分析癌细胞特异性T细胞被抗原特异性激活后高表达的分子即可得到广谱的癌细胞特异性T细胞的信息。The present invention is used to detect cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor infiltrating lymphocytes. The cancer cell-specific T cells are first prepared with activated antigen-presenting cells during detection. Nanoparticles and/or microparticles are co-incubated, and then flow cytometry, enzyme-linked immunospot assay (ELISPOT), or enzyme-linked immunosorbent assay are used to analyze molecules that are highly expressed after cancer cell-specific T cells are specifically activated by antigens. A broad spectrum of cancer cell-specific T cell information can be obtained.
其中,用于制备纳米粒子或微米粒子的抗原提呈细胞先被负载肿瘤组织和/或癌细胞全细胞抗原或其混合物的纳米粒子和/或微米粒子激活。检测癌细胞特异性T细胞的过程及应 用领域如图1所示。Wherein, the antigen-presenting cells used to prepare nanoparticles or microparticles are first activated by nanoparticles and/or microparticles loaded with tumor tissue and/or cancer cell whole cell antigens or mixtures thereof. The process and application fields of detecting cancer cell-specific T cells are shown in Figure 1.
在制备激活抗原提呈细胞的纳米粒子或微米粒子时,可裂解细胞或组织后先分别收集水溶性组分和水不溶性抗原并分别制备纳米或微米粒子系统;或者也可以直接采用含有溶解剂的溶解液直接裂解细胞或组织并溶解癌细胞全细胞抗原并制备纳米或微米粒子系统。本发明所述癌细胞全细胞抗原在裂解前或(和)裂解后既可经过包括但不限于灭活或(和)变性、固化、生物矿化、离子化、化学修饰、核酸酶处理、蛋白酶内切或降解等处理后再制备纳米粒子或微米粒子;也可细胞裂解前或(和)裂解后不经过任何灭活或(和)变性、固化、生物矿化、离子化、化学修饰、核酸酶处理、蛋白酶内切或降解直接制备纳米粒子或微米粒子。本发明部分实施例中,肿瘤组织细胞在裂解前经过了灭活或(和)变性处理,在实际使用过程中也可以在细胞裂解后做灭活或(和)变性处理,或者也可以细胞裂解前和裂解后均做灭活或(和)变性处理;本发明部分实施例中细胞裂解前或(和)裂解后的灭活或(和)变性处理方法为紫外照射和高温加热,在实际使用过程中也可以采用包括但不限于放射线辐照、高压、固化、生物矿化、离子化、化学修饰、核酸酶处理、胶原酶处理、蛋白酶内切或降解、冷冻干燥等处理方法。本领域技术人员可以理解,在实际应用过程中技术人员可根据具体情况进行适当调整。When preparing nanoparticles or microparticles that activate antigen-presenting cells, the cells or tissues can be lysed and the water-soluble components and water-insoluble antigens can be collected separately to prepare nanoparticle or microparticle systems respectively; alternatively, a solution containing a dissolving agent can be directly used. The lysis solution directly lyses cells or tissues and dissolves whole cell antigens of cancer cells to prepare nano or micro particle systems. The cancer cell whole cell antigen of the present invention can be subjected to a process including but not limited to inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, nuclease treatment, protease treatment before or (and) after lysis. Nanoparticles or microparticles can be prepared after endolysis or degradation; or without any inactivation or (and) denaturation, solidification, biomineralization, ionization, chemical modification, nucleic acid treatment before or after cell lysis. Nanoparticles or microparticles are directly prepared by enzymatic treatment, proteolytic digestion or degradation. In some embodiments of the present invention, tumor tissue cells are inactivated or/and denatured before lysis. In actual use, they can also be inactivated or/and denatured after cell lysis, or the cells can also be lysed. Inactivation or (and) denaturation treatment is performed before and after lysis; in some embodiments of the present invention, the inactivation or (and) denaturation treatment method before or (and) after cell lysis is ultraviolet irradiation and high-temperature heating. In actual use Treatment methods including but not limited to radiation irradiation, high pressure, solidification, biomineralization, ionization, chemical modification, nuclease treatment, collagenase treatment, protease endolysis or degradation, freeze-drying and other treatment methods can also be used in the process. Those skilled in the art can understand that during actual application, the skilled person can make appropriate adjustments according to specific circumstances.
在将被激活的抗原提呈细胞制备成纳米粒子或微米粒子时,先对抗原提呈细胞进行机械破坏,然后使用离心和/或一定孔径的滤膜过滤,可选地,与纳米粒子或微米粒子共同作用。When preparing activated antigen-presenting cells into nanoparticles or micron particles, the antigen-presenting cells are first mechanically destroyed, and then centrifuged and/or filtered with a filter membrane of a certain pore size. Optionally, the antigen-presenting cells are combined with the nanoparticles or micron particles. Particles work together.
被激活的抗原提呈细胞在经过机械破坏后含有一定的细胞膜结构。Activated antigen-presenting cells contain a certain cell membrane structure after mechanical destruction.
被激活的抗原提呈细胞在经过机械破坏后,与纳米粒子或微米粒子共作用后所形成的新的纳米粒子或微米粒子中抗原提呈细胞的组分位于粒子外层。After the activated antigen-presenting cells are mechanically destroyed, they interact with nanoparticles or microparticles to form new nanoparticles or microparticles. The components of the antigen-presenting cells are located in the outer layer of the particles.
制备成纳米粒子或微米粒子的抗原提呈细胞以及用来加入与T细胞共孵育的抗原提呈细胞可以来源于自体或者同种异体,也可以来自于细胞系或者干细胞。抗原提呈细胞可以是DC细胞、B细胞、巨噬细胞或者上述三者的任意混合物,也可以是其他具有抗原提呈功能的细胞。The antigen-presenting cells prepared into nanoparticles or microparticles and the antigen-presenting cells used to incubate with T cells can be derived from autologous or allogeneic sources, or from cell lines or stem cells. Antigen-presenting cells can be DC cells, B cells, macrophages, or any mixture of the above three, or other cells with antigen-presenting functions.
在使用负载肿瘤组织和/或癌细胞全细胞组分激活抗原提呈细胞时,体系中可含有细胞因子和/或抗体以提高激活效率。When using tumor tissue and/or whole cell components of cancer cells to activate antigen-presenting cells, the system may contain cytokines and/or antibodies to improve activation efficiency.
在使用被激活的抗原提呈细胞制备成的纳米粒子和/或微米粒子激活癌细胞特异性T细胞时,体系中可含有细胞因子和/或抗体以提高激活效率。When using nanoparticles and/or microparticles prepared from activated antigen-presenting cells to activate cancer cell-specific T cells, the system can contain cytokines and/or antibodies to improve activation efficiency.
在一些实施方案中,采用负载癌细胞全细胞抗原的纳米粒子或微米粒子先激活抗原提呈细胞,再将抗原提呈细胞制备成纳米粒子或微米粒子,使用抗原提呈细胞制备的纳米粒子或微米粒子检测外周血、外周免疫组织或肿瘤浸润淋巴细胞中的癌细胞特异性T细胞的具体方法如下:In some embodiments, nanoparticles or microparticles loaded with cancer cell whole cell antigens are used to first activate antigen-presenting cells, and then the antigen-presenting cells are prepared into nanoparticles or microparticles, and the nanoparticles or microparticles prepared by the antigen-presenting cells are used. The specific method of using micron particles to detect cancer cell-specific T cells in peripheral blood, peripheral immune tissue or tumor-infiltrating lymphocytes is as follows:
步骤1,将第一预定体积的含有第一预定浓度的水相溶液加入第二预定体积的含有第二预定浓度制备粒子原材料的有机相中。Step 1: Add a first predetermined volume of an aqueous phase solution containing a first predetermined concentration to a second predetermined volume of an organic phase containing a second predetermined concentration of the raw material for preparing particles.
在一些实施例中,水相溶液可含有癌细胞/肿瘤组织裂解物中的各组分以及免疫增强佐剂;裂解物中的各组分在制备时分别为水溶性组分或是溶于含有尿素或盐酸胍等溶解剂的溶解液中的原非水溶性组分。水相溶液所含有的水溶性组分的浓度或原非水溶性组分的浓度,也即第一预定浓度要求蛋白质多肽浓度含量大于1ng/mL,能负载足够癌细胞全细胞抗原以激活相关细胞。免疫增强佐剂在初始水相中的浓度为大于0.01ng/mL。In some embodiments, the aqueous solution may contain each component of the cancer cell/tumor tissue lysate and an immune-enhancing adjuvant; each component of the lysate is a water-soluble component during preparation or is dissolved in a solution containing The original non-water-soluble components in the solution of dissolving agents such as urea or guanidine hydrochloride. The concentration of water-soluble components or the original concentration of non-water-soluble components contained in the aqueous phase solution, that is, the first predetermined concentration requires the concentration of protein polypeptide to be greater than 1ng/mL, and can load enough cancer cell whole cell antigens to activate related cells. . The concentration of the immune-enhancing adjuvant in the initial aqueous phase is greater than 0.01ng/mL.
在一些实施例中,有机溶剂选用二氯甲烷。另外,在一些实施例中,制备粒子原材料的第二预定浓度的范围为0.5mg/mL-5000mg/mL,优选为100mg/mL。In some embodiments, methylene chloride is used as the organic solvent. In addition, in some embodiments, the second predetermined concentration of raw materials for preparing particles ranges from 0.5 mg/mL to 5000 mg/mL, preferably 100 mg/mL.
实际中,有机相的第二预定体积根据其和水相的第一预定体积的比例进行设定,在本发明中,水相的第一预定体积和有机相的第二预定体积之比的范围为1:1.1-1:5000,优选地为1:10。在具体实施过程中可根据需要对第一预定体积、第二预定体积和第一预定体积与第二预定体积之比进行调整以调整制备的纳米粒或微米粒的尺寸大小。In practice, the second predetermined volume of the organic phase is set according to its ratio to the first predetermined volume of the aqueous phase. In the present invention, the range of the ratio of the first predetermined volume of the aqueous phase to the second predetermined volume of the organic phase is It is 1:1.1-1:5000, preferably 1:10. During the specific implementation process, the first predetermined volume, the second predetermined volume and the ratio of the first predetermined volume to the second predetermined volume can be adjusted as needed to adjust the size of the prepared nanoparticles or microparticles.
优选地,水相溶液为裂解物组分溶液时,其中蛋白质和多肽的浓度大于1ng/mL,优选1mg/mL~100mg/mL;水相溶液为裂解物组分/免疫佐剂溶液时,其中蛋白质和多肽的浓度大于1ng/mL,优选1mg/mL~100mg/mL,免疫佐剂的浓度大于0.01ng/mL,优选0.01mg/mL~20mg/mL。有机相溶液中,溶剂为DMSO、乙腈、乙醇、氯仿、甲醇、DMF、异丙醇、二氯甲烷、丙醇、乙酸乙酯等,优选二氯甲烷;有机相的浓度为0.5mg/mL~5000mg/mL,优选为100mg/mL。Preferably, when the aqueous phase solution is a lysate component solution, the concentration of protein and polypeptide is greater than 1 ng/mL, preferably 1 mg/mL ~ 100 mg/mL; when the aqueous phase solution is a lysate component/immune adjuvant solution, wherein The concentration of protein and polypeptide is greater than 1ng/mL, preferably 1mg/mL~100mg/mL, and the concentration of immune adjuvant is greater than 0.01ng/mL, preferably 0.01mg/mL~20mg/mL. In the organic phase solution, the solvent is DMSO, acetonitrile, ethanol, chloroform, methanol, DMF, isopropyl alcohol, dichloromethane, propanol, ethyl acetate, etc., preferably dichloromethane; the concentration of the organic phase is 0.5 mg/mL~ 5000mg/mL, preferably 100mg/mL.
步骤2,将步骤1得到的混合液进行大于2秒的超声处理或大于1分钟的搅拌或均质处理或微流控处理。优选地,搅拌为机械搅拌或者磁力搅拌时,搅拌速度大于50rpm,搅拌时间大于1分钟,比如搅拌速度为50rpm~1500rpm,搅拌时间为0.1小时~24小时;超声处理时,超声功率大于5W,时间大于0.1秒,比如2~200秒;均质处理时使用高压/超高压均质机或高剪切均质机,使用高压/超高压均质机时压力大于5psi,比如20psi~100psi,使用高剪切均质机时转速大于100rpm,比如1000rpm~5000rpm;使用微流控处理流速大于0.01mL/min,比如0.1mL/min-100mL/min。超声或者搅拌或者均质处理或者微流控处理进行纳米化和/或微米化,超声时间长短或搅拌速度或均质处理压力及时间能控制制备的微纳粒子大小,过大或过小都会带来粒径大小的变化。Step 2: subject the mixed solution obtained in Step 1 to ultrasonic treatment for more than 2 seconds or stirring or homogenization treatment or microfluidic treatment for more than 1 minute. Preferably, when the stirring is mechanical stirring or magnetic stirring, the stirring speed is greater than 50 rpm, and the stirring time is greater than 1 minute. For example, the stirring speed is 50 rpm ~ 1500 rpm, and the stirring time is 0.1 hour ~ 24 hours; during ultrasonic treatment, the ultrasonic power is greater than 5W, and the time Greater than 0.1 seconds, such as 2 to 200 seconds; use a high-pressure/ultra-high-pressure homogenizer or high-shear homogenizer for homogenization processing. When using a high-pressure/ultra-high-pressure homogenizer, the pressure is greater than 5 psi, such as 20 psi to 100 psi. Use a high-pressure homogenizer. The rotation speed of the shear homogenizer is greater than 100rpm, such as 1000rpm to 5000rpm; the flow rate of microfluidic processing is greater than 0.01mL/min, such as 0.1mL/min-100mL/min. Ultrasonic or stirring or homogenization treatment or microfluidic treatment can be used for nanonization and/or micronization. The length of ultrasonic time or stirring speed or homogenization pressure and time can control the size of the prepared micro-nano particles. Too large or too small will cause to changes in particle size.
步骤3,将步骤2处理后得到的混合物加入第三预定体积的含有第三预定浓度乳化剂的水溶液中并进行大于2秒的超声处理或大于1分钟的搅拌或进行均质处理或微流控处理。该步骤将步骤2得到的混合物加入到乳化剂水溶液中继续超声或搅拌纳米化或微米化。在本发明中,超声时间大于0.1秒,比如2~200秒,搅拌速度大于50rpm,比如50rpm~500rpm,搅拌时间大于1分钟,比如60~6000秒。优选地,搅拌为机械搅拌或者磁力搅拌时,搅拌速度大于50rpm,搅拌时间大于1分钟,比如搅拌速度为50rpm~1500rpm,搅拌时间为0.5小时~5小时;超声处理时,超声功率为50W~500W,时间大于0.1秒,比如2~200秒;均质处理时使用高压/超高压均质机或高剪切均质机,使用高压/超高压均质机时压力大于20psi,比如20psi~100psi,使用高剪切均质机时转速大于1000rpm,比如1000rpm~ 5000rpm;使用微流控处理流速大于0.01mL/min,比如0.1mL/min-100mL/min。超声或者搅拌或者均质处理或者微流控处理进行纳米化或微米化,超声时间长短或搅拌速度或均质处理压力及时间能控制制备的纳米或微米粒子大小,过大或过小都会带来粒径大小的变化。Step 3: Add the mixture obtained after step 2 to a third predetermined volume of aqueous solution containing a third predetermined concentration of emulsifier and perform ultrasonic treatment for more than 2 seconds or stirring for more than 1 minute or perform homogenization or microfluidic treatment. deal with. In this step, the mixture obtained in step 2 is added to the aqueous emulsifier solution and continued to be ultrasonically or stirred to form nanometers or micrometers. In the present invention, the ultrasonic time is greater than 0.1 seconds, such as 2 to 200 seconds, the stirring speed is greater than 50 rpm, such as 50 rpm to 500 rpm, and the stirring time is greater than 1 minute, such as 60 to 6000 seconds. Preferably, when the stirring is mechanical stirring or magnetic stirring, the stirring speed is greater than 50rpm, and the stirring time is greater than 1 minute. For example, the stirring speed is 50rpm to 1500rpm, and the stirring time is 0.5 to 5 hours; during ultrasonic treatment, the ultrasonic power is 50W to 500W. , the time is greater than 0.1 seconds, such as 2 to 200 seconds; when homogenizing, use a high-pressure/ultra-high-pressure homogenizer or high-shear homogenizer. When using a high-pressure/ultra-high-pressure homogenizer, the pressure is greater than 20 psi, such as 20 psi to 100 psi. When using a high-shear homogenizer, the rotation speed is greater than 1000rpm, such as 1000rpm ~ 5000rpm; when using microfluidic processing, the flow rate is greater than 0.01mL/min, such as 0.1mL/min-100mL/min. Ultrasonic or stirring or homogenization treatment or microfluidic treatment can be used to nanonize or micronize the particles. The length of ultrasonic time or stirring speed or homogenization process pressure and time can control the size of the prepared nano or micron particles. Too large or too small will cause Changes in particle size.
在一些实施例中,乳化剂水溶液为聚乙烯醇(PVA)水溶液,第三预定体积为5mL,第三预定浓度为20mg/mL。第三预定体积根据其与第二预定体积的比例进行调整。在本发明中,第二预定体积与第三预定体积之的范围为1:1.1-1:1000进行设定,优选地可以为2:5。在具体实施过程中为了控制纳米粒子或微米粒子的尺寸,可以对第二预定体积和第三预定体积之比进行调整。同样地,本步骤的超声时间或搅拌时间、乳化剂水溶液的体积以及浓度的取值根据,均为了得到尺寸大小合适的纳米粒或微米粒。In some embodiments, the emulsifier aqueous solution is a polyvinyl alcohol (PVA) aqueous solution, the third predetermined volume is 5 mL, and the third predetermined concentration is 20 mg/mL. The third predetermined volume is adjusted according to its ratio to the second predetermined volume. In the present invention, the range between the second predetermined volume and the third predetermined volume is set to 1:1.1-1:1000, preferably 2:5. In order to control the size of nanoparticles or microparticles during specific implementation, the ratio of the second predetermined volume and the third predetermined volume can be adjusted. Similarly, the ultrasonic time or stirring time, the volume and concentration of the emulsifier aqueous solution in this step are all based on obtaining nanoparticles or microparticles of suitable size.
步骤4,将步骤3处理后得到的液体加入第四预定体积的第四预定浓度的乳化剂水溶液中,并进行搅拌直至满足预定搅拌条件。Step 4: Add the liquid obtained after the treatment in Step 3 to a fourth predetermined volume of the emulsifier aqueous solution with a fourth predetermined concentration, and stir until the predetermined stirring conditions are met.
本步骤中,乳化剂水溶液为PVA溶液或其他溶液。In this step, the emulsifier aqueous solution is PVA solution or other solutions.
第四预定浓度为5mg/mL,第四预定浓度的选择,以得到尺寸大小合适的纳米粒或微米粒为依据。第四预定体积的选择依据第三预定体积与第四预定体积之比决定。在本发明中,第三预定体积与第三预定体积之比为范围为1:1.5-1:2000,优选地为1:10。在具体实施过程中为了控制纳米粒子或微米粒子的尺寸可以对第三预定体积和第四预定体积之比进行调整。The fourth predetermined concentration is 5 mg/mL, and the selection of the fourth predetermined concentration is based on obtaining nanoparticles or microparticles of suitable size. The selection of the fourth predetermined volume is determined based on the ratio of the third predetermined volume to the fourth predetermined volume. In the present invention, the ratio of the third predetermined volume to the third predetermined volume ranges from 1:1.5 to 1:2000, preferably 1:10. During specific implementation, the ratio between the third predetermined volume and the fourth predetermined volume can be adjusted in order to control the size of the nanoparticles or microparticles.
在本发明中,本步骤的预定搅拌条件为直至有机溶剂挥发完成,也即步骤1中的二氯甲烷挥发完成。In the present invention, the predetermined stirring condition of this step is until the volatilization of the organic solvent is completed, that is, the volatilization of methylene chloride in step 1 is completed.
步骤5,将步骤4处理满足预定搅拌条件的混合液在以大于100RPM的转速进行大于1分钟的离心后,去除上清液,并将剩下的沉淀物重新混悬于第五预定体积的第五预定浓度的含有冻干保护剂的水溶液中或者第六预定体积的PBS(或生理盐水)中。Step 5: After centrifuging the mixed liquid that meets the predetermined stirring conditions in Step 4 at a rotation speed of greater than 100 RPM for more than 1 minute, remove the supernatant, and resuspend the remaining sediment in a fifth predetermined volume of Five predetermined concentrations of an aqueous solution containing a lyoprotectant or a sixth predetermined volume of PBS (or physiological saline).
步骤6,将步骤5得到的含有冻干保护剂的混悬液进行冷冻干燥处理后,将冻干物质备用。Step 6: After freeze-drying the suspension containing the lyoprotectant obtained in Step 5, the freeze-dried material is used for later use.
步骤7,将第六预定体积的步骤5中得到的重悬于PBS(或生理盐水)中的含纳米粒的混悬液或者采用第六预定体积的PBS(或生理盐水)重悬步骤6得到的冷冻干燥后的含有纳米粒或微米粒和冻干保护剂的冻干物质直接使用;或者上述样品与第七预定体积的水溶性组分或者溶解的原非水溶性组分混合后使用。Step 7: Resuspend a sixth predetermined volume of the nanoparticle-containing suspension obtained in Step 5 in PBS (or physiological saline) or use a sixth predetermined volume of PBS (or physiological saline) to resuspend the nanoparticle-containing suspension obtained in Step 6 The freeze-dried substance containing nanoparticles or microparticles and a lyoprotectant is used directly; or the above sample is mixed with a seventh predetermined volume of water-soluble components or dissolved original non-water-soluble components and used.
在本发明中,第六预定体积与第七预定体积的体积比为1:10000到10000:1,优先体积比为1:100到100:1,最优体积比为1:30到30:1。In the present invention, the volume ratio of the sixth predetermined volume to the seventh predetermined volume is 1:10000 to 10000:1, the preferred volume ratio is 1:100 to 100:1, and the optimal volume ratio is 1:30 to 30:1 .
步骤8,将抗原提呈细胞与上述制备的纳米粒子和/或微米粒子共孵育一定时间。制备纳米粒子和/微米粒子的肿瘤组织和/或癌细胞与抗原提呈细胞可以来自于自体或者同种异体。Step 8: Incubate the antigen-presenting cells with the nanoparticles and/or microparticles prepared above for a certain period of time. The tumor tissue and/or cancer cells and antigen-presenting cells used to prepare nanoparticles and/or microparticles can be from autologous or allogeneic sources.
步骤9,收集共孵育后的细胞,进行超声、均质化、机械搅拌等机械破坏。Step 9: Collect the co-incubated cells and perform mechanical damage such as sonication, homogenization, and mechanical stirring.
步骤10,将超声完的样品进行离心和/或使用一定孔径的滤膜过滤和/或与负载癌细胞 全细胞组分的纳米粒子和/或微米粒子共同作用,制备得到基于抗原提呈细胞的纳米粒子或微米粒子。Step 10: Centrifuge the ultrasonicated sample and/or filter it with a filter membrane of a certain pore size, and/or interact with nanoparticles and/or microparticles loaded with whole cell components of cancer cells to prepare a sample based on antigen-presenting cells. Nanoparticles or microparticles.
步骤11,取得外周血、外周免疫组织或者肿瘤组织,收集上述组织中T细胞或含有T细胞的免疫细胞。以上外周血、外周免疫组织或者肿瘤组织可以来自于自体或者同种异体。Step 11: Obtain peripheral blood, peripheral immune tissue or tumor tissue, and collect T cells or immune cells containing T cells in the above tissues. The above peripheral blood, peripheral immune tissue or tumor tissue can be from autologous or allogeneic.
步骤12,将步骤10制备的纳米和/或微米粒子与步骤11得到的T细胞或含有T细胞的免疫细胞混合后共孵育一定时间。Step 12: Mix the nanoparticles and/or microparticles prepared in step 10 with the T cells or immune cells containing T cells obtained in step 11 and incubate them together for a certain period of time.
步骤13,采用流式细胞术、酶联免疫斑点法、酶联免疫吸附法或磁珠分选法等分析被抗原激活的癌细胞特异性T细胞的含量。Step 13: Use flow cytometry, enzyme-linked immunospot method, enzyme-linked immunosorbent assay, or magnetic bead sorting method to analyze the content of cancer cell-specific T cells activated by the antigen.
实施例1 纳米粒子用于检测外周免疫器官中的癌细胞特异性T细胞Example 1 Nanoparticles are used to detect cancer cell-specific T cells in peripheral immune organs
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米粒子激活的抗原提呈细胞制备的纳米粒子检测小鼠脾细胞中的癌细胞特异性T细胞。本实施例中,裂解B16F10黑色素瘤肿瘤组织以制备肿瘤组织的水溶性组分和非水溶性组分,然后,以有机高分子材料PLGA为纳米粒骨架材料,以Polyinosinic-polycytidylic acid(poly(I:C))为免疫佐剂采用溶剂挥发法制备负载有肿瘤组织的水溶性组分和非水溶性组分的纳米粒子系统,然后使用纳米粒子激活抗原提呈细胞,并将抗原提呈细胞机械破坏后离心制备纳米粒子,使用该纳米粒子辅助检测外周免疫器官中的癌细胞特异性T细胞。This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticles prepared by nanoparticle-activated antigen-presenting cells to detect cancer cell-specific T cells in mouse splenocytes. In this example, B16F10 melanoma tumor tissue was lysed to prepare water-soluble components and non-water-soluble components of the tumor tissue. Then, the organic polymer material PLGA was used as the nanoparticle skeleton material, and Polyinosinic-polycytidylic acid (poly(I :C)) For immune adjuvants, a solvent evaporation method is used to prepare a nanoparticle system loaded with water-soluble components and non-water-soluble components of tumor tissue, and then the nanoparticles are used to activate antigen-presenting cells, and the antigen-presenting cells are mechanically Nanoparticles are prepared by centrifugation after destruction, and the nanoparticles are used to assist in the detection of cancer cell-specific T cells in peripheral immune organs.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量超纯水并反复冻融5次,并伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性组分;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性组分转化为在8M尿素水溶液中可溶。以上即为制备纳米粒子系统的抗原原料来源。 1.5 × 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. The tumor tissue was cut into pieces and then ground. An appropriate amount of ultrapure water was added through a cell filter and frozen and thawed 5 times repeatedly, accompanied by ultrasound to destroy the lysed cells. After the cells are lysed, centrifuge the lysate at 5000g for 5 minutes and take the supernatant, which is the water-soluble component that is soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate and remove the insoluble component from the pure water. The water-insoluble components of water are converted into soluble in 8M urea aqueous solution. The above are the sources of antigen raw materials for preparing nanoparticle systems.
(2)负载全细胞组分的纳米粒子的制备(2) Preparation of nanoparticles loaded with whole cell components
本实施例中纳米粒子1采用溶剂挥发法中的复乳法制备。在制备时负载癌细胞全细胞抗原中水溶性组分的纳米粒子和负载癌细胞全细胞抗原中非水溶性组分的纳米粒子分别制备,然后使用时一起使用。所采用的纳米粒子制备材料PLGA分子量为24KDa-38KDa,所采用的免疫佐剂为poly(I:C)且poly(I:C)只分布于纳米粒子内部。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载细胞组分和佐剂,在内部负载细胞裂解组分后,将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子1平均粒径为280nm左右,每1mg PLGA纳米粒子约负载100μg蛋白质或多肽组分,每1mgPLGA纳米粒所使用的poly(I:C)免疫佐剂为0.02mg。In this embodiment, the nanoparticle 1 is prepared by the double emulsion method in the solvent evaporation method. During preparation, nanoparticles loaded with water-soluble components in whole cell antigens of cancer cells and nanoparticles loaded with non-water-soluble components in whole cell antigens of cancer cells are prepared separately, and then used together. The molecular weight of PLGA, the material used to prepare the nanoparticles, is 24KDa-38KDa. The immune adjuvant used is poly(I:C) and poly(I:C) is only distributed inside the nanoparticles. The preparation method is as described above. During the preparation process, the double emulsion method is first used to load cell components and adjuvants inside the nanoparticles. After loading the cell lysis components inside, 100 mg of nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of nanoparticles containing Resuspend in 4% trehalose ultrapure water and freeze-dry for 48 h. The average particle size of the nanoparticles 1 is about 280nm, and each 1 mg of PLGA nanoparticles is loaded with approximately 100 μg of protein or peptide components. The poly(I:C) immune adjuvant used per 1 mg of PLGA nanoparticles is 0.02 mg.
(3)骨髓来源的树突状细胞(BMDC)的制备(3) Preparation of bone marrow-derived dendritic cells (BMDC)
本实施例以从小鼠骨髓细胞制备树突状细胞为例来说明如何制备BMDC。首先,取1只6-8周龄C57小鼠颈椎脱臼处死,手术取出后腿的胫骨和股骨放入PBS中,用剪刀和镊子 将骨周围的肌肉组织剔除干净。用剪刀剪去骨头两端,再用注射器抽取PBS溶液,针头分别从骨头两端插入骨髓腔,反复冲洗骨髓到培养皿中。收集骨髓溶液,400g离心3min后加入1mL红细胞裂解液裂红。加入3mL RPMI 1640(10%FBS)培养基终止裂解,400g离心3min,弃上清。将细胞放置10mm培养皿中培养,使用RPMI 1640(10%FBS)培养基,同时加入重组小鼠GM-CSF(20ng/mL),37度,5%CO 2培养7天。第3天轻轻摇晃培养瓶,补充同样体积含有GM-CSF(20ng/mL)RPMI 1640(10%FBS)培养基。第6天,对培养基进行半量换液处理。第7天,收集少量悬浮及半贴壁细胞,通过流式检测,当CD86 +CD80 +细胞在CD11c +细胞中的比例为15-20%之间,诱导培养的BMDC即可被用来做下一步实验。 This example uses the preparation of dendritic cells from mouse bone marrow cells as an example to illustrate how to prepare BMDC. First, a 6-8 week old C57 mouse was sacrificed by cervical dislocation. The tibia and femur of the hind legs were surgically removed and placed in PBS. The muscle tissue around the bones was removed with scissors and tweezers. Use scissors to cut off both ends of the bone, and then use a syringe to draw the PBS solution. The needles are inserted into the bone marrow cavity from both ends of the bone, and the bone marrow is repeatedly flushed into the culture dish. Collect the bone marrow solution, centrifuge at 400g for 3 minutes, and then add 1 mL of red blood cell lysis solution to lyse the red blood. Add 3 mL of RPMI 1640 (10% FBS) medium to stop lysis, centrifuge at 400 g for 3 min, and discard the supernatant. The cells were placed in a 10 mm culture dish and cultured in RPMI 1640 (10% FBS) medium with recombinant mouse GM-CSF (20 ng/mL) added at 37 degrees Celsius and 5% CO 2 for 7 days. On the third day, shake the culture bottle gently and add the same volume of RPMI 1640 (10% FBS) medium containing GM-CSF (20ng/mL). On the 6th day, half of the culture medium was replaced. On day 7, collect a small amount of suspended and semi-adherent cells. Through flow cytometry, when the proportion of CD86 + CD80 + cells in CD11c + cells is between 15-20%, the induced cultured BMDC can be used for the following One step experiment.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载来源于肿瘤组织的癌细胞全细胞组分的纳米粒子(250μg负载水溶性组分的纳米粒子+250μg负载非水溶性组分的纳米粒子)与BMDC(1000万个)在15mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有细胞因子组合1:粒细胞-巨噬细胞集落刺激因子(GM-CSF,500U/mL)、IL-2(500U/mL)、IL-7(500U/mL)、IL-12(500U/mL)或者含有细胞因子组分2:GM-CSF(500U/mL)、IL-4(500U/mL)、肿瘤坏死因子α(TNF-α,500U/mL)、IL10(500U/mL)。 Nanoparticles loaded with whole cell components of cancer cells derived from tumor tissue (250 μg nanoparticles loaded with water-soluble components + 250 μg nanoparticles loaded with non-water-soluble components) were completely cultured with BMDC (10 million) in 15 mL RPMI1640. The base was incubated for a total of 48 hours (37°C, 5% CO 2 ); the incubation system contained cytokine combination 1: granulocyte-macrophage colony-stimulating factor (GM-CSF, 500U/mL), IL-2 (500U/mL) ), IL-7 (500U/mL), IL-12 (500U/mL) or containing cytokine component 2: GM-CSF (500U/mL), IL-4 (500U/mL), tumor necrosis factor α ( TNF-α, 500U/mL), IL10 (500U/mL).
(5)DC来源的纳米粒子的制备(5) Preparation of DC-derived nanoparticles
通过在400g离心5分钟收集添加细胞因子组分1的孵育后的DC(1000万个),然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在在4℃和7.5W下超声20分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品在2000g离心20分钟并收集上清液,将上清液在7000g离心20分钟后收集上清液,然后在15000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子2,纳米粒子2粒径为120纳米。Collect the incubated DCs (10 million) with the addition of cytokine component 1 by centrifugation at 400g for 5 minutes, then wash the cells twice with physiological saline, resuspend the cells in physiological saline, and incubate at 4°C and 7.5W. Sonicate for 20 minutes to disrupt cells and prepare samples containing cell membrane components. Then centrifuge the sample at 2000g for 20 minutes and collect the supernatant. Centrifuge the supernatant at 7000g for 20 minutes and collect the supernatant. Then collect the supernatant after centrifugation at 15000g for 120 minutes. Discard the supernatant and collect the pellet. Place the pellet in PBS. After resuspension, nanoparticles 2 are obtained, and the particle size of nanoparticles 2 is 120 nanometers.
或者收集步骤(3)制备的未经任何纳米粒子或微米粒子激活的DC(1000万个),然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在4℃和7.5W下超声20分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品在2000g离心20分钟并收集上清液,将上清液在7000g离心20分钟后收集上清液,将上清液与40mg步骤(2)制备的负载癌细胞全细胞组分的纳米粒子1(负载水溶性组分的纳米粒子20mg+负载非水溶性组分的纳米粒子20mg)共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液在15000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子3,粒径为300nm。Or collect the DCs (10 million) prepared in step (3) without any nanoparticle or microparticle activation, then wash the cells twice with physiological saline, resuspend the cells in physiological saline and incubate at 4°C and 7.5W. Sonicate for 20 minutes to disrupt cells and prepare samples containing cell membrane components. The sample was then centrifuged at 2000g for 20 minutes and the supernatant was collected. The supernatant was centrifuged at 7000g for 20 minutes and the supernatant was collected. The supernatant was mixed with 40 mg of nanoparticles loaded with cancer cell whole cell components prepared in step (2). Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 μm filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticle 3 with a particle size of 300 nm.
