CN115058455B - Preparation method and application of clinical blood immune cell preparation - Google Patents

Preparation method and application of clinical blood immune cell preparation Download PDF

Info

Publication number
CN115058455B
CN115058455B CN202210583135.XA CN202210583135A CN115058455B CN 115058455 B CN115058455 B CN 115058455B CN 202210583135 A CN202210583135 A CN 202210583135A CN 115058455 B CN115058455 B CN 115058455B
Authority
CN
China
Prior art keywords
cells
kir3ds1
target fragment
alpha
receptor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202210583135.XA
Other languages
Chinese (zh)
Other versions
CN115058455A (en
Inventor
王显荣
李素芬
王亮
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Western Medical Biotechnology Chengdu Co ltd Shuangliu Medical Branch
Original Assignee
Western Medical Biotechnology Chengdu Co ltd Shuangliu Medical Branch
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 Western Medical Biotechnology Chengdu Co ltd Shuangliu Medical Branch filed Critical Western Medical Biotechnology Chengdu Co ltd Shuangliu Medical Branch
Priority to CN202210583135.XA priority Critical patent/CN115058455B/en
Publication of CN115058455A publication Critical patent/CN115058455A/en
Application granted granted Critical
Publication of CN115058455B publication Critical patent/CN115058455B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The application discloses a preparation method and application of a clinical blood immune cell preparation. According to the preparation method, T cells expressing CD8 and CD28 are separated, and a lentivirus is used for mediating and transferring a human KIR3DS1 receptor and a human IFN-alpha 1 into the T cells, so that a T cell line stably expressing the KIR3DS1 receptor and the IFN-alpha 1 is obtained. The T cell line not only can kill various tumor cells in vitro, but also has the therapeutic effects of slowing down tumor growth and improving survival rate in vivo, and has small side effect and wide development prospect.