或者通过在400g离心5分钟收集添加细胞因子组分2的孵育后的DC(1000万个),然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在4℃和7.5W下超声20分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品在2000g离心20分钟并收集上清液,将上清液在7000g离心20分钟后收集上清液,将上清液与40mg步骤(2)制备的负载癌细胞全细胞组分的纳米粒子1(负载水溶性组分的纳米粒子20mg+负载非水溶性组分的纳米粒子20mg)共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液在15000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子。其中,使用纳米粒 子1与膜组分共孵育得到的为纳米粒子4,粒径为300nm。Alternatively, collect the incubated DCs (10 million) with the addition of cytokine component 2 by centrifugation at 400g for 5 minutes, then wash the cells twice with physiological saline, resuspend the cells in physiological saline, and incubate at 4°C and 7.5W. Sonicate for 20 minutes to disrupt cells and prepare samples containing cell membrane components. The sample was then centrifuged at 2000g for 20 minutes and the supernatant was collected. The supernatant was centrifuged at 7000g for 20 minutes and the supernatant was collected. The supernatant was mixed with 40 mg of nanoparticles loaded with cancer cell whole cell components prepared in step (2). Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 μm filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticles. Among them, the nanoparticle 4 obtained by incubating the nanoparticle 1 with the membrane component has a particle size of 300 nm.
或者通过在400g离心5分钟收集添加细胞因子组分1的孵育后的DC(1000万个),然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在4℃和7.5W下超声20分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品在2000g离心20分钟并收集上清液,将上清液在7000g离心20分钟后收集上清液,将上清液与40mg步骤(2)制备的负载癌细胞全细胞组分的纳米粒子1(负载水溶性组分的纳米粒子20mg+负载非水溶性组分的纳米粒子20mg)共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液在15000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子5,粒径为300nm。Alternatively, collect the incubated DCs (10 million) added with cytokine component 1 by centrifugation at 400 g for 5 minutes, then wash the cells twice with physiological saline, resuspend the cells in physiological saline, and incubate at 4°C and 7.5W. Sonicate for 20 minutes to disrupt cells and prepare samples containing cell membrane components. The sample was then centrifuged at 2000g for 20 minutes and the supernatant was collected. The supernatant was centrifuged at 7000g for 20 minutes and the supernatant was collected. The supernatant was mixed with 40 mg of nanoparticles loaded with cancer cell whole cell components prepared in step (2). Particle 1 (20 mg of nanoparticles loaded with water-soluble components + 20 mg of nanoparticles loaded with non-water-soluble components) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 μm filter membrane. The extrudate was centrifuged at 15,000g for 120 minutes. Collect and discard the supernatant, collect the precipitate, and resuspend the precipitate in PBS to obtain nanoparticle 5 with a particle size of 300 nm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
在每只C57BL/6小鼠背部皮下接种0.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取脾细胞。将小鼠脾细胞制备成单细胞悬液,然后从小鼠脾细胞单细胞悬液中使用流式细胞术分选出CD3 +T细胞。 0.5 × 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and splenocytes were harvested. Mouse splenocytes were prepared into a single cell suspension, and then CD3 + T cells were sorted from the mouse splenocyte single cell suspension using flow cytometry.
将纳米粒子1(50μg负载水溶性组分的纳米粒子+50μg负载非水溶性组分的纳米粒子)或者纳米粒子2(100μg)或者纳米粒子3(100μg)或者纳米粒子4(100μg)或者纳米粒子5(100μg)与来自脾细胞的T细胞(100万个)在10mL RPMI 1649完全培养基中共孵育72小时(37℃,5%CO 2)。然后收集细胞后在400g离心5分钟,将细胞在PBS中重悬后先采用Fc block处理T细胞以避免非特异性负载。然后采用CD3抗体,CD4抗体,CD8抗体对小鼠脾细胞进行细胞外染色,尔后固定细胞和对细胞进行破膜,并采用FN-γ抗体对T细胞进行细胞内染色。尔后采用流式细胞仪检测样本T细胞。分别分析CD4 +T细胞中被激活后可以分泌IFN-γ的T细胞在所有CD4 +T细胞中所占的比例和CD8 +T细胞中被激活后可以分泌IFN-γ的T细胞在所有CD8 +T细胞中所占的比例。以上CD4 +IFN-γ +T细胞和CD8 +IFN-γ +T细胞即为癌细胞特异性T细胞。 Nanoparticle 1 (50 μg nanoparticles loaded with water-soluble components + 50 μg nanoparticles loaded with non-water-soluble components) or nanoparticles 2 (100 μg) or nanoparticles 3 (100 μg) or nanoparticles 4 (100 μg) or nanoparticles 5 (100 μg) was incubated with splenocyte-derived T cells (1 million) in 10 mL of RPMI 1649 complete medium for 72 hours (37°C, 5% CO2 ). The cells were then collected and centrifuged at 400g for 5 minutes. The cells were resuspended in PBS and the T cells were first treated with Fc block to avoid non-specific loading. Then, CD3 antibody, CD4 antibody, and CD8 antibody were used for extracellular staining of mouse splenocytes, and then the cells were fixed and membrane-broken, and FN-γ antibody was used for intracellular staining of T cells. The sample T cells were then detected using flow cytometry. Analyze the proportion of CD4 + T cells that can secrete IFN-γ after activation among all CD4 + T cells and the proportion of CD8 + T cells that can secrete IFN-γ after activation among all CD8 + T cells. Proportion of T cells. The above CD4 + IFN-γ + T cells and CD8 + IFN-γ + T cells are cancer cell-specific T cells.
或者只将来自脾细胞的T细胞(100万个)在10mL RPMI 1649完全培养基中共孵育72小时(37℃,5%CO 2)。然后收集细胞后在400g离心5分钟,将细胞在PBS中重悬后先采用Fc block处理T细胞以避免非特异性负载。然后采用连接有特定荧光探针的抗小鼠CD3抗体,抗小鼠CD4抗体,抗小鼠CD8抗体对小鼠脾细胞进行细胞外染色,尔后固定细胞和对细胞进行破膜,并采用连接有荧光探针的抗小鼠IFN-γ抗体对T细胞进行细胞内染色。尔后采用流式细胞仪检测样本中含有IFN-γ抗体连接的荧光信号的T细胞。分别分析CD4 +T细胞中被激活后可以分泌IFN-γ的T细胞在所有CD4 +T细胞中所占的比例和CD8 +T细胞中被激活后可以分泌IFN-γ的T细胞在所有CD8 +T细胞中所占的比例。以上CD4 +IFN-γ +T细胞和CD8 +IFN-γ +T细胞即为癌细胞特异性T细胞。 Alternatively, only splenocyte-derived T cells (1 million) were incubated in 10 mL of RPMI 1649 complete medium for 72 hours (37°C, 5% CO 2 ). The cells were then collected and centrifuged at 400g for 5 minutes. The cells were resuspended in PBS and the T cells were first treated with Fc block to avoid non-specific loading. Then use anti-mouse CD3 antibody, anti-mouse CD4 antibody, and anti-mouse CD8 antibody connected with specific fluorescent probes to perform extracellular staining of mouse splenocytes. Then, the cells were fixed and membrane-broken, and the cells were fixed and membrane-broken using the anti-mouse CD4 antibody and anti-mouse CD8 antibody connected with specific fluorescent probes. Intracellular staining of T cells with fluorescently probed anti-mouse IFN-γ antibody. Flow cytometry is then used to detect T cells in the sample that contain fluorescent signals linked to IFN-γ antibodies. Analyze the proportion of CD4 + T cells that can secrete IFN-γ after activation among all CD4 + T cells and the proportion of CD8 + T cells that can secrete IFN-γ after activation among all CD8 + T cells. Proportion of T cells. The above CD4 + IFN-γ + T cells and CD8 + IFN-γ + T cells are cancer cell-specific T cells.
负载肿瘤组织和/或癌细胞全细胞组分的纳米粒子/微米粒子被抗原提呈细胞吞噬后,抗原会被降解成多肽抗原表位并与主要组织相容性复合物(MHC)分子结合后被提呈到抗原提呈细胞膜表面。由于纳米粒子和微米粒子负载的全细胞抗原可以交叉提呈释放,所以癌细胞抗原表位可以通过MHC I和MCH II两个途径被提呈到抗原提呈细胞膜表面。将 抗原提呈细胞细胞膜制备成纳米粒子或微米粒子后,其负载的MHC分子和抗原多肽复合物可以直接与T细胞表面的可以特异性识别癌细胞抗原的T细胞表面受体结合。而且,如果该T细胞为可以杀伤癌细胞的特异性T细胞,该癌细胞会开始分泌IFN-γ、颗粒酶等杀伤性物质,通过分析分泌杀伤性物质的T细胞,就可以得到具有癌细胞识别和杀伤能力的效应性癌细胞特异性T细胞的比例。After nanoparticles/microparticles loaded with tumor tissue and/or cancer cell whole cell components are engulfed by antigen-presenting cells, the antigen will be degraded into polypeptide epitopes and combined with major histocompatibility complex (MHC) molecules. Presented to the surface of antigen-presenting cell membranes. Since whole-cell antigens loaded with nanoparticles and microparticles can be cross-presented and released, cancer cell antigen epitopes can be presented to the surface of antigen-presenting cell membranes through two pathways, MHC I and MCH II. After the antigen-presenting cell membrane is prepared into nanoparticles or microparticles, the MHC molecules and antigen peptide complexes loaded therein can directly bind to T cell surface receptors on the surface of T cells that can specifically recognize cancer cell antigens. Moreover, if the T cells are specific T cells that can kill cancer cells, the cancer cells will begin to secrete killer substances such as IFN-γ and granzymes. By analyzing the T cells secreting killer substances, we can obtain the characteristics of cancer cells. Proportion of effector cancer cell-specific T cells with recognition and killing capabilities.
(8)实验结果(8)Experimental results
如图2所示,单独T细胞对照组和纳米粒子1检测组的癌细胞特异性T细胞很低。而纳米粒子2、纳米粒子3、纳米粒子4和纳米粒子5都能辅助检测到一定含量的癌细胞特异性T细胞。其中,纳米粒子5效果最好,纳米粒子5的效果优于纳米粒子2、纳米粒子3和纳米粒子4。纳米粒子5优于纳米粒子4说明在激活抗原提呈细胞过程中加入细胞因子组合1效果优于细胞因子组分2;纳米粒子5优于纳米粒子2说明辅助检测T细胞的纳米粒子表面负载抗原提呈细胞膜组分,内部为负载细胞组分的实心纳米粒子效果优于只是表面负载膜组分的纳米囊泡结构;纳米粒子4和纳米粒子5优于纳米粒子3说明被负载癌细胞全细胞组分的纳米粒子激活的抗原提呈细胞所制备的纳米粒子效果好于未被激活的抗原提呈细胞制备的纳米粒子。综上所述,本发明所述的粒子系统可以用于检测癌细胞特异性T细胞。被负载癌细胞全细胞组分的纳米粒子激活的抗原提呈细胞会降解和提呈所吞噬纳米粒子负载的癌细胞全细胞组分中的广谱全细胞抗原,被抗原提呈细胞提呈到细胞膜表面的癌细胞抗原表位已经与主要组织相容性复合物(MHC)分子结合。将上述抗原提呈细胞经过机械破坏后,抗原提呈细胞的细胞膜组分中含有与MHC结合的抗原表位。通过离心和/或使用一定孔径的滤膜过滤和/或与纳米粒子或微米粒子共作用,上述抗原提呈细胞中的细胞膜组分会形成纳米粒子或微米粒子,并负载了MHC分子和被降解提呈的癌细胞抗原表位,因而可以不经过抗原提呈细胞的辅助而直接激活癌细胞特异性T细胞用于检测该类细胞。As shown in Figure 2, the cancer cell-specific T cells in the T cell alone control group and the nanoparticle 1 detection group were very low. Nanoparticle 2, nanoparticle 3, nanoparticle 4 and nanoparticle 5 can all assist in detecting a certain amount of cancer cell-specific T cells. Among them, nanoparticle 5 has the best effect, and nanoparticle 5 has a better effect than nanoparticle 2, nanoparticle 3 and nanoparticle 4. Nanoparticle 5 is better than nanoparticle 4, indicating that the effect of adding cytokine combination 1 during the activation of antigen-presenting cells is better than cytokine component 2; nanoparticle 5 is better than nanoparticle 2, indicating that the surface of nanoparticles that assist in the detection of T cells is loaded with antigens. Presenting cell membrane components, the effect of solid nanoparticles loaded with cell components inside is better than that of nanovesicle structures that only load membrane components on the surface; Nanoparticles 4 and 5 are better than Nanoparticles 3, indicating that whole cells of cancer cells are loaded The nanoparticles prepared by the antigen-presenting cells activated by the nanoparticles of the component are more effective than the nanoparticles prepared by the unactivated antigen-presenting cells. In summary, the particle system of the present invention can be used to detect cancer cell-specific T cells. The antigen-presenting cells activated by the nanoparticles loaded with cancer cell whole cell components will degrade and present the broad-spectrum whole-cell antigens in the cancer cell whole cell components loaded with the phagocytosed nanoparticles, which are presented to Cancer cell antigen epitopes on the cell membrane surface have been bound to major histocompatibility complex (MHC) molecules. After the above-mentioned antigen-presenting cells are mechanically destroyed, the cell membrane components of the antigen-presenting cells contain antigen epitopes that bind to MHC. Through centrifugation and/or filtration using a filter membrane with a certain pore size and/or co-operation with nanoparticles or microparticles, the cell membrane components in the above antigen-presenting cells will form nanoparticles or microparticles, and be loaded with MHC molecules and degraded. Therefore, it can directly activate cancer cell-specific T cells for detecting this type of cells without the assistance of antigen-presenting cells.
实施例2 基于抗原提呈细胞的粒子用于癌细胞特异性T细胞的检测Example 2 Antigen-presenting cell-based particles are used for the detection of cancer cell-specific T cells
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米粒子激活的抗原提呈细胞制备纳米粒子辅助检测癌细胞特异性T细胞。本实施例中,裂解B16F10黑色素瘤肿瘤组织以制备肿瘤组织的水溶性组分和非水溶性组分,然后,以有PLGA为纳米粒骨架材料,以poly(I:C)和CpG1018为免疫佐剂采用溶剂挥发法制备负载有肿瘤组织的水溶性组分和非水溶性组分的纳米粒子系统,然后使用纳米粒子激活抗原提呈细胞,并将被激活的抗原提呈细胞制备成纳米粒子检测外周血免疫细胞中的癌细胞特异性T细胞。This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticle-activated antigen-presenting cells to prepare nanoparticles to assist in the detection of cancer cell-specific T cells. In this example, B16F10 melanoma tumor tissue was lysed to prepare water-soluble components and non-water-soluble components of the tumor tissue. Then, PLGA was used as the nanoparticle framework material, and poly(I:C) and CpG1018 were used as immune adjuvants. The agent uses a solvent evaporation method to prepare a nanoparticle system loaded with water-soluble components and non-water-soluble components of tumor tissue, then uses nanoparticles to activate antigen-presenting cells, and prepares the activated antigen-presenting cells into nanoparticle detection Cancer cell-specific T cells among peripheral blood immune cells.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性组分;在所得沉淀部分中加入8M尿素 溶解沉淀部分即可将不溶于纯水的非水溶性组分转化为在8M尿素水溶液中可溶。将水溶性组分和非水溶性组分按质量比1:1混合后即为制备纳米粒子系统的抗原原料来源。 1.5 × 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. Cut the tumor tissue into pieces and then grind it. Add an appropriate amount of pure water through a cell filter and freeze and thaw repeatedly 5 times. Ultrasound can be used to destroy the lysed cells. After the cells are lysed, centrifuge the lysate at 5000g for 5 minutes and take the supernatant, which is the water-soluble component that is soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate and remove the insoluble component from the pure water. The water-insoluble components of water are converted into soluble in 8M urea aqueous solution. Mixing water-soluble components and non-water-soluble components at a mass ratio of 1:1 is the source of antigen raw materials for preparing nanoparticle systems.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子采用溶剂挥发法制备。负载全细胞组分的纳米粒子1(NP1)制备材料PLGA分子量为7Da-17KDa,所采用的免疫佐剂为poly(I:C)和CpG1018且佐剂包载于纳米粒子内部。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载抗原和佐剂,在内部负载抗原(裂解组分)后,将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子1平均粒径为280nm左右;每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,每1mgPLGA纳米粒所使用的poly(I:C)和CpG1018免疫佐剂各0.02mg;纳米粒子1也称纳米疫苗1。本实施例中,采用等质量负载四种多肽新生抗原B16-M20(Tubb3,FRRKAFLHWYTGEAMDEMEFTEAESNM),B16-M24(Dag1,TAVITPPTTTTKKARVSTPKPATPSTD),B16-M46(Actn4,NHSGLVTFQAFIDVMSRETTDTDTADQ)和TRP2:180-188(SVYDFFVWL)的多肽纳米粒子2作为对照纳米粒子使用,其制备材料和制备方法同纳米粒子1,对照纳米粒2的粒径为280nm左右,负载100μg多肽组分,负载等量佐剂。空白纳米粒3的制备材料和制备方法同纳米粒子1,粒径为280nm左右,只负载等量的免疫佐剂却不负载任何抗原组分。In this example, nanoparticles were prepared by solvent evaporation method. The molecular weight of PLGA, the preparation material for nanoparticle 1 (NP1) loaded with whole cell components, is 7Da-17KDa. The immune adjuvants used are poly(I:C) and CpG1018, and the adjuvants are contained inside the nanoparticles. The preparation method is as described above. During the preparation process, the double emulsion method is first used to load the antigen and adjuvant inside the nanoparticles. After loading the antigen (cleavage component) inside, 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL containing Resuspend in 4% trehalose ultrapure water and freeze-dry for 48 h. The average particle size of the nanoparticles 1 is about 280nm; each 1 mg of PLGA nanoparticles is loaded with approximately 100 μg of protein and peptide components, and each 1 mg of PLGA nanoparticles uses 0.02 mg of poly(I:C) and CpG1018 immune adjuvant; nanoparticles 1 Also called nanovaccine1. In this example, four polypeptide neoantigens B16-M20 (Tubb3, FRRKAFLHWYTGEAMDEMEFTEAESNM), B16-M24 (Dag1, TAVITPPTTTTKKARVSTPKPATPSTD), B16-M46 (Actn4, NHSGLVTFQAFIDVMSRETTDTDTADQ) and TRP2:180-188 (SVYDFFVWL) were loaded with equal mass. Peptide nanoparticle 2 is used as a control nanoparticle. Its preparation materials and preparation methods are the same as nanoparticle 1. The particle size of control nanoparticle 2 is about 280 nm, and it is loaded with 100 μg of peptide component and an equal amount of adjuvant. The preparation materials and methods of blank nanoparticle 3 are the same as those of nanoparticle 1. The particle size is about 280 nm. It only carries the same amount of immune adjuvant but does not load any antigen component.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
采用骨髓来源的树突状细胞(BMDC)和B细胞作为抗原提呈细胞。BMDC的制备同实施例1。B细胞提取流程如下:处死小鼠后摘取小鼠脾脏,然后制备小鼠脾细胞单细胞悬液,然后使用磁珠分选法从脾细胞单细胞悬液中分选出CD19 +B细胞。将BMDC和B细胞按数量比1:1混合后作为混合抗原提呈细胞使用。 Bone marrow-derived dendritic cells (BMDC) and B cells were used as antigen-presenting cells. The preparation of BMDC is the same as in Example 1. The B cell extraction process is as follows: kill the mouse and remove the mouse spleen, then prepare a mouse splenocyte single cell suspension, and then use magnetic bead sorting to sort CD19 + B cells from the splenocyte single cell suspension. Mix BMDC and B cells at a ratio of 1:1 and use them as mixed antigen-presenting cells.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将纳米粒子1(500μg)或多肽纳米粒子2(500μg)或空白纳米粒3(500μg)+游离裂解液与2000万个混合抗原提呈细胞(1000万个BMDC+1000万个B细胞)在15mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有细胞因子组合:IL-15(500U/mL)、IL-2(500U/mL)、IL-7(500U/mL)、IL-12(1000U/mL)。 Mix nanoparticle 1 (500 μg) or peptide nanoparticle 2 (500 μg) or blank nanoparticle 3 (500 μg) + free lysate with 20 million mixed antigen-presenting cells (10 million BMDC + 10 million B cells) in 15 mL Incubate in RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contains a combination of cytokines: IL-15 (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL) mL), IL-12 (1000U/mL).
或者纳米粒子1与2000万个BMDC在15mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有细胞因子组合:IL-15(500U/mL)、IL-2(500U/mL)、IL-7(500U/mL)、IL-12(1000U/mL)。 Or nanoparticle 1 and 20 million BMDCs were incubated in 15mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contained a combination of cytokines: IL-15 (500U/mL), IL-2 ( 500U/mL), IL-7 (500U/mL), IL-12 (1000U/mL).
(5)基于抗原提呈细胞的纳米粒子的制备(5) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的2000万个混合抗原提呈细胞(1000万个BMDC+1000万个B细胞),然后使用生理盐水洗涤细胞两遍,将细胞重悬在生理盐水中后在4℃下使用低功率7.5W超声10分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品一次通过滤膜孔径为50μm、10μm、5μm、1μm、0.45μm、0.22μm的滤膜过滤,收集所得滤液后与相对应的步骤(2)制备的负载癌细胞全细胞组分的纳米粒子1(50mg)或者多肽 纳米粒子2(50mg)或者空白纳米粒子3(50mg)共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液并在15000g离心60分钟,弃去上清后使用生理盐水重悬所得沉淀即为纳米粒子。其中,使用纳米粒子1激活的混合抗原提呈细胞膜组分与纳米粒子1共作用后所制得的纳米粒子为纳米粒子4,粒径为300nm;使用多肽纳米粒子2激活的混合抗原提呈细胞膜组分与多肽纳米粒子2共作用后所制得的纳米粒子为纳米粒子5,粒径为300nm;使用空白纳米粒子3激活的混合抗原提呈细胞膜组分与空白纳米粒子3共作用后所制得的纳米粒子为纳米粒子6,粒径为300nm。Collect the incubated 20 million mixed antigen-presenting cells (10 million BMDC + 10 million B cells) by centrifugation at 400g for 5 minutes, then wash the cells twice with physiological saline, resuspend the cells in physiological saline, and Use low-power 7.5W ultrasonic for 10 minutes at 4°C to disrupt cells and prepare samples containing cell membrane components. Then, the sample is filtered once through a filter membrane with pore sizes of 50 μm, 10 μm, 5 μm, 1 μm, 0.45 μm, and 0.22 μm. The obtained filtrate is collected and combined with the nanometer nanoparticles loaded with whole cell components of cancer cells prepared in the corresponding step (2). Particle 1 (50 mg) or peptide nanoparticle 2 (50 mg) or blank nanoparticle 3 (50 mg) was incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 μm filter. The extruded liquid was centrifuged at 15000g for 60 minutes and discarded. After removing the supernatant, resuspend in physiological saline and the resulting precipitate is nanoparticles. Among them, the nanoparticles prepared by using the mixed antigen-presenting cell membrane components activated by nanoparticle 1 and nanoparticle 1 are nanoparticles 4, with a particle size of 300 nm; the mixed antigen-presenting cell membrane activated by peptide nanoparticles 2 The nanoparticles prepared by the interaction between the components and the polypeptide nanoparticles 2 are nanoparticles 5, with a particle size of 300 nm; they are prepared by the interaction between the mixed antigen-presenting cell membrane components activated by the blank nanoparticles 3 and the blank nanoparticles 3. The obtained nanoparticles were nanoparticle 6, with a particle size of 300 nm.
或者通过在400g离心5分钟收集与纳米粒子1孵育后的2000万个BMDC,然后使用生理盐水洗涤BMDC两遍,将细胞重悬在生理盐水中后在4℃下使用低功率7.5W超声10分钟以破坏细胞并制备含有细胞膜组分的样品。然后将样品一次通过滤膜孔径为50μm、10μm、5μm、1μm、0.45μm、0.22μm的滤膜过滤,收集所得滤液后与步骤(2)制备的纳米粒子1(50mg)共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液并在15000g离心60分钟,弃去上清后使用生理盐水重悬所得沉淀即为纳米粒子7,粒径为300nm。Or collect 20 million BMDCs after incubation with nanoparticles 1 by centrifugation at 400g for 5 minutes, then wash the BMDCs twice with physiological saline, resuspend the cells in physiological saline and use low power 7.5W ultrasonic for 10 minutes at 4°C. to disrupt cells and prepare samples containing cell membrane components. Then filter the sample through a filter membrane with pore sizes of 50 μm, 10 μm, 5 μm, 1 μm, 0.45 μm, and 0.22 μm. Collect the filtrate and incubate it with the nanoparticle 1 (50 mg) prepared in step (2) for 10 minutes, and then Repeated co-extrusion using a 0.45 μm filter membrane, centrifuge the extrudate at 15,000 g for 60 minutes, discard the supernatant and resuspend in physiological saline, and the resulting precipitate is Nanoparticle 7, with a particle size of 300 nm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
第0天,在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在第10天和第14天分给小鼠皮下注射100μL负载癌细胞全细胞组分的1mgPLGA纳米疫苗1或者100μL PBS;过程中监测PBS组和疫苗组小鼠肿瘤生长速度。在第18天处死小鼠,收集小鼠的外周血,使用密度梯度离心法从小鼠外周血中分离出外周血单个核细胞(PBMC),然后使用流式细胞术从PBMC中分离CD3 +T细胞。其中使用纳米疫苗1处理组的CD3 +T细胞为疫苗处理组,使用PBS处理组的T细胞为PBS对照组。 On day 0, 1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. On days 10 and 14, the mice were subcutaneously injected with 100 μL of 1 mg PLGA nanovaccine loaded with whole cell components of cancer cells1. Or 100 μL PBS; monitor the tumor growth rate of mice in the PBS group and vaccine group during the process. The mice were sacrificed on day 18, and their peripheral blood was collected. Peripheral blood mononuclear cells (PBMC) were isolated from the mouse peripheral blood using density gradient centrifugation, and then CD3 + T cells were isolated from the PBMC using flow cytometry. . Among them, the CD3 + T cells in the group treated with Nano Vaccine 1 are the vaccine treatment group, and the T cells in the group treated with PBS are the PBS control group.
将步骤(5)制备的基于抗原提呈细胞的100μg纳米粒子(纳米粒子4,或者纳米粒子5,或者纳米粒子6,或者纳米粒子7)与B细胞(500万个)和40万个来自肿瘤浸润淋巴细胞的T细胞(疫苗组或者PBS组)在5mL RPMI1640完全培养基中共孵育24小时(37℃,5%CO 2),然后采用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(mean fluorescence intensity,MFI)。荧光强度越强,说明该癌细胞特异性T细胞所表达的杀伤性物质越多,检测时灵敏度会更高准确性会越好。 Combine 100 μg of antigen-presenting cell-based nanoparticles prepared in step (5) (nanoparticle 4, or nanoparticle 5, or nanoparticle 6, or nanoparticle 7) with B cells (5 million) and 400,000 from tumors Lymphocyte-infiltrating T cells (vaccine group or PBS group) were incubated in 5 mL RPMI1640 complete medium for 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to sort the incubated CD3 + IFN-γ + T cells. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ + in CD3 + IFN-γ + T cells was analyzed. The stronger the fluorescence intensity, the more killer substances expressed by the cancer cell-specific T cells, and the detection sensitivity will be higher and the accuracy will be better.
或者将步骤(2)制备的100μg纳米粒子(纳米粒子1,或者纳米粒子2,或者纳米粒子3)与B细胞(500万个)和40万个来自肿瘤浸润淋巴细胞的T细胞(疫苗组或者PBS组)在5mL RPMI1640完全培养基中共孵育24小时(37℃,5%CO 2),然后采用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(MFI)。 Or combine 100 μg of nanoparticles (nanoparticle 1, or nanoparticle 2, or nanoparticle 3) prepared in step (2) with B cells (5 million) and 400,000 T cells from tumor-infiltrating lymphocytes (vaccine group or PBS group) were incubated in 5 mL RPMI1640 complete medium for a total of 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to sort the incubated CD3 + IFN-γ + T cells. At the same time, the mean fluorescence intensity (MFI) of the IFN-γ + -linked fluorescent probe in CD3 + IFN-γ + T cells was analyzed.
或者将步骤(5)制备的基于抗原提呈细胞的100μg纳米粒子(纳米粒子4,或者纳米粒子5,或者纳米粒子6,或者纳米粒子7)与40万个来自肿瘤浸润淋巴细胞的T细胞(疫苗组或者PBS组)在5mL RPMI1640完全培养基中共孵育24小时(37℃,5%CO 2),然后采 用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(mean fluorescence intensity,MFI)。 Or combine 100 μg of antigen-presenting cell-based nanoparticles prepared in step (5) (nanoparticle 4, or nanoparticle 5, or nanoparticle 6, or nanoparticle 7) with 400,000 T cells derived from tumor-infiltrating lymphocytes ( Vaccine group or PBS group) were incubated in 5 mL RPMI1640 complete medium for a total of 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to sort the incubated CD3 + IFN-γ + T cells. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ + in CD3 + IFN-γ + T cells was analyzed.
或者将步骤(2)制备的100μg纳米粒子(纳米粒子1)与40万个来自肿瘤浸润淋巴细胞的T细胞(疫苗组)在5mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2),然后采用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞。 Or incubate 100 μg of nanoparticles (nanoparticle 1) prepared in step (2) with 400,000 T cells (vaccine group) derived from tumor infiltrating lymphocytes in 5 mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), and then use flow cytometry to sort the incubated CD3 + IFN-γ + T cells.
(7)实验结果(7)Experimental results
如图3中a和b所示,PBS组小鼠肿瘤生长速度明显快于疫苗组,这说明疫苗组小鼠体内癌细胞特异性T细胞明显多于PBS组,因而可以延缓和控制小鼠肿瘤体积的增长。PBS组未经过疫苗诱导,所以含有较少的癌细胞特异性T细胞,而疫苗组经过疫苗诱导后会有更多的癌细胞特异性T细胞,如图3所示,PBS对照组和疫苗组在经过不同纳米粒子检测后结果有差异。纳米粒子4效果最好,所能检测到的癌细胞特异性T细胞最全面。纳米粒子1在没有抗原提呈细胞辅助的情况下,无法检测到癌细胞特异性T细胞。而纳米粒子4、纳米粒子5和纳米粒子7在没有抗原提呈细胞辅助的情况下也能检测到癌细胞特异性T细胞,而纳米粒子4的效果明显好于其他两种纳米粒子。而且,纳米粒子4在没有抗原提呈细胞辅助的情况下检测到的癌细胞特异性T细胞效果好于纳米粒子7在有抗原提呈细胞辅助的情况下检测到的癌细胞特异性T细胞。而且,纳米粒子4、纳米粒子5和纳米粒子7在有无抗原提呈细胞辅助时所能检测到的癌细胞特异性T细胞差异不大。这说明,使用负载癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞制备的纳米粒子有利于更好的检测癌细胞特异性T细胞;而且DC和B细胞的混合抗原提呈细胞效果好于单一DC。而且,本发明所述的检测广谱癌细胞特异性T细胞的方法可以不依赖于抗原提呈细胞。而且,在检测癌细胞特异性T细胞的过程中,不需要添加抗原提呈细胞即可得到与添加抗原提呈细胞检测癌细胞特异性T细胞一样的效果,这也是本发明所述的由被激活的抗原提呈细胞制备的纳米粒子或微米粒子检测癌细胞特异性T细胞的一个优势。As shown in a and b in Figure 3, the tumor growth rate of mice in the PBS group was significantly faster than that of the vaccine group. This shows that there are significantly more cancer cell-specific T cells in the mice in the vaccine group than in the PBS group, and thus the mouse tumors can be delayed and controlled. Volume growth. The PBS group has not been induced by vaccines, so it contains fewer cancer cell-specific T cells, while the vaccine group will have more cancer cell-specific T cells after vaccine induction. As shown in Figure 3, the PBS control group and vaccine group There are differences in the results after testing different nanoparticles. Nanoparticle 4 has the best effect and can detect the most comprehensive cancer cell-specific T cells. Nanoparticle 1 cannot detect cancer cell-specific T cells without the assistance of antigen-presenting cells. Nanoparticle 4, nanoparticle 5 and nanoparticle 7 can also detect cancer cell-specific T cells without the assistance of antigen-presenting cells, and the effect of nanoparticle 4 is significantly better than the other two nanoparticles. Moreover, the effect of nanoparticle 4 on detecting cancer cell-specific T cells without the assistance of antigen-presenting cells was better than that of nanoparticle 7 on detecting cancer cell-specific T cells with the assistance of antigen-presenting cells. Moreover, there is little difference in the cancer cell-specific T cells that can be detected by Nanoparticle 4, Nanoparticle 5 and Nanoparticle 7 with or without the assistance of antigen-presenting cells. This shows that nanoparticles prepared by using antigen-presenting cells activated by nanoparticles loaded with cancer cell whole-cell antigens are conducive to better detection of cancer cell-specific T cells; and the effect of mixed antigen-presenting cells of DC and B cells is better than Single DC. Furthermore, the method of detecting broad-spectrum cancer cell-specific T cells described in the present invention may not rely on antigen-presenting cells. Moreover, in the process of detecting cancer cell-specific T cells, it is not necessary to add antigen-presenting cells to obtain the same effect as adding antigen-presenting cells to detect cancer cell-specific T cells. This is also the method of the present invention. Nanoparticles or microparticles prepared by activated antigen-presenting cells have an advantage in detecting cancer cell-specific T cells.
负载癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞制备的纳米粒子检测癌细胞特异性T细胞的效果优于负载四种抗原多肽的纳米粒子激活的抗原提呈细胞制备的纳米粒子检测癌细胞特异性T细胞。这说明负载四种新生抗原多肽纳米粒子激活的抗原提呈细胞所制备的纳米粒子可以检测到的癌细胞特异性T细胞克隆种类有限。而负载癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞制备的纳米粒子能检测更广谱的癌细胞特异性T细胞,因而扩增后所能得到的T细胞克隆数也就更广谱。Nanoparticles loaded with cancer cell whole cell antigens and activated by antigen-presenting cells are more effective in detecting cancer cell-specific T cells than nanoparticles loaded with four antigen peptides. Nanoparticles prepared by activated antigen-presenting cells are better at detecting cancer cells. Cell-specific T cells. This shows that the types of cancer cell-specific T cell clones that can be detected by nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with four neoantigen peptides are limited. Nanoparticles prepared by activated antigen-presenting cells loaded with nanoparticles loaded with cancer cell whole cell antigens can detect a wider spectrum of cancer cell-specific T cells, so the number of T cell clones that can be obtained after expansion is also broader. .
如图3中c所示,荧光强度越强,说明该癌细胞特异性T细胞所表达的杀伤性物质越多,检测时灵敏度会更高准确性会越好。在使用相同荧光探针标记的抗小鼠IFN-γ抗体标记被激活的癌细胞特异性T细胞时,表面负载激活的抗原提呈细胞膜组分同时负载癌细胞全细胞组分的纳米粒子4所激活的癌细胞特异性T细胞所能检测到的IFN-γ抗体所连接的荧光探针的荧光信号(平均荧光强度,mean fluorescence intensity,MFI)强于只是负载癌细胞全细胞组分的纳米粒子1所激活的癌细胞特异性T细胞所能检测到的IFN-γ抗体所连接 的荧光探针的荧光信号。而且不论检测和激活过程中孵育体系中有无抗原提呈细胞,纳米粒子4激活的癌细胞特异性T细胞所能检测到的荧光信号都强于纳米粒子1所激活的癌细胞特异性T细胞,这说明纳米粒子4激活的癌细胞特异性T细胞表达了更多的特异性标志物,因而在检测时更容易被检测到。As shown in c in Figure 3, the stronger the fluorescence intensity, the more killer substances expressed by the cancer cell-specific T cells, and the detection sensitivity will be higher and the accuracy will be better. When using the same fluorescent probe-labeled anti-mouse IFN-γ antibody to label activated cancer cell-specific T cells, nanoparticles loaded with activated antigen-presenting cell membrane components and whole cell components of cancer cells 4 were used. The fluorescence signal (mean fluorescence intensity, MFI) of the fluorescent probe connected to the IFN-γ antibody that can be detected by activated cancer cell-specific T cells is stronger than that of nanoparticles simply loaded with whole cell components of cancer cells. 1. The activated cancer cell-specific T cells can detect the fluorescent signal of the fluorescent probe connected to the IFN-γ antibody. Moreover, regardless of whether there are antigen-presenting cells in the incubation system during the detection and activation process, the fluorescence signal detected by the cancer cell-specific T cells activated by nanoparticle 4 is stronger than that of the cancer cell-specific T cells activated by nanoparticle 1. , which shows that the cancer cell-specific T cells activated by nanoparticle 4 express more specific markers and are therefore easier to detect during detection.