Description

Preparation method and application of clinical blood immune cell preparation
Technical Field
The application relates to the technical field of cell preparations, in particular to a preparation method and application of a clinical blood immune cell preparation.
Background
In recent decades, great progress has been made in the field of tumor therapy. Cell-based therapies, particularly against tumor-associated antigens (TAAs), have evolved rapidly. T cells were engineered to have the ability to attack tumor cells by generating a CAR construct consisting of a gene encoding an scFv, a costimulatory domain (CD 28 or TNFRSF 9) and a CD247 signaling domain for T cell proliferation and activation. In principle, CAR-T cells are activated by scFv recognition of TAAs on the surface of T cells, and then intracellular signaling domains linked by scFv are subsequently activated to induce downstream signaling pathways involved in T cell proliferation, activation and cytokine production. Currently, the effective function of CAR-T cell therapies has been demonstrated in a variety of diseases including hematological malignancies, solid tumors, autoimmune diseases, and allergic diseases such as asthma. Furthermore, antigen-specific T regulatory cells (Tregs) and genetically edited T cells appear to be beneficial in controlling inflammation of allergic asthma.
Disclosure of Invention
Based on the above, the inventor creatively carries out related genetic engineering on T cells, obtains T cells stably expressing KIR3DS1 receptor and IFN-alpha 1 through lentivirus mediation, and confirms the anti-tumor performance through cell and animal experiments.
In a first aspect, embodiments of the present application disclose a method for preparing a clinical blood immune cell preparation, comprising:
obtaining isolated T cells expressing CD8 and CD 28;
preparing a lentivirus carrying a human KIR3DS1 receptor transcript and a human IFN-alpha 1 transcript, wherein the nucleotide sequence of the human KIR3DS1 receptor transcript is shown as NCBI NM_001368254.1, and the nucleotide sequence of the human IFN-alpha 1 transcript is shown as NCBI NM_ 024013.3;
infecting the cells with a lentivirus to obtain T cells expressing KIR3DS1 receptor and IFN-alpha 1, and dissolving or dispersing the T cells in an electrolyte solution suitable for growth of the T cells to prepare a clinical blood immune cell preparation.
In the examples herein, "preparing a lentivirus carrying a human KIR3DS1 receptor transcript and a human IFN-. Alpha.1 transcript" includes:
synthesizing to obtain a human KIR3DS1 receptor transcript and a connector of a human IFN-alpha 1 transcript as target fragments;
and (3) connecting the target fragment into a vector, and carrying out slow virus packaging to obtain the recombinant slow virus vector.
In the embodiment of the application, the nucleotide sequence of the target fragment is shown as SEQ ID NO.1 or 2.
In the embodiment of the present application, after the target fragment is obtained by synthesis, the method further includes a step of amplifying the target fragment, where the step of amplifying the target fragment specifically includes:
designing a first primer pair to amplify a 1-960 bp region of the target fragment;
designing a second primer pair to amplify the 940 bp-1900 bp region of the target fragment;
designing a third primer pair to amplify the 1890 bp-3' end region of the target fragment; and
and (3) carrying out enzyme digestion on the amplified 1 st to 960 th bp region, the amplified 1 st to 960 th bp region and the amplified 1890 th bp & gt3' end region, purifying, mixing the obtained products to serve as templates, and carrying out PCR amplification by using mixed primers of the first primer pair, the second primer pair and the third primer pair again.
In an embodiment of the present application, the step of "performing lentivirus packaging" includes:
inoculating the recombinant plasmid with the target fragment sequence which is sequenced correctly into LB culture medium containing antibiotics for full culture, extracting bacterial liquid, cracking, purifying, extracting and harvesting the plasmid; and
the plasmids were transferred into 293T cells and the packaged lentivirus supernatants were harvested.
In the examples herein, the step of "isolating clinical blood-derived T cells expressing CD8 and CD 28" includes:
extracting spleen cells of a pregnant female CBA/J pregnant mouse;
the immune should antibody label and magnetic bead are utilized to sort the cells, so that the T cells expressing CD8 and CD28 are obtained.
In an embodiment of the present application, the step of "infecting the cells with a lentivirus" includes:
mixing the T cells expressing CD8 and CD28 obtained by sorting with the lentiviral supernatant, culturing at 37 ℃ for 10 hours, removing the lentiviral supernatant, replacing fresh alpha-MEM culture medium, continuing culturing, and culturing for 5 days after infection, thus obtaining the T cells expressing KIR3DS1 receptor and IFN-alpha 1.
In a second aspect, the present application discloses a clinical blood immune cell preparation prepared by the preparation method of the first aspect, comprising T cells stably expressing KIR3DS1 receptor and IFN- α, said T cells further expressing CD8 and CD28.
In embodiments of the present application, the clinical liquid immune cell preparation further comprises an electrolyte solution suitable for its growth. Among them, the "electrolyte solution suitable for its growth" may be an α -MEM medium suitable for its growth, may be a buffer solution containing serum albumin, may be a sugar solution containing serum albumin, an antibiotic, and DMSO.
In a third aspect, the embodiment of the application also discloses the clinical liquid immune cell preparation prepared by the preparation method in the first aspect or the application of the clinical liquid immune cell preparation in the second aspect in preparing medicines and preparations related to liver cancer, chronic myelogenous leukemia, breast cancer and colon cancer.
Compared with the prior art, the application has at least one of the following beneficial effects:
the embodiment of the application provides an in vitro synthesis method of a human KIR3DS1 receptor transcript and a human IFN-alpha 1 transcript, a lentiviral vector is constructed, a T cell of a mouse origin is infected, and a cell line for stably expressing the KIR3DS1 receptor and the IFN-alpha 1 is constructed.
The application further proves that the T cell line expressing CD8 and CD28 has better tumor cell killing rate and obvious curative effect on liver cancer, chronic myelogenous leukemia, breast cancer and colon cancer through cell tests and animal tests.
In addition, the application also discovers through detecting the expression level of the related tumor biocidal factors that the CD8+CD28+T cells improved by the embodiment of the application can achieve the killing effect through inducing the expression of the performance and FasL in target cells.
Drawings
FIG. 1 is an electrophoresis chart of the target fragment provided in example 1 (lane 1), example 2 (lane 2) and comparative example (1) (lane 3) of the present application, respectively.
FIG. 2 provides an electrophoretogram of recombinant plasmids of example 1 (lane 1), example 2 (lane 2) and comparative example (1) (lane 3), respectively, of the present application.
FIG. 3 shows the results of flow cytometry assays of CD8 (left panel) and CD28 (right panel) expressing T cells isolated in the examples of the present application.
Detailed Description
In order to make the objects, technical solutions and advantages of the present application more apparent, the present application will be further described in detail with reference to examples. It should be understood that the specific embodiments described herein are for purposes of illustration only and are not intended to limit the present application. Reagents not specifically and individually described in this application are all conventional reagents and are commercially available; methods which are not specifically described in detail are all routine experimental methods and are known from the prior art.
It should be noted that, the terms "first," "second," and the like in the description and the claims of the present invention and the above drawings are used for distinguishing similar objects, and are not necessarily used for describing a particular sequence or order, nor do they substantially limit the technical features that follow. It is to be understood that the data so used may be interchanged where appropriate such that the embodiments of the invention described herein may be implemented in sequences other than those illustrated or otherwise described herein. Furthermore, the terms "comprises," "comprising," and "having," and any variations thereof, are intended to cover a non-exclusive inclusion, such that a process, method, system, article, or apparatus that comprises a list of steps or elements is not necessarily limited to those steps or elements expressly listed but may include other steps or elements not expressly listed or inherent to such process, method, article, or apparatus.
Target fragment
1. Target fragment
The target fragment disclosed in the embodiment of the application is obtained by connecting a human KIR3DS1 receptor transcript (NM_ 001368254.1) and a human IFN-alpha 1 transcript (NM_ 024013.3) through an intermediate connecting sequence. Wherein, the human KIR is a killer cell immunoglobulin-like receptor (KIR) which is a member of immunoglobulin superfamily, and the extracellular region of the human KIR-like receptor contains an immunoglobulin-like domain which is a human KIR3DS1 receptor.
In one embodiment, the above fragments of interest (SEQ ID NOS.1-2) are synthesized chemically.
In one embodiment, the intermediate linkage sequence is as shown in SEQ ID NO.3 or as shown in SEQ ID NO. 4. In one comparative example 1, the fragment of interest was a DNA molecule (shown in SEQ ID NO. 5) obtained by direct ligation of a human KIR3DS1 receptor transcript and a human IFN-. Alpha.1 transcript and chemical synthesis.
2. Designing primers
In order to obtain a desired fragment at a proper concentration, it is necessary to perform PCR amplification of the desired fragment, and since the desired fragment is more than 2500bp in each of example 1, example 2 and comparative example 1, the primer set and the PCR amplification method are provided in the examples of the present application. In some embodiments, the primer set provided in the embodiments of the present application includes a first primer pair, a second primer pair and a third primer pair, wherein the first primer pair amplifies a region of 1 to 960bp of the target fragment, the second primer pair amplifies a region of 940 to 1900bp of the target fragment, and the third primer pair amplifies a region of 1890bp→3' of the target fragment. For this purpose, the examples of the present application provide a variety of primer pairs, as specifically shown in table 1. In Table 1, the lower strand is the cleavage site, and the thickened portion is the protecting base.
TABLE 1
Figure GDA0004213437220000061
3. PCR amplification
In order to obtain the objective fragments of examples 1 and 2 and comparative example 1 in large concentrations, respectively, the examples of the present application also provide a PCR amplification method. The method comprises the steps of performing a first amplification, a cleavage and a second amplification, respectively. Wherein, the 2 nd amplification is carried out by using 3 primer pairs as shown in Table 1, respectively using the target fragments of examples 1,2 and comparative example 1 as templates, respectively, to obtain corresponding partial sequences in the respective target fragments, respectively, carrying out enzyme digestion to form purification, mixing the obtained partial sequences, and then respectively mixing with the 3 primer pairs again, thereby carrying out the 2 nd amplification.
Wherein the reaction system for amplification 1 or 2 (for example, 50. Mu.L) comprises: 1.0. Mu.L dNTP (2.5 mmol/L), 9.0. Mu.L upstream primer (10. Mu. Mol/L), 9.0. Mu.L downstream primer (10. Mu. Mol/L), 1.0. Mu.L LPfuDNA polymerase (5U/. Mu.L), 10 Xbuffer 5.0. Mu.L, double distilled water 25. Mu.L. The PCR reaction conditions were 94℃for 5min,94℃for 30s,56℃for 30s,72℃for 1min,28 cycles, and finally 72℃for 10min, and 4℃for termination.
In the above steps, the fragment of interest was recovered using a PCR product purification kit (Guangzhou Novolac, cat# KG 033-1), and the recovered fragment of interest was sequenced and identified in a manner well known to those skilled in the art. The gel electrophoresis of the product obtained by the above procedure is shown in FIG. 1, wherein the concentration of the fragment of interest is significantly higher than the concentration of other non-fragments of interest.
Vector construction
1. Linearization vector and amplification vector
The vector used in the application is GV115 (Ji Kai gene), and AgeI and EcoRI are used for double enzyme digestion to obtain a linearization vector, and the linearization vector is amplified under the action of TransStartFast Pfu DNA polymerase.
2. Lentiviral vector construction
Ligation was performed using Fermentas T4 DNA ligase, for example, a 20. Mu.L reaction system comprising: linearized Vector (100 ng/. Mu.L), 1. Mu.LT 4 DNA library, 2. Mu.L 10 XT 4 DNA ligase Buffer, 1. Mu.L of the fragment of interest (100 ng/. Mu.L) (examples 1,2 and comparative example 1) and make-up double distilled water to 20. Mu.L.
The recombinant vector formed was transformed into TransTl competent cells (catalog number CD501-02, beijing full-size gold Biotechnology Co., ltd.) using conventional bacterial transformation methods well known to those skilled in the art, and the purified vector backbone cleavage product was transformed into a negative control. 2 single clones were picked and plasmids were extracted by conventional methods, and positive clones were identified by AgeI and EcoRI cleavage and PCR. The amplification was carried out using the universal primers (F: cctatttcccatgattccttcata, SEQ ID NO.24; R: gtaatacggttatccacgcg, SEQ ID NO. 25). Both the digested and PCR products were electrophoresed through agarose gel to observe the positions of the bands. Finally, the plasmids positive for both cleavage and PCR identification were sequenced by the Probiotechnological engineering (Shanghai) Co., ltd. Sequencing was performed using the sequencing universal primers T7 and SP6 on the vector for forward and reverse sequencing, respectively.
As shown in FIG. 2, the recombinant plasmid PCR amplification products were subjected to 0.8% agarose gel electrophoresis, and a distinct specific band at about 8.0kb was seen, which was consistent with the expected size.
3. Lentivirus package
(1) Plasmid extraction
Transferring the bacterial liquid with correct sequencing to LB liquid culture medium containing antibiotics (Amp), shaking overnight for culture by a shaking table, and culturing at a constant temperature of 37 ℃ and 220rpm/min for 12-16 h; centrifuging the bacterial liquid at 4000g for 10min, adding Solution I (Ai Mota Weidaul International trade (Shanghai) limited company) containing 10mL RNaseA into the precipitate, stirring uniformly, continuously adding 10mL Solution II, gently mixing upside down for 8-10 times to obtain clear lysate, incubating for 2-3min, adding 5mL precooled N3Buffer (Beijing Bai Albo), gently mixing reversely until egg white floccule appears, standing for 10min at room temperature, filtering, adding 10%v/v times of ETR Solution (Omega) into the supernatant, incubating for 10min on ice until the liquid becomes clear, incubating for 5min at 25 ℃ in a water bath kettle at 42 ℃, centrifuging for 5min at 4000g, capturing the supernatant into 50mL, adding 50%v/v volume ratio absolute ethyl alcohol (standing at room temperature for 96-100%) and incubating for 1-2 min at room temperature; loading to a 50mL filter column (Milbot Co., USA), centrifuging 4000g for 3min, adding 10mL of HBC Buffer (Omega Bio-tek) to the precipitate, centrifuging 4000g for 3min, adding 15mL of DNA Wash Buffer (Omega Bio-tek) to the precipitate, centrifuging 4000g for 3min, adding 10mL of DNA Wash Buffer to the precipitate, centrifuging 4000g for 3min, loading the precipitate to an air-dried filter column, standing the filtrate at room temperature for 5min, and centrifuging 4000g for 5min to obtain the filtrate, wherein the obtained filtrate is the desired plasmid. The concentration and purity of the target plasmid and the packaging plasmid are respectively measured by a Nanodrop ultramicro spectrophotometer, and then the plasmid and the packaging plasmid are stored in a refrigerator at the temperature of minus 20 ℃.
(2) Culture of 293T cells and lentiviral packaging
Resuscitating and subculturing 293T cells (Siemens to obtain suspension cell liquid with the fusion degree of 80%; 10. Mu.g of the plasmid prepared above, 7.0. Mu.g of the packaging plasmid pSPAX2 and 2.0. Mu.g of the packaging plasmid PMD2G were added to a 1.5mL EP tube, and after mixing, 500. Mu.L of CaCl was added 2 Blowing the solutionAfter uniformly mixing, slowly dripping the mixture into BBS balanced salt solution with the same volume, after uniformly blowing and mixing, incubating for 30mi at room temperature until turbidity is visible, adding the mixed solution into 293T cell culture solution, after uniformly mixing, placing the mixed solution into a 37 ℃ incubator for culturing for 8-10 hours, taking precipitate, washing the precipitate for 1-2 times by PBS, replacing 15mL of fresh DMEM culture medium containing 10% fetal bovine serum, continuously culturing the sediment for 48-72 hours, observing the expression intensity of green fluorescent protein under a fluorescent microscope, and thus determining whether to harvest cell supernatant which is slow virus, or adding fresh culture solution for mechanical culturing.
Centrifuging the collected lentivirus supernatant at 3500g for 10min, removing cell debris, filtering with 0.45 μm for sterilization, concentrating, transferring into a sterile ultrafiltration tube, centrifuging at 3500g until concentrating to 500 μl, and packaging in a freezing tube, and storing at-80deg.C. The qPCR lentivirus titer was measured using a qPCR lentivirus titer detection kit (Cat. No. LU900, aibi Meng Biotechnology Co., ltd.). As a result, the virus titers obtained by lentiviral packaging using the recombinant plasmids obtained in example 1, example 2 and comparative example 1 of the present application were all 10 8 The transfection requirement of the T cell for preparing the expressed target gene is met.
T cell for stably expressing target gene
1. Preparation of autologous T cells