由此可见,本发明所述的负载全细胞组分的纳米粒子激活的抗原提呈细胞制备的纳米粒子可以更好的检测具有识别癌细胞和杀伤癌细胞能力的癌细胞特异性T细胞。纳米粒子所负载的癌细胞全细胞抗原在被抗原提呈细胞吞噬后可被降解成抗原表位被提呈到抗原提呈细胞表面,可以识别癌细胞全细胞抗原的特异性T细胞即可以识别癌细胞全细胞抗原表位后被激活并表达特异性表面标志物,通过流式细胞术分析高表达特异性表面标志物的T细胞的比例,即可以知道被激活的可以识别和具有杀伤效能的癌细胞特异性T细胞的数量和比例。It can be seen that the nanoparticles prepared by the antigen-presenting cells activated by the nanoparticles loaded with whole cell components according to the present invention can better detect cancer cell-specific T cells with the ability to recognize and kill cancer cells. The cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen presenting cells and presented to the surface of the antigen presenting cells. Specific T cells that can recognize the cancer cell whole cell antigens can recognize them. Cancer cells are activated after whole-cell antigen epitopes and express specific surface markers. By analyzing the proportion of T cells that highly express specific surface markers through flow cytometry, we can know that the activated T cells can recognize and have killing efficacy. Number and proportion of cancer cell-specific T cells.
实施例3 小鼠外周脾细胞中癌细胞特异性T细胞的检测Example 3 Detection of cancer cell-specific T cells in mouse peripheral spleen cells
本实施例以小鼠黑色素瘤为癌症模型来说明如何使用纳米粒子激活的抗原提呈细胞制备的纳米粒子检测癌细胞特异性T细胞。本实施例中,首先裂解B16F10黑色素瘤肿瘤组织和癌细胞以制备肿瘤组织和癌细胞的水溶性组分混合物(质量比1:1)和非水溶性组分混合物(质量比1:1),并将水溶性组分混合物和非水溶性组分混合物按质量比1:1混合。然后,以PLGA为纳米粒骨架材料,以Poly(I:C)和CpG2006为佐剂制备负载裂解物组分的纳米粒子,然后将纳米粒子与抗原提呈细胞共孵育一段时间后激活抗原提呈细胞,并将抗原提呈细胞制备成纳米粒子检测癌细胞特异性T细胞。This example uses mouse melanoma as a cancer model to illustrate how to use nanoparticles prepared by nanoparticle-activated antigen-presenting cells to detect cancer cell-specific T cells. In this example, B16F10 melanoma tumor tissue and cancer cells are first lysed to prepare a mixture of water-soluble components (mass ratio 1:1) and a mixture of non-water-soluble components (mass ratio 1:1) of tumor tissue and cancer cells, And mix the water-soluble component mixture and the water-insoluble component mixture at a mass ratio of 1:1. Then, PLGA is used as the nanoparticle skeleton material, Poly(I:C) and CpG2006 are used as adjuvants to prepare nanoparticles loaded with lysate components, and then the nanoparticles are incubated with antigen-presenting cells for a period of time to activate antigen presentation. cells, and prepare antigen-presenting cells into nanoparticles to detect cancer cell-specific T cells.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解所得样品;收集培养的B16F10癌细胞系时,先离心去除培养基后使用PBS洗涤两次并离心收集癌细胞,将癌细胞在超纯水中重悬,反复冻融3次,并伴有超声破坏裂解癌细胞。待肿瘤组织或癌细胞裂解后,将裂解物以5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性组分;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性组分转化为在8M尿素水溶液中可溶。将肿瘤组织的水溶性组分和癌细胞的水溶性组分按质量比1:1混合;肿瘤组织的非水溶性组分和癌细胞的非水溶性组分按质量比1:1混合。将水溶性组分混合物和非水溶性组分混合物按质量比1:1混合,即为制备纳米粒子的抗原原料来源。 When collecting tumor tissue, 1.5 × 10 5 B16F10 cells were first subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. The tumor tissue was cut into sections. Grind, add an appropriate amount of pure water through a cell strainer and freeze and thaw repeatedly 5 times, and can be accompanied by ultrasound to destroy the lysed sample; when collecting the cultured B16F10 cancer cell line, first centrifuge to remove the medium, then wash twice with PBS and centrifuge Cancer cells were collected, resuspended in ultrapure water, frozen and thawed three times, and destroyed and lysed by ultrasound. After the tumor tissue or cancer cells are lysed, centrifuge the lysate at 5000g for 5 minutes and take the supernatant to obtain the water-soluble component that is soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate. The non-water-soluble components that are insoluble in pure water are converted into soluble in 8M urea aqueous solution. The water-soluble components of the tumor tissue and the water-soluble components of the cancer cells are mixed at a mass ratio of 1:1; the water-insoluble components of the tumor tissue and the non-water-soluble components of the cancer cells are mixed at a mass ratio of 1:1. Mixing the water-soluble component mixture and the water-insoluble component mixture at a mass ratio of 1:1 is the source of the antigen raw material for preparing nanoparticles.
(2)细菌细胞外囊泡(OMV)的制备(2) Preparation of bacterial extracellular vesicles (OMV)
将长双歧杆菌在5000g离心30分钟,然后弃去沉淀后收集上清液,将上清液使用1μm的滤膜过滤,在4℃下使用20W超声处理5分钟,然后在16000g离心90分钟,将沉淀在PBS中重悬后即为收集到的细菌外囊泡膜组分,然后使用8M尿素水溶液裂解和溶解细菌外囊泡膜组分。Centrifuge Bifidobacterium longum at 5000g for 30 minutes, then discard the precipitate and collect the supernatant. Filter the supernatant with a 1 μm filter, ultrasonicate at 4°C for 5 minutes at 20W, and then centrifuge at 16000g for 90 minutes. Resuspend the pellet in PBS to form the collected bacterial outer vesicle membrane components, and then use 8M urea aqueous solution to lyse and dissolve the bacterial outer vesicle membrane components.
或者将长双歧杆菌在5000g离心30分钟,然后弃去沉淀后收集上清液,将上清液使用1μm的滤膜过滤,在4℃下使用20W超声处理5分钟,然后在16000g离心90分钟,将沉淀在PBS中重悬后即为收集到的细菌外囊泡膜组分,然后使用吐温80水溶液裂解和溶解细菌膜组分。Or centrifuge Bifidobacterium longum at 5000g for 30 minutes, then discard the precipitate and collect the supernatant. Filter the supernatant with a 1 μm filter, ultrasonicate at 4°C for 5 minutes at 20W, and then centrifuge at 16000g for 90 minutes. , resuspend the pellet in PBS to form the collected bacterial outer vesicle membrane components, and then use Tween 80 aqueous solution to lyse and dissolve the bacterial membrane components.
(3)纳米粒子的制备(3) Preparation of nanoparticles
本实施例中纳米粒子1采用复乳法制备,制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为poly(I:C)和CpG2006且佐剂包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解液组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h备用。纳米粒子1平均粒径为250nm左右,每1mg PLGA纳米粒子1约负载130μg蛋白质或多肽组分,每1mg PLGA纳米粒子1所负载的poly(I:C)和CpG2006免疫佐剂各0.02mg,纳米粒子1也在注射时也称为纳米疫苗1。In this example, nanoparticle 1 is prepared by the double emulsion method. The molecular weight of the preparation material PLGA is 7KDa-17KDa. The immune adjuvants used are poly(I:C) and CpG2006 and the adjuvants are encapsulated in the nanoparticles. The preparation method is as mentioned above. During the preparation process, the double emulsion method is first used to load the lysis solution components and adjuvants inside the nanoparticles. Then 100 mg of nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of ultrapure solution containing 4% trehalose is used. Resuspend in water and freeze-dry for 48 hours before use. The average particle size of nanoparticles 1 is about 250nm. Each 1 mg of PLGA nanoparticles 1 is loaded with approximately 130 μg of protein or peptide components. Each 1 mg of PLGA nanoparticles 1 carries 0.02 mg of poly(I:C) and CpG2006 immune adjuvant. Nano Particle 1 is also called nanovaccine 1 when injected.
本实施例中纳米粒子2制备材料和制备方法同纳米粒子1。纳米粒子2内部同时负载步骤(1)所制备的抗原组分和步骤(2)所制备的8M尿素溶解的细菌外囊泡膜组分,且二者质量比为1:1。所采用的免疫佐剂为poly(I:C)和CpG2006且佐剂包载于纳米粒子内。在制备过程中首先采用复乳法在纳米粒子内部负肿瘤组织载裂解液组分、细菌外囊泡组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h备用。纳米粒子2平均粒径为250nm左右,每1mg PLGA纳米粒子2约负载130μg蛋白质或多肽组分,每1mg PLGA纳米粒子2所负载的poly(I:C)和CpG2006免疫佐剂各0.02mg。In this embodiment, the preparation materials and preparation method of nanoparticle 2 are the same as nanoparticle 1. The nanoparticle 2 is internally loaded with the antigen component prepared in step (1) and the 8M urea-dissolved bacterial outer vesicle membrane component prepared in step (2) at the same time, and the mass ratio of the two is 1:1. The immune adjuvants used were poly(I:C) and CpG2006, and the adjuvants were encapsulated in nanoparticles. During the preparation process, the double emulsion method was first used to load the tumor tissue inside the nanoparticles with lysate components, bacterial external vesicle components and adjuvants. Then 100 mg of the nanoparticles were centrifuged at 10,000g for 20 minutes, and 10 mL containing 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 hours before use. The average particle size of nanoparticles 2 is about 250nm. Each 1 mg of PLGA nanoparticles 2 is loaded with approximately 130 μg of protein or peptide components. Each 1 mg of PLGA nanoparticles 2 carries 0.02 mg of poly(I:C) and CpG2006 immune adjuvants each.
本实施例中纳米粒子3制备材料和制备方法同纳米粒子1。纳米粒子3内部同时负载步骤(1)所制备的抗原组分和步骤(2)所制备的吐温80溶解的细菌外囊泡膜组分,且二者质量比为1:1。所采用的免疫佐剂为poly(I:C)和CpG2006且佐剂包载于纳米粒子内。在制备过程中首先采用复乳法在纳米粒子内部负肿瘤组织载裂解液组分、细菌外囊泡组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h备用。纳米粒子3平均粒径为250nm左右,每1mg PLGA纳米粒子3约负载130μg蛋白质或多肽组分,每1mg PLGA纳米粒3所负载的poly(I:C)和CpG2006免疫佐剂各0.02mg。In this embodiment, the preparation materials and preparation methods of nanoparticle 3 are the same as nanoparticle 1. The nanoparticle 3 is simultaneously loaded with the antigen component prepared in step (1) and the bacterial outer vesicle membrane component dissolved in Tween 80 prepared in step (2), and the mass ratio of the two is 1:1. The immune adjuvants used were poly(I:C) and CpG2006, and the adjuvants were encapsulated in nanoparticles. During the preparation process, the double emulsion method was first used to load the tumor tissue inside the nanoparticles with lysate components, bacterial external vesicle components and adjuvants. Then 100 mg of the nanoparticles were centrifuged at 10,000g for 20 minutes, and 10 mL containing 4% trehalose was used. Resuspend in ultrapure water and freeze-dry for 48 hours before use. The average particle size of nanoparticles 3 is about 250nm. Each 1 mg of PLGA nanoparticles 3 is loaded with approximately 130 μg of protein or peptide components. Each 1 mg of PLGA nanoparticles 3 is loaded with 0.02 mg of poly(I:C) and CpG2006 immune adjuvant.
空白纳米粒子4的制备材料和制备方法同纳米粒子1,但是空白纳米粒子4只负载等量的佐剂而不负载任何的肿瘤组织裂解物组分。纳米粒子4的粒径为250nm左右。The preparation materials and methods of blank nanoparticle 4 are the same as nanoparticle 1, but blank nanoparticle 4 only carries the same amount of adjuvant and does not load any tumor tissue lysate components. The particle size of the nanoparticles 4 is approximately 250 nm.
(4)B细胞的分离(4) Isolation of B cells
处死C57BL/6小鼠后摘取小鼠脾脏,制备小鼠脾细胞单细胞悬液,使用磁珠分选法分离脾细胞中的CD19 +B细胞。 After the C57BL/6 mice were sacrificed, the spleens of the mice were removed, a single cell suspension of mouse splenocytes was prepared, and CD19 + B cells in the splenocytes were isolated using magnetic bead sorting.
(5)抗原提呈细胞的激活(5)Activation of antigen-presenting cells
将500μg的纳米粒子1,或者500μg的纳米粒子2,或者500μg的纳米粒子3,或者500μg的纳米粒子4分别与B细胞(1000万个)在15mL RPMI1640完全培养基中共孵育48小时 (37℃,5%CO 2),孵育体系中含GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、白蛋白(50ng/mL)、和CD80抗体(10ng/mL)。 500 μg of nanoparticle 1, or 500 μg of nanoparticle 2, or 500 μg of nanoparticle 3, or 500 μg of nanoparticle 4 were incubated with B cells (10 million cells) in 15 mL of RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), albumin (50ng /mL), and CD80 antibody (10ng/mL).
(6)抗原提呈细胞来源的纳米粒子的制备(6) Preparation of nanoparticles derived from antigen-presenting cells
通过在400g离心5分钟收集孵育后的B细胞,然后使用PBS洗涤细胞三遍,将细胞重悬在PBS水中后在低功率(10W)超声15分钟。然后将样品在500g离心5分钟并收集上清液,将上清液依次过孔径为30μm、10μm、5μm、0.45μm、0.22μm的膜过滤后,将所得滤液样品在18000g离心60分钟后弃去上清液并将沉淀使用PBS重悬后即得纳米粒子。其中,使用纳米粒子1激活的抗原提呈细胞制备的纳米粒子为纳米粒子5,粒径为110纳米;使用纳米粒子2激活的抗原提呈细胞制备的纳米粒子为纳米粒子6,粒径为110纳米;使用纳米粒子3激活的抗原提呈细胞制备的纳米粒子为纳米粒子7,粒径为110纳米;使用纳米粒子4激活的抗原提呈细胞制备的纳米粒子为纳米粒子8,粒径为110纳米。The incubated B cells were collected by centrifugation at 400g for 5 minutes, then washed with PBS three times, resuspended in PBS water and sonicated at low power (10W) for 15 minutes. Then centrifuge the sample at 500g for 5 minutes and collect the supernatant. The supernatant is filtered through membranes with pore diameters of 30 μm, 10 μm, 5 μm, 0.45 μm, and 0.22 μm. The resulting filtrate sample is centrifuged at 18000g for 60 minutes and discarded. The supernatant and the pellet were resuspended in PBS to obtain nanoparticles. Among them, the nanoparticles prepared by using antigen-presenting cells activated by nanoparticle 1 are nanoparticles 5, with a particle size of 110 nanometers; the nanoparticles prepared by using antigen-presenting cells activated by nanoparticles 2 are nanoparticles 6, with a particle size of 110 nanometers. Nanoparticles; the nanoparticles prepared using antigen-presenting cells activated by nanoparticle 3 are nanoparticles 7, with a particle size of 110 nanometers; the nanoparticles prepared using antigen-presenting cells activated with nanoparticles 4 are nanoparticles 8, with a particle size of 110 nanometers nanometer.
(7)癌细胞特异性T细胞的检测(7) Detection of cancer cell-specific T cells
在第0天给每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在第7天,第12天和第17天分别给小鼠皮下注射0.7mg步骤(3)制备的PLGA纳米粒子1(纳米疫苗1)或者100μL PBS。第21天处死小鼠,摘取小鼠脾脏并制备小鼠脾脏单细胞悬液,使用磁珠分选法分选脾脏细胞中的CD3 +T细胞。将分离得到的T细胞(400万个)与步骤(6)制备的100μg的纳米粒子(纳米粒子5,或者纳米粒子6,或者纳米粒子7,或者纳米粒子8)在40mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2),然后在400g离心5分钟收集细胞,然后进行抗体标记后采用流式细胞术分析孵育后的T细胞中CD3 +IFNγ +T细胞,即为被癌细胞全细胞抗原特异性激活的癌细胞特异性T细胞的数量和比例。 Inoculate 1.5×10 5 B16F10 cells subcutaneously on the back of each C57BL/6 mouse on day 0, and subcutaneously inject 0.7 mg of PLGA prepared in step (3) into the mice on day 7, day 12, and day 17 respectively. Nanoparticle 1 (Nanovaccine 1) or 100μL PBS. The mice were sacrificed on the 21st day, the mouse spleens were removed and mouse spleen single cell suspension was prepared, and the CD3 + T cells in the spleen cells were sorted using magnetic bead sorting method. Put the isolated T cells (4 million) and 100 μg of nanoparticles prepared in step (6) (nanoparticle 5, or nanoparticle 6, or nanoparticle 7, or nanoparticle 8) in 40 mL of high-glucose DMEM complete medium The CCP was incubated for 48 hours (37°C, 5% CO 2 ), and then centrifuged at 400 g for 5 minutes to collect the cells. After antibody labeling, flow cytometry was used to analyze the CD3 + IFNγ + T cells in the incubated T cells, which were the The number and proportion of cancer cell-specific T cells specifically activated by cancer cell whole-cell antigens.
纳米粒子所负载的癌细胞全细胞抗原在被抗原提呈细胞吞噬后可被降解成抗原表位被提呈到抗原提呈细胞膜表面,抗原提呈细胞制备的纳米粒子负载有上述降解提呈后的抗原表位,可以被癌细胞特异性T细胞识别并激活癌细胞特异性T细胞,被激活后分泌杀伤性细胞因子。IFN-γ是抗原特异性T细胞识别抗原后被激活所分泌的最主要的细胞因子。使用流式细胞术分析的CD3 +IFN-γ +T细胞即为可以识别和杀伤癌细胞的癌细胞特异性T细胞。 The cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen-presenting cells and presented to the surface of the antigen-presenting cell membrane. The nanoparticles prepared by the antigen-presenting cells are loaded with the above-mentioned degraded and presented products. The antigenic epitope can be recognized by cancer cell-specific T cells and activate cancer cell-specific T cells. After activation, they secrete killer cytokines. IFN-γ is the most important cytokine secreted by antigen-specific T cells that are activated after recognizing antigens. CD3 + IFN-γ + T cells analyzed using flow cytometry are cancer cell-specific T cells that can recognize and kill cancer cells.
(5)实验结果(5)Experimental results
如图4所示,PBS对照组小鼠的肿瘤生长速度很快,而纳米粒子处理组肿瘤生长速度较慢,这说明纳米粒子诱导了癌细胞特异性T细胞可以控制肿瘤的生长。纳米粒子8几乎不能激活癌细胞特异性T细胞;而纳米粒子5、纳米粒子6和纳米粒子7能检测到更多的癌细胞特异性T细胞。而且,纳米粒子6的效果优于纳米粒子5和纳米粒子7,这说明使用适当方法裂解和溶解的细菌外囊泡组分负载到纳米粒子后所激活的抗原提呈细胞有利于检测癌细胞特异性T细胞。As shown in Figure 4, the tumor growth rate of mice in the PBS control group was very fast, while the tumor growth rate of the nanoparticle-treated group was slower, which shows that the nanoparticles induce cancer cell-specific T cells and can control the growth of tumors. Nanoparticle 8 could hardly activate cancer cell-specific T cells; while nanoparticle 5, nanoparticle 6, and nanoparticle 7 could detect more cancer cell-specific T cells. Moreover, the effect of Nanoparticle 6 is better than that of Nanoparticle 5 and Nanoparticle 7, which shows that the activated antigen-presenting cells after loading the bacterial outer vesicle components lysed and dissolved using appropriate methods into the nanoparticles are beneficial to the specific detection of cancer cells. Sexual T cells.
实施例4 纳米粒子或微米粒子检测癌细胞特异性T细胞Example 4 Nanoparticles or microparticles detect cancer cell-specific T cells
本实施例中,首先使用6M盐酸胍裂解B16F10黑色素瘤癌细胞全细胞抗原。然后,以PLGA为微米粒骨架材料,以CpG BW006(B类)、CPG2216(A类)和Poly ICLC为免疫 佐剂制备负载有癌细胞全细胞抗原的微米粒子系统。使用微米粒子激活抗原提呈细胞后,将抗原提呈细胞制备成纳米粒子或微米粒子检测癌细胞特异性T细胞。In this example, 6M guanidine hydrochloride was first used to cleave the whole cell antigen of B16F10 melanoma cancer cells. Then, a micron particle system loaded with cancer cell whole cell antigens was prepared using PLGA as the micron particle skeleton material, CpG BW006 (B type), CPG2216 (A type) and Poly ICLC as immune adjuvants. After using micron particles to activate antigen-presenting cells, the antigen-presenting cells are prepared into nanoparticles or micron particles to detect cancer cell-specific T cells.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的B16F10黑色素瘤癌细胞系收集后在350g离心5分钟,然后弃去上清并用PBS洗涤两遍,然后采用6M盐酸胍重悬和裂解癌细胞,癌细胞全细胞抗原裂解并溶于6M盐酸胍后即为制备微米粒子系统的抗原原料来源。The cultured B16F10 melanoma cancer cell line was collected and centrifuged at 350g for 5 minutes, then the supernatant was discarded and washed twice with PBS, and then the cancer cells were resuspended and lysed with 6M guanidine hydrochloride. The whole cell antigen of the cancer cells was lysed and dissolved in 6M Guanidine hydrochloride is the source of antigen raw materials for preparing micron particle systems.
(2)微米粒子系统的制备(2) Preparation of micron particle system
本实施例中微米粒子采用复乳法制备。所采用的微米粒子1制备材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为CpG BW006、CPG2216和Poly ICLC。Poly ICLC是toll样受体3激动剂,而各类CpG是Toll样受体9激动剂,而Toll样受体3和Toll样受体9均位于细胞内的内吞体膜结构中。首先将裂解物组分和免疫佐剂共负载于微米粒子内,然后在10000g离心15分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h;在粒子使用前将其用7mL PBS重悬然后加入3mL癌细胞裂解液组分(蛋白质浓度50mg/mL)并室温作用10min,得到内外都负载裂解物的微米粒子1。该微米粒子平均粒径为2.50μm左右,表面电位为-2mV左右;每1mg PLGA微米粒子1约负载140μg蛋白质或多肽组分,负载的CpG BW006(B类)、CPG2216(A类)和Poly ICLC各0.02mg。In this embodiment, the micron particles are prepared by the double emulsion method. The molecular weight of PLGA, the material used to prepare micron particles 1, is 38KDa-54KDa, and the immune adjuvants used are CpG BW006, CPG2216 and Poly ICLC. Poly ICLC is a toll-like receptor 3 agonist, while various CpGs are Toll-like receptor 9 agonists, and both Toll-like receptor 3 and Toll-like receptor 9 are located in the endosome membrane structure within the cell. First, the lysate components and immune adjuvant were loaded into the micron particles, and then centrifuged at 10,000g for 15 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and then freeze-dried for 48 hours; before use, the particles were Resuspend in 7 mL of PBS and then add 3 mL of cancer cell lysate component (protein concentration 50 mg/mL) and incubate at room temperature for 10 min to obtain micron particles 1 loaded with lysate both inside and outside. The average particle size of the microparticles is about 2.50μm, and the surface potential is about -2mV; each 1 mg of PLGA microparticles is loaded with approximately 140 μg of protein or peptide components, and the loaded CpG BW006 (Class B), CPG2216 (Class A) and Poly ICLC 0.02mg each.
对照微米粒子2制备材料和制备方法相同,负载的免疫佐剂为CpG2336(A类)、CPG2216(A类)和Poly ICLC。对照微米粒子2粒径为2.50μm左右,表面电位为-2mV左右,每1mg PLGA微米粒子约负载140μg蛋白质或多肽组分,每1mgPLGA微米粒所负载的CpG2336(A类)、CPG2216(A类)和Poly ICLC免疫佐剂各为0.02mg。The preparation materials and preparation methods of control microparticle 2 are the same, and the loaded immune adjuvants are CpG2336 (Class A), CPG2216 (Class A) and Poly ICLC. The diameter of the control micron particles 2 is about 2.50 μm, and the surface potential is about -2mV. Each 1 mg PLGA micron particle is loaded with approximately 140 μg of protein or peptide components. Each 1 mg PLGA micron particle is loaded with CpG2336 (Class A) and CPG2216 (Class A). and Poly ICLC immune adjuvant are 0.02mg each.
对照微米粒子3制备材料和制备方法相同,负载的免疫佐剂为CpG BW006(B类)和CPG2216(A类)。对照微米粒子3每1mgPLGA微米粒所使用的佐剂为0.02mg,粒径为2.50μm左右,表面电位为-2mV左右,每1mg PLGA微米粒子约负载140μg蛋白质或多肽组分,每1mgPLGA微米粒所负载的CpG BW006(B类)和CPG2216(A类)各0.03mg。The preparation materials and preparation methods of control microparticle 3 are the same, and the loaded immune adjuvants are CpG BW006 (category B) and CPG2216 (category A). The adjuvant used in control microparticle 3 is 0.02 mg per 1 mg PLGA microparticle, the particle size is about 2.50 μm, and the surface potential is about -2mV. Each 1 mg PLGA microparticle is loaded with approximately 140 μg of protein or peptide components. Each 1 mg PLGA microparticle contains The loaded CpG BW006 (Category B) and CPG2216 (Category A) are 0.03mg each.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
处死小鼠后收集小鼠淋巴结和脾脏,将小鼠淋巴结或者脾脏切碎研磨分别通过细胞筛网过滤制备单细胞悬液,将淋巴结单细胞悬液和脾脏单细胞悬液混合后,使用流式细胞术从中分选出CD19 +B细胞和CD11c +的DC。 After the mice were killed, mouse lymph nodes and spleens were collected. The mouse lymph nodes or spleens were minced and ground, and filtered through a cell mesh to prepare a single cell suspension. After mixing the lymph node single cell suspension and the spleen single cell suspension, flow cytometry was used. CD19 + B cells and CD11c + DC were sorted by cytometry.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的微米粒子(500μg)与制备的DC(1000万个)和B细胞(1000万个)在20mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO2),孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。Micron particles (500 μg) loaded with whole cell components of cancer cells were incubated with prepared DCs (10 million) and B cells (10 million) in 20 mL high-glucose DMEM complete medium for 72 hours (37°C, 5% CO2 ), the incubation system contains granulocyte-macrophage colony-stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL ) and CD86 antibody (10ng/mL).
(5)抗原提呈细胞来源的纳米粒子或微米粒子的制备(5) Preparation of nanoparticles or microparticles derived from antigen-presenting cells
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷 酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(22.5W)超声1分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子。其中微米粒子1激活的混合抗原提呈细胞制备的纳米粒为纳米粒子1,粒径为110纳米;其中微米粒子2激活的混合抗原提呈细胞制备的纳米粒为纳米粒子2,粒径为110纳米;其中微米粒子3激活的混合抗原提呈细胞制备的纳米粒为纳米粒子3,粒径为110纳米。The incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors. The cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 22.5W) Ultrasonic for 1 minute. Then centrifuge the sample at 3000g for 15 minutes and collect the supernatant. Centrifuge the supernatant at 8000g for 15 minutes and collect the supernatant. Then collect the supernatant after centrifugation at 16000g for 90 minutes. Discard the supernatant and collect the pellet. Place the pellet in PBS. After resuspension, nanoparticles are obtained. Among them, the nanoparticles prepared by the mixed antigen-presenting cells activated by micron particles 1 are nanoparticles 1, and the particle size is 110 nanometers; among them, the nanoparticles prepared by the mixed antigen-presenting cells activated by micron particles 2 are nanoparticles 2, and the particle size is 110 nanometers. Nano; the nanoparticles prepared by mixed antigen-presenting cells activated by micron particles 3 are nanoparticles 3, with a particle size of 110 nanometers.
或者通过在400g离心5分钟收集孵育后的DC和B细胞(采用微米粒子1激活),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(22.5W)超声1分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在16000g离心90分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子。将20mg纳米粒子与100mg微米粒子1混合后在室温共孵育15分钟,然后在10W超声处理1分钟,然后在在8000g离心15分钟后弃去上清液并收集沉淀,将沉淀重悬后即为微米粒子4,粒径为2.55μm左右。Or collect the incubated DC and B cells (activated with micron particles 1) by centrifugation at 400g for 5 minutes, then wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, and resuspend the cells in PBS Then ultrasonicate at 4°C for 1 minute at low power (22.5W) in water. Then centrifuge the sample at 3000g for 15 minutes and collect the supernatant. Centrifuge the supernatant at 8000g for 15 minutes and collect the supernatant. Then collect the supernatant after centrifugation at 16000g for 90 minutes. Discard the supernatant and collect the pellet. Place the pellet in PBS. After resuspension, nanoparticles are obtained. Mix 20 mg nanoparticles and 100 mg micron particles 1 and incubate at room temperature for 15 minutes, then ultrasonicate at 10W for 1 minute, then centrifuge at 8000g for 15 minutes, discard the supernatant and collect the precipitate, and resuspend the precipitate. Micron particles 4, the particle size is about 2.55μm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
第0天,在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞。在第10天,第15天,第20天使用放射线照射肿瘤部位对小鼠进行放射线照射治疗(Radiotherapy)或者注射100μL PBS。在第24天处死小鼠,收集各组小鼠的外周血,然后制备外周血单个核细胞(PBMC),使用磁珠分选法分选出CD3 +T细胞。将分选所得T细胞(500万个)、步骤(5)所制备的纳米粒子1-3(100μg)或微米粒子4(100μg)在2mL RPMI1640完全培养基中共孵育24小时(37℃,5%CO 2),尔后采用流式细胞术检测T细胞中的CD3 +IFN-γ +T细胞,即为被癌细胞全细胞抗原激活的癌细胞特异性T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(MFI)。 On day 0, 1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. On days 10, 15, and 20, the mice were subjected to radiotherapy or injected with 100 μL PBS at the tumor site. The mice were sacrificed on the 24th day, and the peripheral blood of mice in each group was collected. Peripheral blood mononuclear cells (PBMC) were then prepared, and CD3 + T cells were sorted using magnetic bead sorting. The sorted T cells (5 million), nanoparticles 1-3 (100 μg) or microparticles 4 (100 μg) prepared in step (5) were incubated in 2 mL RPMI1640 complete medium for 24 hours (37°C, 5% CO 2 ), and then flow cytometry was used to detect CD3 + IFN-γ + T cells in T cells, which are cancer cell-specific T cells activated by cancer cell whole cell antigens. At the same time, the mean fluorescence intensity (MFI) of the IFN-γ + -linked fluorescent probe in CD3 + IFN-γ + T cells was analyzed.
(7)实验结果(7)Experimental results
如图5a所示,负载CpG佐剂和Poly ICLC混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米粒子检测的癌细胞特异性T细胞效果优于负载两种CpG混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米粒子。而且,负载一种B类CpG、一种A类CpG和Poly ICLC混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米粒子效果好于使用负载两种A类CpG和PolyICLC混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米粒子;而且,由被激活的抗原提呈细胞膜制备的粒子同时内部负载癌细胞全细胞组分效果更佳。CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(MFI)的分析结果与上述趋势一致(图5b)。这说明负载两种不同toll样受体的混合佐剂的微米粒子激活的抗原提呈细胞能制备的纳米粒子效果更好,而且,含有B类CpG与Toll样受体3激动剂作为混合佐剂的微米粒子激活的抗原提呈细胞制备的纳米粒子效果更好。 As shown in Figure 5a, nanoparticles prepared from antigen-presenting cells activated by microparticles loaded with CpG adjuvant and Poly ICLC mixed adjuvant were better at detecting cancer cell-specific T cells than microparticles loaded with two CpG mixed adjuvants. Nanoparticles prepared from activated antigen-presenting cells. Moreover, nanoparticles prepared by activating antigen-presenting cells using microparticles loaded with a type B CpG, a type A CpG, and PolyICLC mixed adjuvant were better than those prepared using micron particles loaded with two type A CpG and PolyICLC mixed adjuvants. Nanoparticles prepared from particle-activated antigen-presenting cells; moreover, particles prepared from activated antigen-presenting cell membranes are more effective at loading whole cell components of cancer cells internally. The analysis results of the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ + in CD3 + IFN-γ + T cells were consistent with the above trend (Figure 5b). This shows that the nanoparticles activated by antigen-presenting cells loaded with mixed adjuvants of two different toll-like receptors can produce better nanoparticles, and that nanoparticles containing class B CpG and Toll-like receptor 3 agonists are used as mixed adjuvants. Nanoparticles prepared by activating antigen-presenting cells with micron particles are more effective.
实施例5 检测肿瘤组织中的癌细胞特异性T细胞Example 5 Detection of cancer cell-specific T cells in tumor tissue
本实施例中,首先使用8M尿素裂解B16F10黑色素瘤肿瘤组织,并溶解肿瘤组织裂解物组分。然后,以PLGA为纳米粒骨架材料,以Poly(I:C)、CpG2006(B类)和CpGSL01(B类)为免疫佐剂制备负载有癌细胞全细胞抗原的纳米粒子,使用纳米粒子激活抗原提呈细胞后制备纳米粒子,然后检测肿瘤组织中的癌细胞特异性T细胞。In this example, 8M urea was first used to lyse B16F10 melanoma tumor tissue and dissolve the tumor tissue lysate components. Then, PLGA was used as the nanoparticle skeleton material, and Poly(I:C), CpG2006 (B type) and CpGSL01 (B type) were used as immune adjuvants to prepare nanoparticles loaded with cancer cell whole cell antigens, and the nanoparticles were used to activate the antigens. After presenting the cells, the nanoparticles are prepared and then detected for cancer cell-specific T cells in the tumor tissue.
(1)肿瘤组织的收集及裂解(1) Collection and lysis of tumor tissue
在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量8M尿素裂解细胞,并溶解细胞裂解物。以上即为制备纳米粒子系统的抗原原料来源。 1.5 × 10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. Cut the tumor tissue into pieces and grind it. Add an appropriate amount of 8M urea through a cell filter to lyse the cells and dissolve the cell lysate. The above are the sources of antigen raw materials for preparing nanoparticle systems.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子采用溶剂挥发法制备。纳米粒子1所采用制备材料PLGA分子量为7KDa-17KDa,所采用的免疫佐剂为Poly(I:C)、CpG2006和CpGSL01,且裂解物组分和佐剂包载于纳米粒子内部。制备方法如前所述,在纳米粒子内部负载裂解物组分和佐剂后,将100mg纳米粒子在12000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h,得冻干粉后备用。该纳米粒子平均粒径为270nm左右,纳米粒子表面电位为-3mV左右;每1mg PLGA纳米粒子约负载80μg蛋白质或多肽组分,每1mg PLGA纳米粒所使用的Poly(I:C)、CpG2006和CpGSL01各为0.02mg。In this example, nanoparticles were prepared by solvent evaporation method. The molecular weight of PLGA, the material used to prepare nanoparticle 1, is 7KDa-17KDa. The immune adjuvants used are Poly(I:C), CpG2006 and CpGSL01, and the lysate components and adjuvants are encapsulated inside the nanoparticles. The preparation method is as described above. After loading the lysate components and adjuvants inside the nanoparticles, 100 mg of the nanoparticles are centrifuged at 12,000 g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and then freeze-dried for 48 h to obtain Freeze-dry the powder for later use. The average particle size of the nanoparticles is about 270nm, and the surface potential of the nanoparticles is about -3mV; each 1 mg of PLGA nanoparticles is loaded with approximately 80 μg of protein or peptide components, and each 1 mg of PLGA nanoparticles uses Poly(I:C), CpG2006 and CpGSL01 is 0.02 mg each.
对照纳米粒子2制备材料和制备方法同上,粒径为270nm左右,负载等量的裂解物组分,负载免疫佐剂为Poly(I:C),每1mg PLGA负载Poly(I:C)0.06mg。The preparation materials and preparation method of control nanoparticle 2 are the same as above. The particle size is about 270nm. It is loaded with an equal amount of lysate components. The loaded immune adjuvant is Poly(I:C). Each 1mg of PLGA is loaded with 0.06mg of Poly(I:C). .
对照纳米粒子3制备材料和制备方法同上,粒径为270nm左右,负载等量的裂解物组分,负载免疫佐剂为Poly(I:C)、CpG1585(A类)和CpG2216(A类),每1mg PLGA负载Poly(I:C)、CpG1585(A类)和CpG2216(A类)各0.02mg。The preparation materials and preparation method of control nanoparticle 3 are the same as above. The particle size is about 270nm. It is loaded with equal amounts of lysate components. The loaded immune adjuvants are Poly(I:C), CpG1585 (Class A) and CpG2216 (Class A). Each 1mg of PLGA loads 0.02mg each of Poly (I:C), CpG1585 (Class A) and CpG2216 (Class A).
(3)DC和B细胞的制备(3)Preparation of DCs and B cells
处死C57BL/6后摘取小鼠淋巴结,制备小鼠淋巴结单细胞悬液,然后使用流式细胞术从淋巴结细胞单细胞悬液中分选出CD11c +DC和CD19 +B细胞。 After C57BL/6 were sacrificed, mouse lymph nodes were removed to prepare mouse lymph node single cell suspension, and then flow cytometry was used to sort CD11c + DC and CD19 + B cells from the lymph node cell single cell suspension.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的纳米粒子1(500μg)、纳米粒子2(500μg)或纳米粒子3(500μg)与DC(500万个)和B细胞(500万个)在20mL高糖DMEM完全培养基中共孵育72小时(37℃,5%CO 2);孵育体系中含有粒细胞-巨噬细胞集落刺激因子(GM-CSF,2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。 Nanoparticles 1 (500 μg), nanoparticles 2 (500 μg), or nanoparticles 3 (500 μg) loaded with cancer cell whole cell components were mixed with DCs (5 million cells) and B cells (5 million cells) in 20 mL of high-glucose DMEM. The culture medium was incubated for a total of 72 hours (37°C, 5% CO 2 ); the incubation system contained granulocyte-macrophage colony-stimulating factor (GM-CSF, 2000U/mL), IL-2 (500U/mL), IL- 7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL).
(5)基于抗原提呈细胞的纳米粒子的制备(5) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃使用匀浆机在2000rpm搅拌破坏处理25分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,然后在15000g离心30分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米例子。其中,使用纳米粒子1激活的抗原提呈细胞制备的为纳米粒子4, 粒径为150纳米;使用纳米粒子2激活的抗原提呈细胞制备的为纳米粒子5,粒径为150纳米;使用纳米粒子3激活的抗原提呈细胞制备的为纳米粒子6,粒径为150纳米。Collect the incubated DC and B cells by centrifugation at 400g for 5 minutes, then wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water and homogenize at 4°C. The machine was stirred and destroyed at 2000rpm for 25 minutes. Then the sample was centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected. Then the supernatant was discarded after centrifugation at 15000g for 30 minutes to collect the pellet. The pellet was resuspended in PBS. After suspension, nanometer samples are obtained. Among them, nanoparticles 4 were prepared using antigen-presenting cells activated by nanoparticles 1, with a particle size of 150 nanometers; nanoparticles 5 were prepared using antigen-presenting cells activated with nanoparticles 2, and the particle size was 150 nanometers; The antigen-presenting cells activated by particle 3 were prepared as nanoparticles 6, with a particle size of 150 nanometers.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
第0天,在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在第8天,第10天,第12天,第14天,第16天分别给小鼠皮下注射100μL的αPD-1抗体(10mg/kg)或100μL PBS。在第20天处死小鼠,分别收集小鼠的肿瘤组织,将肿瘤组织切成小块后通过细胞筛网过滤制备成肿瘤组织单细胞悬液,然后使用磁珠分选法从中分选得到CD3 +T细胞。然后,将分选得到的T细胞(50万个)与来源于同种异体的B细胞(250万个)、步骤(5)制备的纳米粒子(100μg)或在20mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2)。尔后在400g离心5分钟,收集上清液,并采用ELISA法分析上清液中IFN-γ的浓度。 On day 0, 1.5×10 5 B16F10 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. On days 8, 10, 12, 14, and 16, mice were injected subcutaneously with 100 μL. αPD-1 antibody (10 mg/kg) or 100 μL PBS. The mice were sacrificed on the 20th day, and the tumor tissues of the mice were collected respectively. The tumor tissues were cut into small pieces and filtered through a cell mesh to prepare a single cell suspension of tumor tissue, and then CD3 was sorted from it using a magnetic bead sorting method. + T cells. Then, the sorted T cells (500,000) were co-incubated with allogeneic B cells (2.5 million), the nanoparticles prepared in step (5) (100 μg), or in 20 mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ). Then centrifuge at 400g for 5 minutes, collect the supernatant, and analyze the concentration of IFN-γ in the supernatant using ELISA.
在ELISA检测方法中,癌细胞特异性T细胞被激活后会分泌杀伤性物质如IFN-γ。该类杀伤性物质浓度的高低代表着被激活的癌症特异性T细胞的含量的多少及癌细胞杀伤能力的强弱。In the ELISA detection method, cancer cell-specific T cells secrete killer substances such as IFN-γ after being activated. The concentration of this type of killer substance represents the amount of activated cancer-specific T cells and the strength of the killing ability of cancer cells.
(7)实验结果(7)Experimental results
如图6所示,与PBS对照组相比,负载癌细胞全细胞抗原的纳米粒子激活的抗原提呈细胞制备的纳米粒子共孵育后可以检测到更多的IFN-γ。而且,负载两种B类CpG与Poly(I:C)作为混合佐剂的纳米粒子激活的抗原提呈细胞所制备的纳米粒子效果好于负载两种A类CpG与Poly(I:C)作为混合佐剂的纳米粒子或者只负载Poly(I:C)作为佐剂的纳米粒子激活的抗原提呈细胞所制备的纳米粒子。As shown in Figure 6, compared with the PBS control group, more IFN-γ could be detected after co-incubation with nanoparticles prepared by antigen-presenting cells activated by nanoparticles loaded with cancer cell whole cell antigens. Moreover, the effect of nanoparticles loaded with two types of B CpG and Poly(I:C) as mixed adjuvants to activate antigen-presenting cells is better than that of two types of type A CpG with Poly(I:C) as a mixed adjuvant. Nanoparticles prepared by mixing adjuvant nanoparticles or nanoparticles loading only Poly(I:C) as an adjuvant to activate antigen-presenting cells.
实施例6 结肠癌中癌细胞特异性T细胞的检测Example 6 Detection of cancer cell-specific T cells in colon cancer
本实施例以MC38小鼠结肠癌为癌症模型来说明如何使用纳米粒子激活的抗原提呈细胞制备的纳米粒子检测广谱的癌细胞特异性T细胞。首先裂解结肠癌肿瘤组织和肺癌癌细胞以制备水溶性组分和非水溶性组分,并将抗原使用蛋白酶在体外先降解为多肽。在实际应用中也可以使用其他酶或者其他方法先将全细胞组分中的蛋白质降解为多肽。然后再制备水溶性组分混合物(质量比1:1)和非水溶性组分(质量比1:1)混合物,并将水溶性组分混合物和非水溶性组分混合物按质量比1:1混合。然后,以PLA为纳米粒骨架材料,以CpGM362、CPG1018和Poly ICLC为免疫佐剂制备纳米粒子,并用该纳米粒子体外检测癌细胞特异性T细胞。This example uses MC38 mouse colon cancer as a cancer model to illustrate how to use nanoparticles prepared from nanoparticle-activated antigen-presenting cells to detect a broad spectrum of cancer cell-specific T cells. Colon cancer tumor tissue and lung cancer cancer cells are first lysed to prepare water-soluble components and water-insoluble components, and the antigen is first degraded into polypeptides using protease in vitro. In practical applications, other enzymes or other methods can also be used to first degrade the proteins in the whole cell fraction into peptides. Then prepare a mixture of water-soluble components (mass ratio 1:1) and non-water-soluble components (mass ratio 1:1), and mix the water-soluble component mixture and the non-water-soluble component mixture in a mass ratio of 1:1 mix. Then, PLA was used as the nanoparticle skeleton material, and CpGM362, CPG1018 and Poly ICLC were used as immune adjuvants to prepare nanoparticles, and the nanoparticles were used to detect cancer cell-specific T cells in vitro.
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
在每只C57BL/6小鼠背部皮下接种2×10 6个MC38细胞在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织。将肿瘤组织切块后研磨,通过细胞过滤网加入适量纯水并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以大于5000g的转速离心5分钟并取上清液即为可溶于纯水的水溶性组分;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性组分转化为在8M尿素水溶液中可溶。在水溶性组分(80mg/mL)中加入胰蛋白酶(Trypsin,0.5mg/mL)和糜蛋白酶(Chymotrypsin,0.5mg/mL) 共孵育1小时,然后在95℃加热10分钟灭活蛋白酶备用。 2 × 10 6 MC38 cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. Cut the tumor tissue into pieces and then grind it. Add an appropriate amount of pure water through a cell filter and freeze and thaw repeatedly 5 times. Ultrasound can be used to destroy the lysed cells. After the cells are lysed, centrifuge the lysate at a speed greater than 5000g for 5 minutes and take the supernatant, which is the water-soluble component soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate and remove the insoluble components. The water-insoluble components of pure water are converted into soluble in 8M urea aqueous solution. Add trypsin (0.5 mg/mL) and chymotrypsin (Chymotrypsin, 0.5 mg/mL) to the water-soluble component (80 mg/mL), incubate for 1 hour, and then heat at 95°C for 10 minutes to inactivate the proteases for later use.
将培养的LLC肺癌细胞系收集后在350g离心5分钟,然后弃去上清并用PBS洗涤两遍,然后采用超纯水重悬细胞并反复冻融5次,并可伴有超声以破坏裂解细胞。待细胞裂解后,将裂解物以3000g的转速离心6分钟并取上清液即为可溶于纯水的水溶性组分;在所得沉淀部分中加入8M尿素溶解沉淀部分即可将不溶于纯水的非水溶性组分转化为在8M尿素水溶液中可溶。在水溶性组分(80mg/mL)中加入胰蛋白酶(Trypsin,0.5mg/mL)和糜蛋白酶(Chymotrypsin,0.5mg/mL)共孵育1小时,然后在95℃加热10分钟灭活蛋白酶备用。Collect the cultured LLC lung cancer cell lines and centrifuge them at 350g for 5 minutes. Then discard the supernatant and wash twice with PBS. Then resuspend the cells in ultrapure water and freeze and thaw repeatedly 5 times. Ultrasound can be used to destroy the lysed cells. . After the cells are lysed, centrifuge the lysate at 3000g for 6 minutes and take the supernatant to obtain the water-soluble components that are soluble in pure water; add 8M urea to the resulting precipitate to dissolve the precipitate and remove the insoluble components in pure water. The water-insoluble components of water are converted into soluble in 8M urea aqueous solution. Add trypsin (0.5 mg/mL) and chymotrypsin (Chymotrypsin, 0.5 mg/mL) to the water-soluble component (80 mg/mL) and incubate for 1 hour, then heat at 95°C for 10 minutes to inactivate the proteases for later use.
将来自结肠癌肿瘤组织的和肺癌癌细胞的水溶性组分按质量比1:1混合;溶解于8M尿素中的非水溶性组分也按质量比1:1混合。然后将水溶性组分混合物和非水溶性组分混合物按照质量比1:1混合,该混合物为制备纳米粒子的原料来源。The water-soluble components from colon cancer tumor tissue and lung cancer cancer cells were mixed at a mass ratio of 1:1; the water-insoluble components dissolved in 8M urea were also mixed at a mass ratio of 1:1. Then, the water-soluble component mixture and the water-insoluble component mixture are mixed at a mass ratio of 1:1, and this mixture is the source of raw materials for preparing nanoparticles.
(2)卡介苗(BCG)的裂解和溶解(2) Lysis and dissolution of Bacillus Calmette-Guérin (BCG)
收集BCG,使用8M尿素水溶液裂解BCG后溶解裂解组分备用。Collect BCG, use 8M urea aqueous solution to cleave BCG, and then dissolve the cleavage components for later use.
(3)纳米粒子的制备(3) Preparation of nanoparticles
本实施例中纳米粒子1采用溶剂挥发法制备。纳米粒子1制备材料PLA分子量为20KDa,纳米粒子内部负载肿瘤组织和癌细胞裂解物、细菌裂解物以及免疫佐剂,表面负载肿瘤组织和癌细胞裂解物组分。所采用的免疫佐剂为CpGM362、CPG1018和poly ICLC,且佐剂负载于纳米粒子内部,制备纳米粒子时使用的肿瘤组织和癌细胞裂解物与细菌裂解物的质量比为1:1。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解物混合物、细菌裂解物组分和佐剂,在内部负载裂解物和佐剂后,将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。使用前将20mg纳米粒重悬于0.9mL PBS中,并于0.1mL含有等量癌细胞和肿瘤组织裂解物混合物和细菌裂解物组分(80mg/mL)的样品室温混合孵育5分钟后即可使用。纳米粒子1平均粒径为290nm左右,每1mg PLGA纳米粒子1约负载140μg蛋白质或多肽组分,每1mgPLGA纳米粒含有CpGM362、CPG1018和Poly ICLC免疫佐剂各0.04mg。In this embodiment, nanoparticle 1 is prepared by solvent evaporation method. The molecular weight of PLA, the material for preparing nanoparticle 1, is 20KDa. The nanoparticles are loaded with tumor tissue and cancer cell lysate, bacterial lysate and immune adjuvant inside, and the tumor tissue and cancer cell lysate components are loaded on the surface. The immune adjuvants used were CpGM362, CPG1018 and poly ICLC, and the adjuvants were loaded inside the nanoparticles. The mass ratio of tumor tissue and cancer cell lysate to bacterial lysate used in preparing the nanoparticles was 1:1. The preparation method is as mentioned above. During the preparation process, the lysate mixture, bacterial lysate components and adjuvants are first loaded inside the nanoparticles using the double emulsion method. After loading the lysates and adjuvants inside, 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose and freeze-dried for 48 h. Before use, resuspend 20 mg of nanoparticles in 0.9 mL of PBS and mix and incubate at room temperature for 5 minutes with 0.1 mL of samples containing equal amounts of cancer cell and tumor tissue lysate mixture and bacterial lysate components (80 mg/mL). use. The average particle size of nanoparticles 1 is about 290nm. Each 1 mg of PLGA nanoparticles 1 is loaded with approximately 140 μg of protein or peptide components. Each 1 mg of PLGA nanoparticles contains 0.04 mg each of CpGM362, CPG1018 and Poly ICLC immune adjuvant.
(4)抗原提呈细胞的制备(4) Preparation of antigen-presenting cells
处死C57BL/6后收集小鼠外周血,从外周血中分离外周血单核细胞(PBMC),然后使用流式细胞术从PBMC中分选出CD11c +DC和CD19 +B细胞。本实施例中同时使用BMDC和BMDM作为抗原提呈细胞。BMDC制备方法同实施例1。BMDM制备方法如下: Peripheral blood mononuclear cells (PBMC) were isolated from the peripheral blood of mice after killing C57BL/6, and then CD11c + DC and CD19 + B cells were sorted from PBMC using flow cytometry. In this example, BMDC and BMDM were used simultaneously as antigen-presenting cells. The preparation method of BMDC is the same as in Example 1. The preparation method of BMDM is as follows:
将C57小鼠麻醉后脱臼处死,将小鼠使用75%乙醇的消毒,然后用剪刀在小鼠背部剪开一小口,用手直接撕开皮肤至小鼠小腿关节处,去除小鼠足关节以及皮肤。用剪刀沿着小鼠大腿根部大转子将后肢拆下来,去掉肌肉组织后放置在含有75%乙醇的培养皿内浸泡5min,更换新的75%乙醇的培养皿移入超净台中。将乙醇浸泡的腿骨移入冷的PBS浸泡,洗去胫骨、股骨表面的乙醇,此过程可重复3次。将清洗好的股骨、胫骨分开,并用剪刀分别将股骨、胫骨两端剪断,使用1mL注射器吸取冷的诱导培养基将骨髓从股骨、胫骨中吹出,反复吹洗3次,直至腿骨内看不到明显的红色为止。用5mL移液枪将含有骨髓细胞的 培养基反复吹打,使细胞团块分散,然后使用70μm细胞滤器将细胞过筛,转移至15mL离心管内,1500rpm/min离心5min,弃上清,加入红细胞裂解液重悬静置5min后1500rpm/min离心5min,弃上清用冷的配置好的骨髓巨噬细胞诱导培养基(含有15%L929培养基的DMEM高糖培养基)重悬,铺板。将细胞培养过夜,以去除贴壁较快的其他杂细胞如纤维细胞等等。收集未贴壁细胞按实验设计安排种入皿或细胞培养板内。巨噬细胞集落刺激因子(M-CSF)以40ng/mL浓度刺激使骨髓细胞向单核巨噬细胞分化。培养8天,光镜下观察巨噬细胞形态变化。8天后消化收集细胞,用抗小鼠F4/80抗体和抗小鼠CD11b抗体,4℃避光孵育30min后,使用流式细胞术鉴定所诱导成功的巨噬细胞的比例即可。C57 mice were anesthetized and sacrificed by dislocation. The mice were disinfected with 75% ethanol. Then, a small opening was made on the back of the mouse with scissors. The skin was directly torn open to the calf joint of the mouse by hand, and the foot joint and foot joint of the mouse were removed. skin. Use scissors to remove the hind limbs along the greater trochanter at the root of the mouse's thigh, remove the muscle tissue and place it in a petri dish containing 75% ethanol to soak for 5 minutes. Replace the petri dish with a new one with 75% ethanol and move it to a clean bench. Move the leg bones soaked in ethanol into cold PBS and soak them to wash off the ethanol on the surfaces of the tibia and femur. This process can be repeated three times. Separate the cleaned femur and tibia, and use scissors to cut off both ends of the femur and tibia respectively. Use a 1mL syringe to draw cold induction medium and blow out the bone marrow from the femur and tibia. Repeat the blowing 3 times until the inside of the leg bone is no longer visible. to a distinct red color. Use a 5mL pipette to pipe the culture medium containing bone marrow cells repeatedly to disperse the cell clumps, then use a 70μm cell strainer to sieve the cells, transfer to a 15mL centrifuge tube, centrifuge at 1500rpm/min for 5 minutes, discard the supernatant, and add red blood cells to lyse Resuspend the solution and let stand for 5 minutes, then centrifuge at 1500 rpm/min for 5 minutes. Discard the supernatant and resuspend in cold prepared bone marrow macrophage induction medium (DMEM high-glucose medium containing 15% L929 medium), and plate. Culture the cells overnight to remove other miscellaneous cells that adhere quickly, such as fibroblasts, etc. Collect non-adherent cells and seed them into dishes or cell culture plates according to the experimental design. Macrophage colony-stimulating factor (M-CSF) stimulates bone marrow cells to differentiate into mononuclear macrophages at a concentration of 40ng/mL. After culturing for 8 days, the morphological changes of macrophages were observed under a light microscope. Digest and collect the cells after 8 days, use anti-mouse F4/80 antibody and anti-mouse CD11b antibody, incubate at 4°C for 30 minutes in the dark, and use flow cytometry to identify the proportion of successfully induced macrophages.
(5)抗原提呈细胞的激活(5)Activation of antigen-presenting cells
将纳米粒子1(1000μg)与外周血来源的DC(2000万个)、BMDC(2000万个)在RPMI1640完全培养基中共孵育72小时(37℃,5%CO 2);孵育体系中含有GM-CSF(500U/mL)、IL-2(500U/mL)、IL-7(500U/mL)、IL-12(500U/mL)和CD80抗体(10ng/mL)。 Nanoparticle 1 (1000 μg) was incubated with peripheral blood-derived DC (20 million) and BMDC (20 million) in RPMI1640 complete medium for 72 hours (37°C, 5% CO 2 ); the incubation system contained GM- CSF (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL), IL-12 (500U/mL) and CD80 antibody (10ng/mL).
或者将纳米粒子1(1000μg)与外周血来源的DC(1000万个)、BMDC(1000万个)、BMDM(1000万个)、B细胞(1000万个)在20mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2),孵育体系中含有GM-CSF(500U/mL)、IL-2(500U/mL)、IL-7(500U/mL)、IL-12(500U/mL)和CD80抗体(10ng/mL)。 Or incubate nanoparticle 1 (1000 μg) with peripheral blood-derived DCs (10 million), BMDC (10 million), BMDM (10 million), and B cells (10 million) in 20 mL RPMI1640 complete medium for 48 hours (37°C, 5% CO 2 ), the incubation system contains GM-CSF (500U/mL), IL-2 (500U/mL), IL-7 (500U/mL), and IL-12 (500U/mL) and CD80 antibody (10ng/mL).
(6)基于抗原提呈细胞的纳米粒子的制备(6) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的外周血来源的DC(2000万个)和BMDC(2000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在高压均质机中(5000bar)中处理5分钟。然后将样品在2000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液与步骤(3)所制备的纳米粒子1(50mg)在4℃共孵育16小时,然后使用0.45μm的滤膜反复共挤出,将挤出液在13000g离心20分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子2,粒径为310纳米。The incubated peripheral blood-derived DCs (20 million) and BMDCs (20 million) were collected by centrifugation at 400g for 5 minutes, and then the cells were washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors. The cells were resuspended in PBS water and treated in a high-pressure homogenizer (5000 bar) for 5 minutes. The sample was then centrifuged at 2000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected. The supernatant was mixed with the nanoparticle 1 (50mg) prepared in step (3) at 4°C. Incubate for a total of 16 hours, and then use a 0.45 μm filter to repeatedly co-extrude. Centrifuge the extruded liquid at 13000g for 20 minutes, discard the supernatant and collect the precipitate. Resuspend the precipitate in PBS to obtain nanoparticle 2. diameter is 310 nm.
或者通过在400g离心5分钟收集孵育后的外周血来源的DC(1000万个)、BMDC(1000万个)、B细胞(1000万个)及BMDM(1000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在高压均质机中(5000bar)中处理5分钟。然后将样品在2000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液与步骤(3)所制备的纳米粒子1(50mg)在4℃共孵育16小时,然后使用0.45μm的滤膜反复共挤出,将挤出液在13000g离心20分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子3,粒径为310纳米。Or collect the incubated peripheral blood-derived DC (10 million), BMDC (10 million), B cells (10 million), and BMDM (10 million) by centrifugation at 400g for 5 minutes, and then use protease inhibitor-containing Wash the cells twice with 4°C phosphate buffer solution (PBS), resuspend the cells in PBS water and treat them in a high-pressure homogenizer (5000bar) for 5 minutes. The sample was then centrifuged at 2000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected. The supernatant was mixed with the nanoparticle 1 (50mg) prepared in step (3) at 4°C. Incubate for a total of 16 hours, and then use a 0.45 μm filter to repeatedly co-extrude. Centrifuge the extruded liquid at 13000g for 20 minutes, discard the supernatant and collect the precipitate. Resuspend the precipitate in PBS to obtain nanoparticle 3. diameter is 310 nm.
(7)癌细胞特异性T细胞的检测(7) Detection of cancer cell-specific T cells
第0天每只C57BL/6小鼠背部皮下接种1.5×10 5个MC38细胞,在第10天,第15天和第21天分别给小鼠皮下注射100μL的1mg PLGA纳米粒子1以激活小鼠体内的癌细胞特异性T细胞。在第24天处死小鼠,收集小鼠的引流淋巴结和脾脏,制备引流淋巴结和脾细胞的单细 胞悬液并使用磁珠法从中分选出T细胞。 On day 0, each C57BL/6 mouse was subcutaneously inoculated with 1.5 × 10 5 MC38 cells on the back. On days 10, 15 and 21, the mice were subcutaneously injected with 100 μL of 1 mg PLGA nanoparticles 1 to activate the mice. Cancer cell-specific T cells in the body. The mice were sacrificed on day 24, and the draining lymph nodes and spleens of the mice were collected. Single-cell suspensions of draining lymph nodes and spleen cells were prepared and T cells were isolated from them using the magnetic bead method.
将所得T细胞(400万个)、200μg的纳米粒子(纳米粒子1,或者纳米粒子2,或者纳米粒子3)、IL-2(500U/mL)、IL-7(500U/mL)及IL-15(5000U/mL)在5mLDMEM完全培养基中共孵育24小时,然后采用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞,即为被癌细胞全细胞抗原激活的癌细胞特异性T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(MFI)。荧光强度越强,说明该癌细胞特异性T细胞所表达的杀伤性物质越多,检测时灵敏度会更高准确性会越好。 The obtained T cells (4 million), 200 μg of nanoparticles (nanoparticle 1, or nanoparticle 2, or nanoparticle 3), IL-2 (500 U/mL), IL-7 (500 U/mL) and IL- 15 (5000U/mL) was incubated in 5mL DMEM complete medium for 24 hours, and then flow cytometry was used to sort the incubated CD3 + IFN-γ + T cells, which are cancer cell-specific cells activated by cancer cell whole cell antigens. T cells. At the same time, the mean fluorescence intensity (MFI) of the IFN-γ + -linked fluorescent probe in CD3 + IFN-γ + T cells was analyzed. The stronger the fluorescence intensity, the more killer substances expressed by the cancer cell-specific T cells, and the detection sensitivity will be higher and the accuracy will be better.
或者将所得T细胞(400万个)、外周血来源的DC(100万个)、BMDC(100万个)、B细胞(100万个)、BMDM(100万个)以及200μg的纳米粒子(纳米粒子1,或者纳米粒子3)、IL-2(500U/mL)、IL-7(500U/mL)及IL-15(5000U/mL)在5mLDMEM完全培养基中共孵育24小时,然后采用流式细胞术分选孵育后的CD3 +IFN-γ +T细胞,即为被癌细胞全细胞抗原激活的癌细胞特异性T细胞。与此同时,分析CD3 +IFN-γ +T细胞中IFN-γ +所连接荧光探针的平均荧光强度(MFI)。 Alternatively, the obtained T cells (4 million), peripheral blood-derived DCs (1 million), BMDC (1 million), B cells (1 million), BMDM (1 million), and 200 μg of nanoparticles (nano Particle 1, or nanoparticle 3), IL-2 (500U/mL), IL-7 (500U/mL) and IL-15 (5000U/mL) were incubated in 5mL DMEM complete medium for 24 hours, and then flow cytometry was used CD3 + IFN-γ + T cells after surgical sorting and incubation are cancer cell-specific T cells activated by cancer cell whole cell antigens. At the same time, the mean fluorescence intensity (MFI) of the IFN-γ + -linked fluorescent probe in CD3 + IFN-γ + T cells was analyzed.
(8)实验结果(8)Experimental results
如图7所示,纳米粒子3优于纳米粒子1和纳米粒子2。在不含有抗原提呈细胞检测T细胞时,纳米粒子1无法检测到,而纳米粒子2和纳米粒子3都可以检测到。在含有抗原提呈细胞检测T细胞时,纳米粒子1、纳米粒子2和纳米粒子3都可以检测到T细胞,而且,纳米粒子2和纳米粒子3好于纳米粒子1,纳米粒子3好于纳米粒子2。而且,即使是在不含有抗原提呈细胞检测T细胞时,纳米粒子2的效果也好于纳米粒子1和纳米粒子2在含有抗原提呈细胞时检测T细胞的效果。平均荧光强度的分析结果与上述趋势一致。这说明粒子表面负载被激活的抗原提呈细胞膜组分以及使用混合抗原提呈细胞膜组分均可以提高纳米粒子或微米粒子检测癌细胞特异性T细胞的效果。As shown in Figure 7, Nanoparticle 3 outperforms Nanoparticle 1 and Nanoparticle 2. When detecting T cells without antigen-presenting cells, nanoparticle 1 cannot be detected, but both nanoparticle 2 and nanoparticle 3 can be detected. When detecting T cells containing antigen-presenting cells, nanoparticle 1, nanoparticle 2 and nanoparticle 3 can all detect T cells. Furthermore, nanoparticle 2 and nanoparticle 3 are better than nanoparticle 1, and nanoparticle 3 is better than nanoparticle 3. Particle 2. Moreover, even when detecting T cells without antigen-presenting cells, the effect of nanoparticle 2 is better than that of nanoparticles 1 and 2 when detecting T cells when they contain antigen-presenting cells. The analysis results of average fluorescence intensity are consistent with the above trend. This shows that loading activated antigen-presenting cell membrane components on the particle surface and using mixed antigen-presenting cell membrane components can improve the effectiveness of nanoparticles or microparticles in detecting cancer cell-specific T cells.
实施例7 乳腺癌小鼠中癌细胞特异性T细胞的检测Example 7 Detection of cancer cell-specific T cells in breast cancer mice
本实施例以4T1小鼠三阴性乳腺癌为癌症模型来说明如何负载癌细胞全细胞抗原的微米粒子激活抗原提呈细胞后制备成微米粒子检测来自外周免疫器官的癌细胞特异性T细胞。This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how micron particles loaded with cancer cell whole cell antigens activate antigen-presenting cells and then are prepared into micron particles to detect cancer cell-specific T cells from peripheral immune organs.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中,然后反复冻融5遍并伴有超声以裂解癌细胞。在所裂解系细胞中加入1mg/mL的核酸酶降解裂解物中的核酸,然后在95℃加热10分钟灭活核酸酶,然后在5000g离心5分钟收集上清液即为水溶性组分组分,沉淀使用10%脱氧胆酸钠(含10M精氨酸)溶解即为非水溶性组分组分,将水溶性组分组分与非水溶性组分组分按质量比3:1混合即为制备粒子系统的原料来源。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water, and then repeatedly frozen and thawed 5 times with ultrasound to lyse cancer cells. Add 1mg/mL nuclease to the lysed cells to degrade the nucleic acid in the lysate, then heat at 95°C for 10 minutes to inactivate the nuclease, and then centrifuge at 5000g for 5 minutes to collect the supernatant, which is the water-soluble component. Precipitate using 10% sodium deoxycholate (containing 10M arginine) to dissolve the non-water-soluble components. Mix the water-soluble components and the non-water-soluble components at a mass ratio of 3:1 to prepare the particle system. sources of raw materials.
(2)微米粒子系统的制备(2) Preparation of micron particle system
本实施例中制备微米粒子采用复乳法。微米粒子1骨架材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为CpG2395(C类)、CpGM362(C类)和Poly(I:C)。制备时采用复乳法制备内部负载裂解物组分和佐剂的微米粒子,然后将100mg微米粒子在 9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后备用。该微米粒子系统平均粒径为2.5μm左右,表面电位为-6mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,负载CpG2395、CpGM362和Poly(I:C)各0.02mg。对照微米粒子2制备材料和制备方法同上,粒径为2.5μm左右,表面电位为-6mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,每1mgPLGA负载CpG1585(A类)、CpG2336(A类)和Poly(I:C)各0.02mg。In this embodiment, the double emulsion method is used to prepare micron particles. The molecular weight of the micron particle 1 framework material PLGA is 38KDa-54KDa, and the immune adjuvants used are CpG2395 (Category C), CpGM362 (Category C) and Poly (I:C). During preparation, the double emulsion method is used to prepare micron particles loaded with lysate components and adjuvants internally, and then 100 mg of micron particles are centrifuged at 9000g for 20 minutes, resuspended in 10 mL of ultrapure water containing 4% trehalose, and dried for 48 hours before use. The average particle size of this microparticle system is about 2.5 μm, and the surface potential is about -6mV; each 1 mg of PLGA micron particles is loaded with approximately 110 μg of protein or polypeptide components, and each of CpG2395, CpGM362 and Poly(I:C) is loaded with 0.02 mg. The preparation materials and preparation method of control micron particles 2 are the same as above, the particle size is about 2.5 μm, and the surface potential is about -6mV; every 1 mg of PLGA micron particles is loaded with approximately 110 μg of protein or peptide components, and every 1 mg of PLGA is loaded with CpG1585 (Class A), CpG2336 ( Class A) and Poly(I:C) 0.02mg each.
(3)B细胞的制备(3) Preparation of B cells
使用来自外周脾细胞中的B细胞。处死小鼠后摘取脾脏,然后制备小鼠脾细胞单细胞悬液,使用磁珠分选法分选单细胞悬液中的CD19 +B细胞。 B cells from peripheral splenocytes were used. After the mice were sacrificed, the spleens were removed, and then a single cell suspension of mouse spleen cells was prepared, and the CD19 + B cells in the single cell suspension were sorted using magnetic bead sorting.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞抗原组分的微米粒子(800μg)与步骤(3)制备的B细胞(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2),孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)和CD86抗体(10ng/mL)。 The micron particles (800 μg) loaded with cancer cell whole cell antigen components were incubated with the B cells (10 million) prepared in step (3) in 15 mL high-sugar DMEM complete medium for 48 hours (37°C, 5% CO 2 ) , the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL) and CD86 antibody (10ng/mL).
(5)基于抗原提呈细胞的微米粒子的制备(5) Preparation of microparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的B细胞(1000万个),然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声1分钟后使用匀浆机在1000rpm处理3分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在8000g离心15分钟后收集上清液,将上清液与步骤(2)所制备的微米粒子(60mg)以及DSPE-PEG-甘露糖(1mg)在50W超声处理2分钟,然后在8000g离心20分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得微米粒子。其中使用微米粒子1激活的抗原提呈细胞膜组分与微米粒子1共作用所制备的微米粒子为微米粒子3,粒径为2.6μm;使用微米粒子2激活的抗原提呈细胞膜组分与微米粒子2共作用所制备的微米粒子为微米粒子4,粒径为2.6μm。The incubated B cells (10 million) were collected by centrifugation at 400g for 5 minutes, then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, and resuspended in PBS water at 4°C. Ultrasonicate at low power (20W) for 1 minute and then use a homogenizer at 1000rpm for 3 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 8000g for 15 minutes and the supernatant was collected. The supernatant was mixed with the microparticles (60mg) prepared in step (2) and DSPE-PEG. -Mannose (1mg) was sonicated at 50W for 2 minutes, then centrifuged at 8000g for 20 minutes and then collected. The supernatant was discarded to collect the precipitate. The precipitate was resuspended in PBS to obtain micron particles. The microparticles prepared by using the antigen-presenting cell membrane component activated by micron particle 1 and micron particle 1 are micron particles 3, with a particle size of 2.6 μm; using the antigen-presenting cell membrane component activated by micron particle 2 and micron particle The micron particles prepared by the combined action of 2 are micron particles 4, with a particle size of 2.6 μm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