The autologous T cells provided in the examples herein were derived from pregnant female CBA/J pregnant mice (Beijing Botai Honda Biotechnology Co., ltd.). The pregnant mice are killed by cervical dislocation on the 9 th day of pregnancy, soaked in 75% alcohol for 3min, placed on sterile paper with the left abdomen side facing upwards, and placed in a sterile super clean bench; cutting a pregnant opening in the middle of the left abdominal side of a pregnant mouse, tearing open the skin, exposing the abdominal wall, and displaying red long-strip spleen; lifting the peritoneum on the lower side of the spleen, cutting, turning up, exposing the spleen, lifting the spleen by forceps, aseptically separating connective tissue below the spleen by using an ophthalmic scissors, taking out the spleen, placing the spleen in a culture dish containing 5mL of Hank's liquid, placing the spleen on a screen, lightly grinding the spleen by using a 2mL syringe needle core, and obtaining a cell suspension; the collected spleen cell suspension is centrifuged at 1800rpm and 4 ℃ for 8min; discard supernatant and resuspend cellsAdding 5mLELS (erythrocyte lysate), mixing, and standing at room temperature for 5min; adding Hank's solution to 10mL, mixing well, centrifuging at 1800rpm and 4 ℃ for 8min; discarding the supernatant, adding Hank's solution to 10mL, mixing, filtering with a screen, centrifuging at 1800rpm at 4deg.C for 8min, discarding the supernatant; resuspend cells with 5ml RPMI 1640; adjusting the concentration to 10 8 mol/L。
2. Immunofluorescence antibody mark and magnetic bead sorting magnetic bead positive sorting
Cd8+ T cells: taking 1×10 8 The individual cells were resuspended in 100. Mu.L of 1640, 10. Mu.L of CD8-FITC was added, mixed well, incubated at 4-8℃for 15min, then 1ml RPM was added, mixed well, centrifuged at 1000r/min for 10min, the supernatant was discarded, and resuspended in 80. Mu.L of 1640. Adding 40 mu LCD8 magnetic beads into the cell suspension, uniformly mixing, incubating for 20min at 4-8 ℃, centrifuging for 10min at 1000r/min after buffer washing, discarding the supernatant, precipitating, and re-suspending by using 500 mu L buffer. The LD magnetic bead sorting column was placed in a sorter, 2mL buffer was passed through the column, and then the cell suspension was added thereto, and the cells flowing out of the sorting column were collected to obtain CD8+ cells. Similarly, magnetic bead fractions were used to sort CD8+CD28+ T cells, and CD8-CD 28-cells. The results are shown in FIG. 3.
3. Lentivirus-infected cells
The CD8+CD28+ T cells and the medium for CD8-CD 28-cells obtained by the above-mentioned sorting were adjusted to 10 5 The cells were placed in 24-well plates, and the lentivirus stock solutions (multiplicity of infection 150) obtained in examples 1 and 2 and comparative example 1 were added, respectively, and polybrene was added at a final concentration of 8. Mu.g/mL, and after mixing, they were placed in an incubator at 37℃for 10 hours, centrifuged, the lentivirus supernatant was removed, and fresh alpha-MEM medium was replaced, followed by further culture. After 48 hours and 72 hours of infection culture, the expression of Green Fluorescent Protein (GFP) was observed under a fluorescence microscope, respectively, to examine the effect of lentivirus infection. After 5 days of infection culture, the target fragment T cells are obtained.
4. Detection of expression of Gene of interest
(1) Cell preparation
The lentiviruses obtained in examples 1,2 and comparative example 1, respectively, infest CD8+CD28+ T cells and CD8-CD 28-cells 1X 10 6 Washing with PBS buffer containing 1% fetal bovine serum (washing buffer), and then washing with 100% fetal bovine serummu.L of wash buffer resuspended cells to give a cell suspension.
(2) Detection of T cell proliferation levels
The lentiviruses obtained in examples 1,2 and comparative example 1 were used to infect CD8+CD28+ T cells and CD8-CD 28-cells, respectively, and the cell concentration was adjusted to 10 6 Each cell proliferation rate was calculated by adding 100. Mu.L of each well, culturing at 37℃for 24 hours, measuring the proliferation level of each cell by CCK-8 method, adding 10. Mu. LCCK-8 solution to the cell solution, incubating in an incubator at 37℃for 2.5 hours, measuring absorbance at 450nm by using an ELISA reader, and calculating cell proliferation rate (%) = (experimental OD value-blank OD value)/(experimental OD value-blank OD value) ×100% by using CD8+CD28+ T cells and CD8-CD 28-cells which were not infected with lentivirus as controls and well plates without adding cell solution (buffer only) as blank.
(3)RT-PCR
KIR3DS1 receptor expression assay: total RNA of each cell was extracted by TRIZOL method, and cDNA was synthesized by reverse transcription, and PCR amplification was performed using the following primers.
KIR3DS1F:cagcgctgtggtgcctcgc,SEQ ID NO.26;
KIR3DS1R:ctgtgaccatgatcaccat,SEQ ID NO.27;
GAPDHF:tgcttdcaacagcgacaccca,SEQ ID NO.28,
GAPDHR:caccctgttgctgtagccaaa,SEQ ID NO.29;
IFN-ɑF:ttaggatccatggcctcgcccttt,SEQ ID NO.30,
IFN-ɑR:cgcgaattcgttattccttcctcc,SEQ ID NO.31;
PerformF:agtacagcttcagcactgaca,SEQ ID NO.32;
PerformR:atgaagtgggtgccgtagtt,SEQ ID NO.33;
FasLF:cactttgggattctttccat,SEQ ID NO.34;
FasLR:gtgagttgaggagctacaga,SEQ ID NO.35。
Reverse transcription is carried out by adopting a Promega M-MLV kit to obtain cDNA, and the reaction comprises 0.4 mu l of Rnasin (40U/. Mu.l), 2.6 mu l of Rnasin (40U/. Mu.l) and 2.0 mu L of dNTPs10mM, 0.1. Mu.LM-MLV-RTase and 5 XRT buffer 4. Mu.L. PCR expansion is carried out by taking synthesized cDNA as a template, and a reaction system comprises: 1.0. Mu.L cDNA, 10. Mu. L SYBR prmix ex taq, 5. Mu.L upstream and downstream primers 0.5. Mu.L and 20. Mu.L RNase-Free H 2 O. The reaction condition is 95 ℃ for 15 seconds; the melting curve was prepared by keeping the reaction temperature at 95℃for 5 seconds at 95℃and 60℃at 95℃from 55℃to 95℃for 4 seconds at each 0.5℃increase, and reading the absorbance. The relative expression levels of KIR3DS1 receptor and IFN-a mRNA were calculated using control group 2- ΔΔct assay.
(4) ELISA detection
Further taking 40mL of cell suspension (containing 10mM Tris-HCl, 0.1% deoxycholate sodium, pH 8.0), adding 12.5mg of lysozyme and 32 mu L of 0.1M PMSF, standing at room temperature for 30min, performing ultrasonic sterilization, freezing and storing at 4 ℃ and 12000r/min multiplied by 10min, and the supernatant at-70 ℃; adding washing solution (10 mM Tris,0.5M urea, pH 8.0), re-suspending the precipitate, performing ultrasonic sterilization, centrifuging, repeating washing for 3 times, dissolving in BufferB (100 mM NaH) 2 PO 4 After SDS-PAGE was performed for 2 hours in 10mM Tris.HCl, 8M urea, pH 8.0), the blocking solution was 5% skim milk powder, murine anti-human KIR3DS1 monoclonal antibody DX9 (bioleged) was used as primary antibody, goat anti-murine IgG was used as secondary antibody, DAB was used as chromogenic substrate, and ELISA was performed to determine the expression level of KIR3DS1 in the cells.
Further, 40. Mu.L of the cell suspension was taken, the supernatant was taken, 160. Mu.L of the sample diluent was added, blank wells were set, and ELISA was performed to determine the IFN-. Alpha.expression level (eBioscience) in the cell supernatant using murine anti-human KIR3DS1 monoclonal antibody DX9 (Biolegend) as primary antibody, goat anti-mouse IgG as secondary antibody, DAB as chromogenic substrate.
(5) Results
Table 2 shows the relative levels of KIR3DS1 receptor mRNA and IFN-alpha mRNA, and the amount of KIR3DS1 receptor expression in cells and IFN-alpha expression in cell supernatants, respectively, produced by example 1, example 2 and comparative example 1 for lentiviruses transfected respectively, "-" in Table 2 indicates undetectable. As can be seen from Table 2, the lentiviruses provided in example 1, example 2 and comparative example 1 respectively infect CD8+CD28+T and CD8-CD28-T cells of female CBA/J pregnant mouse origin respectively, and T cells stably expressing KIR3DS1 receptor and IFN-alpha are sequentially prepared, and have no obvious trend of decreasing proliferation level compared with cells which are not infected by the lentiviruses. Neither comparative example 1 nor the control group showed expression of KIR3DS1 receptor and IFN-alpha. The CD8+CD28+T and CD8-CD28-T cells provided in examples 1 and 2 both have KIR3DS1 receptor and IFN-alpha expression, and IFN-alpha is expressed extracellularly, while KIR3DS1 receptor is expressed in or on the cell surface. To verify whether the cell line obtained in the application stably expresses the related target genes, normal T cells of examples 1 and 2 and comparative example 1 and a control group are respectively passaged for 5 generations, and then the related levels of KIR3DS1 receptor mRNA and IFN-alpha mRNA are detected again, wherein the levels are not obviously reduced, so that the target genes are stably inherited by the cells.
TABLE 2
Figure GDA0004213437220000131
Figure GDA0004213437220000141
Note that P <0.01 compared to "CD8-CD28-T
In vitro cell assay
1. MTT assay
T cells prepared in examples 1 and 2 and comparative example 1 of the present application were used as effector cells (CD8+CD28+T and CD8-CD 28-T), and HepG2 (human hepatoma cell, accession No. SNL-083, shang En Bio), K562 (human chronic myelogenous leukemia cell, SNL-042, shang En bio) and EMT6 cells (mouse breast cancer cell, SNL-298, shang En bio) were used as target cells, respectively. Collecting target cells growing in log phase, washing, and adjusting cell concentration to 5×10 4 transfer/mL into 96-well plates, 100. Mu.L per well, 5% CO at 37deg.C 2 After culturing in an incubator for 12h, effector cells (0.5:1, 2.5:1, 5:1) were added according to different target ratios, 5 compound wells were set per target ratio, the total volume of each well was 200. Mu.L, a complete medium was used as a zeroing control well, and 5% CO was used at 37 ℃ 2 After the incubator continues to cultivate for 6 hours, MTT is added to continue to cultivateAfter 4h, the supernatant was removed by centrifugation, 100u1DMSO was added to each well, the OD value at 570nm was determined, and T cell killing activity was calculated according to the following formula: killing rate (%) = [1- (OD) E+T -OD E )/OD T ]×100%。
2. Data processing
All test data are expressed in mean and standard deviation, data were processed using SPSS13.0 software and multiple comparisons and significance signatures were performed for each column or row.
5. Results
Table 3 shows the results of killing of each target cell by T cells provided in example 1, example 2, comparative example 1 and control at an effect ratio of 0.5:1,2.5:1,5:1, multiple comparisons and significant difference labeling were performed for each row of data in table 3. As can be seen from table 3, as the effector ratio increases, the T cells provided in examples 1,2, comparative example 1 and control group increased the killing rate of each target cell, and the cd8+cd28+ T cells provided in examples 1 and 2 had higher killing rate than the CD8-CD28-T cells, while the cd8+cd28+ T cells or CD8-CD28-T cells provided in comparative example 1 were not significantly different from the control group. It can be seen from Table 2 that the effect of killing target cells is not enhanced due to the fact that CD8+CD28+ T cells or CD8-CD28-T cells provided in comparative example 1 are not expressed by KIR3DS1 receptor and IFN-alpha.
TABLE 3 killing Rate (%)
Figure GDA0004213437220000151
Note that P <0.05 compared to "CD8-CD28-T", delta indicates P <0.05 compared to "control"
TABLE 4 mRNA expression of related genes
Figure GDA0004213437220000152
Figure GDA0004213437220000161
Note that P <0.05 compared to "CD8-CD28-T
Table 4 shows mRNA expression levels of relevant tumor killing molecules after T cells provided in example 1, example 2, and comparative example 1, respectively, act on target cells. Wherein, the relative tumor killing molecule expression of the T cells provided by the comparative example 1 and the control group is not changed significantly, while the expression level of the performance and the FasL in the CD8+CD28+T cells provided by the example 1 and the example 2 is increased significantly. Thus, it was demonstrated that T cells engineered with KIR3DS and IFN-alpha induced and promoted expression of both perform and FasL.
Animal experiment
1. Materials and methods
The experimental example adopts NOD tumor-bearing mice (HT-29 colon cancer, kernel-derived organisms), SPF grade feeding is started when the tumor diameter reaches 4-5 mm.
The injections were given 1 time every 3 days after tumor inoculation, 14 times consecutively, and a blank group was set. At each treatment, each mouse in the blank group was intraperitoneally injected with 100 μlpbs buffer; each mouse of the experimental group was injected intraperitoneally 10 6 T cells provided in example 1, example 2, comparative example 1 and control group. According to the formula V = length x width 2 Tumor volume was calculated by/2, data was entered into GraphPadPrism5 software, and tumor volume change rate after treatment was completed and survival rate of mice was calculated. Wherein, tumor volume change rate = (pre-treatment tumor volume-post-treatment tumor volume)/post-treatment tumor volume x 100%, and mouse survival rate was calculated as 100 days post-treatment mouse survival rate.
2. Statistical analysis
All results are presented as mean ± standard deviation, statistical analysis detected with Student's t test, p <0.05, p < 0.01).
3. Results
TABLE 5
Figure GDA0004213437220000171
Table 5 shows the rate of change of tumor volume and the survival of mice after intervention of tumor-bearing mice with T cells provided in example 1, example 2 and comparative example 1, respectively. In table 5, p <0.05, # indicates p <0.01 compared to corresponding CD8-CD28-T cells; delta represents the blank versus p <0.05; and (d) represents p <0.05 compared to the corresponding comparative example 1. As can be seen from table 5, the T cell intervention tumor-bearing mice provided in example 1, example 2 and comparative example 1 respectively have more remarkable therapeutic effects. In particular, the cd8+cd28+ T cells provided in each of the examples and comparative examples have a more pronounced therapeutic effect relative to the CD8-CD28-T cells; examples 1 and 2 have a more pronounced therapeutic effect relative to comparative example 1.
In summary, the examples of the present application provide for in vitro synthesis of human KIR3DS1 receptor transcripts and human IFN- α1 transcripts, and construction of lentiviral vectors, infecting T cells of mouse origin, and constructing cell lines stably expressing KIR3DS1 receptor and IFN- α1.
The application further proves that the T cell line expressing CD8 and CD28 has better tumor cell killing rate and obvious curative effect on liver cancer, chronic myelogenous leukemia, breast cancer and colon cancer through cell tests and animal tests.
In addition, the application also discovers through detecting the expression level of the related tumor biocidal factors that the CD8+CD28+T cells improved by the embodiment of the application can achieve the killing effect through inducing the expression of the performance and FasL in target cells.
The foregoing is merely a preferred embodiment of the present application, but the scope of the present application is not limited thereto, and any changes or substitutions easily contemplated by those skilled in the art within the technical scope of the present application should be covered by the scope of the present application.
Sequence listing
<110> Western medical and biomedical science and technology Co., ltd, double-flow medical division
Preparation method and application of <120> clinical blood immune cell preparation
<141> 2022-05-23
<160> 35
<170> SIPOSequenceListing 1.0
<210> 1
<211> 2817
<212> DNA
<213> Artificial Sequence
<400> 1
agagaaccta gagcccaagg ttcagagtca cccatctcag caagcccaga agtatctgca 60
atatctacga tggcctcgcc ctttgcttta ctgatggtcc tggtggtgct cagctgcaag 120
tcaagctgct ctctgggctg tgatctccct gagacccaca gcctggataa caggaggacc 180
ttgatgctcc tggcacaaat gagcagaatc tctccttcct cctgtctgat ggacagacat 240
gactttggat ttccccagga ggagtttgat ggcaaccagt tccagaaggc tccagccatc 300
tctgtcctcc atgagctgat ccagcagatc ttcaacctct ttaccacaaa agattcatct 360
gctgcttggg atgaggacct cctagacaaa ttctgcaccg aactctacca gcagctgaat 420
gacttggaag cctgtgtgat gcaggaggag agggtgggag aaactcccct gatgaatgcg 480
gactccatct tggctgtgaa gaaatacttc cgaagaatca ctctctatct gacagagaag 540
aaatacagcc cttgtgcctg ggaggttgtc agagcagaaa tcatgagatc cctctcttta 600
tcaacaaact tgcaagaaag attaaggagg aaggaataac atctggtcca acatgaaaac 660
aattcttatt gactcataca ccaggtcacg ctttcatgaa ttctgtcatt tcaaagactc 720
tcacccctgc tataactatg accatgctga taaactgatt tatctattta aatatttatt 780
taactattca taagatttaa attatttttg ttcatataac gtcatgtgca cctttacact 840
gtggttagtg taataaaaca tgttccttat atttactcga gaaccatccc agtaacccga 900
ccgccgctgg tcttcgctgg acaccgctga gctgagctgg ggcgcagccg cctgtctgca 960
ccggcagcac catgttgctc atggtcgtca gcatggcgtg tgttgggttg ttcttggtcc 1020
agagggccgg tccacacatg ggtggtcagg acaagccctt cctgtctgcc tggcccagcg 1080
ctgtggtgcc tcgcggagga cacgtgactc ttcggtgtca ctatcgtcat aggtttaaca 1140
atttcatgct atacaaagaa gacagaatcc acgttcccat cttccatggc agaatattcc 1200
aggagggctt caacatgagc cctgtgacca cagcacatgc agggaactac acatgtcggg 1260
gttcacaccc acactccccc actgggtggt cggcacccag caaccccatg gtgatcatgg 1320
tcacaggaaa ccacagaaaa ccttccctcc tggcccaccc aggtcccctg gtgaaatcag 1380
gagagagagt catcctgcaa tgttggtcag atatcatgtt tgagcacttc tttctgcaca 1440
aagagtggat ctctaaggac ccctcacgcc tcgttggaca gatccatgat ggggtctcca 1500
aggccaattt ctccatcggt tccatgatgc gtgcccttgc agggacctac agatgctacg 1560
gttctgttac tcacaccccc tatcagttgt cagctcccag tgatcccctg gacatcgtgg 1620
tcacaggtct atatgagaaa ccttctctct cagcccagcc gggccccaag gttcaggcag 1680
gagagagcgt gaccttgtcc tgtagctccc ggagctccta tgacatgtac catctatcca 1740
gggagggggg agcccatgaa cgtaggctcc ctgcagtgcg caaggtcaac agaacattcc 1800
aggcagattt ccctctgggc cctgccaccc acggagggac ctacagatgc ttcggctctt 1860
tccgtcactc tccctacgag tggtcagacc cgagtgaccc actgcttgtt tctgtcacag 1920
gaaacccttc aagtagttgg ccttcaccca cagaaccaag ctccaaatct ggtaacctca 1980
gacacctgca cattctgatt gggacctcag tggtcaaaat ccctttcacc atcctcctct 2040
tctttctcct tcatcgctgg tgctccaaca aaaaaaaatg ctgctgtaat ggaccaagag 2100
cctgcaggga acagaagtga acagcgagga ttctgatgaa caagaccatc aggaggtgtc 2160
atacgcataa ttggaacact gtgttttcac acagagaaaa atcactcgcc cttctcagag 2220
gcccaagaca cccccaacag ataccagcat gtacatagaa cttccaaatg ctgagcccag 2280
atccaaagtt gtcttctgtc cacgagcacc acagtcaggc cttgagggga tcttctaggg 2340
agacaacagc cctgtctcaa aactgggttg ccagctccca tgtaccagca gctggaatct 2400
gaaggcatca gtcttcatct tagggcatcg ctcttcctca caccacaaat ctgaatgtgc 2460
ctctcacttg cttacaaatg tctaaggtcc ccactgcctg ctggagaaaa aacacactcc 2520
tttgcttagc ccacagttct ccatttcact tgacccctgc ccacctctcc aacctaactg 2580
gcttacttcc tagtctactt gaggctgcaa tcacactgag gaactcacaa ttccacacat 2640
acaagaggct ccgtcttaac gcagcactta gacacgtgct gttccacctt ccctcatgct 2700
gttccacctc ccctcagact agctttcagc cttctgtcag cagtaaaact tatatacttt 2760
ttaaaataac ttcaatgtag ttttccatcc ttcaaataaa catgtctgcc cccatgg 2817
<210> 2
<211> 2858
<212> DNA
<213> Artificial Sequence
<400> 2
agagaaccta gagcccaagg ttcagagtca cccatctcag caagcccaga agtatctgca 60
atatctacga tggcctcgcc ctttgcttta ctgatggtcc tggtggtgct cagctgcaag 120