第0天每只BALB/c小鼠背部皮下接种1×10 6个4T1细胞,在第10天,第14天和第18天分别给小鼠皮下注射100μL的1mg PLGA微米粒子1(微米疫苗1)或者100μL的PBS。在第22天处死小鼠,收集小鼠的脾脏,制备脾细胞单细胞悬液。将脾细胞单细胞悬液(600万个)、DC2.4(200万个)以及微米粒子(50μg)在2mL DMEM完全培养基中共孵育72小时(37℃,5%CO 2),然后采用流式细胞术从中分选出CD3 +IFN-γ +T细胞,即为被癌细胞全细胞抗原激活的癌细胞特异性T细胞。 On day 0, each BALB/c mouse was subcutaneously inoculated with 1 × 10 6 4T1 cells on the back. On days 10, 14 and 18, the mice were subcutaneously injected with 100 μL of 1 mg PLGA micron particles 1 (micron vaccine 1). ) or 100 μL of PBS. The mice were sacrificed on day 22, their spleens were collected, and a single cell suspension of splenocytes was prepared. Spleen cell single cell suspension (6 million cells), DC2.4 (2 million cells) and micron particles (50 μg) were incubated in 2 mL DMEM complete medium for 72 hours (37°C, 5% CO 2 ), and then used flow CD3 + IFN-γ + T cells, which are cancer cell-specific T cells activated by whole-cell antigens of cancer cells, are sorted out by cytometry.
(7)实验结果(7)Experimental results
如图8所示,与对照组相比,微米粒子激活的抗原提呈细胞所制备的微米粒子可以在处理组小鼠中有效检测到更多的癌细胞特异性T细胞。而且,使用两种C类CpG与Poly(I:C)作为混合佐剂的微米粒子1激活的抗原提呈细胞制备的微米粒子3效果好于两种A类CpG 与Poly(I:C)作为混合佐剂的微米粒子2激活的抗原提呈细胞制备的微米粒子4。本实施例微米疫苗中使用甘露糖作为主动靶向的靶头,在实际应用中也可以使用CD32抗体、甘露聚糖、CD205抗体、CD19抗体等任何具有靶向靶细胞能力的靶头。As shown in Figure 8, compared with the control group, micron particles prepared by micron particle-activated antigen-presenting cells can effectively detect more cancer cell-specific T cells in mice in the treatment group. Moreover, the effect of microparticles 3 prepared by using two types of CpG and Poly(I:C) as mixed adjuvants was better than that of two types of A CpG and Poly(I:C). The micron particles 4 were prepared by mixing the micron particles 2 with the adjuvant to activate the antigen-presenting cells. Mannose is used as an active target in the micron vaccine of this embodiment. In practical applications, any target with the ability to target target cells, such as CD32 antibodies, mannans, CD205 antibodies, and CD19 antibodies, can also be used.
实施例8 胰腺癌中癌细胞特异性T细胞的检测Example 8 Detection of cancer cell-specific T cells in pancreatic cancer
本实施例以甘露糖为靶头说明如何使用主动靶向纳米粒激活抗原提呈细胞制备的纳米粒子检测外周血中的癌细胞特异性T细胞。This example uses mannose as the target to illustrate how to detect cancer cell-specific T cells in peripheral blood using nanoparticles prepared by activating antigen-presenting cells using active targeting nanoparticles.
(1)癌细胞的裂解(1) Lysis of cancer cells
收集培养的Pan02胰腺癌癌细胞后采用10%辛基葡萄糖苷裂解癌细胞和溶解来源于癌细胞的癌细胞全细胞抗原。The cultured Pan02 pancreatic cancer cells were collected and 10% octylglucoside was used to lyse the cancer cells and dissolve the whole cell antigens derived from the cancer cells.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例中纳米粒子系统使用复乳法制备。纳米粒子制备材料为PLGA和甘露糖修饰的PLGA,二者分子量都为7KDa-17KDa。制备带有靶头的纳米粒子时二者一起使用时质量比为4:1。所采用的免疫佐剂为Poly(I:C)和CpG SL03。制备方法如前所述,采用复乳法将裂解物组分和佐剂共负载于纳米粒子内部,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用。带有靶头的纳米粒子1的平均粒径均为270nm左右,每1mg PLGA纳米粒子约负载80μg蛋白质和多肽组分,含Poly(I:C)和CpGSL03各0.04mg。不负载佐剂但带有甘露糖靶头的纳米粒子2粒径也为270nm左右,制备时采用等量细胞裂解组分但是不含任何免疫佐剂,每1mg PLGA纳米粒子约负载80μg蛋白质和多肽组分。In this example, the nanoparticle system was prepared using the double emulsion method. The nanoparticle preparation materials are PLGA and mannose-modified PLGA, both of which have molecular weights of 7KDa-17KDa. When the two are used together to prepare nanoparticles with a target, the mass ratio is 4:1. The immune adjuvants used were Poly(I:C) and CpG SL03. The preparation method is as described above. The lysate components and adjuvants are loaded into the nanoparticles using the double emulsion method. Then 100 mg of the nanoparticles are centrifuged at 10,000 g for 20 minutes and resuspended in 10 mL of ultrapure water containing 4% trehalose. Then freeze-dry for 48 hours before use. The average particle size of the nanoparticles 1 with the target is about 270nm. Each 1 mg of PLGA nanoparticles is loaded with approximately 80 μg of protein and peptide components, including 0.04 mg each of Poly(I:C) and CpGSL03. Nanoparticles 2 that are not loaded with adjuvants but have a mannose target are also about 270nm in size. They are prepared using the same amount of cell lysis components but do not contain any immune adjuvants. Each 1 mg of PLGA nanoparticles is loaded with approximately 80 μg of protein and peptides. components.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC和BMDM作为抗原提呈细胞。BMDC和BMDM制备方法同上。This example uses BMDC and BMDM as antigen-presenting cells. The preparation methods of BMDC and BMDM are the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将纳米粒子1(1000μg)或者纳米粒子2(1000μg)分别与BMDC(1000万个)、BMDM(1000万个)及IL-7(500U/mL)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2)。 Nanoparticle 1 (1000 μg) or nanoparticle 2 (1000 μg) were incubated with BMDC (10 million pcs), BMDM (10 million pcs) and IL-7 (500 U/mL) respectively in 15 mL high-glucose DMEM complete medium for 48 hours. (37°C, 5% CO 2 ).
或者将BMDC(1000万个)、BMDM(1000万个)及IL-7(500U/mL)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO2)。上述两种孵育系统中都含有IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)。Or incubate BMDC (10 million pcs), BMDM (10 million pcs) and IL-7 (500U/mL) in 15mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO2). Both of the above incubation systems contain IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10ng/mL ).
(5)抗原提呈细胞来源的纳米粒子的制备(5) Preparation of nanoparticles derived from antigen-presenting cells
通过在400g离心5分钟收集孵育后的DC和巨噬细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(10W)超声20分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液依次过孔径为30μm、10μm、5μm、2μm、1μm、0.45μm、0.22μm的膜过滤后,收集滤液,然后将滤液在18000g离心50分钟后收集弃去上清液收集沉淀,将沉淀在4%海藻糖水溶液中重悬,尔后冷冻干燥48小时后即得纳米粒子。其中,使用未被纳米粒子激活的混合抗原提呈细胞制备的为纳米 粒子3,粒径为110纳米;使用纳米粒子2激活的混合抗原提呈细胞制备的为纳米粒子4,粒径为110纳米;使用纳米粒子1激活的混合抗原提呈细胞制备的为纳米粒子5,粒径为110纳米。Collect the incubated DCs and macrophages by centrifugation at 400g for 5 minutes, then wash the cells twice using 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water and incubate at 4°C on low power. (10W) Ultrasound for 20 minutes. Then centrifuge the sample at 3000g for 15 minutes and collect the supernatant. The supernatant is filtered through membranes with pore sizes of 30 μm, 10 μm, 5 μm, 2 μm, 1 μm, 0.45 μm, and 0.22 μm. The filtrate is collected, and then the filtrate is filtered at 18000 g. After centrifugation for 50 minutes, collect and discard the supernatant to collect the precipitate, resuspend the precipitate in 4% trehalose aqueous solution, and then freeze-dry for 48 hours to obtain nanoparticles. Among them, the one prepared using mixed antigen-presenting cells that have not been activated by nanoparticles is nanoparticle 3, with a particle size of 110 nanometers; the one prepared using mixed antigen-presenting cells activated with nanoparticles 2 is nanoparticle 4, with a particle size of 110 nanometers. ; Nanoparticle 5 was prepared using mixed antigen-presenting cells activated by nanoparticle 1, with a particle size of 110 nanometers.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
在第0天给每只C57BL/6小鼠背部皮下接种1×10 6个Pan02胰腺癌细胞,第10天,第15天、第20天和第27天分别给小鼠皮下注射100μL的1mg PLGA纳米粒子。第24天处死小鼠并收集小鼠外周血,从外周血中制备外周血单个核细胞(PBMC),然后从PBMC中使用流式细胞术分离CD3 +T细胞。将T细胞(500万个)与100μg纳米粒子(纳米粒子1,或者纳米粒子3,或者纳米粒子4,或者纳米粒子5)在DMEM高糖培养基中共孵育72小时(37℃,5%CO 2),孵育体系中含有IL-2(500U/mL)、IL-7(500U/mL)。然后采用流式细胞术从孵育后细胞中分选出CD3 +IFN-γ +T细胞,即为癌细胞特异性T细胞。 On day 0, each C57BL/6 mouse was subcutaneously inoculated with 1 × 10 6 Pan02 pancreatic cancer cells on the back. On day 10, day 15, day 20, and day 27, mice were injected subcutaneously with 100 μL of 1 mg PLGA. Nanoparticles. The mice were sacrificed on day 24 and peripheral blood was collected. Peripheral blood mononuclear cells (PBMC) were prepared from the peripheral blood, and then CD3 + T cells were isolated from the PBMC using flow cytometry. T cells (5 million) were incubated with 100 μg of nanoparticles (nanoparticle 1, or nanoparticle 3, or nanoparticle 4, or nanoparticle 5) in DMEM high-glucose medium for 72 hours (37°C, 5% CO 2 ), the incubation system contains IL-2 (500U/mL) and IL-7 (500U/mL). Then flow cytometry is used to sort out CD3 + IFN-γ + T cells from the incubated cells, which are cancer cell-specific T cells.
(7)实验结果(7)Experimental results
如图9所示,纳米粒子4和纳米粒子5效果好于纳米粒子1和纳米粒子3。而且纳米粒子5好于纳米粒子4。综上所述,不管是否被带有佐剂的纳米粒子激活的抗原提呈细胞,其所制备的纳米粒子都可以有效检测癌细胞特异性T细胞,但是带有佐剂的纳米粒子激活的抗原提呈细胞制备的纳米粒子效果更佳。As shown in Figure 9, nanoparticles 4 and 5 are better than nanoparticles 1 and 3. And Nanoparticle 5 is better than Nanoparticle 4. In summary, regardless of whether the antigen-presenting cells are activated by nanoparticles with adjuvants, the prepared nanoparticles can effectively detect cancer cell-specific T cells, but the antigens activated by nanoparticles with adjuvants Nanoparticles prepared by presenting cells are more effective.
实施例9 纳米粒子检测肺癌癌细胞特异性T细胞Example 9 Nanoparticles detect lung cancer cancer cell-specific T cells
本实施例说明钙化的纳米粒子检测小鼠脾细胞中的癌细胞特异性T细胞,在实际使用时也可以使用其他生物矿化技术、交联、凝胶化等修饰粒子。本实施例中,将小鼠肺癌肿瘤组织以8M尿素(含200mM氯化钠)裂解后溶解并负载于纳米粒子系统,使用该粒子激活抗原提呈细胞后,将抗原提呈细胞制备成纳米粒子检测癌细胞特异性T细胞。This example illustrates the detection of cancer cell-specific T cells in mouse splenocytes using calcified nanoparticles. In actual use, other biomineralization technologies, cross-linking, gelation and other modified particles can also be used. In this example, mouse lung cancer tumor tissue was lysed with 8M urea (containing 200mM sodium chloride), dissolved and loaded into a nanoparticle system. After using the particles to activate antigen-presenting cells, the antigen-presenting cells were prepared into nanoparticles. Detection of cancer cell-specific T cells.
(1)肿瘤组织和癌细胞的裂解(1) Lysis of tumor tissue and cancer cells
在6-8周的雌性C57BL/6小鼠后背接种1×10 6个LLC小鼠肺癌细胞,待肿瘤体积长到1000mm 3时处死小鼠摘取小鼠肿瘤组织,将肿瘤组织切块研磨后通过细胞筛网过滤制备成单细胞悬液,使用紫外照射5分钟后在80℃高温加热10分钟,然后采用8M尿素(含200mM氯化钠)裂解和溶解肿瘤组织单细胞悬液,即得癌细胞全细胞抗原。 Inoculate10 LLC mouse lung cancer cells on the back of female C57BL/6 mice aged 6 to 8 weeks. When the tumor volume reaches 1000 mm3 , the mice will be sacrificed. The mouse tumor tissue will be removed, cut into pieces and ground. Then filter it through a cell mesh to prepare a single cell suspension, use ultraviolet irradiation for 5 minutes and then heat at 80°C for 10 minutes, and then use 8M urea (containing 200mM sodium chloride) to lyse and dissolve the tumor tissue single cell suspension to obtain Cancer cell whole cell antigen.
(2)纳米粒子的制备(2) Preparation of nanoparticles
本实施例在纳米粒子内部和表面负载癌细胞全细胞抗原后生物钙化纳米粒子。本实施例中纳米粒子采用溶剂挥发法制备,所采用的纳米粒子制备材料PLGA分子量为7KDa-17KDa,所采用免疫佐剂CpG2006和Poly(I:C)负载于纳米粒子内部。制备方法如下所述,在制备过程中首先采用复乳法在纳米粒子内部负载抗原,在内部负载裂解组分后,将100mg PLGA纳米粒子在13000g离心20min后使用18mL PBS重悬,然后加入2mL溶解于8M尿素的肿瘤组织和癌细胞裂解液(60mg/mL),在室温作用10分钟后在12000g离心20分钟后收集沉淀。然后将该100mg PLGA纳米粒子重悬于20mL DMEM培养基中,然后加入200μL CaCl 2(1mM)并在37℃反应两小时。然后在10000g离心20分钟后收集沉淀, 并采用超纯水重悬后离心洗涤两遍即得纳米粒子1,平均粒径为290nm左右。每1mg PLGA纳米粒子1约负载140μg蛋白质或多肽组分,CpG2006和Poly(I:C)各0.03mg。 In this embodiment, the nanoparticles are loaded with cancer cell whole cell antigens inside and on the surface, and then the nanoparticles are biocalcified. In this example, the nanoparticles are prepared by a solvent evaporation method. The molecular weight of the nanoparticle preparation material PLGA is 7KDa-17KDa. The immune adjuvants CpG2006 and Poly(I:C) are loaded inside the nanoparticles. The preparation method is as follows. During the preparation process, the double emulsion method is first used to load the antigen inside the nanoparticles. After loading the lysis components inside, 100mg PLGA nanoparticles are centrifuged at 13000g for 20 minutes and resuspended in 18mL PBS. Then 2mL is added to dissolve Act in 8M urea tumor tissue and cancer cell lysate (60mg/mL) at room temperature for 10 minutes, then centrifuge at 12000g for 20 minutes to collect the precipitate. The 100 mg PLGA nanoparticles were then resuspended in 20 mL DMEM medium, and then 200 μL CaCl 2 (1 mM) was added and reacted at 37°C for two hours. Then, collect the precipitate after centrifugation at 10,000 g for 20 minutes, resuspend in ultrapure water, and centrifuge and wash twice to obtain nanoparticles 1, with an average particle size of about 290 nm. Each 1 mg of PLGA nanoparticles 1 is loaded with approximately 140 μg of protein or peptide components, including 0.03 mg of CpG2006 and Poly(I:C).
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例1。B细胞来自小鼠外周血PBMC,制备方法同上。将BMDC和B细胞按数量比1:1混合即得混合抗原提呈细胞。This example uses BMDC and B as antigen-presenting cells. The preparation method of BMDC is the same as in Example 1. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above. Mix BMDC and B cells at a ratio of 1:1 to obtain mixed antigen-presenting cells.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有细胞因子组分1:IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)。 Nanoparticles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains cytokine component 1: IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL).
或者作为对照将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);在孵育体系中不含有任何细胞因子和抗体。 Or as a control, nanoparticles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); does not contain any cytokines or antibodies in the incubation system.
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有细胞因子组合2:IL-4(500U/mL)、IL-10(200U/mL)、IL-37(200U/mL)、TGF-β(500U/mL)。 Nanoparticles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains cytokine combination 2: IL-4 (500U/mL), IL-10 (200U/mL), IL-37 (200U/mL), and TGF-β (500U/mL).
(5)基于抗原提呈细胞的纳米粒子的制备(5) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品与步骤(2)制备的纳米粒子1混合后使用高压均质机(10000bar)处理3分钟,然后在13000g离心30分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子。其中使用粒子激活抗原提呈细胞时体系中含有细胞因子组合1的抗原提呈细胞所制备的为纳米粒子2,粒径为300纳米;使用粒子激活抗原提呈细胞时体系中不含有细胞因子的抗原提呈细胞所制备的为纳米粒子3,粒径为300纳米;其中使用粒子激活抗原提呈细胞时体系中含有细胞因子组合2的抗原提呈细胞所制备的为纳米粒子4,粒径为300纳米。The incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors. The cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 20W) Ultrasonic for 2 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45μm membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, filter and concentrate. Mix the filtered and concentrated sample with the nanoparticles 1 prepared in step (2) and treat it with a high-pressure homogenizer (10000bar) for 3 minutes. Then centrifuge at 13000g for 30 minutes and discard the supernatant for collection. Precipitate and resuspend the precipitate in PBS to obtain nanoparticles. Among them, when particles are used to activate antigen-presenting cells, the antigen-presenting cells containing cytokine combination 1 in the system are prepared as nanoparticles 2, with a particle size of 300 nanometers; when particles are used to activate antigen-presenting cells, the system does not contain cytokines. The nanoparticles 3 prepared by the antigen-presenting cells have a particle size of 300 nanometers; the nanoparticles 4 prepared by the antigen-presenting cells containing the cytokine combination 2 in the system when activating the antigen-presenting cells using particles are nanoparticles 4 with a particle size of 300 nanometers. 300 nm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
在第0天给每只C57BL/6小鼠背部皮下接种0.5×10 6个LLC肺癌细胞,第10天,第15天和第20天分别给小鼠皮下注射100μL的1mg PLGA纳米粒子。第24天处死小鼠并摘取小鼠脾细胞并制备成单细胞悬液,然后使用流式细胞术分离CD45 +CD3 +T细胞。 On day 0, each C57BL/6 mouse was subcutaneously inoculated with 0.5 × 10 6 LLC lung cancer cells on the back. On days 10, 15, and 20, the mice were subcutaneously injected with 100 μL of 1 mg PLGA nanoparticles. On day 24, the mice were sacrificed and spleen cells were harvested and prepared into single cell suspension, and then flow cytometry was used to isolate CD45 + CD3 + T cells.
将T细胞(500万个)与抗原提呈细胞制备的纳米粒子2(100μg)或纳米粒子3(100μg)或纳米粒子4(100μg)在DMEM高糖培养基中共孵育12小时(37℃,5%CO 2),在孵育过 程中系统中含有IL-2(500U/mL)和IL-7(500U/mL); T cells (5 million) were incubated with nanoparticles 2 (100 μg) or nanoparticles 3 (100 μg) or nanoparticles 4 (100 μg) prepared by antigen-presenting cells in DMEM high-glucose medium for 12 hours (37°C, 5 %CO 2 ), the system contains IL-2 (500U/mL) and IL-7 (500U/mL) during the incubation process;
或者T细胞(500万个)与抗原提呈细胞制备的纳米粒子2(100μg)在DMEM高糖培养基中共孵育12小时(37℃,5%CO2),且孵育体系中不含任何细胞因子或抗体;Or T cells (5 million) and nanoparticles 2 (100 μg) prepared from antigen-presenting cells were incubated in DMEM high-glucose medium for 12 hours (37°C, 5% CO2), and the incubation system did not contain any cytokines or Antibody;
或者将T细胞(500万个),1000万个步骤(3)制备的混合抗原提呈细胞以及纳米粒子1(100μg)在DMEM高糖培养基中共孵育12小时(37℃,5%CO 2),在孵育过程中系统中含有IL-2(500U/mL)和IL-7(500U/mL)。 Or incubate T cells (5 million), 10 million mixed antigen-presenting cells prepared in step (3) and nanoparticle 1 (100 μg) in DMEM high-glucose medium for 12 hours (37°C, 5% CO 2 ) , the system contained IL-2 (500U/mL) and IL-7 (500U/mL) during the incubation process.
然后收集上述孵育后的细胞,在400g离心5分钟后使用相应的流式抗体标记孵育后的细胞,尔后采用流式细胞术检测CD3 +IFN-γ +T细胞,即为癌细胞特异性T细胞。 Then collect the above-incubated cells, centrifuge at 400g for 5 minutes and use the corresponding flow cytometry antibody to label the incubated cells, and then use flow cytometry to detect CD3 + IFN-γ + T cells, which are cancer cell-specific T cells. .
(7)实验结果(7)Experimental results
如图10所示,与对照组相比,钙化纳米粒激活的抗原提呈细胞制备的纳米粒子可以检测到更多的癌细胞特异性T细胞。而且,负载癌细胞全细胞抗原纳米粒子激活抗原提呈细胞时体系中含有细胞因子组合1优于体系中不含细胞因子和/或抗体的;纳米粒子激活抗原提呈细胞时体系中含有细胞因子组合1或体系中不含细胞因子的效果优于体系中含有细胞因子组合2;而且,抗原提呈细胞制备的纳米粒子与T细胞共孵育时体系中含有细胞因子组合2的优于体系中不含有细胞因子的。As shown in Figure 10, compared with the control group, nanoparticles prepared from antigen-presenting cells activated by calcified nanoparticles could detect more cancer cell-specific T cells. Moreover, when nanoparticles loaded with whole cell antigens of cancer cells activate antigen-presenting cells, the system containing cytokine combination 1 is better than the system without cytokines and/or antibodies; when nanoparticles activate antigen-presenting cells, the system contains cytokines. The effect of combination 1 or the system without cytokines is better than that of the system with cytokine combination 2; moreover, when the nanoparticles prepared by antigen-presenting cells are co-incubated with T cells, the effect of the system with cytokine combination 2 is better than that of the system without cytokine combination 2. Contains cytokines.
实施例10 纳米粒子检测黑色素瘤中的癌细胞特异性T细胞Example 10 Nanoparticles detect cancer cell-specific T cells in melanoma
(1)肿瘤组织和癌细胞的裂解及各组分的收集(1) Lysis of tumor tissue and cancer cells and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种1.5×10 5个B16F10细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网后制备单细胞悬液,加入超纯水后反复冻融并伴有超声裂解上述细胞,然后加入核酸酶作用5分钟,再在95℃作用10分钟灭活核酸酶。尔后在8000g离心3分钟,上清液部分即为水溶性组分;沉淀部分使用10%脱氧胆酸钠水溶液溶解非水溶性组分。将水溶性组分和脱氧胆酸钠溶解后的非水溶性组分按质量比1:1混溶即为制备纳米粒子系统的抗原原料来源。 When collecting tumor tissue, 1.5 × 10 5 B16F10 cells were first subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm 3 , the mice were sacrificed and the tumor tissue was removed. The tumor tissue was cut into sections. Grind and pass through the cell strainer to prepare a single cell suspension. Add ultrapure water and freeze and thaw repeatedly with ultrasound to lyse the above cells. Then add nuclease and incubate for 5 minutes, and then inactivate the nuclease at 95°C for 10 minutes. Then centrifuge at 8000g for 3 minutes, the supernatant part is the water-soluble component; the precipitate part uses 10% sodium deoxycholate aqueous solution to dissolve the non-water-soluble component. The water-soluble component and the non-water-soluble component after dissolving sodium deoxycholate are miscible at a mass ratio of 1:1, which is the source of the antigen raw material for preparing the nanoparticle system.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒采用复乳法制备,具有靶向树突状细胞的能力。所采用的纳米粒子制备材料为PLGA和甘露聚糖修饰的PLGA,二者分子量都为24KDa-38KDa,使用时未修饰PLGA和甘露聚糖修饰PLGA的质量比为9:1。所采用的免疫佐剂为poly(I:C)、CpG1018和CpG2216,增加溶酶体免疫逃逸的物质为KALA多肽(WEAKLAKALAKALAKHLAKALAKALKACEA),且佐剂、KALA多肽包载于纳米粒子内。制备方法如前所述,在制备过程中首先采用复乳法在纳米粒子内部负载裂解液组分、佐剂、KALA多肽,在内部负载上述组分后,将100mg纳米粒子在12000g离心25分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h。该纳米粒子平均粒径为250nm左右,表面电位为-5mV左右;每1mg PLGA纳米粒子约负载100μg蛋白质或多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)、CpG1018和CpG2216免疫佐剂各0.02mg,负载KALA多肽0.05mg。纳 米粒2的制备材料和方法相同,其粒径为250nm左右,表面电位为-5mV左右,不负载KALA多肽,负载等量佐剂和细胞裂解组分。纳米粒子3的制备材料和制备方法相同,为250nm左右,表面电位为-5mV左右;每1mg PLGA纳米粒子约负载100μg蛋白质和多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)0.02mg,负载CpG1018为0.04mg,负载KALA多肽0.05mg。In this embodiment, the nanoparticles are prepared by the double emulsion method and have the ability to target dendritic cells. The nanoparticle preparation materials used are PLGA and mannan-modified PLGA, both of which have molecular weights of 24KDa-38KDa. When used, the mass ratio of unmodified PLGA to mannan-modified PLGA is 9:1. The immune adjuvants used are poly(I:C), CpG1018 and CpG2216. The substance that increases lysosomal immune escape is KALA polypeptide (WEAKLAKALAKALAKHLAKALAKALKACEA), and the adjuvants and KALA polypeptide are encapsulated in nanoparticles. The preparation method is as mentioned above. During the preparation process, the double emulsion method is first used to load the lysis solution components, adjuvants, and KALA polypeptide inside the nanoparticles. After loading the above components inside, 100 mg of the nanoparticles are centrifuged at 12,000g for 25 minutes. And resuspended in 10 mL of ultrapure water containing 4% trehalose and freeze-dried for 48 h. The average particle size of the nanoparticles is about 250nm, and the surface potential is about -5mV; each 1 mg of PLGA nanoparticles is loaded with approximately 100 μg of protein or peptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I:C), CpG1018 and CpG2216 immune The adjuvants are 0.02mg each, and the loaded KALA polypeptide is 0.05mg. The preparation materials and methods of Nanoparticle 2 are the same. Its particle size is about 250nm, its surface potential is about -5mV, it does not load KALA polypeptide, and it loads equal amounts of adjuvants and cell lysis components. The preparation materials and preparation methods of nanoparticle 3 are the same, about 250nm, and the surface potential is about -5mV; each 1 mg of PLGA nanoparticles is loaded with approximately 100 μg of protein and peptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I:C) 0.02mg, the loaded CpG1018 is 0.04mg, and the loaded KALA polypeptide is 0.05mg.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC和BMDM作为抗原提呈细胞。BMDC和BMDM制备方法同上。This example uses BMDC and BMDM as antigen-presenting cells. The preparation methods of BMDC and BMDM are the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(1000万个)与BMDM(1000万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有GM-CSF(2000U/mL)、M-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)。 Nanoparticles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (10 million) and BMDM (10 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); The incubation system contains GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN -γ (500U/mL) and CD80 antibody (10ng/mL).
(5)基于抗原提呈细胞的纳米粒子的制备(5) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的BMDC和BMDM,然后使用含有0.0759M蔗糖和0.225M甘露醇的30mM pH 7.0Tris-HCl缓冲液中1200rpm 3min离心清洗三次,然后在磷酸酶抑制剂和蛋白酶抑制剂的存在下超声机械破坏抗原提呈细胞。经过离心后所获细胞膜用10mM pH 7.5的Tris-HCl和1mM EDTA的溶液清洗。然后将样品依次过孔径为30μm、10μm、5μm、2μm、0.45μm的膜过滤后,将滤液在16000g离心35分钟后弃去上清液收集沉淀,将沉淀使用PBS重悬后与步骤(2)制备的纳米粒子共孵育10分钟,然后使用0.45μm的滤膜反复共挤出,将挤出液在12000g离心25分钟后弃去上清液收集沉淀,将沉淀在含有的4%海藻糖水溶液中重悬后冷冻干燥,即得纳米粒子。其中使用纳米粒子1激活的抗原提呈细胞膜组分与纳米粒子1共作用制备的为纳米粒子4,粒径为260纳米;其中使用纳米粒子2激活的抗原提呈细胞膜组分与纳米粒子2共作用制备的为纳米粒子5,粒径为260纳米;其中使用纳米粒子3激活的抗原提呈细胞膜组分与纳米粒子3共作用制备的为纳米粒子6,粒径为260纳米。Collect the incubated BMDC and BMDM by centrifugation at 400g for 5 minutes, then wash three times using 1200rpm for 3min in 30mM pH 7.0 Tris-HCl buffer containing 0.0759M sucrose and 0.225M mannitol, and then incubate with phosphatase inhibitors and protease inhibitors. Ultrasound mechanically destroys antigen-presenting cells in the presence of the agent. After centrifugation, the cell membrane obtained was washed with a solution of 10mM Tris-HCl pH 7.5 and 1mM EDTA. Then filter the sample through membranes with pore sizes of 30 μm, 10 μm, 5 μm, 2 μm, and 0.45 μm in sequence. Centrifuge the filtrate at 16,000g for 35 minutes and discard the supernatant to collect the precipitate. Resuspend the precipitate in PBS and proceed with step (2) The prepared nanoparticles were incubated for 10 minutes, and then repeatedly co-extruded using a 0.45 μm filter membrane. The extruded liquid was centrifuged at 12,000g for 25 minutes and the supernatant was discarded to collect the precipitate. The precipitate was placed in a 4% trehalose aqueous solution. Resuspend and freeze-dry to obtain nanoparticles. Among them, the antigen-presenting cell membrane component activated by nanoparticle 1 and nanoparticle 1 were used to prepare nanoparticle 4, with a particle size of 260 nanometers; the antigen-presenting cell membrane component activated by nanoparticle 2 was used together with nanoparticle 2. The nanoparticle 5 prepared by the interaction has a particle size of 260 nanometers; the nanoparticle 6 is prepared by using the antigen-presenting cell membrane component activated by the nanoparticle 3 and the nanoparticle 3 to interact with the nanoparticle 3, and the particle size is 260 nanometers.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
选取6-8周的雌性C57BL/6小鼠,在第0天、第7天、第14天、第21天和第28天分别给小鼠皮下注射0.5mg的PLGA纳米粒子(负载裂解物组分、Poly(I:C)和两种CpG佐剂及KALA多肽)。在第32天处死小鼠,收集小鼠的外周血和淋巴结。使用梯度离心法分离小鼠外周血中的PBMC,将淋巴结切成小块后研磨通过细胞筛网制备单细胞悬液,尔后把PBMC和淋巴结细胞单细胞悬液混合。然后使用磁珠分选法分选出CD45 +CD3 +的T细胞。将分选得到的CD3 +T细胞(500万个)和纳米粒子(40μg)以及IL-7(10ng/mL)在2mL RPMI1640完全培养基中共孵育96小时。然后采用流式细胞术分选孵育后的T细胞中的CD3 +IFN-γ +T细胞,即为可识别癌细胞全细胞抗原的癌细胞特异性T细胞。 Female C57BL/6 mice aged 6-8 weeks were selected and injected subcutaneously with 0.5 mg of PLGA nanoparticles (lysate-loaded group) on days 0, 7, 14, 21 and 28. points, Poly(I:C) and two CpG adjuvants and KALA polypeptide). The mice were sacrificed on day 32, and their peripheral blood and lymph nodes were collected. Gradient centrifugation was used to isolate PBMCs from the peripheral blood of mice. The lymph nodes were cut into small pieces and ground through a cell mesh to prepare a single cell suspension. The PBMCs were then mixed with the single cell suspension of lymph node cells. CD45 + CD3 + T cells were then sorted using magnetic bead sorting. The sorted CD3 + T cells (5 million cells) were incubated with nanoparticles (40 μg) and IL-7 (10 ng/mL) in 2 mL RPMI1640 complete medium for 96 hours. Then flow cytometry is used to sort the CD3 + IFN-γ + T cells in the incubated T cells, which are cancer cell-specific T cells that can recognize cancer cell whole cell antigens.
(7)实验结果(7)Experimental results
如图11所示,与对照组相比,负载全细胞组分的纳米粒子激活的抗原提呈细胞制备的 纳米粒子可以检测到更多的癌细胞特异性T细胞。而且,加入增加溶酶体逃逸物质的纳米粒子激活的抗原提呈细胞制备的纳米粒子4效果好于未加入溶酶体逃逸的纳米粒子激活的抗原提呈细胞制备的纳米粒子5;使用两种CpG和Poly(I:C)作为混合佐剂的纳米粒子激活的抗原提呈细胞制备的纳米粒子4效果好于只使用一种CpG和Poly(I:C)混合佐剂的纳米粒子激活的抗原提呈细胞制备的纳米粒子6。As shown in Figure 11, compared with the control group, nanoparticles prepared by activated antigen-presenting cells loaded with nanoparticles loaded with whole cell components can detect more cancer cell-specific T cells. Moreover, the effect of nanoparticles 4 prepared by adding antigen-presenting cells activated by nanoparticles that increase lysosome escape substances is better than that of nanoparticles prepared by adding nanoparticles activated by antigen-presenting cells without adding lysosome escape substances 5; using two Nanoparticles prepared with CpG and Poly(I:C) as mixed adjuvants activated antigen-presenting cells. Nanoparticles 4 are more effective than nanoparticles activated with only one CpG and Poly(I:C) mixed adjuvant. Presentation of Cell-Prepared Nanoparticles 6.
实施例11 乳腺癌中癌细胞特异性T细胞的检测Example 11 Detection of cancer cell-specific T cells in breast cancer
本实施例以4T1小鼠三阴性乳腺癌为癌症模型来说明如何采用负载癌细胞全细胞抗原的微米粒子激活的抗原提呈细胞膜组分和癌细胞膜组分制备的微米粒子检测癌细胞特异性T细胞。本实施例中,首先对乳腺癌细胞进行灭活和变性处理,尔后裂解细胞,并以辛基葡萄糖苷溶解裂解癌细胞中的非水溶性组分。然后,以PLGA为微米粒子骨架材料,以CpG2007、CpG1018、和Poly ICLC为免疫佐剂,以聚精氨酸和聚赖氨酸为增强溶酶体逃逸的物质,制备负载有癌细胞全细胞抗原的微米粒子,并使用该粒子激活抗原提呈细胞后制备基于抗原提呈细胞膜组分和癌细胞膜组分混合微米粒子,尔后使用该微米粒子检测癌细胞特异性T细胞。This example uses 4T1 mouse triple-negative breast cancer as a cancer model to illustrate how to use microparticles loaded with cancer cell whole cell antigens to activate the antigen-presenting cell membrane component and microparticles prepared from cancer cell membrane components to detect cancer cell-specific T cell. In this embodiment, breast cancer cells are first inactivated and denatured, and then the cells are lysed, and octylglucoside is used to dissolve and lyse the non-water-soluble components in the cancer cells. Then, PLGA was used as the micron particle skeleton material, CpG2007, CpG1018, and Poly ICLC were used as immune adjuvants, and polyarginine and polylysine were used as substances that enhance lysosomal escape to prepare whole cell antigens loaded with cancer cells. Micron particles are used to activate antigen-presenting cells to prepare mixed micron particles based on antigen-presenting cell membrane components and cancer cell membrane components, and then the micron particles are used to detect cancer cell-specific T cells.
(1)癌细胞的裂解(1) Lysis of cancer cells
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中。所得癌细胞分别采用紫外线和60℃高温加热进行灭活和变性处理5分钟,然后加入超纯水并反复冻融5次辅以超声裂解癌细胞,将细胞裂解物在5000g离心10分钟,上清液即为水溶性组分,将沉淀物使用10%辛基葡萄糖苷溶解后即为溶解后的原非水溶性组分,将水溶性组分和非水溶性组分按质量比2:1混合,即为制备微米粒子所需的裂解物组分。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water. The obtained cancer cells were inactivated and denatured using ultraviolet light and high-temperature heating at 60°C for 5 minutes, and then ultrapure water was added and repeatedly frozen and thawed 5 times, supplemented by ultrasound to lyse the cancer cells. The cell lysate was centrifuged at 5000g for 10 minutes, and the supernatant The liquid is the water-soluble component. Dissolve the precipitate with 10% octyl glucoside to become the original dissolved non-water-soluble component. Mix the water-soluble component and the non-water-soluble component at a mass ratio of 2:1 , which is the lysate component required to prepare micron particles.
(2)微米粒子的制备(2) Preparation of micron particles
本实施例中制备微米粒子1制备采用复乳法,微米粒子骨架材料PLGA分子量为38KDa-54KDa,所采用的免疫佐剂为CpG2007、CpG1018和Poly ICLC,所采用的溶酶体逃逸增加物质为聚精氨酸和聚赖氨酸。制备时先采用复乳法制备内部负载裂解物组分、佐剂和KALA多肽的微米粒子,在内部负载裂解物和佐剂后,将100mg微米粒子在9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后备用。该微米粒子1系统平均粒径为3.1μm左右,微米粒子系统表面电位为-7mV左右;每1mg PLGA微米粒子约负载110μg蛋白质或多肽组分,含CpG2007、CpG1018和Poly ICLC各0.01mg,含聚精氨酸和聚赖氨酸各0.02mg。In this example, the micron particles 1 are prepared using the double emulsion method. The molecular weight of the micron particle skeleton material PLGA is 38KDa-54KDa. The immune adjuvants used are CpG2007, CpG1018 and Poly ICLC. The lysosomal escape increasing substance used is poly. Arginine and polylysine. During preparation, first use the double emulsion method to prepare microparticles internally loaded with lysate components, adjuvants and KALA polypeptides. After loading lysate and adjuvants internally, centrifuge 100mg of microparticles at 9000g for 20 minutes, and use 10mL containing 4% trehalose. Resuspend in ultrapure water and dry for 48 hours before use. The average particle size of the micron particle 1 system is about 3.1 μm, and the surface potential of the micron particle system is about -7mV; every 1 mg of PLGA micron particles is loaded with approximately 110 μg of protein or peptide components, including 0.01 mg each of CpG2007, CpG1018 and Poly ICLC. 0.02mg each of arginine and polylysine.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC和DC2.4作为抗原提呈细胞。BMDC制备方法同实施例1。This example uses BMDC and DC2.4 as antigen-presenting cells. The preparation method of BMDC is the same as in Example 1.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的微米粒子(1000μg)与BMDC(500万个)及DC2.4细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有GM-CSF(2000U/mL)、M-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、 IL-12(200U/mL)、IFN-γ(500U/mL)和CD80抗体(10ng/mL)。 Micron particles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (5 million cells) and DC2.4 cells (5 million cells) in 15 mL of high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ); the incubation system contains GM-CSF (2000U/mL), M-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL ), IFN-γ (500U/mL) and CD80 antibody (10ng/mL).
(5)基于提呈细胞和癌细胞混合膜组分的微米粒子的制备(5) Preparation of microparticles based on mixed membrane components of presenting cells and cancer cells
通过在400g离心5分钟收集孵育后的DC1000万个,与1000万4T1细胞混合后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.22μm的膜反复共挤出,然后加入30mg步骤(2)制备的微米粒子1后在10W超声10秒钟后共孵育10分钟,然后使用5μm的膜反复共挤出,挤出液在9000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得微米粒子2,粒径为3.15μm。Collect 10 million DCs after incubation by centrifugation at 400 g for 5 minutes, mix with 10 million 4T1 cells, wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water, and Ultrasound at 4°C for 2 minutes at low power (20W). The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was collected after centrifugation at 5000g for 10 minutes. The supernatant was repeatedly co-extruded through a 0.22μm membrane, and then 30mg of step (2) was added. The prepared micron particles 1 were ultrasonicated at 10W for 10 seconds and incubated for 10 minutes, and then repeatedly co-extruded using a 5 μm membrane. The extruded liquid was collected after centrifugation at 9000g for 120 minutes. The supernatant was discarded to collect the precipitate, and the precipitate was placed in PBS. After medium resuspension, micron particles 2 were obtained, with a particle size of 3.15 μm.
或者通过在400g离心5分钟收集孵育后的DC2000万个,使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.22μm的膜反复共挤出,然后加入30mg步骤(2)制备的微米粒子1后在10W超声10秒钟后共孵育10分钟,然后使用5μm的膜反复共挤出,挤出液在9000g离心120分钟后收集弃去上清液收集沉淀,将沉淀在PBS中重悬后即得微米粒子3,粒径为3.15μm。Alternatively, collect 20 million DCs after incubation by centrifugation at 400 g for 5 minutes, wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water and incubate at 4°C with low power (20W ) Ultrasound for 2 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was collected after centrifugation at 5000g for 10 minutes. The supernatant was repeatedly co-extruded through a 0.22μm membrane, and then 30mg of step (2) was added. The prepared micron particles 1 were ultrasonicated at 10W for 10 seconds and incubated for 10 minutes, and then repeatedly co-extruded using a 5 μm membrane. The extruded liquid was collected after centrifugation at 9000g for 120 minutes. The supernatant was discarded to collect the precipitate, and the precipitate was placed in PBS. After medium resuspension, micron particles 3 were obtained, with a particle size of 3.15 μm.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
选取6-8周的雌性BALB/c小鼠,在第0天小鼠后背皮下接种2×10 6个4T1乳腺癌细胞;在第7天、第14天和第21天分别皮下注射0.3mg PLGA的微米粒子1。第25天处死小鼠,收集小鼠外周血,从外周血中分离PBMC,并从PBMC中将分选得到CD3 +T细胞。将CD3 +T细胞(100万个)、100μg微米粒子2或者微米粒子3在10mL RPMI1640完全培养基中共孵育24小时(37℃,5%CO 2)。然后先进行不同荧光探针修饰的抗小鼠CD4抗体和抗小鼠CD8抗体的胞膜染色,固定细胞后再进行荧光探针修饰的抗小鼠颗粒酶B(Granzyme B)抗体的胞内染色,然后采用流式细胞术检测孵育后细胞中的CD8 +Granzyme B +T细胞以及CD4 +Granzyme B +T细胞,即为可识别癌细胞全细胞抗原的癌细胞特异性T细胞。癌细胞特异性T细胞被抗原激活后会开始合成和表达杀伤性物质,颗粒酶B即为该类杀伤性物质之一,其诱发细胞凋亡的活力最强。与此同时,分析CD8 +Granzyme B +T细胞中Granzyme B所连接的荧光探针的平均荧光强度(MFI)。 Select 6-8 week old female BALB/c mice and inoculate 2×10 6 4T1 breast cancer cells subcutaneously on the back of the mice on day 0; subcutaneously inject 0.3mg on days 7, 14 and 21 respectively. PLGA micron particles1. On the 25th day, the mice were sacrificed, peripheral blood was collected, PBMC were isolated from the peripheral blood, and CD3 + T cells were sorted from the PBMC. CD3 + T cells (1 million), 100 μg of microparticles 2 or microparticles 3 were incubated in 10 mL of RPMI1640 complete medium for 24 hours (37°C, 5% CO 2 ). Then, the cell membrane was stained with anti-mouse CD4 antibodies and anti-mouse CD8 antibodies modified with different fluorescent probes. After the cells were fixed, intracellular staining with anti-mouse granzyme B (Granzyme B) antibodies modified with fluorescent probes was performed. , and then use flow cytometry to detect CD8 + Granzyme B + T cells and CD4 + Granzyme B + T cells in the incubated cells, which are cancer cell-specific T cells that can recognize cancer cell whole cell antigens. Cancer cell-specific T cells will begin to synthesize and express killer substances after being activated by antigens. Granzyme B is one of these killer substances and has the strongest activity in inducing cell apoptosis. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to Granzyme B in CD8 + Granzyme B + T cells was analyzed.
(7)实验结果(7)Experimental results
如图12所示,微米粒子激活的抗原提呈细胞膜组分与癌细胞膜组分混合后制备的微米粒子2比微米粒子3可更好的检测癌细胞特异性T细胞,且其所激活的癌细胞特异性T细胞可合成更多的杀伤性物质。而且,检测时,微米粒子2激活的癌细胞特异性T细胞可以合成更多的杀伤性物质,因为更易被检测到,检测也会更准确。As shown in Figure 12, micron particles 2 prepared by mixing the antigen-presenting cell membrane components activated by micron particles and the cancer cell membrane components can better detect cancer cell-specific T cells than micron particles 3, and the cancer cells activated by them are better than those of micron particles 3. Cell-specific T cells can synthesize more killer substances. Moreover, during detection, cancer cell-specific T cells activated by micron particles 2 can synthesize more killer substances, because they are easier to detect and the detection will be more accurate.
实施例12 乳腺癌中癌细胞特异性T细胞的检测Example 12 Detection of cancer cell-specific T cells in breast cancer
(1)癌细胞及细菌外囊泡的裂解(1) Cleavage of cancer cell and bacterial external vesicles
将培养的4T1细胞在400g离心5分钟,然后用PBS洗涤两遍后重悬于超纯水中。所得癌细胞分别采用紫外线和高温加热进行灭活和变性处理,然后使用8M尿素水溶液(含500mM氯化钠)裂解癌细胞并溶解裂解物组分,即为制备微米粒子系统的抗原组分。The cultured 4T1 cells were centrifuged at 400g for 5 minutes, then washed twice with PBS and resuspended in ultrapure water. The obtained cancer cells were inactivated and denatured using ultraviolet light and high-temperature heating respectively, and then an 8M urea aqueous solution (containing 500mM sodium chloride) was used to lyse the cancer cells and dissolve the lysate components, which are the antigen components for preparing the micron particle system.
将嗜酸乳杆菌在5000g离心30分钟,然后弃去沉淀后收集上清液,将上清液使用1μm的滤膜过滤,然后在16000g离心90分钟,将沉淀使用8M尿素水溶液(含500mM氯化钠)裂解和溶解细菌外囊泡组分。Centrifuge Lactobacillus acidophilus at 5000g for 30 minutes, then discard the precipitate and collect the supernatant. Filter the supernatant with a 1 μm filter membrane, and then centrifuge at 16000g for 90 minutes. The precipitate is treated with 8M urea aqueous solution (containing 500mM chloride). Sodium) cleaves and solubilizes bacterial outer vesicle components.
(2)微米粒子的制备(2) Preparation of micron particles
本实施例中制备微米粒子采用复乳法。微米粒子1骨架材料为未修饰的PLA和甘露糖修饰的PLA,分子量都为40KDa,未修饰的PLA和甘露糖修饰的PLA的比例为4:1。所采用的免疫佐剂为CpG2006、CpG2216和Poly ICLC,所采用的溶酶体逃逸增加物质为精氨酸和组氨酸。微米粒子制备时所使用的癌细胞裂解物组分和细菌外囊泡组分质量比为1:1。制备时先采用复乳法制备内部负载癌细胞裂解物组分、细菌外囊泡组分、佐剂、精氨酸和组氨酸的微米粒子,尔后,将100mg微米粒子在9000g离心20分钟,使用10mL含4%海藻糖的超纯水重悬后干燥48h后即得微米粒子1,平均粒径为1.5μm左右,每1mg PLGA微米粒子1约负载100μg蛋白质或多肽组分,含CpG2006,CpG2216和Poly ICLC各0.02mg,含精氨酸和组氨酸各0.05mg。对照微米粒子2制备材料和制备方法同微米粒子1,粒径为1.5μm左右,负载等量精氨酸、组氨酸和等量的癌细胞裂解物组分和细菌外囊泡组分,但是不负载任何佐剂。In this embodiment, the double emulsion method is used to prepare micron particles. The skeleton material of micron particle 1 is unmodified PLA and mannose-modified PLA, both with a molecular weight of 40KDa. The ratio of unmodified PLA to mannose-modified PLA is 4:1. The immune adjuvants used were CpG2006, CpG2216 and Poly ICLC, and the lysosomal escape-increasing substances used were arginine and histidine. The mass ratio of cancer cell lysate components and bacterial external vesicle components used in the preparation of micron particles is 1:1. During preparation, the double emulsion method is first used to prepare micron particles that are internally loaded with cancer cell lysate components, bacterial external vesicle components, adjuvants, arginine and histidine. Then, 100mg of micron particles are centrifuged at 9000g for 20 minutes. Use 10 mL of ultrapure water containing 4% trehalose to resuspend and dry for 48 hours to obtain micron particles 1. The average particle size is about 1.5 μm. Each 1 mg of PLGA micron particles 1 is loaded with approximately 100 μg of protein or peptide components, including CpG2006 and CpG2216. and Poly ICLC 0.02mg each, containing 0.05mg each of arginine and histidine. The preparation materials and preparation methods of control micron particle 2 are the same as those of micron particle 1. The particle size is about 1.5 μm, and it is loaded with equal amounts of arginine, histidine and equal amounts of cancer cell lysate components and bacterial outer vesicle components. However, Does not contain any adjuvants.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC、B细胞以及BMDM作为抗原提呈细胞。BMDC及BMDM制备制备方法同上。B细胞来自小鼠外周血PBMC,制备方法同上。将BMDC、B细胞和BMDM按数量比2:1:1混合后即为混合抗原提呈细胞。This example uses BMDC, B cells and BMDM as antigen-presenting cells. The preparation methods of BMDC and BMDM are the same as above. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above. Mix BMDC, B cells and BMDM in a quantity ratio of 2:1:1 to form mixed antigen-presenting cells.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将1000μg的微米粒子1或微米粒子2分别与4000万个混合抗原提呈细胞(含2000万个BMDC,1000万个B细胞及1000万个BMDM)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、IFN-γ(500U/mL)、CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。 1000 μg of microparticles 1 or 2 were incubated with 40 million mixed antigen-presenting cells (containing 20 million BMDCs, 10 million B cells and 10 million BMDMs) in 15 mL of high-glucose DMEM complete medium for 48 hours. (37°C, 5% CO 2 ); the incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL), CD80 antibody (10ng/mL) and CD40 antibody (20mg/mL).
(5)抗原提呈细胞来源的粒子的制备(5) Preparation of particles derived from antigen-presenting cells
通过在400g离心5分钟收集孵育后的4000万个混合抗原提呈细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品使用高压均质机(10000bar)处理3分钟,然后在13000g离心30分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米 粒子。其中,使用微米粒子1激活的混合抗原提呈细胞制备的为纳米粒子1,粒径为250纳米;使用微米粒子2激活的混合抗原提呈细胞制备的为纳米粒子2,粒径为250纳米。Collect the incubated 40 million mixed antigen-presenting cells by centrifugation at 400g for 5 minutes, then wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors, resuspend the cells in PBS water and incubate at 4 ℃ low power (20W) ultrasonic for 2 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45μm membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, filter and concentrate. Use a high-pressure homogenizer (10,000 bar) to process the filtered and concentrated sample for 3 minutes. Then, centrifuge at 13,000g for 30 minutes. Discard the supernatant to collect the precipitate. Resuspend the precipitate in PBS. Nanoparticles. Among them, the mixed antigen-presenting cells activated using micron particles 1 were prepared as nanoparticles 1, with a particle size of 250 nanometers; the mixed antigen-presenting cells activated using micron particles 2 were prepared as nanoparticles 2, with a particle size of 250 nanometers.
或者通过在400g离心5分钟收集与微米粒子1或者微米粒子2孵育后的4000万个混合抗原提呈细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品使用高压均质机(10000bar)处理3分钟,然后与60mg相对应的步骤(2)制备的微米粒子1或微米粒子2共作用10分钟后使用2μm滤膜反复共挤出,然后将挤出液在10000g离心20分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得微米粒子。其中,使用微米粒子1激活的混合抗原提呈细胞膜组分与微米粒子1共作用制备的为微米粒子3,粒径为1.6μm;使用微米粒子2激活的混合抗原提呈细胞膜组分与微米粒子2共作用制备的为微米粒子4,粒径为1.6μm。Or collect 40 million mixed antigen-presenting cells after incubation with microparticles 1 or 2 by centrifugation at 400g for 5 minutes, and then wash the cells twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors. The cells were resuspended in PBS water and sonicated at 4°C for 2 minutes at low power (20W). The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45μm membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, and concentrate. Use a high-pressure homogenizer (10,000 bar) to treat the filtered and concentrated sample for 3 minutes, and then react it with 60 mg of micron particles 1 or 2 prepared in step (2) for 10 minutes before use. The 2 μm filter membrane was repeatedly co-extruded, and then the extruded liquid was centrifuged at 10,000 g for 20 minutes, the supernatant was discarded, and the precipitate was collected. The precipitate was resuspended in PBS to obtain micron particles. Among them, the mixed antigen-presenting cell membrane component activated by micron particle 1 and the micron particle 1 are combined to prepare micron particle 3, with a particle size of 1.6 μm; the mixed antigen-presenting cell membrane component activated by micron particle 2 and micron particle 2 are prepared by the combined action of micron particles 4, with a particle size of 1.6 μm.
(6)癌细胞特异性T细胞的分析(6) Analysis of cancer cell-specific T cells
选取6-8周的雌性BALB/c小鼠,在第0天、第7天、第14天、第21天和第28天分别皮下注射100μL含0.4mg步骤(2)制备的PLGA的微米粒子1。在第32天处死小鼠,收集外周血和脾脏,然后制备PBMC和脾细胞单细胞悬液并将二者混合,然后使用磁珠分选法从中分选出CD3 +T细胞。将分选得到的CD3 +T细胞(200万个)、100μg纳米粒子或微米粒子(纳米粒子1、或者纳米粒子2、或者微米粒子3或者微米粒子4)、DC2.4细胞(100万个)在10mL RPMI1640完全培养基中共孵育48小时(37℃,5%CO 2),孵育体系中含有IL-2(200U/mL)、IL-7(200U/mL)、IL-15(200U/mL)以及CD80抗体(10ng/mL)。然后采用流式细胞术分析孵育后的CD3 +T细胞中的CD3 +IFN-γ +T细胞,即为可识别癌细胞全细胞抗原的癌细胞特异性T细胞。与此同时,分析CD3 +IFN-γ +T细胞中的IFN-γ所连接的荧光探针的平均荧光强度(MFI)。 Select female BALB/c mice that are 6-8 weeks old and subcutaneously inject 100 μL of micron particles containing 0.4 mg of PLGA prepared in step (2) on days 0, 7, 14, 21, and 28. 1. The mice were sacrificed on day 32, peripheral blood and spleen were collected, and then a single cell suspension of PBMC and spleen cells was prepared and mixed, and then CD3 + T cells were sorted out using magnetic bead sorting method. The sorted CD3 + T cells (2 million), 100 μg nanoparticles or microparticles (nanoparticle 1, or nanoparticle 2, or microparticle 3, or microparticle 4), DC2.4 cells (1 million) Incubate in 10mL RPMI1640 complete medium for a total of 48 hours (37°C, 5% CO 2 ). The incubation system contains IL-2 (200U/mL), IL-7 (200U/mL), and IL-15 (200U/mL). and CD80 antibody (10ng/mL). Then flow cytometry was used to analyze the CD3 + IFN-γ + T cells in the incubated CD3 + T cells, which are cancer cell-specific T cells that can recognize cancer cell whole cell antigens. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ in CD3 + IFN-γ + T cells was analyzed.
纳米粒子所负载的癌细胞全细胞抗原在被抗原提呈细胞吞噬后可被降解成抗原表位被提呈到抗原提呈细胞膜表面,抗原提呈细胞制备的纳米粒子负载有上述降解提呈后的抗原表位,可以被癌细胞特异性T细胞识别并激活癌细胞特异性T细胞,被激活后分泌杀伤性细胞因子。IFN-γ是抗原特异性T细胞识别抗原后被激活所分泌的最主要的细胞因子。使用流式细胞术分析的CD3 +IFN-γ +T细胞即为可以识别和杀伤癌细胞的癌细胞特异性T细胞。与此同时,分析CD3+IFN-γ+T细胞中的IFN-γ所连接的荧光探针的平均荧光强度(MFI)。 The cancer cell whole cell antigens loaded by the nanoparticles can be degraded into antigen epitopes after being phagocytosed by the antigen-presenting cells and presented to the surface of the antigen-presenting cell membrane. The nanoparticles prepared by the antigen-presenting cells are loaded with the above-mentioned degraded and presented products. The antigenic epitope can be recognized by cancer cell-specific T cells and activate cancer cell-specific T cells. After activation, they secrete killer cytokines. IFN-γ is the most important cytokine secreted by antigen-specific T cells that are activated after recognizing antigens. CD3 + IFN-γ + T cells analyzed using flow cytometry are cancer cell-specific T cells that can recognize and kill cancer cells. At the same time, the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ in CD3+IFN-γ+T cells was analyzed.
(7)实验结果(7)Experimental results
如图13所示,癌细胞特异性T细胞的比例高低与图中的疗效相关,这说明使用本发明所述粒子检测T细胞就是可以特异性识别和杀伤癌细胞的癌细胞特异性T细胞。而且,纳米粒子1好于纳米粒子2;微米粒子3好于微米粒子4。这说明含有增加溶酶体逃逸功能的物质和混合佐剂的微米粒子激活的抗原提呈细胞制备的粒子检测癌细胞特异性T细胞效果好于只含有溶酶体逃逸功能的物质而不含有混合佐剂的微米粒子激活的抗原提呈细胞制备的 粒子。而且,微米粒子3好于纳米粒子1,微米粒子4好于纳米粒子2,这说明内部负载癌细胞裂解组分而表面负载被激活的抗原提呈细胞组分的实心粒子检测癌细胞特异性T细胞效果好于只是负载被激活的抗原提呈细胞组分的囊泡粒子。而且,IFN-γ所连接的荧光探针的平均荧光强度(MFI)的分析结果与上述结果一致。由此可见,混合佐剂的使用以及内部负载癌细胞全细胞组分均有助于检测癌细胞特异性T细胞。As shown in Figure 13, the proportion of cancer cell-specific T cells is related to the efficacy in the figure, which shows that the T cells detected using the particles of the present invention are cancer cell-specific T cells that can specifically recognize and kill cancer cells. Moreover, nanoparticle 1 is better than nanoparticle 2; microparticle 3 is better than microparticle 4. This shows that the particles prepared by activated antigen-presenting cells containing micron particles containing substances that increase lysosomal escape function and mixed adjuvants are more effective in detecting cancer cell-specific T cells than those containing only substances with lysosomal escape function without mixed adjuvants. Adjuvant micron particles are prepared by activating antigen-presenting cells. Moreover, micron particles 3 are better than nanoparticles 1, and microparticles 4 are better than nanoparticles 2, which shows that solid particles loaded with cancer cell lysis components inside and activated antigen-presenting cell components on the surface detect cancer cell-specific T The cellular effect is better than vesicle particles simply loaded with activated antigen-presenting cell components. Moreover, the analysis results of the mean fluorescence intensity (MFI) of the fluorescent probe connected to IFN-γ were consistent with the above results. It can be seen that the use of mixed adjuvants and the internal loading of cancer cell whole cell components are helpful in detecting cancer cell-specific T cells.
实施例13 结肠癌中癌细胞特异性T的检测Example 13 Detection of cancer cell-specific T in colon cancer
本实施例以小鼠结肠癌为癌症模型来说明如何使用负载结肠癌全细胞抗原的纳米粒子激活的抗原提呈细胞制备的纳米粒子检测癌细胞特异性T细胞。本实施例中,首先使用8M尿素水溶液裂解结肠癌肿瘤组织并溶解裂解组分,然后,以PLGA为骨架材料,以Poly(I:C)、CpG2336和CpG2006为佐剂,以NH 4HCO 3为增加溶酶体逃逸物质,制备纳米粒子,使用该纳米粒子激活抗原提呈细胞后将抗原提呈细胞制备成纳米粒子,然后使用纳米粒子检测癌细胞特异性T细胞。 This example uses mouse colon cancer as a cancer model to illustrate how to detect cancer cell-specific T cells using nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with colon cancer whole-cell antigens. In this example, 8M urea aqueous solution is first used to lyse colon cancer tumor tissue and dissolve the lysed components. Then, PLGA is used as the skeleton material, Poly(I:C), CpG2336 and CpG2006 are used as adjuvants, and NH 4 HCO 3 is used as the adjuvant. Add lysosomal escape substances to prepare nanoparticles, use the nanoparticles to activate antigen-presenting cells, prepare the antigen-presenting cells into nanoparticles, and then use the nanoparticles to detect cancer cell-specific T cells.
(1)肿瘤组织的裂解及各组分的收集(1) Lysis of tumor tissue and collection of components
收集肿瘤组织时先在每只C57BL/6小鼠背部皮下接种2×10 6个MC38结肠癌细胞,在肿瘤长到体积分别为约1000mm 3时处死小鼠并摘取肿瘤组织,将肿瘤组织切块后研磨,通过细胞过滤网加入8M尿素水溶液理解肿瘤组织并溶解裂解后组分。以上即为制备纳米粒子系统的抗原原料来源。 When collecting tumor tissue, 2 × 10 MC38 colon cancer cells were subcutaneously inoculated on the back of each C57BL/6 mouse. When the tumor grew to a volume of approximately 1000 mm, the mice were sacrificed and the tumor tissue was removed and cut into sections. After the block was ground, an 8M urea aqueous solution was added through a cell strainer to digest the tumor tissue and dissolve the lysed components. The above are the sources of antigen raw materials for preparing nanoparticle systems.
(2)纳米粒子系统的制备(2) Preparation of nanoparticle system
本实施例中纳米粒子采用复乳法制备。纳米粒子的制备材料PLGA分子量为7KDa-17KDa,以Poly(I:C)和CpG为佐剂,以NH 4HCO 3为增加溶酶体逃逸物质,且佐剂和NH 4HCO 3负载于纳米粒子内;制备方法如前所述,在制备过程中首先在纳米粒子内部负载裂解液组分和佐剂,然后将100mg纳米粒子在10000g离心20分钟,并使用10mL含4%海藻糖的超纯水重悬后冷冻干燥48h后备用;该纳米粒子平均粒径为260nm左右,表面电位为-7mV左右;每1mg PLGA纳米粒子约负载90μg蛋白质和多肽组分,每1mg PLGA纳米粒所负载的poly(I:C)、CpG2336和CpG2006免疫佐剂各0.02mg,负载NH 4HCO 3 0.01mg。 In this example, the nanoparticles were prepared using the double emulsion method. The preparation material of nanoparticles is PLGA with a molecular weight of 7KDa-17KDa, Poly(I:C) and CpG as adjuvants, NH 4 HCO 3 as a substance that increases lysosomal escape, and the adjuvants and NH 4 HCO 3 are loaded on the nanoparticles Within; the preparation method is as described above. During the preparation process, the lysis solution components and adjuvants are first loaded inside the nanoparticles, and then 100 mg of the nanoparticles are centrifuged at 10,000g for 20 minutes, and 10 mL of ultrapure water containing 4% trehalose is used to rehydrate the nanoparticles. Suspend and freeze-dry for 48 hours before use; the average particle size of the nanoparticles is about 260nm, and the surface potential is about -7mV; each 1 mg of PLGA nanoparticles is loaded with approximately 90 μg of protein and polypeptide components, and each 1 mg of PLGA nanoparticles is loaded with poly(I :C), CpG2336 and CpG2006 immune adjuvant 0.02mg each, loaded with NH 4 HCO 3 0.01mg.
纳米粒子2的制备材料和制备方法同纳米粒子1,粒径为260nm左右,表面电位为-7mV左右,每1mg PLGA纳米粒子约负载90μg蛋白质和多肽组分,每1mg PLGA纳米粒负载NH 4HCO 3 0.01mg,负载CpG2336和CpG2006各0.03mg。 The preparation materials and methods of nanoparticle 2 are the same as nanoparticle 1. The particle size is about 260nm and the surface potential is about -7mV. Each 1 mg PLGA nanoparticle is loaded with approximately 90 μg of protein and peptide components. Each 1 mg PLGA nanoparticle is loaded with NH 4 HCO. 3 0.01mg, loaded with 0.03mg each of CpG2336 and CpG2006.
(3)抗原提呈细胞的制备(3) Preparation of antigen-presenting cells
本实施例使用BMDC和B作为抗原提呈细胞。BMDC制备方法同实施例1。B细胞来自小鼠外周血PBMC,制备方法同上。This example uses BMDC and B as antigen-presenting cells. The preparation method of BMDC is the same as in Example 1. B cells were derived from mouse peripheral blood PBMC, and the preparation method was the same as above.
(4)抗原提呈细胞的激活(4) Activation of antigen-presenting cells
将负载癌细胞全细胞组分的纳米粒子(1000μg)与BMDC(500万个)及B细胞(500万个)在15mL高糖DMEM完全培养基中共孵育48小时(37℃,5%CO 2);孵育体系中含有GM-CSF(2000U/mL)、IL-2(500U/mL)、IL-7(200U/mL)、IL-12(200U/mL)、 IFN-γ(500U/mL)和CD80抗体(10ng/mL)和CD40抗体(20mg/mL)。 Nanoparticles (1000 μg) loaded with cancer cell whole cell components were incubated with BMDC (5 million) and B cells (5 million) in 15 mL high-glucose DMEM complete medium for 48 hours (37°C, 5% CO 2 ) ; The incubation system contains GM-CSF (2000U/mL), IL-2 (500U/mL), IL-7 (200U/mL), IL-12 (200U/mL), IFN-γ (500U/mL) and CD80 antibody (10ng/mL) and CD40 antibody (20mg/mL).
(5)基于抗原提呈细胞的纳米粒子的制备(5) Preparation of nanoparticles based on antigen-presenting cells
通过在400g离心5分钟收集孵育后的DC和B细胞,然后使用含有蛋白酶抑制剂的4℃磷酸盐缓冲溶液(PBS)洗涤细胞两遍,将细胞重悬在PBS水中后在4℃低功率(20W)超声2分钟。然后将样品在3000g离心15分钟并收集上清液,将上清液在5000g离心10分钟后收集上清液,将上清液通过0.45μm的膜过滤后使用超滤膜(截留分子量50KDa)超滤离心过滤和浓缩,将过滤和浓缩后的样品与步骤(2)制备的纳米粒子混合后使用高压均质机(10000bar)处理3分钟,然后在13000g离心30分钟后弃去上清液收集沉淀,将沉淀在PBS中重悬后即得纳米粒子。其中,使用纳米粒子1激活的抗原提呈细胞与纳米粒子1共作用所制备的为纳米粒子3,粒径为300纳米;使用纳米粒子2激活的抗原提呈细胞与纳米粒子2共作用所制备的为纳米粒子4,粒径为300纳米。The incubated DC and B cells were collected by centrifugation at 400g for 5 minutes, and then washed twice with 4°C phosphate buffer solution (PBS) containing protease inhibitors. The cells were resuspended in PBS water and incubated at 4°C at low power (PBS). 20W) Ultrasonic for 2 minutes. The sample was then centrifuged at 3000g for 15 minutes and the supernatant was collected. The supernatant was centrifuged at 5000g for 10 minutes and the supernatant was collected. The supernatant was filtered through a 0.45μm membrane and ultrafiltrated using an ultrafiltration membrane (molecular weight cutoff 50KDa). Filter, centrifuge, filter and concentrate. Mix the filtered and concentrated sample with the nanoparticles prepared in step (2) and treat it with a high-pressure homogenizer (10000bar) for 3 minutes. Then centrifuge at 13000g for 30 minutes and then discard the supernatant to collect the precipitate. , resuspend the precipitate in PBS to obtain nanoparticles. Among them, the one prepared by using the antigen-presenting cells activated by the nanoparticle 1 and the nanoparticle 1 is nanoparticle 3, with a particle size of 300 nanometers; the one prepared by using the antigen-presenting cells activated by the nanoparticle 2 and the nanoparticle 2. is nanoparticle 4, with a particle size of 300 nanometers.
(6)癌细胞特异性T细胞的检测(6) Detection of cancer cell-specific T cells
选取6-8周的雌性C57BL/6小鼠,在第0天、第7天、第14天和第28天分别皮下注射100μL含0.5mg PLGA的纳米粒子(负载裂解物组分、混合佐剂及增加溶酶体逃逸的物质)或者100μL PBS。在第32天处死小鼠,收集小鼠外周血并分离外周血单个核细胞(PBM C),然后使用流式细胞术从PBMC中分选CD3 +T细胞。 Female C57BL/6 mice aged 6-8 weeks were selected and subcutaneously injected with 100 μL of nanoparticles containing 0.5 mg PLGA (loaded lysate component, mixed adjuvant) on days 0, 7, 14, and 28. and substances that increase lysosomal escape) or 100 μL PBS. Mice were sacrificed on day 32, peripheral blood was collected and peripheral blood mononuclear cells (PBM C) were isolated, and then flow cytometry was used to sort CD3 + T cells from PBMC.
本实施例使用酶联免疫斑点法(ELISPOT)检测癌细胞特异性T细胞。首先在96孔板中包被抗小鼠IFN-γ抗体a(capture antibody,包被抗体)12小时,然后加入培养基封闭处理1小时后使用PBS洗涤,然后将100μL RPMI 1640完全培养基中的上述分选得到T细胞(5000个)加入每孔并在其中加入50μg抗原提呈细胞制备的纳米粒子3或者纳米粒子4,并在37℃(5%CO 2)条件下孵育24小时。尔后,弃去细胞和粒子的混合物,洗涤96孔板并在加入抗小鼠IFN-γ抗体b(detection antibody,检测抗体)后在37℃(5%CO 2)培养箱中培养2小时以上。弃去含有抗小鼠IFN-γ抗体b的溶液,洗涤96孔板后并采用相应的方法显色以在96孔板表面形成斑点。采用ELISPOT分析仪读取数据并分析每孔中形成斑点的数量,即为可识别癌细胞全细胞抗原的癌细胞特异性T细胞。 This example uses enzyme-linked immunospot method (ELISPOT) to detect cancer cell-specific T cells. First, anti-mouse IFN-γ antibody a (capture antibody, coating antibody) was coated in a 96-well plate for 12 hours, then the medium was added for blocking treatment for 1 hour, and then washed with PBS, and then 100 μL of RPMI 1640 complete medium was added. T cells (5000) obtained by the above sorting were added to each well and 50 μg of nanoparticles 3 or 4 prepared from antigen-presenting cells were added, and incubated at 37°C (5% CO 2 ) for 24 hours. Afterwards, the mixture of cells and particles was discarded, and the 96-well plate was washed and cultured in a 37° C. (5% CO 2 ) incubator for more than 2 hours after adding anti-mouse IFN-γ antibody b (detection antibody). Discard the solution containing anti-mouse IFN-γ antibody b, wash the 96-well plate, and use the corresponding method to develop color to form spots on the surface of the 96-well plate. Use an ELISPOT analyzer to read the data and analyze the number of spots formed in each well, which are cancer cell-specific T cells that can recognize cancer cell whole-cell antigens.
(7)实验结果(7)Experimental results
如图14所示,与PBS对照组相比,注射负载全细胞组分的粒子组小鼠体内含有更多的癌细胞特异性T细胞,而本发明所述的纳米粒子激活的抗原提呈细胞制备的纳米粒子可以有效的检测癌细胞特异性T细胞。而且,负载混合佐剂、裂解物组分和溶酶体逃逸物质的纳米粒子激活的抗原提呈细胞制备成的纳米粒子辅助分离的癌细胞特异性T细胞效果好于负载裂解物组分、两种CpG佐剂和溶酶体逃逸物质的纳米粒子。As shown in Figure 14, compared with the PBS control group, the mice injected with particles loaded with whole cell components contained more cancer cell-specific T cells, and the antigen-presenting cells activated by the nanoparticles of the present invention The prepared nanoparticles can effectively detect cancer cell-specific T cells. Moreover, nanoparticles prepared from antigen-presenting cells activated by nanoparticles loaded with mixed adjuvants, lysate components and lysosomal escape substances are more effective in assisting in the isolation of cancer cell-specific T cells than those loaded with lysate components and both. Nanoparticles of CpG adjuvants and lysosomal escape substances.
显然,上述实施例仅仅是为清楚地说明所作的举例,并非对实施方式的限定。对于所属领域的普通技术人员来说,在上述说明的基础上还可以做出其他不同形式变化或变动。这里无需也无法对所有的实施方式予以穷举。而由此所引申出的显而易见的变化或变动仍处于本发明创造的保护范围之中。Obviously, the above-mentioned embodiments are only examples for clear explanation and are not intended to limit the implementation. For those of ordinary skill in the art, other changes or modifications may be made based on the above description. An exhaustive list of all implementations is neither necessary nor possible. The obvious changes or modifications derived therefrom are still within the protection scope of the present invention.

Claims (10)

  1. 一种由激活的抗原提呈细胞制备的粒子检测癌细胞特异性T细胞的方法,其特征在于,包括以下步骤:A method for detecting cancer cell-specific T cells using particles prepared from activated antigen-presenting cells, which is characterized by including the following steps:
    S1、将抗原提呈细胞与第一粒子共孵育,得到激活后的抗原提呈细胞;其中,第一粒子为负载组织和/或细胞全细胞组分的纳米粒子或微米粒子;S1. Incubate the antigen-presenting cells with the first particles to obtain activated antigen-presenting cells; wherein the first particles are nanoparticles or microparticles loaded with tissue and/or whole cell components;
    S2、将激活后的抗原提呈细胞的细胞膜组分制备成纳米囊泡;或将激活后的抗原提呈细胞的细胞膜组分与第二粒子共作用,得到负载细胞膜组分的第二粒子;其中,第二粒子为负载组织和/或细胞全细胞组分的纳米粒子或微米粒子;S2. Preparing the activated cell membrane components of the antigen-presenting cells into nanovesicles; or cooperating the activated cell membrane components of the antigen-presenting cells with second particles to obtain second particles loaded with cell membrane components; Wherein, the second particles are nanoparticles or microparticles loaded with whole cell components of tissues and/or cells;
    S3、将步骤S2所述的纳米囊泡和/或负载细胞膜组分的第二粒子与待测细胞共孵育,检测待测细胞内部或表面的标志物,通过分析含有标志物的T细胞实现所述癌细胞特异性T细胞的检测。S3. Incubate the nanovesicles and/or the second particles loaded with cell membrane components described in step S2 with the cells to be tested, detect the markers inside or on the surface of the cells to be tested, and achieve the desired results by analyzing the T cells containing the markers. Detection of cancer cell-specific T cells.
  2. 根据权利要求1所述的方法,其特征在于:所述标志物包括蛋白质或核酸。The method of claim 1, wherein the marker includes protein or nucleic acid.
  3. 根据权利要求1所述的方法,其特征在于:所述纳米囊泡和/或负载细胞膜组分的第二粒子与待测细胞共孵育之前,还包括对待测细胞中的T细胞进行分选的步骤。The method according to claim 1, characterized in that: before the nanovesicles and/or the second particles loaded with cell membrane components are co-incubated with the cells to be tested, the method further includes the step of sorting the T cells in the cells to be tested. step.
  4. 根据权利要求1所述的方法,其特征在于:在步骤S1或步骤S3中,共孵育体系中含有细胞因子和/或抗体。The method according to claim 1, characterized in that in step S1 or step S3, the co-incubation system contains cytokines and/or antibodies.
  5. 根据权利要求1所述的方法,其特征在于:所述第一粒子或第二粒子上负载有细菌组分或细菌外囊泡组分。The method according to claim 1, wherein the first particle or the second particle is loaded with bacterial components or bacterial outer vesicle components.
  6. 根据权利要求5所述的方法,其特征在于:所述细菌组分或细菌外囊泡组分经含有裂解剂的裂解液裂解细菌或细菌外囊泡得到;所述裂解剂为尿素、盐酸胍、脱氧胆酸盐、十二烷基硫酸盐、甘油、蛋白质降解酶、白蛋白、卵磷脂、Triton、吐温、氨基酸、糖苷和胆碱中的一种或多种。The method according to claim 5, characterized in that: the bacterial component or bacterial outer vesicle component is obtained by lysing the bacteria or bacterial outer vesicles with a lysis solution containing a lysing agent; the lytic agent is urea, guanidine hydrochloride , one or more of deoxycholate, lauryl sulfate, glycerol, protein degrading enzymes, albumin, lecithin, Triton, Tween, amino acids, glycosides and choline.
  7. 根据权利要求1所述的方法,其特征在于:在步骤S2中,还包括将癌细胞细胞膜组分或细胞外囊泡膜组分与激活后的抗原提呈细胞的细胞膜组分混合,制备成纳米囊泡或负载于第二粒子上的步骤。The method according to claim 1, characterized in that: in step S2, it also includes mixing the cell membrane components of cancer cells or extracellular vesicle membrane components with the cell membrane components of activated antigen-presenting cells to prepare Nanovesicles or the step of loading onto a second particle.
  8. 根据权利要求1所述的方法,其特征在于:所述第一粒子或第二粒子上负载有免疫增强佐剂。The method of claim 1, wherein the first particle or the second particle is loaded with an immune-enhancing adjuvant.
  9. 根据权利要求8所述的方法,其特征在于:所述免疫增强佐剂包括两种或两种以上Toll样受体激动剂。The method of claim 8, wherein the immune-enhancing adjuvant includes two or more Toll-like receptor agonists.
  10. 一种用于检测癌细胞特异性T细胞的试剂盒,其特征在于,所述试剂盒中包括以下(1)-(2)中的至少一种:A kit for detecting cancer cell-specific T cells, characterized in that the kit includes at least one of the following (1)-(2):
    (1)由激活的抗原提呈细胞制备的纳米囊泡;(1) Nanovesicles prepared from activated antigen-presenting cells;
    (2)负载激活的抗原提呈细胞细胞膜组分的粒子;(2) Particles loaded with activated antigen-presenting cell membrane components;
    其中,in,
    所述纳米囊泡通过将抗原提呈细胞与第一粒子共孵育,得到激活后的抗原提呈细胞,再提取激活后的抗原提呈细胞的细胞膜组分制备得到;The nanovesicles are prepared by co-incubating the antigen-presenting cells with the first particles to obtain activated antigen-presenting cells, and then extracting the cell membrane components of the activated antigen-presenting cells;
    所述负载激活的抗原提呈细胞细胞膜组分的粒子通过将激活后的抗原提呈细胞的细胞膜组分与第二粒子共作用,使所述细胞膜组分负载于第二粒子上得到;The particles loaded with cell membrane components of activated antigen-presenting cells are obtained by cooperating the cell membrane components of activated antigen-presenting cells with second particles, so that the cell membrane components are loaded on the second particles;
    所述第一粒子或第二粒子分别独立地选自负载肿瘤组织和/或癌细胞全细胞组分的纳米粒子或微米粒子。The first particles or second particles are independently selected from nanoparticles or microparticles loaded with tumor tissue and/or whole cell components of cancer cells.
PCT/CN2022/108956 2022-07-08 2022-07-29 Method and kit for detecting cancer cell-specific t cell WO2024007388A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210801405.XA CN115561145A (en) 2022-07-08 2022-07-08 Detection method and kit for cancer cell specific T cells
CN202210801405.X 2022-07-08

Publications (1)

Publication Number Publication Date
WO2024007388A1 true WO2024007388A1 (en) 2024-01-11

Family

ID=84739509

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/108956 WO2024007388A1 (en) 2022-07-08 2022-07-29 Method and kit for detecting cancer cell-specific t cell

Country Status (2)

Country Link
CN (1) CN115561145A (en)
WO (1) WO2024007388A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160144009A1 (en) * 2013-04-29 2016-05-26 The Board Of Trustees Of The Leland Stanford Junior University Use of Anti-CD47 Agents to Enhance Immunization
CN107427466A (en) * 2015-01-29 2017-12-01 浦项工科大学校产学协力团 From nano vesicle and application thereof derived from cell membrane
CN110836966A (en) * 2018-08-15 2020-02-25 王镕 Detection nanoparticles, detection method and kit for detecting content of antigen-specific T cells and the like
CN112114129A (en) * 2020-09-25 2020-12-22 苏州大学 Method for detecting tumor specific T cells
CN114404580A (en) * 2021-12-24 2022-04-29 苏州大学 Dendritic cell cancer vaccine and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160144009A1 (en) * 2013-04-29 2016-05-26 The Board Of Trustees Of The Leland Stanford Junior University Use of Anti-CD47 Agents to Enhance Immunization
CN107427466A (en) * 2015-01-29 2017-12-01 浦项工科大学校产学协力团 From nano vesicle and application thereof derived from cell membrane
CN110836966A (en) * 2018-08-15 2020-02-25 王镕 Detection nanoparticles, detection method and kit for detecting content of antigen-specific T cells and the like
CN112114129A (en) * 2020-09-25 2020-12-22 苏州大学 Method for detecting tumor specific T cells
CN114404580A (en) * 2021-12-24 2022-04-29 苏州大学 Dendritic cell cancer vaccine and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HWANG INKYU, SHEN XUEFEI, SPRENT JONATHAN, MILLER JACQUES F A P, HALL ELIZA: "Direct stimulation of naı ¨ve T cells by membrane vesicles from antigen-presenting cells: Distinct roles for CD54 and B7 molecules", PNAS, vol. 100, no. 11, 27 May 2003 (2003-05-27), pages 6670 - 6675, XP093124805 *

Also Published As

Publication number Publication date
CN115561145A (en) 2023-01-03

Similar Documents

Publication Publication Date Title
US20240122984A1 (en) Compositions and methods for delivery of immune cells to treat un-resectable or non-resected tumor cells and tumor relapse
WO2023115676A1 (en) Dendritic cell cancer vaccine and use thereof
WO2023087441A1 (en) Vaccine system for preventing or treating cancer and use thereof
CN112114129A (en) Method for detecting tumor specific T cells
CN114288397A (en) Vaccine system for preventing or treating cancer based on multiple cancer cells and/or tumor tissue whole cell components, preparation and application thereof
Dong et al. Hybrid M13 bacteriophage-based vaccine platform for personalized cancer immunotherapy
WO2023236330A1 (en) Cancer vaccine based on antigen-presenting cell membrane components, method for preparing same, and use thereof
WO2024000724A1 (en) Preparation method for vaccine loaded with cancer cell whole-cell component and mixed membrane component and use thereof
JP6029677B2 (en) Vaccines for tumor immunotherapy
CN113181354B (en) Foot-and-mouth disease bionic nanometer vaccine based on dendritic cells, and preparation method and application thereof
WO2023236331A1 (en) Method for preparing vaccine of autoimmune disease derived from pre-activated antigen-presenting cell and use thereof
Li et al. A" trained immunity" inducer-adjuvanted nanovaccine reverses the growth of established tumors in mice
WO2024007388A1 (en) Method and kit for detecting cancer cell-specific t cell
US20230288404A1 (en) Detection method for tumor-specific t cells
WO2024000725A1 (en) Cancer cell-specific t cell vaccine and method for activating cancer cell-specific t cells
WO2023227085A1 (en) Specific t cell for preventing or treating cancer, and preparation method therefor
WO2023206684A1 (en) Cell system and use thereof, and method for activating broad-spectrum cancer cell-specific t cells
WO2024016688A1 (en) Nucleic acid delivery particle based on activated antigen-presenting cells, nucleic acid delivery system and preparation method
WO2023201787A1 (en) Cancer-specific t cell-based cell system, lymphocyte drug and use thereof
AU3298100A (en) Immunostimulant bacterial membrane fractions in cancer treatment
CN117122678A (en) Method for preparing antigen delivery particles
CN117653720A (en) Extraction method of antigen component, delivery particle containing antigen component and application of delivery particle
CN117618382A (en) Preparation method and application of antigen delivery particles
CN116449006A (en) Detection kit for cancer cell specific T cells
CN117618602A (en) Terminal sterilization method of cancer particle vaccine loaded with cancer cell lysate component and application of terminal sterilization method

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22949966

Country of ref document: EP

Kind code of ref document: A1