tcaagctgct ctctgggctg tgatctccct gagacccaca gcctggataa caggaggacc 180
ttgatgctcc tggcacaaat gagcagaatc tctccttcct cctgtctgat ggacagacat 240
gactttggat ttccccagga ggagtttgat ggcaaccagt tccagaaggc tccagccatc 300
tctgtcctcc atgagctgat ccagcagatc ttcaacctct ttaccacaaa agattcatct 360
gctgcttggg atgaggacct cctagacaaa ttctgcaccg aactctacca gcagctgaat 420
gacttggaag cctgtgtgat gcaggaggag agggtgggag aaactcccct gatgaatgcg 480
gactccatct tggctgtgaa gaaatacttc cgaagaatca ctctctatct gacagagaag 540
aaatacagcc cttgtgcctg ggaggttgtc agagcagaaa tcatgagatc cctctcttta 600
tcaacaaact tgcaagaaag attaaggagg aaggaataac atctggtcca acatgaaaac 660
aattcttatt gactcataca ccaggtcacg ctttcatgaa ttctgtcatt tcaaagactc 720
tcacccctgc tataactatg accatgctga taaactgatt tatctattta aatatttatt 780
taactattca taagatttaa attatttttg ttcatataac gtcatgtgca cctttacact 840
gtggttagtg taataaaaca tgttccttat atttactcat caggaggcat cactgaggcc 900
aggagtatca gccctttatc cacgcttttc tacaatggca ttcaataaag tgcacgtgtt 960
tctggtgctg agctgagctg gggcgcagcc gcctgtctgc accggcagca ccatgttgct 1020
catggtcgtc agcatggcgt gtgttgggtt gttcttggtc cagagggccg gtccacacat 1080
gggtggtcag gacaagccct tcctgtctgc ctggcccagc gctgtggtgc ctcgcggagg 1140
acacgtgact cttcggtgtc actatcgtca taggtttaac aatttcatgc tatacaaaga 1200
agacagaatc cacgttccca tcttccatgg cagaatattc caggagggct tcaacatgag 1260
ccctgtgacc acagcacatg cagggaacta cacatgtcgg ggttcacacc cacactcccc 1320
cactgggtgg tcggcaccca gcaaccccat ggtgatcatg gtcacaggaa accacagaaa 1380
accttccctc ctggcccacc caggtcccct ggtgaaatca ggagagagag tcatcctgca 1440
atgttggtca gatatcatgt ttgagcactt ctttctgcac aaagagtgga tctctaagga 1500
cccctcacgc ctcgttggac agatccatga tggggtctcc aaggccaatt tctccatcgg 1560
ttccatgatg cgtgcccttg cagggaccta cagatgctac ggttctgtta ctcacacccc 1620
ctatcagttg tcagctccca gtgatcccct ggacatcgtg gtcacaggtc tatatgagaa 1680
accttctctc tcagcccagc cgggccccaa ggttcaggca ggagagagcg tgaccttgtc 1740
ctgtagctcc cggagctcct atgacatgta ccatctatcc agggaggggg gagcccatga 1800
acgtaggctc cctgcagtgc gcaaggtcaa cagaacattc caggcagatt tccctctggg 1860
ccctgccacc cacggaggga cctacagatg cttcggctct ttccgtcact ctccctacga 1920
gtggtcagac ccgagtgacc cactgcttgt ttctgtcaca ggaaaccctt caagtagttg 1980
gccttcaccc acagaaccaa gctccaaatc tggtaacctc agacacctgc acattctgat 2040
tgggacctca gtggtcaaaa tccctttcac catcctcctc ttctttctcc ttcatcgctg 2100
gtgctccaac aaaaaaaaat gctgctgtaa tggaccaaga gcctgcaggg aacagaagtg 2160
aacagcgagg attctgatga acaagaccat caggaggtgt catacgcata attggaacac 2220
tgtgttttca cacagagaaa aatcactcgc ccttctcaga ggcccaagac acccccaaca 2280
gataccagca tgtacataga acttccaaat gctgagccca gatccaaagt tgtcttctgt 2340
ccacgagcac cacagtcagg ccttgagggg atcttctagg gagacaacag ccctgtctca 2400
aaactgggtt gccagctccc atgtaccagc agctggaatc tgaaggcatc agtcttcatc 2460
ttagggcatc gctcttcctc acaccacaaa tctgaatgtg cctctcactt gcttacaaat 2520
gtctaaggtc cccactgcct gctggagaaa aaacacactc ctttgcttag cccacagttc 2580
tccatttcac ttgacccctg cccacctctc caacctaact ggcttacttc ctagtctact 2640
tgaggctgca atcacactga ggaactcaca attccacaca tacaagaggc tccgtcttaa 2700
cgcagcactt agacacgtgc tgttccacct tccctcatgc tgttccacct cccctcagac 2760
tagctttcag ccttctgtca gcagtaaaac ttatatactt tttaaaataa cttcaatgta 2820
gttttccatc cttcaaataa acatgtctgc ccccatgg 2858
<210> 3
<211> 47
<212> DNA
<213> Artificial Sequence
<400> 3
gagaaccatc ccagtaaccc gaccgccgct ggtcttcgct ggacacc 47
<210> 4
<211> 88
<212> DNA
<213> Artificial Sequence
<400> 4
atcaggaggc atcactgagg ccaggagtat cagcccttta tccacgcttt tctacaatgg 60
cattcaataa agtgcacgtg tttctggt 88
<210> 5
<211> 2770
<212> DNA
<213> Artificial Sequence
<400> 5
agagaaccta gagcccaagg ttcagagtca cccatctcag caagcccaga agtatctgca 60
atatctacga tggcctcgcc ctttgcttta ctgatggtcc tggtggtgct cagctgcaag 120
tcaagctgct ctctgggctg tgatctccct gagacccaca gcctggataa caggaggacc 180
ttgatgctcc tggcacaaat gagcagaatc tctccttcct cctgtctgat ggacagacat 240
gactttggat ttccccagga ggagtttgat ggcaaccagt tccagaaggc tccagccatc 300
tctgtcctcc atgagctgat ccagcagatc ttcaacctct ttaccacaaa agattcatct 360
gctgcttggg atgaggacct cctagacaaa ttctgcaccg aactctacca gcagctgaat 420
gacttggaag cctgtgtgat gcaggaggag agggtgggag aaactcccct gatgaatgcg 480
gactccatct tggctgtgaa gaaatacttc cgaagaatca ctctctatct gacagagaag 540
aaatacagcc cttgtgcctg ggaggttgtc agagcagaaa tcatgagatc cctctcttta 600
tcaacaaact tgcaagaaag attaaggagg aaggaataac atctggtcca acatgaaaac 660
aattcttatt gactcataca ccaggtcacg ctttcatgaa ttctgtcatt tcaaagactc 720
tcacccctgc tataactatg accatgctga taaactgatt tatctattta aatatttatt 780
taactattca taagatttaa attatttttg ttcatataac gtcatgtgca cctttacact 840
gtggttagtg taataaaaca tgttccttat atttactcgc tgagctgagc tggggcgcag 900
ccgcctgtct gcaccggcag caccatgttg ctcatggtcg tcagcatggc gtgtgttggg 960
ttgttcttgg tccagagggc cggtccacac atgggtggtc aggacaagcc cttcctgtct 1020
gcctggccca gcgctgtggt gcctcgcgga ggacacgtga ctcttcggtg tcactatcgt 1080
cataggttta acaatttcat gctatacaaa gaagacagaa tccacgttcc catcttccat 1140
ggcagaatat tccaggaggg cttcaacatg agccctgtga ccacagcaca tgcagggaac 1200
tacacatgtc ggggttcaca cccacactcc cccactgggt ggtcggcacc cagcaacccc 1260
atggtgatca tggtcacagg aaaccacaga aaaccttccc tcctggccca cccaggtccc 1320
ctggtgaaat caggagagag agtcatcctg caatgttggt cagatatcat gtttgagcac 1380
ttctttctgc acaaagagtg gatctctaag gacccctcac gcctcgttgg acagatccat 1440
gatggggtct ccaaggccaa tttctccatc ggttccatga tgcgtgccct tgcagggacc 1500
tacagatgct acggttctgt tactcacacc ccctatcagt tgtcagctcc cagtgatccc 1560
ctggacatcg tggtcacagg tctatatgag aaaccttctc tctcagccca gccgggcccc 1620
aaggttcagg caggagagag cgtgaccttg tcctgtagct cccggagctc ctatgacatg 1680
taccatctat ccagggaggg gggagcccat gaacgtaggc tccctgcagt gcgcaaggtc 1740
aacagaacat tccaggcaga tttccctctg ggccctgcca cccacggagg gacctacaga 1800
tgcttcggct ctttccgtca ctctccctac gagtggtcag acccgagtga cccactgctt 1860
gtttctgtca caggaaaccc ttcaagtagt tggccttcac ccacagaacc aagctccaaa 1920
tctggtaacc tcagacacct gcacattctg attgggacct cagtggtcaa aatccctttc 1980
accatcctcc tcttctttct ccttcatcgc tggtgctcca acaaaaaaaa atgctgctgt 2040
aatggaccaa gagcctgcag ggaacagaag tgaacagcga ggattctgat gaacaagacc 2100
atcaggaggt gtcatacgca taattggaac actgtgtttt cacacagaga aaaatcactc 2160
gcccttctca gaggcccaag acacccccaa cagataccag catgtacata gaacttccaa 2220
atgctgagcc cagatccaaa gttgtcttct gtccacgagc accacagtca ggccttgagg 2280
ggatcttcta gggagacaac agccctgtct caaaactggg ttgccagctc ccatgtacca 2340
gcagctggaa tctgaaggca tcagtcttca tcttagggca tcgctcttcc tcacaccaca 2400
aatctgaatg tgcctctcac ttgcttacaa atgtctaagg tccccactgc ctgctggaga 2460
aaaaacacac tcctttgctt agcccacagt tctccatttc acttgacccc tgcccacctc 2520
tccaacctaa ctggcttact tcctagtcta cttgaggctg caatcacact gaggaactca 2580
caattccaca catacaagag gctccgtctt aacgcagcac ttagacacgt gctgttccac 2640
cttccctcat gctgttccac ctcccctcag actagctttc agccttctgt cagcagtaaa 2700
acttatatac tttttaaaat aacttcaatg tagttttcca tccttcaaat aaacatgtct 2760
gcccccatgg 2770
<210> 6
<211> 26
<212> DNA
<213> Artificial Sequence
<400> 6
ggaattcaga gaacctagag cccaag 26
<210> 7
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 7
ggaattcggt gtccagcgaa g 21
<210> 8
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 8
ggaattcaga gaacctagag cccaagg 27
<210> 9
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 9
ggaattcgca ccagaaacac gtgc 24
<210> 10
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 10
ggaattcaga gaacctagag cccaagg 27
<210> 11
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 11
ggaattccag ctcagctcag cgag 24
<210> 12
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 12
ggaattcctt cgctggacac c 21
<210> 13
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 13
ggaattcagg gagagtgacg gaaagag 27
<210> 14
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 14
ggaattcgca cgtgtttctg gtgc 24
<210> 15
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 15
ggaattcgac cactcgtagg gagagtg 27
<210> 16
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 16
ggaattcctc gctgagctga gctg 24
<210> 17
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 17
ggaattcgac cactcgtagg gagagtg 27
<210> 18
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 18
ggaattcctc tttccgtcac tctccct 27
<210> 19
<211> 19
<212> DNA
<213> Artificial Sequence
<400> 19
ggaattccca tgggggcag 19
<210> 20
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 20
ggaattccac tctccctacg agtggtc 27
<210> 21
<211> 19
<212> DNA
<213> Artificial Sequence
<400> 21
ggaattccca tgggggcag 19
<210> 22
<211> 27
<212> DNA
<213> Artificial Sequence
<400> 22
ggaattccac tctccctacg agtggtc 27
<210> 23
<211> 18
<212> DNA
<213> Artificial Sequence
<400> 23
ggaattccca tgggggca 18
<210> 24
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 24
cctatttccc atgattcctt cata 24
<210> 25
<211> 20
<212> DNA
<213> Artificial Sequence
<400> 25
gtaatacggt tatccacgcg 20
<210> 26
<211> 19
<212> DNA
<213> Artificial Sequence
<400> 26
cagcgctgtg gtgcctcgc 19
<210> 27
<211> 19
<212> DNA
<213> Artificial Sequence
<400> 27
ctgtgaccat gatcaccat 19
<210> 28
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 28
tgcttdcaac agcgacaccc a 21
<210> 29
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 29
caccctgttg ctgtagccaa a 21
<210> 30
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 30
ttaggatcca tggcctcgcc cttt 24
<210> 31
<211> 24
<212> DNA
<213> Artificial Sequence
<400> 31
cgcgaattcg ttattccttc ctcc 24
<210> 32
<211> 21
<212> DNA
<213> Artificial Sequence
<400> 32
agtacagctt cagcactgac a 21
<210> 33
<211> 20
<212> DNA
<213> Artificial Sequence
<400> 33
atgaagtggg tgccgtagtt 20
<210> 34
<211> 20
<212> DNA
<213> Artificial Sequence
<400> 34
cactttggga ttctttccat 20
<210> 35
<211> 20
<212> DNA
<213> Artificial Sequence
<400> 35
gtgagttgag gagctacaga 20

Claims (6)

1. A method for preparing a clinical blood immune cell preparation, comprising:
obtaining isolated T cells expressing CD8 and CD 28;
preparing a lentivirus carrying a human KIR3DS1 receptor transcript and a human IFN-alpha 1 transcript;
infecting the cells with a lentivirus to obtain T cells expressing KIR3DS1 receptor and IFN-alpha 1, and dissolving or dispersing the T cells in an electrolyte solution suitable for growth of the T cells to prepare a clinical blood immune cell preparation.
2. The method of claim 1, wherein preparing a lentivirus carrying a human KIR3DS1 receptor transcript and a human IFN- α1 transcript comprises:
synthesizing nucleotide sequences of a human KIR3DS1 receptor transcript and a human IFN-alpha 1 transcript to obtain target fragments;
and (3) connecting the target fragment into a vector, and carrying out slow virus packaging to obtain the recombinant slow virus vector.
3. The preparation method of claim 2, wherein the nucleotide sequence of the target fragment is as shown in SEQ ID NO:1 or 2.
4. The production method according to claim 2, wherein, after the synthesis of the target fragment, the production method further comprises a step of amplifying the target fragment, "the step of amplifying the target fragment" specifically comprises:
designing a first primer pair to amplify a 1-960 bp region of the target fragment;
designing a second primer pair to amplify the 940 bp-1900 bp region of the target fragment;
designing a third primer pair to amplify the 1890 bp-3' end region of the target fragment; and
and (3) carrying out enzyme digestion on the amplified 1-960 bp region, the 940 bp-190 bp region and the 1890 bp-3' end region, purifying, mixing to serve as templates, and carrying out PCR amplification by using mixed primers of the first primer pair, the second primer pair and the third primer pair.
5. The method of claim 2, wherein the step of "packaging lentiviruses" comprises:
inoculating the recombinant plasmid carrying the target fragment with correct sequencing into LB culture medium containing antibiotics for full culture, extracting bacterial liquid, cracking, purifying, extracting and harvesting the recombinant plasmid; and
transferring the recombinant plasmid into 293T cells, and harvesting the packaged lentivirus supernatant.
6. The method of claim 1, wherein the step of infecting the cells with a lentivirus comprises:
mixing the T cells expressing CD8 and CD28 obtained by sorting with the lentiviral supernatant, culturing at 37 ℃ for 10 hours, removing the lentiviral supernatant, replacing fresh alpha-MEM culture medium, continuing culturing, and infecting and culturing for 5 days to obtain the T cells expressing KIR3DS1 receptor and IFN-alpha 1.
CN202210583135.XA 2022-05-26 2022-05-26 Preparation method and application of clinical blood immune cell preparation Active CN115058455B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210583135.XA CN115058455B (en) 2022-05-26 2022-05-26 Preparation method and application of clinical blood immune cell preparation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210583135.XA CN115058455B (en) 2022-05-26 2022-05-26 Preparation method and application of clinical blood immune cell preparation

Publications (2)

Publication Number Publication Date
CN115058455A CN115058455A (en) 2022-09-16
CN115058455B true CN115058455B (en) 2023-07-14

Family

ID=83198965

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210583135.XA Active CN115058455B (en) 2022-05-26 2022-05-26 Preparation method and application of clinical blood immune cell preparation

Country Status (1)

Country Link
CN (1) CN115058455B (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101948806A (en) * 2010-10-14 2011-01-19 山东大学 Natural killer cell line modified by human interferon-alpha gene and establishing method thereof
US10731128B2 (en) * 2016-11-22 2020-08-04 Alloplex Biotherapeutics, Inc. Compositions and methods for in vitro activation and expansion of serial killer T cell populations and passive immunization of a cancer patient with tumor cell killing cells

Also Published As

Publication number Publication date
CN115058455A (en) 2022-09-16

Similar Documents

Publication Publication Date Title
CN105602992B (en) It is a kind of based on the CAR-T transgene carrier and its construction method of replication defective recombinant slow virus and application
CN106279434B (en) Engineered CD20 targeted NKT cell and preparation method and application thereof
WO2018014384A1 (en) Tcr-/pd-1- double negative t-cell and construction method thereof
CN109678965B (en) Chimeric antigen receptor, gene and recombinant expression vector thereof, CD22-CD19 dual-targeting T cell and application thereof
KR20200000495A (en) Nucleic acid for coding chimeric antigen feceptor protein and t lymphocyte for expression of chimeric antigen receprof prorein
CN109721659B (en) Novel Chimeric Antigen Receptor (CAR) targeting CD19 and application thereof
CN111925451B (en) BCMA (brain cell activating antigen) -targeted Chimeric Antigen Receptor (CAR) and application thereof
CN105949316B (en) Anti- EGFRvIII Chimeric antigen receptor, encoding gene, recombinant expression carrier and its construction method and application
CN105950664B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD123 and its construction method and application
CN107033248A (en) Recognize Chimeric antigen receptor and its application of carcinomebryonic antigen
CN110964697A (en) Anti-tumor NK cell and preparation method and application thereof
WO2018006880A1 (en) Co-expression of recombinant immune-checkpoint receptor and immune-checkpoint inhibitor and application
CN105820255A (en) Anti-CD33 chimeric antigen receptor, coding gene, recombinant expression vector and construction method and application of recombinant expression vector
CN105950663B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD30 and its construction method and application
CN105950662B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting CD22 and its construction method and application
CN117126280B (en) Anti-human BCMA nanobody with hydrophilic amino acid residues, CAR-T and application
CN110760480A (en) Anti-tumor NK (Natural killer) cell and preparation method thereof
CN115058455B (en) Preparation method and application of clinical blood immune cell preparation
JPH03505039A (en) Retroviral vector expressing soluble CD4, gene therapy for AIDS
CN116396385A (en) anti-BCMA nano antibody and preparation method and application thereof
CN105969805B (en) A kind of replication defective recombinant slow virus CAR-T transgene carrier targeting Mesothelin and its construction method and application
WO2021136415A1 (en) Method for purifying ucart cell and use thereof
JP7088902B2 (en) Nucleic acid encoding the chimeric antigen receptor protein and T lymphocytes expressing the chimeric antigen receptor protein
CN110093359B (en) Separable nucleic acid containing CD3 promoter sequence and CAR sequence and application thereof
CN105585637B (en) Tumor therapeutic agent based on IL-12 stable membrane expression and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant